Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 515
Filtrar
Más filtros

Publication year range
1.
BMC Neurol ; 23(1): 217, 2023 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-37280540

RESUMEN

BACKGROUND: Extraaxial cerebellopontine angle cavernous hemangiomas are rare and their diagnosis and treatment are challenging. CASE PRESENTATION: A 43-year-old female was admitted to the hospital who had repeated hearing loss in her left ear accompanied by tinnitus. Magnetic resonance imaging revealed a hemangioma-like lesion in the left cerebellopontine angle extra-axial cisternal segment. During the surgery, it was found that the lesion was located in the cisternal segment of the root of the auditory nerve. Postoperative pathological examination confirmed that the lesion was a cavernous hemangioma. CONCLUSION: We report a case of cavernous hemangioma in the brain spatula cisternal segment of the left auditory nerve. For cranial nerve CMs early diagnosis and surgical removal may maximize the chance of a positive outcome.


Asunto(s)
Hemangioma Cavernoso , Humanos , Femenino , Adulto , Hemangioma Cavernoso/diagnóstico por imagen , Hemangioma Cavernoso/cirugía , Nervio Coclear/patología , Imagen por Resonancia Magnética
2.
J Neurosci ; 41(20): 4439-4447, 2021 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-33883202

RESUMEN

Animal studies suggest that cochlear nerve degeneration precedes sensory cell degeneration in both noise-induced hearing loss (NIHL) and age-related hearing loss (ARHL), producing a hearing impairment that is not reflected in audiometric thresholds. Here, we investigated the histopathology of human ARHL and NIHL by comparing loss of auditory nerve fibers (ANFs), cochlear hair cells and the stria vascularis in a group of 52 cases with noise-exposure history against an age-matched control group. Although strial atrophy increased with age, there was no effect of noise history. Outer hair cell (OHC) loss also increased with age throughout the cochlea but was unaffected by noise history in the low-frequency region (<2 kHz), while greatly exacerbated at high frequencies (≥2 kHz). Inner hair cell (IHC) loss was primarily seen at high frequencies but was unaffected by noise at either low or high frequencies. ANF loss was substantial at all cochlear frequencies and was exacerbated by noise throughout. According to a multivariable regression model, this loss of neural channels contributes to poor word discrimination among those with similar audiometric threshold losses. The histopathological patterns observed also suggest that, whereas the low-frequency OHC loss may be an unavoidable consequence of aging, the high-frequency loss, which produces the classic down-sloping audiogram of ARHL, may be partially because of avoidable ear abuse, even among those without a documented history of acoustic overexposure.SIGNIFICANCE STATEMENT As regenerative therapeutics in sensorineural hearing loss enter clinical trials, it becomes critical to infer which cochlear pathologies are present in addition to hair cell loss. Here, by analyzing human autopsy material, we show that acoustic injury accelerates age-related primary neural degeneration, but not strial degeneration, neither of which can be inferred from audiometric thresholds. It exacerbates outer hair cell (OHC) loss only in the high-frequency half of the cochlea, suggesting that the apical loss is age-related, whereas the basal loss is partially noise induced, and therefore avoidable. Statistical analysis suggests that neural loss helps explain differences in word-recognition ability among individuals with similar audiometric thresholds. The surprising correlation between neural loss and OHC loss in the cochlea's speech region also implicates neural loss in the well-known decline in word scores as thresholds deteriorate with age.


Asunto(s)
Cóclea/patología , Células Ciliadas Auditivas Externas/patología , Pérdida Auditiva Provocada por Ruido/patología , Degeneración Nerviosa/patología , Ruido/efectos adversos , Adulto , Anciano , Anciano de 80 o más Años , Envejecimiento/patología , Umbral Auditivo/fisiología , Nervio Coclear/patología , Femenino , Células Ciliadas Auditivas Internas , Humanos , Masculino , Persona de Mediana Edad , Degeneración Nerviosa/etiología
3.
BMC Neurosci ; 22(1): 18, 2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-33752606

RESUMEN

BACKGROUND: The SCN11A gene, encoded Nav1.9 TTX resistant sodium channels, is a main effector in peripheral inflammation related pain in nociceptive neurons. The role of SCN11A gene in the auditory system has not been well characterized. We therefore examined the expression of SCN11A in the murine cochlea, the morphological and physiological features of Nav1.9 knockout (KO) ICR mice. RESULTS: Nav1.9 expression was found in the primary afferent endings beneath the inner hair cells (IHCs). The relative quantitative expression of Nav1.9 mRNA in modiolus of wild-type (WT) mice remains unchanged from P0 to P60. The number of presynaptic CtBP2 puncta in Nav1.9 KO mice was significantly lower than WT. In addition, the number of SGNs in Nav1.9 KO mice was also less than WT in the basal turn, but not in the apical and middle turns. There was no lesion in the somas and stereocilia of hair cells in Nav1.9 KO mice. Furthermore, Nav1.9 KO mice showed higher and progressive elevated ABR threshold at 16 kHz, and a significant increase in CAP thresholds. CONCLUSIONS: These data suggest a role of Nav1.9 in regulating the function of ribbon synapses and the auditory nerves. The impairment induced by Nav1.9 gene deletion mimics the characters of cochlear synaptopathy.


Asunto(s)
Nervio Coclear/patología , Pérdida Auditiva Sensorineural/genética , Canal de Sodio Activado por Voltaje NAV1.9/genética , Sinapsis/patología , Animales , Nervio Coclear/metabolismo , Eliminación de Gen , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Internas/patología , Pérdida Auditiva Sensorineural/metabolismo , Pérdida Auditiva Sensorineural/patología , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Sinapsis/metabolismo
4.
Int J Mol Sci ; 22(6)2021 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-33799503

RESUMEN

Growth hormone (GH) plays an important role in auditory development during the embryonic stage. Exogenous agents such as sound, noise, drugs or trauma, can induce the release of this hormone to perform a protective function and stimulate other mediators that protect the auditory pathway. In addition, GH deficiency conditions hearing loss or central auditory processing disorders. There are promising animal studies that reflect a possible regenerative role when exogenous GH is used in hearing impairments, demonstrated in in vivo and in vitro studies, and also, even a few studies show beneficial effects in humans presented and substantiated in the main text, although they should not exaggerate the main conclusions.


Asunto(s)
Vías Auditivas/metabolismo , Hormona del Crecimiento/genética , Pérdida Auditiva Funcional/genética , Pérdida Auditiva Sensorineural/genética , Hipocampo/metabolismo , Factor I del Crecimiento Similar a la Insulina/genética , Animales , Corteza Auditiva/metabolismo , Corteza Auditiva/patología , Vías Auditivas/patología , Cóclea/metabolismo , Cóclea/patología , Nervio Coclear/metabolismo , Nervio Coclear/patología , Regulación de la Expresión Génica , Hormona del Crecimiento/metabolismo , Pérdida Auditiva Funcional/metabolismo , Pérdida Auditiva Funcional/fisiopatología , Pérdida Auditiva Sensorineural/metabolismo , Pérdida Auditiva Sensorineural/fisiopatología , Hipocampo/patología , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Regeneración Nerviosa/fisiología , Ruido/prevención & control
5.
Neuropediatrics ; 51(2): 170-172, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31698482

RESUMEN

Unilateral sensorineural hearing loss is a common symptom of vestibular schwannomas in adolescent patients with neurofibromatosis type 2 or sporadic vestibular schwannomas and is often the initial clinical feature. While rare cases of sensorineural impairment presenting as vision or hearing loss due to metastatic medulloblastoma are known, hearing loss as an isolated presenting symptom of primary malignant neuroepithelial tumors of the central nervous system has not been reported in the pediatric population so far. We present two adolescents with unilateral hearing loss due to cochlear nerve dysfunction as the only symptom of a primary nonmetastatic medulloblastoma of the WNT signaling pathway family members subgroup.


Asunto(s)
Neoplasias Cerebelosas/complicaciones , Nervio Coclear/patología , Pérdida Auditiva Unilateral/etiología , Meduloblastoma/complicaciones , Adolescente , Neoplasias Cerebelosas/diagnóstico por imagen , Nervio Coclear/diagnóstico por imagen , Femenino , Humanos , Imagen por Resonancia Magnética , Meduloblastoma/diagnóstico por imagen
6.
Am J Otolaryngol ; 41(3): 102435, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32107054

RESUMEN

The purpose of this study is to examine various preoperative factors that can play a role in the auditory rehabilitation outcome of cochlear implant (CI) recipients. In order to determine the level of integrity of central processing preoperatively, special attention was given to residual hearing, duration of deafness, and cochlear nerve diameter as prognostic factors. A cohort of 232 (272 CI implantations) postlingually deafened adults was evaluated in this study. Hearing results at 1, 2 and up to 3 years postoperatively were compared with various preoperative factors: promontory stimulation testing, residual hearing, duration of deafness, and magnetic resonance imaging of the cochlear nerve. Postoperative hearing performance was measured based on the German Freiburg monosyllabic word test and the Oldenburg sentence test. Postoperative hearing performance showed a significant improvement in each consecutive year after implantation. Duration of deafness showed a negative correlation to word recognition and a positive correlation to increased signal-to-noise-ratio in sentence testing. A significant decline in hearing outcome was shown starting around the second decade of deafness corresponding to 66% of life spent in deafness. MR imaging of cochlear nerve diameter shows a positive correlation of larger nerve diameter to better speech understanding. Promontory stimulation testing did not show any prognostic value. In this retrospective review it could be shown that there is an intricate interaction in the preoperative variables: duration of deafness - as well as the ratio of life spent in deafness; residual hearing; and cochlear nerve diameter.


Asunto(s)
Implantación Coclear/rehabilitación , Audición , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Nervio Coclear/diagnóstico por imagen , Nervio Coclear/patología , Estudios de Cohortes , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Periodo Posoperatorio , Pronóstico , Estudios Retrospectivos , Habla , Adulto Joven
7.
J Neurosci ; 38(10): 2551-2568, 2018 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-29437856

RESUMEN

Noise exposure causes auditory nerve (AN) degeneration and hearing deficiency, though the proximal biological consequences are not entirely understood. Most AN fibers and spiral ganglion neurons are ensheathed by myelinating glia that provide insulation and ensure rapid transmission of nerve impulses from the cochlea to the brain. Here we show that noise exposure administered to mice of either sex rapidly affects myelinating glial cells, causing molecular and cellular consequences that precede nerve degeneration. This response is characterized by demyelination, inflammation, and widespread expression changes in myelin-related genes, including the RNA splicing regulator Quaking (QKI) and numerous QKI target genes. Analysis of mice deficient in QKI revealed that QKI production in cochlear glial cells is essential for proper myelination of spiral ganglion neurons and AN fibers, and for normal hearing. Our findings implicate QKI dysregulation as a critical early component in the noise response, influencing cochlear glia function that leads to AN demyelination and, ultimately, to hearing deficiency.SIGNIFICANCE STATEMENT Auditory glia cells ensheath a majority of spiral ganglion neurons with myelin, protect auditory neurons, and allow for fast conduction of electrical impulses along the auditory nerve. Here we show that noise exposure causes glial dysfunction leading to myelin abnormality and altered expression of numerous genes in the auditory nerve, including QKI, a gene implicated in regulating myelination. Study of a conditional mouse model that specifically depleted QKI in glia showed that QKI deficiency alone was sufficient to elicit myelin-related abnormality and auditory functional declines. These results establish QKI as a key molecular target in the noise response and a causative agent in hearing loss.


Asunto(s)
Nervio Coclear/patología , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/patología , Pérdida Auditiva Provocada por Ruido/genética , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/patología , Ratones Quaking/genética , Proteínas de Unión al ARN/genética , Animales , Cóclea/patología , Femenino , Regulación de la Expresión Génica , Inmunohistoquímica , Masculino , Ratones Endogámicos CBA , Neuroglía/patología , Neuronas/patología , Ganglio Espiral de la Cóclea/patología
8.
FASEB J ; 32(6): 3005-3019, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29401591

RESUMEN

Systemic corticosteroids have been the mainstay of treatment for various hearing disorders for more than 30 yr. Accordingly, numerous studies have described glucocorticoids (GCs) and stressors to be protective in the auditory organ against damage associated with a variety of health conditions, including noise exposure. Conversely, stressors are also predictive risk factors for hearing disorders. How both of these contrasting stress actions are linked has remained elusive. Here, we demonstrate that higher corticosterone levels during acoustic trauma in female rats is highly correlated with a decline of auditory fiber responses in high-frequency cochlear regions, and that hearing thresholds and the outer hair cell functions (distortion products of otoacoustic emissions) are left unaffected. Moreover, when GC receptor (GR) or mineralocorticoid receptor (MR) activation was antagonized by mifepristone or spironolactone, respectively, GR, but not MR, inhibition significantly and permanently attenuated trauma-induced effects on auditory fiber responses, including inner hair cell ribbon loss and related reductions of early and late auditory brainstem responses. These findings strongly imply that higher corticosterone stress levels profoundly impair auditory nerve processing, which may influence central auditory acuity. These changes are likely GR mediated as they are prevented by mifepristone.-Singer, W., Kasini, K., Manthey, M., Eckert, P., Armbruster, P., Vogt, M. A., Jaumann, M., Dotta, M., Yamahara, K., Harasztosi, C., Zimmermann, U., Knipper, M., Rüttiger, L. The glucocorticoid antagonist mifepristone attenuates sound-induced long-term deficits in auditory nerve response and central auditory processing in female rats.


Asunto(s)
Nervio Coclear/fisiopatología , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Glucocorticoides/antagonistas & inhibidores , Trastornos de la Audición/fisiopatología , Pérdida Auditiva Provocada por Ruido/fisiopatología , Mifepristona/farmacología , Animales , Cóclea/metabolismo , Cóclea/patología , Cóclea/fisiopatología , Nervio Coclear/metabolismo , Nervio Coclear/patología , Femenino , Glucocorticoides/efectos adversos , Glucocorticoides/farmacología , Trastornos de la Audición/inducido químicamente , Trastornos de la Audición/tratamiento farmacológico , Trastornos de la Audición/metabolismo , Pérdida Auditiva Provocada por Ruido/inducido químicamente , Pérdida Auditiva Provocada por Ruido/tratamiento farmacológico , Pérdida Auditiva Provocada por Ruido/metabolismo , Ratas , Ratas Wistar , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/metabolismo
9.
Int J Neurosci ; 129(6): 580-587, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30475092

RESUMEN

AIMS: Hearing loss is the most common form of sensory impairment in humans. Short impulses of a high intensity noise can trigger sudden hearing loss, which is generally irreversible and associated with structural tissue damage of the cochlea and auditory nerve. It is well established that myelination is essential for the rapid propagation of action potentials along axons, and that Schwann cells are responsible for myelin sheath production in the peripheral nervous system. In the cochlea, spiral ganglion neuron axons are myelinated by Schwann cells. This myelin contributes to axonal protection and allows for efficient action potential transmission along the auditory nerve. For this reason, here we studie the morphological changes on cochlear hair cells and myelin sheaths of the auditory nerve, directly linked to hearing impairment induced by acoustic trauma. MATERIAL AND METHODS: To study the auditory functions, auditory brainstem responses and distortion products were measured at baseline, 2 days, and 21 days after trauma in rats. Then, scanning and transmission electron microscopy techniques were performed to analyze cochleae and the auditory nerve at 21 days after trauma. RESULTS: We observed that acoustic trauma induced cochlear outer hair cell loss and fusion of inner hair cell stereocilia. We also observed an axonal loss and myelin sheath disorganization of the auditory nerve. CONCLUSIONS: These data confirm that a strong acoustic trauma induced histological changes in the cochlea and auditory nerve, leading to permanent hearing loss.


Asunto(s)
Nervio Coclear/patología , Células Ciliadas Auditivas/patología , Pérdida Auditiva Provocada por Ruido/patología , Vaina de Mielina/patología , Animales , Nervio Coclear/ultraestructura , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Células Ciliadas Auditivas/ultraestructura , Pérdida Auditiva Provocada por Ruido/fisiopatología , Masculino , Vaina de Mielina/ultraestructura , Degeneración Nerviosa/patología , Ratas
10.
Audiol Neurootol ; 23(6): 326-334, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30677751

RESUMEN

Charcot-Marie-Tooth (CMT) syndrome is a clinically and genetically heterogeneous group of neuropathies affecting both peripheral motor and sensory nerves. Progressive sensorineural hearing loss, vestibular abnormalities, and dysfunction of other cranial nerves have been described. This is the second case report of otopathology in a patient with CMT syndrome. Molecular genetic testing of DNA obtained at autopsy revealed a missense variant in the MPZ gene (p.Thr65Ala), pathogenic for an autosomal-dominant form of CMT1B. The temporal bones were also prepared for light microscopy by hematoxylin and eosin and Gömöri trichome stains, and immunostaining for anti-myelin protein zero. Pathology was consistent with a myelinopathy of the auditory, vestibular, and facial nerves bilaterally. The pathophysiology of cranial nerve dysfunction in CMT is unknown. Findings in the current case suggested, at least in cranial nerves 7 and 8, that a myelinopathy may be causative.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/genética , Enfermedad de Charcot-Marie-Tooth/patología , Nervio Coclear/patología , Oído Interno/inervación , Variación Genética/genética , Mutación Missense/genética , Proteína P0 de la Mielina/genética , Anciano , Alanina/genética , Aberraciones Cromosómicas , Nervio Facial/patología , Genes Dominantes/genética , Humanos , Masculino , Vaina de Mielina/patología , Treonina/genética , Nervio Vestibular/patología , Secuenciación del Exoma
11.
Proc Natl Acad Sci U S A ; 112(26): E3431-40, 2015 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-26080415

RESUMEN

Cell transplantation therapy has long been investigated as a therapeutic intervention for neurodegenerative disorders, including spinal cord injury, Parkinson's disease, and amyotrophic lateral sclerosis. Indeed, patients have high hopes for a cell-based therapy. However, there are numerous practical challenges for clinical translation. One major problem is that only very low numbers of donor cells survive and achieve functional integration into the host. Glial scar tissue in chronic neurodegenerative disorders strongly inhibits regeneration, and this inhibition must be overcome to accomplish successful cell transplantation. Intraneural cell transplantation is considered to be the best way to deliver cells to the host. We questioned this view with experiments in vivo on a rat glial scar model of the auditory system. Our results show that intraneural transplantation to the auditory nerve, preceded by chondroitinase ABC (ChABC)-treatment, is ineffective. There is no functional recovery, and almost all transplanted cells die within a few weeks. However, when donor cells are placed on the surface of a ChABC-treated gliotic auditory nerve, they autonomously migrate into it and recapitulate glia- and neuron-guided cell migration modes to repair the auditory pathway and recover auditory function. Surface transplantation may thus pave the way for improved functional integration of donor cells into host tissue, providing a less invasive approach to rescue clinically important neural tracts.


Asunto(s)
Proliferación Celular , Nervio Coclear/patología , Regeneración Nerviosa , Neuroglía/patología , Animales , Condroitina ABC Liasa/administración & dosificación , Modelos Animales de Enfermedad , Humanos , Masculino , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/terapia , Ratas , Ratas Sprague-Dawley
12.
J Neurosci ; 36(39): 10214-27, 2016 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-27683915

RESUMEN

UNLABELLED: Sound deprivation by conductive hearing loss increases hearing thresholds, but little is known about the response of the auditory brainstem during and after conductive hearing loss. Here, we show in young adult rats that 10 d of monaural conductive hearing loss (i.e., earplugging) leads to hearing deficits that persist after sound levels are restored. Hearing thresholds in response to clicks and frequencies higher than 8 kHz remain increased after a 10 d recovery period. Neural output from the cochlear nucleus measured at 10 dB above threshold is reduced and followed by an overcompensation at the level of the lateral lemniscus. We assessed whether structural and molecular substrates at auditory nerve (endbulb of Held) synapses in the cochlear nucleus could explain these long-lasting changes in hearing processing. During earplugging, vGluT1 expression in the presynaptic terminal decreased and synaptic vesicles were smaller. Together, there was an increase in postsynaptic density (PSD) thickness and an upregulation of GluA3 AMPA receptor subunits on bushy cells. After earplug removal and a 10 d recovery period, the density of synaptic vesicles increased, vesicles were also larger, and the PSD of endbulb synapses was larger and thicker. The upregulation of the GluA3 AMPAR subunit observed during earplugging was maintained after the recovery period. This suggests that GluA3 plays a role in plasticity in the cochlear nucleus. Our study demonstrates that sound deprivation has long-lasting alterations on structural and molecular presynaptic and postsynaptic components at the level of the first auditory nerve synapse in the auditory brainstem. SIGNIFICANCE STATEMENT: Despite being the second most prevalent form of hearing loss, conductive hearing loss and its effects on central synapses have received relatively little attention. Here, we show that 10 d of monaural conductive hearing loss leads to an increase in hearing thresholds, to an increased central gain upstream of the cochlear nucleus at the level of the lateral lemniscus, and to long-lasting presynaptic and postsynaptic structural and molecular effects at the endbulb of the Held synapse. Knowledge of the structural and molecular changes associated with decreased sensory experience, along with their potential reversibility, is important for the treatment of hearing deficits, such as hyperacusis and chronic otitis media with effusion, which is prevalent in young children with language acquisition or educational disabilities.


Asunto(s)
Nervio Coclear/patología , Nervio Coclear/fisiopatología , Núcleo Coclear/patología , Núcleo Coclear/fisiopatología , Pérdida Auditiva Conductiva/patología , Pérdida Auditiva Conductiva/fisiopatología , Sinapsis/patología , Animales , Percepción Auditiva , Potenciación a Largo Plazo , Depresión Sináptica a Largo Plazo , Masculino , Terminales Presinápticos/metabolismo , Terminales Presinápticos/patología , Ratas , Ratas Sprague-Dawley , Sinapsis/metabolismo , Potenciales Sinápticos
13.
PLoS Genet ; 10(12): e1004823, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25473838

RESUMEN

Tonotopy is a fundamental organizational feature of the auditory system. Sounds are encoded by the spatial and temporal patterns of electrical activity in spiral ganglion neurons (SGNs) and are transmitted via tonotopically ordered processes from the cochlea through the eighth nerve to the cochlear nuclei. Upon reaching the brainstem, SGN axons bifurcate in a stereotyped pattern, innervating target neurons in the anteroventral cochlear nucleus (aVCN) with one branch and in the posteroventral and dorsal cochlear nuclei (pVCN and DCN) with the other. Each branch is tonotopically organized, thereby distributing acoustic information systematically along multiple parallel pathways for processing in the brainstem. In mice with a mutation in the receptor guanylyl cyclase Npr2, this spatial organization is disrupted. Peripheral SGN processes appear normal, but central SGN processes fail to bifurcate and are disorganized as they exit the auditory nerve. Within the cochlear nuclei, the tonotopic organization of the SGN terminal arbors is blurred and the aVCN is underinnervated with a reduced convergence of SGN inputs onto target neurons. The tonotopy of circuitry within the cochlear nuclei is also degraded, as revealed by changes in the topographic mapping of tuberculoventral cell projections from DCN to VCN. Nonetheless, Npr2 mutant SGN axons are able to transmit acoustic information with normal sensitivity and timing, as revealed by auditory brainstem responses and electrophysiological recordings from VCN neurons. Although most features of signal transmission are normal, intermittent failures were observed in responses to trains of shocks, likely due to a failure in action potential conduction at branch points in Npr2 mutant afferent fibers. Our results show that Npr2 is necessary for the precise spatial organization typical of central auditory circuits, but that signals are still transmitted with normal timing, and that mutant mice can hear even with these deficits.


Asunto(s)
Vías Auditivas/anomalías , Tipificación del Cuerpo/genética , Nervio Coclear/anomalías , Mutación , Receptores del Factor Natriurético Atrial/genética , Potenciales de Acción , Animales , Vías Auditivas/embriología , Vías Auditivas/metabolismo , Percepción Auditiva/fisiología , Axones/fisiología , Tronco Encefálico/anomalías , Tronco Encefálico/citología , Tronco Encefálico/patología , Cóclea/anomalías , Cóclea/citología , Cóclea/patología , Nervio Coclear/embriología , Nervio Coclear/patología , Embrión de Mamíferos , Femenino , Ratones , Ratones Transgénicos , Neuronas Aferentes/fisiología , Embarazo
14.
J Clin Monit Comput ; 31(1): 123-134, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26852030

RESUMEN

Auditory steady state responses (ASSR) may offer an alternative to brainstem auditory evoked potentials for monitoring of the auditory nerve during surgical procedures. In the current study, we evaluated the influence of noise on ASSR characteristics in total intravenous anesthesia (TIVA). Simulated ASSR in real noise recorded during surgery under TIVA were constructed with known parameters. Influence of amplitude, modulation frequency, averaging sweeps and detection threshold on ASSR were evaluated. High amplitude, more sweeps and a liberal threshold facilitated detection. High amplitude ASSR (80 nV) were detected in up to 45 % with 16 s of data, in 80-90 % with 112 s. Near-threshold ASSR were detected in 0.8-25 %. False positives ranged between 0.3 and 10.3 %. Number of sweeps did not influence false positives. Amplitude errors varied between -61 and +39 % and improved with more averages but not with different thresholds. Modulation rate demonstrated the strongest influence on all parameters. 110 Hz yielded best, 90 Hz the worst results. Choice of parameters strongly influences detection and characteristics of ASSR. Optimal parameters enabled detection after 16 s in 45 %. Due to specific noise characteristics, modulation has a critical impact, which is currently not sufficiently recognized in ASSR studies.


Asunto(s)
Umbral Auditivo , Nervio Coclear/patología , Monitoreo Intraoperatorio/métodos , Estimulación Acústica/métodos , Adulto , Anestesia/métodos , Tronco Encefálico/patología , Potenciales Evocados Auditivos , Potenciales Evocados Auditivos del Tronco Encefálico , Neoplasias Faciales/cirugía , Reacciones Falso Positivas , Femenino , Audición , Humanos , Masculino , Meningioma/cirugía , Persona de Mediana Edad , Neurilemoma/cirugía , Neuroma Acústico/cirugía , Ruido
15.
J Neurosci ; 35(36): 12331-45, 2015 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-26354903

RESUMEN

After substantial loss of cochlear hair cells, exogenous neurotrophins prevent degeneration of the auditory nerve. Because cochlear implantation, the current therapy for profound sensorineural hearing loss, depends on a functional nerve, application of neurotrophins is being investigated. We addressed two questions important for fundamental insight into the effects of exogenous neurotrophins on a degenerating neural system, and for translation to the clinic. First, does temporary treatment with brain-derived neurotrophic factor (BDNF) prevent nerve degeneration on the long term? Second, how does a BDNF-treated nerve respond to electrical stimulation? Deafened guinea pigs received a cochlear implant, and their cochleas were infused with BDNF for 4 weeks. Up to 8 weeks after treatment, their cochleas were analyzed histologically. Electrically evoked compound action potentials (eCAPs) were recorded using stimulation paradigms that are informative of neural survival. Spiral ganglion cell (SGC) degeneration was prevented during BDNF treatment, resulting in 1.9 times more SGCs than in deafened untreated cochleas. Importantly, SGC survival was almost complete 8 weeks after treatment cessation, when 2.6 times more SGCs were observed. In four eCAP characteristics (three involving alteration of the interphase gap of the biphasic current pulse and one involving pulse trains), we found large and statistically significant differences between normal-hearing and deaf controls. Importantly, for BDNF-treated animals, these eCAP characteristics were near normal, suggesting healthy responsiveness of BDNF-treated SGCs. In conclusion, clinically practicable short-term neurotrophin treatment is sufficient for long-term survival of SGCs, and it can restore or preserve SGC function well beyond the treatment period. Significance statement: Successful restoration of hearing in deaf subjects by means of a cochlear implant requires a healthy spiral ganglion cell population. Deafness-induced degeneration of these cells can be averted with neurotrophic factors. In the present study in deafened guinea pigs, we investigated the long-term effects of temporary (i.e., clinically practicable) treatment with brain-derived neurotrophic factor (BDNF). We show that, after treatment cessation, the neuroprotective effect remains for at least 8 weeks. Moreover, for the first time, it is shown that the electrical responsiveness of BDNF-treated spiral ganglion cells is preserved during this period as well. These findings demonstrate that treatment of the auditory nerve with neurotrophic factors may be relevant for cochlear implant users.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/farmacología , Nervio Coclear/efectos de los fármacos , Sordera/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Potenciales de Acción , Animales , Factor Neurotrófico Derivado del Encéfalo/uso terapéutico , Nervio Coclear/patología , Nervio Coclear/fisiología , Sordera/patología , Femenino , Cobayas , Fármacos Neuroprotectores/uso terapéutico , Ganglio Espiral de la Cóclea/citología , Ganglio Espiral de la Cóclea/efectos de los fármacos , Ganglio Espiral de la Cóclea/fisiología
16.
J Virol ; 90(6): 2920-7, 2015 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-26719273

RESUMEN

UNLABELLED: Approximately one-third of Lassa virus (LASV)-infected patients develop sensorineural hearing loss (SNHL) in the late stages of acute disease or in early convalescence. With 500,000 annual cases of Lassa fever (LF), LASV is a major cause of hearing loss in regions of West Africa where LF is endemic. To date, no animal models exist that depict the human pathology of LF with associated hearing loss. Here, we aimed to develop an animal model to study LASV-induced hearing loss using human isolates from a 2012 Sierra Leone outbreak. We have recently established a murine model for LF that closely mimics many features of human disease. In this model, LASV isolated from a lethal human case was highly virulent, while the virus isolated from a nonlethal case elicited mostly mild disease with moderate mortality. More importantly, both viruses were able to induce SNHL in surviving animals. However, utilization of the nonlethal, human LASV isolate allowed us to consistently produce large numbers of survivors with hearing loss. Surviving mice developed permanent hearing loss associated with mild damage to the cochlear hair cells and, strikingly, significant degeneration of the spiral ganglion cells of the auditory nerve. Therefore, the pathological changes in the inner ear of the mice with SNHL supported the phenotypic loss of hearing and provided further insights into the mechanistic cause of LF-associated hearing loss. IMPORTANCE: Sensorineural hearing loss is a major complication for LF survivors. The development of a small-animal model of LASV infection that replicates hearing loss and the clinical and pathological features of LF will significantly increase knowledge of pathogenesis and vaccine studies. In addition, such a model will permit detailed characterization of the hearing loss mechanism and allow for the development of appropriate diagnostic approaches and medical care for LF patients with hearing impairment.


Asunto(s)
Modelos Animales de Enfermedad , Pérdida Auditiva Sensorineural/patología , Fiebre de Lassa/complicaciones , Animales , Nervio Coclear/patología , Brotes de Enfermedades , Oído Interno/patología , Pérdida Auditiva Sensorineural/epidemiología , Histocitoquímica , Humanos , Fiebre de Lassa/epidemiología , Virus Lassa/aislamiento & purificación , Ratones , Microscopía , Sierra Leona/epidemiología , Virulencia
17.
Eur Arch Otorhinolaryngol ; 273(9): 2373-83, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26498948

RESUMEN

The aim of the study was to investigate the option of cochlear implantation (CI) in resultant single-sided deafness associated with unilateral translabyrinthine resection of sporadic vestibular schwannoma (VS). This is a retrospective study performed at Tertiary Care Academic Centre. Following extensive counselling regarding the potential for delayed CI, translabyrinthine VS resection was performed and an intracochlear placeholder was inserted to allow later CI in 11 patients who showed intraoperative microscopic confirmation of preserved cochlear nerve anatomy. Follow-up magnetic resonance imaging (MRI) and promontory testing were performed 1 year after surgery to confirm the absence of VS recurrence and viable cochlea. Confirmed CI candidates underwent a second procedure where the placeholder was removed and the CI inserted (4/11). Preimplant unaided and CI-aided evaluations at 12 and 24 months were performed for subjective and objective hearing outcomes. Tinnitus suppression was also measured for implant on and off effects. Available audiological data for three patients demonstrated significant hearing benefits for 'speech from deaf/implanted side, noise from the normal-hearing side' in all three patients and localisation ability improved for 2/3 patients. Subjective findings presented similar results. For the two patients with preimplant tinnitus, complete suppression occurred during active CI. CI is beneficial for hearing rehabilitation and tinnitus reduction in SSD patients with remaining viable cochlear nerve after translabyrinthine VS surgery. Counselling on the risks of intracochlear placeholder insertion and the inherent limitations for ongoing MRI investigations of VS recurrence is essential.


Asunto(s)
Implantación Coclear , Disección/efectos adversos , Pérdida Auditiva Unilateral , Recurrencia Local de Neoplasia/prevención & control , Neuroma Acústico , Complicaciones Posoperatorias , Acúfeno , Adulto , Anciano , Cóclea/patología , Cóclea/cirugía , Implantación Coclear/instrumentación , Implantación Coclear/métodos , Implantes Cocleares , Nervio Coclear/patología , Nervio Coclear/fisiopatología , Disección/métodos , Femenino , Francia , Pérdida Auditiva Sensorineural/diagnóstico , Pérdida Auditiva Sensorineural/etiología , Pérdida Auditiva Sensorineural/fisiopatología , Pérdida Auditiva Sensorineural/cirugía , Pérdida Auditiva Unilateral/diagnóstico , Pérdida Auditiva Unilateral/etiología , Pérdida Auditiva Unilateral/fisiopatología , Pérdida Auditiva Unilateral/cirugía , Pruebas Auditivas/métodos , Humanos , Masculino , Persona de Mediana Edad , Neuroma Acústico/patología , Neuroma Acústico/cirugía , Complicaciones Posoperatorias/diagnóstico , Complicaciones Posoperatorias/fisiopatología , Complicaciones Posoperatorias/cirugía , Estudios Retrospectivos , Percepción del Habla , Acúfeno/diagnóstico , Acúfeno/etiología , Acúfeno/cirugía , Resultado del Tratamiento
18.
J Neurosci ; 34(7): 2684-8, 2014 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-24523557

RESUMEN

Exposure to loud sound (acoustic overexposure; AOE) induces hearing loss and damages cellular structures at multiple locations in the auditory pathway. Whether AOE can also induce changes in myelin sheaths of the auditory nerve (AN) is an important issue particularly because these changes can be responsible for impaired action potential propagation along the AN. Here we investigate the effects of AOE on morphological and electrophysiological features of the centrally directed part of the rat AN projecting from the cochlear spiral ganglion to brainstem cochlear nuclei. Using electron microscopy and immunocytochemistry, we show that AOE elongates the AN nodes of Ranvier and triggers notable perinodal morphological changes. Compound action potential recordings of the AN coupled to biophysical modeling demonstrated that these nodal and perinodal structural changes were associated with decreased conduction velocity and conduction block. Furthermore, AOE decreased the number of release sites in the cochlear nuclei associated with the reduced amplitudes of EPSCs evoked by AN stimulation. In conclusion, AN dysmyelination may be of fundamental importance in auditory impairment following exposure to loud sound.


Asunto(s)
Nervio Coclear/patología , Pérdida Auditiva Provocada por Ruido/patología , Vaina de Mielina/patología , Nódulos de Ranvier/patología , Potenciales de Acción/fisiología , Animales , Nervio Coclear/fisiopatología , Modelos Animales de Enfermedad , Potenciales Evocados Auditivos/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Pérdida Auditiva Provocada por Ruido/fisiopatología , Masculino , Técnicas de Placa-Clamp , Ratas , Ratas Wistar
19.
J Neurosci ; 34(2): 434-45, 2014 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-24403143

RESUMEN

The auxiliary subunit α2δ3 modulates the expression and function of voltage-gated calcium channels. Here we show that α2δ3 mRNA is expressed in spiral ganglion neurons and auditory brainstem nuclei and that the protein is required for normal acoustic responses. Genetic deletion of α2δ3 led to impaired auditory processing, with reduced acoustic startle and distorted auditory brainstem responses. α2δ3(-/-) mice learned to discriminate pure tones, but they failed to discriminate temporally structured amplitude-modulated tones. Light and electron microscopy analyses revealed reduced levels of presynaptic Ca(2+) channels and smaller auditory nerve fiber terminals contacting cochlear nucleus bushy cells. Juxtacellular in vivo recordings of sound-evoked activity in α2δ3(-/-) mice demonstrated impaired transmission at these synapses. Together, our results identify a novel role for the α2δ3 auxiliary subunit in the structure and function of specific synapses in the mammalian auditory pathway and in auditory processing disorders.


Asunto(s)
Trastornos de la Percepción Auditiva/metabolismo , Canales de Calcio/metabolismo , Nervio Coclear/metabolismo , Aprendizaje Discriminativo/fisiología , Sinapsis/metabolismo , Animales , Trastornos de la Percepción Auditiva/genética , Trastornos de la Percepción Auditiva/fisiopatología , Tronco Encefálico/metabolismo , Tronco Encefálico/patología , Canales de Calcio/genética , Nervio Coclear/patología , Electrofisiología , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ganglio Espiral de la Cóclea/metabolismo , Ganglio Espiral de la Cóclea/fisiología , Sinapsis/patología , Transmisión Sináptica/fisiología
20.
Proc Natl Acad Sci U S A ; 109(21): 8292-7, 2012 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-22566618

RESUMEN

Exposure to loud sound causes cochlear damage resulting in hearing loss and tinnitus. Tinnitus has been related to hyperactivity in the central auditory pathway occurring weeks after loud sound exposure. However, central excitability changes concomitant to hearing loss and preceding those periods of hyperactivity, remain poorly explored. Here we investigate mechanisms contributing to excitability changes in the dorsal cochlear nucleus (DCN) shortly after exposure to loud sound that produces hearing loss. We show that acoustic overexposure alters synaptic transmission originating from the auditory and the multisensory pathway within the DCN in different ways. A reduction in the number of myelinated auditory nerve fibers leads to a reduced maximal firing rate of DCN principal cells, which cannot be restored by increasing auditory nerve fiber recruitment. In contrast, a decreased membrane resistance of DCN granule cells (multisensory inputs) leads to a reduced maximal firing rate of DCN principal cells that is overcome when additional multisensory fibers are recruited. Furthermore, gain modulation by inhibitory synaptic transmission is disabled in both auditory and multisensory pathways. These cellular mechanisms that contribute to decreased cellular excitability in the central auditory pathway are likely to represent early neurobiological markers of hearing loss and may suggest interventions to delay or stop the development of hyperactivity that has been associated with tinnitus.


Asunto(s)
Nervio Coclear/fisiopatología , Núcleo Coclear/fisiopatología , Pérdida Auditiva Provocada por Ruido/fisiopatología , Acúfeno/fisiopatología , Animales , Vías Auditivas/patología , Vías Auditivas/fisiopatología , Umbral Auditivo/fisiología , Cóclea/patología , Cóclea/fisiopatología , Nervio Coclear/patología , Núcleo Coclear/patología , Potenciales Postsinápticos Excitadores/fisiología , Pérdida Auditiva Provocada por Ruido/patología , Humanos , Potenciales Postsinápticos Inhibidores/fisiología , Fibras Nerviosas Mielínicas/patología , Fibras Nerviosas Mielínicas/fisiología , Inhibición Neural/fisiología , Técnicas de Placa-Clamp , Ratas , Ratas Wistar , Acúfeno/patología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda