Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 169
Filtrar
1.
Neural Plast ; 2021: 6678863, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34257641

RESUMO

Patients with profound bilateral deafness (BD) are prone to suffering from tinnitus, which further leads to psychological comorbidities and makes it more difficult for patients to communicate with people. This study was aimed at investigating the effect of cochlear implants (CIs) on tinnitus distress and psychological comorbidities in patients with profound BD. This multicenter retrospective study reviewed 51 patients with severe postlingual BD who underwent cochlear implantation; 49 patients underwent unilateral cochlear implantation, and 2 patients underwent bilateral cochlear implantation. The patients were asked to complete all the questionnaires, including the tinnitus handicap inventory (THI), the visual analog scale (VAS) score, the Hospital Anxiety and Depression Scale Questionnaire (HADS), the Categories of Auditory Performance (CAP), and the Speech Intelligibility Rating (SIR), at least 4 months after implantation when the CI was on or off, in approximately May-June 2019. In our study, 94% (48/51) of BD patients suffered from tinnitus before CI, and 77% (37/48) of them suffered from bilateral tinnitus. In addition, 50.9% (26/51) of the CI patients were suffering from anxiety, 52.9% (27/51) of them were suffering from depression (score ≥ 8), and 66.7% (34/51) (27/51) of them were suffering from anxiety or depression. Cochlear implantation could reduce tinnitus more obviously when the CI was on than when the CI was off. Cochlear implantation also reduced anxiety/depression severity. There were significantly positive correlations between tinnitus severity and anxiety/depression severity before and after surgery. Moreover, hearing improvement is positively correlated with reduction level of tinnitus, the better hearing, and the lesser severity of tinnitus. Thus, along with effective restoration of deafferentation, cochlear implantation shows positive therapeutic effects on tinnitus and psychological comorbidities, providing a reference for future clinical and research work.


Assuntos
Ansiedade/terapia , Implante Coclear , Implantes Cocleares , Depressão/terapia , Perda Auditiva Bilateral/complicações , Zumbido/terapia , Adulto , Vias Aferentes/fisiopatologia , Idoso , Ansiedade/etiologia , Vias Auditivas/fisiopatologia , Núcleo Coclear/fisiopatologia , Depressão/etiologia , Feminino , Perda Auditiva Bilateral/cirurgia , Humanos , Colículos Inferiores/fisiopatologia , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Inteligibilidade da Fala , Inquéritos e Questionários , Zumbido/etiologia , Zumbido/fisiopatologia , Zumbido/psicologia , Escala Visual Analógica
2.
Trends Hear ; 23: 2331216519835080, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30868944

RESUMO

Deep brain stimulation of the central auditory pathway is emerging as a promising treatment modality for tinnitus. Within this pathway, the dorsal cochlear nucleus (DCN) plays a key role in the pathophysiology of tinnitus and is believed to be a tinnitus generator. We hypothesized that high-frequency stimulation (HFS) of the DCN would influence tinnitus-related abnormal neuronal activity within the auditory pathway and hereby suppress tinnitus. To this end, we assessed the effect of HFS of the DCN in a noise-induced rat model of tinnitus. The presence of tinnitus was verified using the gap prepulse inhibition of the acoustic startle response paradigm. Hearing thresholds were determined before and after noise trauma by measuring the auditory brainstem responses. In addition, changes in neuronal activity induced by noise trauma and HFS were assessed using c-Fos immunohistochemistry in related structures. Results showed tinnitus development after noise trauma and hearing loss ipsilateral to the side exposed to noise trauma. During HFS of the DCN, tinnitus was suppressed. There was no change in c-Fos expression within the central auditory pathway after HFS. These findings suggest that DCN-HFS changes patterns of activity and results in information lesioning within the network and hereby blocking the relay of abnormal tinnitus-related neuronal activity.


Assuntos
Núcleo Coclear/fisiopatologia , Estimulação Encefálica Profunda/métodos , Zumbido/fisiopatologia , Zumbido/terapia , Animais , Vias Auditivas/fisiopatologia , Modelos Animais de Doenças , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Masculino , Ruído , Ratos , Ratos Sprague-Dawley , Reflexo de Sobressalto
3.
Hear Res ; 374: 1-4, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30669034

RESUMO

The Scn8amedJ mutation of the gene for sodium channels at the nodes of Ranvier slows nerve conduction, resulting in motor abnormalities. This mutation is also associated with loss of spontaneous bursting activity in the dorsal cochlear nucleus. However initial tests of auditory sensitivity in mice homozygous for this mutation, using standard 400-ms tones, demonstrated normal hearing sensitivity. Further testing, reported here, revealed a severely compromised sensitivity to short-duration tones of 10 and 2 ms durations. Such a deficit might be expected to interfere with auditory functions that depend on rapid processing of auditory signals.


Assuntos
Limiar Auditivo/fisiologia , Mutação , Canal de Sódio Disparado por Voltagem NAV1.6/deficiência , Canal de Sódio Disparado por Voltagem NAV1.6/genética , Estimulação Acústica , Animais , Audiometria de Tons Puros , Núcleo Coclear/fisiopatologia , Testes Auditivos , Homozigoto , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Canal de Sódio Disparado por Voltagem NAV1.6/fisiologia , Condução Nervosa/genética , Condução Nervosa/fisiologia , Nós Neurofibrosos/fisiologia , Fatores de Tempo
4.
Neuroscience ; 399: 184-198, 2019 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-30593923

RESUMO

The cochlear nucleus, located in the brainstem, receives its afferent auditory input exclusively from the auditory nerve fibers of the ipsilateral cochlea. Noise-induced neurodegenerative changes occurring in the auditory nerve stimulate a cascade of neuroplastic changes in the cochlear nucleus resulting in major changes in synaptic structure and function. To identify some of the key molecular mechanisms mediating this synaptic reorganization, we unilaterally exposed rats to a high-intensity noise that caused significant hearing loss and then measured the resulting changes in a synaptic plasticity gene array targeting neurogenesis and synaptic reorganization. We compared the gene expression patterns in the dorsal cochlear nucleus (DCN) and ventral cochlear nucleus (VCN) on the noise-exposed side versus the unexposed side using a PCR gene array at 2 d (early) and 28 d (late) post-exposure. We discovered a number of differentially expressed genes, particularly those related to synaptogenesis and regeneration. Significant gene expression changes occurred more frequently in the VCN than the DCN and more changes were seen at 28  d versus 2 d post-exposure. We confirmed the PCR findings by in situ hybridization for Brain-derived neurotrophic factor (Bdnf), Homer-1, as well as the glutamate NMDA receptor Grin1, all involved in neurogenesis and plasticity. These results suggest that Bdnf, Homer-1 and Grin1 play important roles in synaptic remodeling and homeostasis in the cochlear nucleus following severe noise-induced afferent degeneration.


Assuntos
Núcleo Coclear/fisiopatologia , Perda Auditiva Provocada por Ruído/fisiopatologia , Plasticidade Neuronal/fisiologia , Sinapses/fisiologia , Estimulação Acústica/efeitos adversos , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Núcleo Coclear/patologia , Modelos Animais de Doenças , Potenciais Evocados Auditivos do Tronco Encefálico , Regulação da Expressão Gênica , Perda Auditiva Provocada por Ruído/patologia , Proteínas de Arcabouço Homer/metabolismo , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/patologia , Fatores de Tempo
5.
Neuroscience ; 400: 1-16, 2019 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-30594562

RESUMO

Worldwide, almost 500 million people are hearing impaired, making hearing loss the most common sensory impairment among humans. For people with single-sided deafness (SSD), cochlear implants (CIs) can be enormously beneficial by providing binaural information. However, binaural benefits in CI users have been only incompletely realized. Overcoming these limitations requires a better knowledge of how neuronal circuits adapt to SSD and how unilateral CI stimulation can compensate a deaf ear. We investigated effects of neonatal SSD on auditory brainstem circuitry using acoustic (AS), electric (ES), or acoustic stimulation on one ear and electric stimulation on the other ear (AS + ES). The molecular marker Fos was used to investigate changes in interneuronal communication due to SSD. To induce SSD, neonatal rats obtained a unilateral intracochlear injection of neomycin. In adulthood, rats were acutely stimulated by AS, ES, or AS + ES. AS and ES were applied correspondingly in terms of intracochlear stimulation side and intensity resulting in bilaterally comparable Fos expression in hearing rats. In contrast, SSD rats showed a loss of tonotopic order along the deafened pathway, indicated by a massive increase and spread of Fos expressing neurons. We report three major results: First, AS of the hearing ear of SSD rats resulted in bilateral activation of neurons in the cochlear nucleus (CN). Second, ES of the deaf ear did not activate contralateral CN. Third, AS + ES of SSD rats resulted in bilateral reduced Fos expression in the auditory brainstem compared to monaural stimulations. These findings indicate changes in inhibitory interactions among neuronal networks as a result of monaural deafness.


Assuntos
Núcleo Coclear/fisiopatologia , Surdez/fisiopatologia , Neurônios/fisiologia , Estimulação Acústica , Animais , Vias Auditivas/fisiopatologia , Implante Coclear , Estimulação Elétrica , Potenciais Evocados Auditivos do Tronco Encefálico , Feminino , Lateralidade Funcional , Masculino , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos Wistar
6.
Neuroscience ; 407: 170-181, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-30217755

RESUMO

Following noise overexposure and tinnitus-induction, fusiform cells of the dorsal cochlear nucleus (DCN) show increased spontaneous firing rates (SFR), increased spontaneous synchrony and altered stimulus-timing-dependent plasticity (StDP), which correlate with behavioral measures of tinnitus. Sodium salicylate, the active ingredient in aspirin, which is commonly used to induce tinnitus, increases SFR and activates NMDA receptors in the ascending auditory pathway. NMDA receptor activation is required for StDP in many brain regions, including the DCN. Blocking NMDA receptors can alter StDP timing rules and decrease synchrony in DCN fusiform cells. Thus, systemic activation of NMDA receptors with sodium salicylate should elicit pathological changes to StDP, thereby increasing SFR and synchrony and induce tinnitus. Herein, we examined the action of salicylate in tinnitus generation in guinea pigs in vivo by measuring tinnitus using two behavioral measures and recording single-unit responses from DCN fusiform cells pre- and post-salicylate administration in the same animals. First, we show that animals administered salicylate show evidence of tinnitus using both behavioral paradigms, cross-validating the tests. Second, fusiform cells in animals with tinnitus showed increased SFR, synchrony and altered StDP timing rules, like animals with noise-induced tinnitus. These findings suggest that alterations to fusiform-cell plasticity are an essential component of tinnitus, regardless of induction technique.


Assuntos
Plasticidade Celular/fisiologia , Núcleo Coclear/fisiopatologia , Plasticidade Neuronal/fisiologia , Zumbido/fisiopatologia , Animais , Vias Auditivas/efeitos dos fármacos , Vias Auditivas/fisiologia , Plasticidade Celular/efeitos dos fármacos , Potenciais Evocados Auditivos/efeitos dos fármacos , Potenciais Evocados Auditivos/fisiologia , Potenciais Evocados Auditivos do Tronco Encefálico/efeitos dos fármacos , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Cobaias , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Ruído , Salicilato de Sódio/farmacologia
7.
eNeuro ; 5(4)2018.
Artigo em Inglês | MEDLINE | ID: mdl-30123822

RESUMO

Noise exposure is one of the most common causes of hearing loss and peripheral damage to the auditory system. A growing literature suggests that the auditory system can compensate for peripheral loss through increased central neural activity. The current study sought to investigate the link between noise exposure, increases in central gain, synaptic reorganization, and auditory function. All axons of the auditory nerve project to the cochlear nucleus, making it a requisite nucleus for sound detection. As the first synapse in the central auditory system, the cochlear nucleus is well positioned to respond plastically to loss of peripheral input. To investigate noise-induced compensation in the central auditory system, we measured auditory brainstem responses (ABRs) and auditory perception and collected tissue from mice exposed to broadband noise. Noise-exposed mice showed elevated ABR thresholds, reduced ABR wave 1 amplitudes, and spiral ganglion neuron loss. Despite peripheral damage, noise-exposed mice were hyperreactive to loud sounds and showed nearly normal behavioral sound detection thresholds. Ratios of late ABR peaks (2-4) relative to the first ABR peak indicated that brainstem pathways were hyperactive in noise-exposed mice, while anatomical analysis indicated there was an imbalance between expression of excitatory and inhibitory proteins in the ventral cochlear nucleus. The results of the current study suggest that a reorganization of excitation and inhibition in the ventral cochlear nucleus may drive hyperactivity in the central auditory system. This increase in central gain can compensate for peripheral loss to restore some aspects of auditory function.


Assuntos
Percepção Auditiva/fisiologia , Limiar Auditivo/fisiologia , Comportamento Animal/fisiologia , Núcleo Coclear/fisiopatologia , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Hipercinese/fisiopatologia , Ruído/efeitos adversos , Gânglio Espiral da Cóclea/citologia , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos CBA
8.
J Neurosurg ; 128(1): 296-303, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28298014

RESUMO

OBJECTIVE The risk of injury of the cochlear nerve during angle (CPA) surgery is high. Granulocyte colony-stimulating factor (G-CSF) has been found in various experimental models of peripheral and CNS injury to have a neuroprotective effect by inhibiting apoptosis and inflammation. However, to the authors' knowledge, the influence of G-CSF on cochlear nerve regeneration has not been reported. This study investigated the neuroprotective effect of G-CSF after a partial cochlear nerve lesion in rats. METHODS A lesion of the right cochlear nerve in adult male Sprague-Dawley rats was created using a water-jet dissector with a pressure of 8 bar. In the first group (G-CSF-post), G-CSF was administrated on Days 1, 3, and 5 after the surgery. The second group (G-CSF-pre/post) was treated with G-CSF 1 day before and 1, 3, and 5 days after applying the nerve injury. The control group received sodium chloride after nerve injury at the various time points. Brainstem auditory evoked potentials (BAEPs) were measured directly before and after nerve injury and on Days 1 and 7 to evaluate the acoustic function of the cochlear nerve. The animals were sacrificed 1 week after the operation, and their brains were fixed in formalin. Nissl staining of the cochlear nuclei was performed, and histological sections were analyzed with a light microscope and an image-processing program. The numbers of neurons in the cochlear nuclei were assessed. RESULTS The values for Waves 2 and 4 of the BAEPs decreased abruptly in all 3 groups in the direct postoperative measurement. Although the amplitude in the control group did not recover, it increased in both treatment groups. According to 2-way ANOVA, groups treated with G-CSF had a significant increase in BAEP Wave II amplitudes on the right side (p = 0.0401) after the applied cochlear nerve injury. With respect to Wave IV, a trend toward better recovery in the G-CSF groups was found, but this difference did not reach statistical significance. In the histological analysis, higher numbers of neurons were found in the G-CSF groups. In the statistical analysis, the difference in the numbers of neurons between the control and G-CSF-post groups reached significance (p = 0.0086). The difference in the numbers of neurons between the control and G-CSF-pre/post groups and between the G-CSF-post and G-CSF-pre/post groups did not reach statistical significance. CONCLUSIONS The use of G-CSF improved the function of the eighth cranial nerve and protected cochlear nucleus cells from destruction after a controlled partial injury of the nerve. These findings might be relevant for surgery that involves CPA tumors. The use of G-CSF in patients with a lesion in the CPA might improve postoperative outcomes.


Assuntos
Nervo Coclear/efeitos dos fármacos , Nervo Coclear/lesões , Núcleo Coclear/efeitos dos fármacos , Núcleo Coclear/lesões , Fator Estimulador de Colônias de Granulócitos/farmacologia , Fármacos Neuroprotetores/farmacologia , Animais , Nervo Coclear/fisiopatologia , Núcleo Coclear/fisiopatologia , Modelos Animais de Doenças , Potenciais Evocados Auditivos do Tronco Encefálico/efeitos dos fármacos , Masculino , Distribuição Aleatória , Ratos Sprague-Dawley
9.
Biomed Res Int ; 2018: 8624745, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30599000

RESUMO

Single-sided deafness (SSD) induces cortical neural plastic changes according to duration of deafness. However, it is still unclear how the auditory cortical changes accompany the subcortical neural changes. The present study aimed to find the neural plastic changes in the cortical and subcortical auditory system following adult-onset single-sided deafness (SSD) using Mn-enhanced magnetic resonance imaging (MEMRI). B57BL/6 mice (postnatal 8-week-old) were divided into three groups: the SSD-4-week group (postnatal 12-week-old, n = 11), the SSD-8-week group (postnatal 16-week-old, n = 11), and a normal-hearing control group (postnatal 8-week-old, n = 9). The left cochlea was ablated in the SSD groups. White Gaussian noise was delivered for 24 h before MEMRI acquisition. T1-weighted MRI data were analyzed from the cochlear nucleus (CN), superior olivary complex (SOC), lateral lemniscus (LL), inferior colliculus (IC), medial geniculate body (MG), and auditory cortex (AC). The differences in relative Mn2+-enhanced signal intensities (Mn2+SI) and laterality were analyzed between the groups. Four weeks after the SSD procedure, the ipsilateral side of the SSD showed significantly lower Mn2+SI in the CN than the control group. On the other hand, the contralateral side of the SSD demonstrated significantly lower Mn2+SI in the SOC, LL, and IC. These decreased Mn2+SI values were partially recovered at 8 weeks after the SSD procedure. The interaural Mn2+SI differences representing the interaural dominance were highest in CN and then became less prominently higher in the auditory neural system. The SSD-8-week group still showed interaural differences in the CN, LL, and IC. In contrast, the MG and AC did not show any significant intergroup or interaural differences in Mn2+SI. In conclusion, subcortical auditory neural activities were decreased after SSD, and the interaural differences were diluted in the higher auditory nervous system. These findings were attenuated with time. Subcortical auditory neural changes after SSD may contribute to the change in tinnitus severity and the outcomes of cochlear implantation in SSD patients.


Assuntos
Córtex Auditivo/fisiopatologia , Vias Auditivas/fisiopatologia , Surdez/fisiopatologia , Vias Neurais/fisiopatologia , Plasticidade Neuronal/fisiologia , Animais , Cóclea/fisiopatologia , Implante Coclear/métodos , Implantes Cocleares , Núcleo Coclear/fisiopatologia , Modelos Animais de Doenças , Lateralidade Funcional/fisiologia , Testes Auditivos/métodos , Colículos Inferiores/fisiopatologia , Imageamento por Ressonância Magnética/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Zumbido/fisiopatologia
10.
Hear Res ; 354: 28-37, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28843833

RESUMO

This investigation compared the development of neuronal excitability in the ventral nucleus of the trapezoid body (VNTB) between two strains of mice with differing progression rates for age-related hearing loss. In contrast to CBA/Ca (CBA) mice, the C57BL/6J (C57) strain are subject to hearing loss from a younger age and are more prone to damage from sound over-exposure. Higher firing rates in the medial olivocochlear system (MOC) are associated with protection from loud sounds and these cells are located in the VNTB. We postulated that reduced neuronal firing of the MOC in C57 mice could contribute to hearing loss in this strain by reducing efferent protection. Whole cell patch clamp was used to compare the electrical properties of VNTB neurons from the two strains initially in two age groups: before and after hearing onset at âˆ¼ P9 and ∼P16, respectively. Prior to hearing onset VNTB neurons electrophysiological properties were identical in both strains, but started to diverge after hearing onset. One week after hearing onset VNTB neurons of C57 mice had larger amplitude action potentials but in contrast to CBA mice, their waveform failed to accelerate with increasing age, consistent with the faster inactivation of voltage-gated potassium currents in C57 VNTB neurons. The lower frequency action potential firing of C57 VNTB neurons at P16 was maintained to P28, indicating that this change was not a developmental delay. We conclude that C57 VNTB neurons fire at lower frequencies than in the CBA strain, supporting the hypothesis that reduced MOC firing could contribute to the greater hearing loss of the C57 strain.


Assuntos
Potenciais Evocados Auditivos do Tronco Encefálico , Audição , Presbiacusia/fisiopatologia , Corpo Trapezoide/fisiopatologia , Fatores Etários , Envelhecimento , Animais , Vias Auditivas/metabolismo , Vias Auditivas/fisiopatologia , Núcleo Coclear/metabolismo , Núcleo Coclear/fisiopatologia , Estimulação Elétrica , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Neurônios/metabolismo , Núcleo Olivar/metabolismo , Núcleo Olivar/fisiopatologia , Técnicas de Patch-Clamp , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Presbiacusia/metabolismo , Tempo de Reação , Especificidade da Espécie , Fatores de Tempo , Corpo Trapezoide/metabolismo
11.
Hear Res ; 350: 139-151, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28478300

RESUMO

Animal model research has shown that the central features of tinnitus, the perception of sound without an acoustic correlate, include elevated spontaneous and stimulus-driven activity, enhanced burst-mode firing, decreased variance of inter-spike intervals, and distortion of tonotopic frequency representation. Less well documented are cell-specific correlates of tinnitus. Unipolar brush cell (UBC) alterations in animals with psychophysical evidence of tinnitus has recently been reported. UBCs are glutamatergic interneurons that appear to function as local-circuit signal amplifiers. UBCs are abundant in the dorsal cochlear nucleus (DCN) and very abundant in the flocculus (FL) and paraflocculus (PFL) of the cerebellum. In the present research, two indicators of UBC structure and function were examined: Doublecortin (DCX) and epidermal growth factor receptor substrate 8 (Eps8). DCX is a protein that binds to microtubules where it can modify their assembly and growth. Eps8 is a cell-surface tyrosine kinase receptor mediating the response to epidermal growth factor; it appears to have a role in actin polymerization as well as cytoskeletal protein interactions. Both functions could contribute to synaptic remodeling. In the present research UBC Eps8 and DCX immunoreactivity (IR) were determined in 4 groups of rats distinguished by their exposure to high-level sound and psychophysical performance: Unexposed, exposed to high-level sound with behavioral evidence of tinnitus, and two exposed groups without behavioral evidence of tinnitus. Compared to unexposed controls, exposed animals with tinnitus had Eps8 IR elevated in their PFL; other structures were not affected, nor was DCX IR affected. This was interpreted as UBC upregulation in animals with tinnitus. Exposure that failed to produce tinnitus did not increase either Eps8 or DCX IR. Rather Eps8 IR was decreased in the FL and DCN of one subgroup (Least-Tinnitus), while DCX IR decreased in the FL of the other subgroup (No-Tinnitus). Neuron degeneration was also documented in the cochlear nucleus and PFL of exposed animals, both with and without tinnitus. Degeneration was not found in unexposed animals. Implications for tinnitus neuropathy are discussed in the context of synaptic remodeling and cerebellar sensory modulation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cerebelo/metabolismo , Núcleo Coclear/metabolismo , Interneurônios/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Neuropeptídeos/metabolismo , Zumbido/metabolismo , Animais , Percepção Auditiva , Comportamento Animal , Biomarcadores/metabolismo , Cerebelo/patologia , Cerebelo/fisiopatologia , Doença Crônica , Núcleo Coclear/patologia , Núcleo Coclear/fisiopatologia , Modelos Animais de Doenças , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Potenciais Evocados Auditivos do Tronco Encefálico , Audição , Interneurônios/patologia , Masculino , Degeneração Neural , Ruído , Ratos Long-Evans , Zumbido/patologia , Zumbido/fisiopatologia , Zumbido/psicologia
12.
Hear Res ; 347: 28-40, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27989950

RESUMO

High dose of salicylate, the active ingredient in aspirin, has long been known to induce transient hearing loss, tinnitus and hyperacusis making it a powerful experimental tool. These salicylate-induced perceptual disturbances are associated with a massive reduction in the neural output of the cochlea. Paradoxically, the diminished neural output of the cochlea is accompanied by a dramatic increase in sound-evoked activity in the auditory cortex (AC) and several other parts of the central nervous system. Exactly where the increase in neural activity begins and builds up along the central auditory pathway are not fully understood. To address this issue, we measured sound-evoked neural activity in the cochlea, cochlear nucleus (CN), inferior colliculus (IC), and AC before and after administering a high dose of sodium salicylate (SS, 300 mg/kg). The SS-treatment abolished low-level sound-evoked responses along the auditory pathway resulting in a 20-30 dB threshold shift. While the neural output of the cochlea was substantially reduced at high intensities, the neural responses in the CN were only slightly reduced; those in the IC were nearly normal or slightly enhanced while those in the AC considerably enhanced, indicative of a progress increase in central gain. The SS-induced increase in central response in the IC and AC was frequency-dependent with the greatest increase occurring in the mid-frequency range the putative pitch of SS-induced tinnitus. This frequency-dependent hyperactivity appeared to result from shifts in the frequency receptive fields (FRF) such that the response areas of many FRF shifted/expanded toward the mid-frequencies. Our results suggest that the SS-induced threshold shift originates in the cochlea. In contrast, enhanced central gain is not localized to one region, but progressively builds up at successively higher stage of the auditory pathway either through a loss of inhibition and/or increased excitation.


Assuntos
Vias Auditivas/fisiopatologia , Limiar Auditivo , Comportamento Animal , Transtornos da Audição/fisiopatologia , Transtornos da Audição/psicologia , Audição , Plasticidade Neuronal , Salicilato de Sódio , Estimulação Acústica , Adaptação Fisiológica , Adaptação Psicológica , Animais , Córtex Auditivo/fisiopatologia , Fadiga Auditiva , Cóclea/fisiopatologia , Núcleo Coclear/fisiopatologia , Modelos Animais de Doenças , Potenciais Evocados Auditivos , Transtornos da Audição/induzido quimicamente , Transtornos da Audição/prevenção & controle , Colículos Inferiores/fisiopatologia , Masculino , Ratos Sprague-Dawley , Fatores de Tempo
13.
Med Hypotheses ; 98: 2-4, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28012597

RESUMO

Subjective tinnitus and cervical spine disorders (CSD) are among the most common complaints encountered by physicians. Although the relationship between tinnitus and CSD has attracted great interest during the past several years, the pathogenesis of tinnitus induced by CSD remains unclear. Conceivably, CSD could trigger a somatosensory pathway-induced disinhibition of dorsal cochlear nucleus (DCN) activity in the auditory pathway; furthermore, CSD can cause inner ear blood impairment induced by vertebral arteries hemodynamic alterations and trigeminal irritation. In genetically -predisposed CSD patients with reduced serotoninergic tone, signals from chronically stimulated DCNs could activate specific cortical neuronal networks and plastic neural changes resulting in tinnitus. Therefore, an early specific tailored CSD treatments and/or boosting serotoninergic activity may be required to prevent the creation of 'tinnitus memory circuits' in CSD patients.


Assuntos
Vértebras Cervicais/patologia , Zumbido/complicações , Zumbido/fisiopatologia , Estimulação Acústica , Animais , Núcleo Coclear/fisiopatologia , Hemodinâmica , Humanos , Modelos Neurológicos , Modelos Teóricos , Rede Nervosa , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Tomografia por Emissão de Pósitrons , Serotonina/metabolismo , Transdução de Sinais , Transmissão Sináptica , Nervo Trigêmeo/fisiopatologia
14.
Hear Res ; 344: 284-294, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28011083

RESUMO

AMPA glutamate receptor complexes with fast kinetics conferred by subunits like GluA3 and GluA4 are essential for temporal precision of synaptic transmission. The specific role of GluA3 in auditory processing and experience related changes in the auditory brainstem remain unknown. We investigated the role of the GluA3 in auditory processing by using wild type (WT) and GluA3 knockout (GluA3-KO) mice. We recorded auditory brainstem responses (ABR) to assess auditory function and used electron microscopy to evaluate the ultrastructure of the auditory nerve synapse on bushy cells (AN-BC synapse). Since labeling for GluA3 subunit increases on auditory nerve synapses within the cochlear nucleus in response to transient sound reduction, we investigated the role of GluA3 in experience-dependent changes in auditory processing. We induced transient sound reduction by plugging one ear and evaluated ABR threshold and peak amplitude recovery for up to 60 days after ear plug removal in WT and GluA3-KO mice. We found that the deletion of GluA3 leads to impaired auditory signaling that is reflected in decreased ABR peak amplitudes, an increased latency of peak 2, early onset hearing loss and reduced numbers and sizes of postsynaptic densities (PSDs) of AN-BC synapses. Additionally, the lack of GluA3 hampers ABR threshold recovery after transient ear plugging. We conclude that GluA3 is required for normal auditory signaling, normal ultrastructure of AN-BC synapses in the cochlear nucleus and normal experience-dependent changes in auditory processing after transient sound reduction.


Assuntos
Percepção Auditiva , Comportamento Animal , Nervo Coclear/metabolismo , Núcleo Coclear/metabolismo , Perda Auditiva de Alta Frequência/metabolismo , Audição , Receptores de AMPA/deficiência , Sinapses/metabolismo , Estimulação Acústica , Adaptação Fisiológica , Animais , Nervo Coclear/fisiopatologia , Nervo Coclear/ultraestrutura , Núcleo Coclear/fisiopatologia , Núcleo Coclear/ultraestrutura , Potenciais Evocados Auditivos do Tronco Encefálico , Predisposição Genética para Doença , Perda Auditiva de Alta Frequência/genética , Perda Auditiva de Alta Frequência/patologia , Perda Auditiva de Alta Frequência/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Tempo de Reação , Receptores de AMPA/genética , Sinapses/ultraestrutura , Fatores de Tempo
15.
Hear Res ; 343: 14-33, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27473502

RESUMO

Studies of congenital and early-onset deafness have demonstrated that an absence of peripheral sound-evoked activity in the auditory nerve causes pathological changes in central auditory structures. The aim of this study was to establish whether progressive acquired hearing loss could lead to similar brain changes that would degrade the precision of signal transmission. We used complementary physiologic hearing tests and microscopic techniques to study the combined effect of both magnitude and duration of hearing loss on one of the first auditory synapses in the brain, the endbulb of Held (EB), along with its bushy cell (BC) target in the anteroventral cochlear nucleus. We compared two hearing mouse strains (CBA/Ca and heterozygous shaker-2+/-) against a model of early-onset progressive hearing loss (DBA/2) and a model of congenital deafness (homozygous shaker-2-/-), examining each strain at 1, 3, and 6 months of age. Furthermore, we employed a frequency model of the mouse cochlear nucleus to constrain our analyses to regions most likely to exhibit graded changes in hearing function with time. No significant differences in the gross morphology of EB or BC structure were observed in 1-month-old animals, indicating uninterrupted development. However, in animals with hearing loss, both EBs and BCs exhibited a graded reduction in size that paralleled the hearing loss, with the most severe pathology seen in deaf 6-month-old shaker-2-/- mice. Ultrastructural pathologies associated with hearing loss were less dramatic: minor changes were observed in terminal size but mitochondrial fraction and postsynaptic densities remained relatively stable. These results indicate that acquired progressive hearing loss can have consequences on auditory brain structure, with prolonged loss leading to greater pathologies. Our findings suggest a role for early intervention with assistive devices in order to mitigate long-term pathology and loss of function.


Assuntos
Nervo Coclear/ultraestrutura , Núcleo Coclear/ultraestrutura , Perda Auditiva/patologia , Audição , Sinapses/ultraestrutura , Estimulação Acústica , Fatores Etários , Animais , Limiar Auditivo , Comportamento Animal , Nervo Coclear/fisiopatologia , Núcleo Coclear/fisiopatologia , Modelos Animais de Doenças , Progressão da Doença , Potenciais Evocados Auditivos do Tronco Encefálico , Feminino , Predisposição Genética para Doença , Audição/genética , Perda Auditiva/genética , Perda Auditiva/fisiopatologia , Perda Auditiva/psicologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Endogâmicos DBA , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Miosinas/deficiência , Miosinas/genética , Fenótipo , Índice de Gravidade de Doença , Fatores de Tempo
16.
J Neurosci ; 36(39): 10214-27, 2016 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-27683915

RESUMO

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.


Assuntos
Nervo Coclear/patologia , Nervo Coclear/fisiopatologia , Núcleo Coclear/patologia , Núcleo Coclear/fisiopatologia , Perda Auditiva Condutiva/patologia , Perda Auditiva Condutiva/fisiopatologia , Sinapses/patologia , Animais , Percepção Auditiva , Potenciação de Longa Duração , Depressão Sináptica de Longo Prazo , Masculino , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/patologia , Ratos , Ratos Sprague-Dawley , Sinapses/metabolismo , Potenciais Sinápticos
17.
Mol Cell Neurosci ; 75: 101-12, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27473923

RESUMO

Severe noise-induced damage to the inner ear leads to auditory nerve fiber degeneration thereby reducing the neural input to the cochlear nucleus (CN). Paradoxically, this leads to a significant increase in spontaneous activity in the CN which has been linked to tinnitus, hyperacusis and ear pain. The biological mechanisms that lead to an increased spontaneous activity are largely unknown, but could arise from changes in glutamatergic or GABAergic neurotransmission or neuroinflammation. To test this hypothesis, we unilaterally exposed rats for 2h to a 126dB SPL narrow band noise centered at 12kHz. Hearing loss measured by auditory brainstem responses exceeded 55dB from 6 to 32kHz. The mRNA from the exposed CN was harvested at 14 or 28days post-exposure and qRT-PCR analysis was performed on 168 genes involved in neural inflammation, neuropathic pain and glutamatergic or GABAergic neurotransmission. Expression levels of mRNA of Slc17a6 and Gabrg3, involved in excitation and inhibition respectively, were significantly increased at 28days post-exposure, suggesting a possible role in the CN spontaneous hyperactivity associated with tinnitus and hyperacusis. In the pain and inflammatory array, noise exposure upregulated mRNA expression levels of four pain/inflammatory genes, Tlr2, Oprd1, Kcnq3 and Ntrk1 and decreased mRNA expression levels of two more genes, Ccl12 and Il1ß. Pain/inflammatory gene expression changes via Ntrk1 signaling may induce sterile inflammation, neuropathic pain, microglial activation and migration of nerve fibers from the trigeminal, cuneate and vestibular nuclei into the CN. These changes could contribute to somatic tinnitus, hyperacusis and otalgia.


Assuntos
Núcleo Coclear/metabolismo , Perda Auditiva Provocada por Ruído/metabolismo , Neuralgia/metabolismo , Receptor trkA/genética , Transdução de Sinais , Animais , Núcleo Coclear/fisiopatologia , Perda Auditiva Provocada por Ruído/fisiopatologia , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Canal de Potássio KCNQ3/genética , Canal de Potássio KCNQ3/metabolismo , Proteínas Quimioatraentes de Monócitos/genética , Proteínas Quimioatraentes de Monócitos/metabolismo , Neuralgia/fisiopatologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor trkA/metabolismo , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/genética , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo
18.
Clin Perinatol ; 43(2): 313-23, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27235210

RESUMO

Although hyperbilirubinemia is extremely common among neonates and is usually mild and transient, it sometimes leads to bilirubin-induced neurologic damage (BIND). The auditory pathway is highly sensitive to the effects of elevated total serum/plasma bilirubin (TB) levels, with damage manifesting clinically as auditory neuropathy spectrum disorder. Compared to full-term neonates, preterm neonates are more susceptible to BIND and suffer adverse effects at lower TB levels with worse long-term outcomes. Furthermore, although standardized guidelines for management of hyperbilirubinemia exist for term and late preterm neonates, similar guidelines for neonates less than 35 weeks gestational age are limited.


Assuntos
Nervo Coclear/fisiopatologia , Núcleo Coclear/fisiopatologia , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Perda Auditiva Central/fisiopatologia , Hiperbilirrubinemia Neonatal/fisiopatologia , Kernicterus/fisiopatologia , Perda Auditiva Central/etiologia , Humanos , Hiperbilirrubinemia Neonatal/complicações , Hiperbilirrubinemia Neonatal/terapia , Recém-Nascido , Recém-Nascido Prematuro , Kernicterus/etiologia
19.
Nat Rev Neurol ; 12(3): 150-60, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26868680

RESUMO

Tinnitus is a phantom auditory sensation that reduces quality of life for millions of people worldwide, and for which there is no medical cure. Most cases of tinnitus are associated with hearing loss caused by ageing or noise exposure. Exposure to loud recreational sound is common among the young, and this group are at increasing risk of developing tinnitus. Head or neck injuries can also trigger the development of tinnitus, as altered somatosensory input can affect auditory pathways and lead to tinnitus or modulate its intensity. Emotional and attentional state could be involved in the development and maintenance of tinnitus via top-down mechanisms. Thus, military personnel in combat are particularly at risk owing to combined risk factors (hearing loss, somatosensory system disturbances and emotional stress). Animal model studies have identified tinnitus-associated neural changes that commence at the cochlear nucleus and extend to the auditory cortex and other brain regions. Maladaptive neural plasticity seems to underlie these changes: it results in increased spontaneous firing rates and synchrony among neurons in central auditory structures, possibly generating the phantom percept. This Review highlights the links between animal and human studies, and discusses several therapeutic approaches that have been developed to target the neuroplastic changes underlying tinnitus.


Assuntos
Vias Auditivas/fisiopatologia , Plasticidade Neuronal/fisiologia , Zumbido/diagnóstico , Zumbido/fisiopatologia , Animais , Córtex Auditivo/patologia , Córtex Auditivo/fisiopatologia , Vias Auditivas/patologia , Núcleo Coclear/patologia , Núcleo Coclear/fisiopatologia , Humanos , Ruído/efeitos adversos , Zumbido/terapia , Resultado do Tratamento
20.
Hear Res ; 332: 188-198, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26548740

RESUMO

High doses of salicylate induce reversible tinnitus in experimental animals and humans, and is a common tinnitus model. Salicylate probably acts centrally and induces hyperactivity in specific auditory brainstem areas like the dorsal cochlear nucleus (DCN). However, little is known about the effect of high doses of salicylate in synapses and neurons of the DCN. Here we investigated the effects of salicylate on the excitability and evoked and spontaneous neurotransmission in the main neurons (fusiform, cartwheel and tuberculoventral) and synapses of the DCN using whole cell recordings in slices containing the DCN. For this, we incubate the slices for at least 1 h in solution with 1.4 mM salicylate, and recorded action potentials and evoked and spontaneous synaptic currents in fusiform, cartwheel (CW) and putative tuberculoventral (TBV) neurons. We found that incubation with salicylate did not affect the firing of fusiform and TBV neurons, but decreased the spontaneous firing of cartwheel neurons, without affecting AP threshold or complex spikes. Evoked and spontaneous glutamatergic neurotransmission on the fusiform and CW neurons cells was unaffected by salicylate and evoked glycinergic neurotransmission on fusiform neurons was also unchanged by salicylate. On the other hand spontaneous glycinergic transmission on fusiform neurons was reduced in the presence of salicylate. We conclude that high doses of salicylate produces a decreased inhibitor drive on DCN fusiform neurons by reducing the spontaneous firing of cartwheel neurons, but this effect is not able to increase the excitability of fusiform neurons. So, the mechanisms of salicylate-induced tinnitus are probably more complex than simple changes in the neuronal firing and basal synaptic transmission in the DCN.


Assuntos
Núcleo Coclear/efeitos dos fármacos , Glicina/metabolismo , Inibição Neural/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Salicilato de Sódio/toxicidade , Transmissão Sináptica/efeitos dos fármacos , Zumbido/induzido quimicamente , Animais , Núcleo Coclear/metabolismo , Núcleo Coclear/fisiopatologia , Potenciais Evocados Auditivos , Técnicas In Vitro , Masculino , Neurônios/metabolismo , Ratos Wistar , Zumbido/metabolismo , Zumbido/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...