Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 310
Filtrar
1.
Hear Res ; 449: 109033, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38797036

RESUMO

Hearing loss is well known to cause plastic changes in the central auditory system and pathological changes such as tinnitus and hyperacusis. Impairment of inner ear functions is the main cause of hearing loss. In aged individuals, not only inner ear dysfunction but also senescence of the central nervous system is the cause of malfunction of the auditory system. In most cases of hearing loss, the activity of the auditory nerve is reduced, but that of the successive auditory centers is increased in a compensatory way. It has been reported that activity changes occur in the inferior colliculus (IC), a critical nexus of the auditory pathway. The IC integrates the inputs from the brainstem and drives the higher auditory centers. Since abnormal activity in the IC is likely to affect auditory perception, it is crucial to elucidate the neuronal mechanism to induce the activity changes of IC neurons with hearing loss. This review outlines recent findings on hearing-loss-induced plastic changes in the IC and brainstem auditory neuronal circuits and discusses what neuronal mechanisms underlie hearing-loss-induced changes in the activity of IC neurons. Considering the different causes of hearing loss, we discuss age-related hearing loss separately from other forms of hearing loss (non-age-related hearing loss). In general, the main plastic change of IC neurons caused by both age-related and non-age-related hearing loss is increased central gain. However, plastic changes in the IC caused by age-related hearing loss seem to be more complex than those caused by non-age-related hearing loss.


Assuntos
Vias Auditivas , Colículos Inferiores , Plasticidade Neuronal , Neurônios , Colículos Inferiores/fisiopatologia , Animais , Humanos , Neurônios/patologia , Vias Auditivas/fisiopatologia , Audição , Presbiacusia/fisiopatologia , Presbiacusia/patologia , Percepção Auditiva , Fatores Etários , Perda Auditiva/fisiopatologia , Perda Auditiva/patologia , Envelhecimento/patologia , Potenciais Evocados Auditivos do Tronco Encefálico , Estimulação Acústica
2.
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
3.
J Neurophysiol ; 124(4): 1165-1182, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32845200

RESUMO

Sensorineural hearing loss (SNHL) causes an overall deficit in binaural hearing, including the abilities to localize sound sources, discriminate interaural time and level differences (ITDs and ILDs, respectively), and utilize binaural cues to aid signal detection and comprehension in noisy environments. Few studies have examined the effect of SNHL on binaural coding in the central auditory system, and those that have focused on age-related hearing loss. We induced hearing loss in male and female Dutch-belted rabbits via noise overexposure and compared unanesthetized single-unit responses of their inferior colliculi [hearing loss (HL) neurons] with those of unexposed rabbits. Sound-level thresholds of HL neurons to diotic noise were elevated by 75 dB, on average. Sensitivity of firing rates of HL neurons to the azimuth of a broadband noise stimulus was reduced, on average, but was confounded by differences in sound level with respect to detection threshold between groups. We independently manipulated ITD and ILD in virtual acoustic space and found directional sensitivity in binaurally sensitive HL neurons was entirely due to ILD sensitivity and no different than that for unexposed rabbits. However, ITD sensitivity was completely absent in binaurally sensitive HL neurons for noise stimuli both in virtual acoustic space and with ITDs extending to ±3 ms. HL neurons also had weaker spike-timing precision and slightly increased spontaneous rates. Overall, ILD sensitivity was uncompromised, whereas ITD sensitivity was completely lost, implying a specific inability to use information in the timing or correlation of acoustic noise waveforms between the two ears following severe SNHL.NEW & NOTEWORTHY Sensorineural hearing loss compromises perceptual abilities that arise from hearing with two ears, yet its effects on binaural aspects of neural responses are largely unknown. We found that, following severe hearing loss because of acoustic trauma, auditory midbrain neurons specifically lost the ability to encode time differences between the arrival of a broadband noise stimulus to the two ears, whereas the encoding of sound level differences between the two ears remained uncompromised.


Assuntos
Potenciais Evocados Auditivos , Lateralidade Funcional , Perda Auditiva Provocada por Ruído/fisiopatologia , Colículos Inferiores/fisiopatologia , Animais , Limiar Auditivo , Feminino , Masculino , Ruído , Coelhos , Tempo de Reação
4.
Acta Otolaryngol ; 140(8): 682-686, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32401101

RESUMO

Background: Vestibular vertigo is a common clinical symptom; however, the central neural mechanism of it is still poorly understood.Objective: To demonstrate the changes of neural excitability and ascorbate in inferior colliculus (IC) in a rat vertigo model induced by water caloric irrigation.Methods: In vertigo model induced by water caloric irrigation, we recorded the changes of spontaneous firing rate (SFR) of IC. Then a technique that combining in vivo microanalysis with an online electrochemical system (OECS) was employed to monitor the changes of extracellular ascorbate in IC.Results: Electrophysiological studies showed that after vestibular ice water stimulation, the level of SFR in IC significantly increased, reaching (989 ± 9) % and (941 ± 62) % respectively at 2.0 h after contralateral ice water vestibular stimulation and ipsilateral ice water vestibular stimulation. However, the level of ascorbate in IC dramatically decreased after ice stimulation, decreased to (30 ± 12) % and (57 ± 24) % of the basal level respectively in the contralateral group and ipsilateral group.Conclusions and significance: These findings suggest that inferior colliculus plays a role in peripheral vertigo, which would appear useful for uncovering neural mechanisms of vertigo and help finding novel therapeutic targets for vertigo.


Assuntos
Colículos Inferiores/fisiopatologia , Vertigem/fisiopatologia , Animais , Testes Calóricos , Modelos Animais de Doenças , Eletrofisiologia , Masculino , Ratos , Ratos Sprague-Dawley , Vertigem/etiologia
5.
J Neurophysiol ; 123(6): 2101-2121, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32319849

RESUMO

Sensory processing abnormalities are frequently associated with autism spectrum disorders, but the underlying mechanisms are unclear. Here we studied auditory processing in a mouse model of Fragile X Syndrome (FXS), a leading known genetic cause of autism and intellectual disability. Both humans with FXS and the Fragile X mental retardation gene (Fmr1) knockout (KO) mouse model show auditory hypersensitivity, with the latter showing a strong propensity for audiogenic seizures (AGS) early in development. Because midbrain abnormalities cause AGS, we investigated whether the inferior colliculus (IC) of the Fmr1 KO mice shows abnormal auditory processing compared with wild-type (WT) controls at specific developmental time points. Using antibodies against neural activity marker c-Fos, we found increased density of c-Fos+ neurons in the IC, but not auditory cortex, of Fmr1 KO mice at P21 and P34 following sound presentation. In vivo single-unit recordings showed that IC neurons of Fmr1 KO mice are hyperresponsive to tone bursts and amplitude-modulated tones during development and show broader frequency tuning curves. There were no differences in rate-level responses or phase locking to amplitude-modulated tones in IC neurons between genotypes. Taken together, these data provide evidence for the development of auditory hyperresponsiveness in the IC of Fmr1 KO mice. Although most human and mouse work in autism and sensory processing has centered on the forebrain, our new findings, along with recent work on the lower brainstem, suggest that abnormal subcortical responses may underlie auditory hypersensitivity in autism spectrum disorders.NEW & NOTEWORTHY Autism spectrum disorders (ASD) are commonly associated with sensory sensitivity issues, but the underlying mechanisms are unclear. This study presents novel evidence for neural correlates of auditory hypersensitivity in the developing inferior colliculus (IC) in Fmr1 knockout (KO) mouse, a mouse model of Fragile X Syndrome (FXS), a leading genetic cause of ASD. Responses begin to show genotype differences between postnatal days 14 and 21, suggesting an early developmental treatment window.


Assuntos
Transtornos da Percepção Auditiva/fisiopatologia , Síndrome do Cromossomo X Frágil/fisiopatologia , Colículos Inferiores/crescimento & desenvolvimento , Colículos Inferiores/fisiopatologia , Animais , Transtornos da Percepção Auditiva/etiologia , Modelos Animais de Doenças , Fenômenos Eletrofisiológicos/fisiologia , Epilepsia Reflexa/etiologia , Epilepsia Reflexa/fisiopatologia , Proteína do X Frágil da Deficiência Intelectual , Síndrome do Cromossomo X Frágil/complicações , Masculino , Camundongos , Camundongos Knockout , Neurônios/fisiologia
6.
Hear Res ; 391: 107957, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32278945

RESUMO

We examined the effect of acoustic trauma on the spontaneous activities of the glutamatergic and GABAergic neurons in the inferior colliculus (IC) of mice. Optogenetics was used to identify the neuron type. In control animals, the spontaneous firing rate was higher in GABAergic neurons than in glutamatergic neurons. However, in the animals with acoustic trauma, the balance of spontaneous activities between glutamatergic and GABAergic neurons was inverted. The spontaneous firing rate was enhanced in glutamatergic neurons only, with bursting episodes occurring frequently. Moreover, the spike shapes of GABAergic and glutamatergic neurons were modified differently in both cell types. These results suggested that the acoustic trauma induced plastic changes in the neuronal circuits in the IC and altered the balance of the activities of excitatory and inhibitory neurons. This aberrant excitatory-inhibitory balance in the IC might underpin tinnitus perception.


Assuntos
Neurônios GABAérgicos/metabolismo , Ácido Glutâmico/metabolismo , Perda Auditiva Provocada por Ruído/metabolismo , Colículos Inferiores/metabolismo , Inibição Neural , Plasticidade Neuronal , Transmissão Sináptica , Animais , Modelos Animais de Doenças , Potenciais Evocados Auditivos do Tronco Encefálico , Feminino , Neurônios GABAérgicos/patologia , Perda Auditiva Provocada por Ruído/etiologia , Perda Auditiva Provocada por Ruído/patologia , Perda Auditiva Provocada por Ruído/fisiopatologia , Colículos Inferiores/patologia , Colículos Inferiores/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ruído
7.
Schizophr Bull ; 46(1): 193-201, 2020 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-31220318

RESUMO

The mismatch negativity is a cortical response to auditory changes and its reduction is a consistent finding in schizophrenia. Recent evidence revealed that the human brain detects auditory changes already at subcortical stages of the auditory pathway. This finding, however, raises the question where in the auditory hierarchy the schizophrenic deficit first evolves and whether the well-known cortical deficit may be a consequence of dysfunction at lower hierarchical levels. Finally, it should be resolved whether mismatch profiles differ between schizophrenia and affective disorders which exhibit auditory processing deficits as well. We used functional magnetic resonance imaging to assess auditory mismatch processing in 29 patients with schizophrenia, 27 patients with major depression, and 31 healthy control subjects. Analysis included whole-brain activation, region of interest, path and connectivity analysis. In schizophrenia, mismatch deficits emerged at all stages of the auditory pathway including the inferior colliculus, thalamus, auditory, and prefrontal cortex. In depression, deficits were observed in the prefrontal cortex only. Path analysis revealed that activation deficits propagated from subcortical to cortical nodes in a feed-forward mechanism. Finally, both patient groups exhibited reduced connectivity along this processing stream. Auditory mismatch impairments in schizophrenia already manifest at the subcortical level. Moreover, subcortical deficits contribute to the well-known cortical deficits and show specificity for schizophrenia. In contrast, depression is associated with cortical dysfunction only. Hence, schizophrenia and major depression exhibit different neural profiles of sensory processing deficits. Our findings add to a converging body of evidence for brainstem and thalamic dysfunction as a hallmark of schizophrenia.


Assuntos
Córtex Auditivo/fisiopatologia , Vias Auditivas/fisiopatologia , Percepção Auditiva/fisiologia , Conectoma , Transtorno Depressivo Maior/fisiopatologia , Colículos Inferiores/fisiopatologia , Córtex Pré-Frontal/fisiopatologia , Esquizofrenia/fisiopatologia , Tálamo/fisiopatologia , Adulto , Córtex Auditivo/diagnóstico por imagem , Vias Auditivas/diagnóstico por imagem , Transtorno Depressivo Maior/diagnóstico por imagem , Feminino , Humanos , Colículos Inferiores/diagnóstico por imagem , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Córtex Pré-Frontal/diagnóstico por imagem , Esquizofrenia/diagnóstico por imagem , Tálamo/diagnóstico por imagem
8.
J Neurosci ; 39(49): 9852-9863, 2019 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-31666356

RESUMO

Fragile X syndrome (FXS) is the most common form of inherited intellectual disability and the leading monogenetic cause of autism. One symptom of FXS and autism is sensory hypersensitivity (also called sensory over-responsivity). Perhaps related to this, the audiogenic seizure (AGS) is arguably the most robust behavioral phenotype in the FXS mouse model-the Fmr1 knock-out (KO) mouse. Therefore, the AGS may be considered a mouse model of sensory hypersensitivity. Hyperactive circuits are hypothesized to underlie dysfunction in a number of brain regions in patients with FXS and Fmr1 KO mice, and the AGS may be a result of this. But the specific cell types and brain regions underlying AGSs in the Fmr1 KO are unknown. We used conditional deletion or expression of Fmr1 in different cell populations to determine whether Fmr1 deletion in those cells was sufficient or necessary, respectively, for the AGS phenotype in males. Our data indicate that Fmr1 deletion in glutamatergic neurons that express vesicular glutamate transporter 2 (VGlut2) and are located in subcortical brain regions is sufficient and necessary to cause AGSs. Furthermore, the deletion of Fmr1 in glutamatergic neurons of the inferior colliculus is necessary for AGSs. When we demonstrate necessity, we show that Fmr1 expression in either the larger population of VGlut2-expressing glutamatergic neurons or the smaller population of inferior collicular glutamatergic neurons-in an otherwise Fmr1 KO mouse-eliminates AGSs. Therefore, targeting these neuronal populations in FXS and autism may be part of a therapeutic strategy to alleviate sensory hypersensitivity.SIGNIFICANCE STATEMENT Sensory hypersensitivity in fragile X syndrome (FXS) and autism patients significantly interferes with quality of life. Audiogenic seizures (AGSs) are arguably the most robust behavioral phenotype in the FXS mouse model-the Fmr1 knockout-and may be considered a model of sensory hypersensitivity in FXS. We provide the clearest and most precise genetic evidence to date for the cell types and brain regions involved in causing AGSs in the Fmr1 knockout and, more broadly, for any mouse mutant. The expression of Fmr1 in these same cell types in an otherwise Fmr1 knockout eliminates AGSs indicating possible cellular targets for alleviating sensory hypersensitivity in FXS and other forms of autism.


Assuntos
Epilepsia Reflexa/genética , Epilepsia Reflexa/fisiopatologia , Proteína do X Frágil da Deficiência Intelectual/genética , Colículos Inferiores/fisiopatologia , Neurônios/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/biossíntese , Animais , Síndrome do Cromossomo X Frágil/genética , Síndrome do Cromossomo X Frágil/fisiopatologia , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Knockout , Órgão Espiral/metabolismo , Órgão Espiral/fisiopatologia , Proteína Vesicular 2 de Transporte de Glutamato/genética
9.
J Neurophysiol ; 122(2): 451-465, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31116647

RESUMO

The CHRNA7 gene that encodes the α7-subunit of the nicotinic acetylcholine receptor (α7-nAChR) has been associated with some autism spectrum disorders and other neurodevelopmental conditions characterized, in part, by auditory and language impairment. These conditions may include auditory processing disorders that represent impaired timing of neural activity, often accompanied by problems understanding speech. Here, we measure timing properties of sound-evoked activity via the auditory brainstem response (ABR) of α7-nAChR knockout mice of both sexes and wild-type colony controls. We find a significant timing delay in evoked ABR signals that represents midbrain activity in knockouts. We also examine spike-timing properties of neurons in the inferior colliculus, a midbrain nucleus that exhibits high levels of α7-nAChR during development. We find delays of evoked responses along with degraded spiking precision in knockout animals. We find similar timing deficits in responses of neurons in the superior paraolivary nucleus and ventral nucleus of the lateral lemniscus, which are brainstem nuclei thought to shape temporal precision in the midbrain. In addition, we find that other measures of temporal acuity including forward masking and gap detection are impaired for knockout animals. We conclude that altered temporal processing at the level of the brainstem in α7-nAChR-deficient mice may contribute to degraded spike timing in the midbrain, which may underlie the observed timing delay in the ABR signals. Our findings are consistent with a role for the α7-nAChR in types of neurodevelopmental and auditory processing disorders and we identify potential neural targets for intervention.NEW & NOTEWORTHY Disrupted signaling via the α7-nicotinic acetylcholine receptor (α7-nAChR) is associated with neurodevelopmental disorders that include impaired auditory processing. The underlying causes of dysfunction are not known but a common feature is abnormal timing of neural activity. We examined temporal processing of α7-nAChR knockout mice and wild-type controls. We found degraded spike timing of neurons in knockout animals, which manifests at the level of the auditory brainstem and midbrain.


Assuntos
Transtornos da Percepção Auditiva/fisiopatologia , Tronco Encefálico/fisiopatologia , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Percepção do Tempo/fisiologia , Receptor Nicotínico de Acetilcolina alfa7/deficiência , Animais , Doenças Auditivas Centrais/fisiopatologia , Transtorno do Espectro Autista/fisiopatologia , Modelos Animais de Doenças , Feminino , Colículos Inferiores/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexo Olivar Superior/fisiopatologia , Fatores de Tempo
10.
Hear Res ; 377: 318-329, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30878270

RESUMO

It is well known that quality of hearing decreases with increasing age due to changes in the peripheral or central auditory pathway. Along with the decrease in the number of neurons the neurotransmitter profile is also affected in the various parts of the auditory system. Particularly, changes in the inhibitory neurons in the inferior colliculus (IC) are known to affect quality of hearing with aging. To date, there is no information about the status of the inhibitory neurotransmitter GABA in the human IC during aging. We have collected and processed inferior colliculi of persons aged 11-97 years at the time of death for morphometry and immunohistochemical expression of glutamic acid decarboxylase (GAD67) and parvalbumin. We used unbiased stereology to estimate the number of cresyl-violet and immunostained neurons. Quantitative real-time PCR was used to measure the relative expression of the GAD67 mRNA. We found that the number of total, GABAergic and PV-positive neurons significantly decreased with increasing age (p < 0.05). The proportion of GAD67-ir neurons to total number of neurons was also negatively associated with increasing age (p = 0.004), but there was no change observed in the proportion of PV-ir neurons relative to GABAergic neurons (p = 0.25). Further, the fold change in the levels of GAD67 mRNA was negatively correlated to age (p = 0.024). We conclude that the poorer quality of hearing with increasing age may be due to decreased expression of inhibitory neurotransmitters and the decline in the number of inhibitory neurons in the IC.


Assuntos
Envelhecimento/patologia , Vias Auditivas/patologia , Neurônios GABAérgicos/patologia , Colículos Inferiores/patologia , Presbiacusia/patologia , Adolescente , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/metabolismo , Vias Auditivas/química , Vias Auditivas/fisiopatologia , Morte Celular , Criança , Feminino , Neurônios GABAérgicos/química , Glutamato Descarboxilase/análise , Glutamato Descarboxilase/genética , Audição , Humanos , Colículos Inferiores/química , Colículos Inferiores/fisiopatologia , Masculino , Pessoa de Meia-Idade , Parvalbuminas/análise , Presbiacusia/metabolismo , Presbiacusia/fisiopatologia , Adulto Jovem , Ácido gama-Aminobutírico/análise
11.
Neurobiol Aging ; 73: 30-40, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30316050

RESUMO

Hearing impairment in older people is thought to arise from impaired temporal processing in auditory circuits. We used a systems-level (scalp recordings) and a microcircuit-level (extracellular recordings) approach to investigate how aging affects the sensitivity to temporal envelopes of speech-like sounds in rats. Scalp-recorded potentials suggest an age-related increase in sensitivity to temporal regularity along the ascending auditory pathway. The underlying cellular changes in the midbrain were examined using extracellular recordings from inferior colliculus neurons. We observed an age-related increase in sensitivity to the sound's onset and temporal regularity (i.e., periodicity envelope) in the spiking output of inferior colliculus neurons, relative to their synaptic inputs (local field potentials). This relative enhancement for aged animals was most prominent for multi-unit (relative to single-unit) spiking activity. Spontaneous multi-unit, but not single-unit, activity was also enhanced in aged compared with young animals. Our results suggest that aging is associated with altered sensitivity to a sound's temporal regularities, and that these effects may be due to increased gain of neural network activity in the midbrain.


Assuntos
Estimulação Acústica , Envelhecimento/fisiologia , Envelhecimento/psicologia , Percepção Auditiva/fisiologia , Encéfalo/fisiopatologia , Colículos Inferiores/fisiopatologia , Neurônios/fisiologia , Presbiacusia/fisiopatologia , Presbiacusia/psicologia , Som , Fala , Animais , Estudos Transversais , Modelos Animais de Doenças , Fenômenos Eletrofisiológicos , Potenciais Evocados Auditivos do Tronco Encefálico , Masculino , Ratos Endogâmicos F344
12.
Neuroscience ; 407: 93-107, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-30292765

RESUMO

The central gain model of hyperacusis proposes that loss of auditory input can result in maladaptive neuronal gain increases in the central auditory system, leading to the over-amplification of sound-evoked activity and excessive loudness perception. Despite the attractiveness of this model, and supporting evidence for it, a critical test of the central gain theory requires that changes in sound-evoked activity be explicitly linked to perceptual alterations of loudness. Here we combined an operant conditioning task that uses a subject's reaction time to auditory stimuli to produce reliable measures of loudness growth with chronic electrophysiological recordings from the auditory cortex and inferior colliculus of awake, behaviorally-phenotyped animals. In this manner, we could directly correlate daily assessments of loudness perception with neurophysiological measures of sound encoding within the same animal. We validated this novel psychophysical-electrophysiological paradigm with a salicylate-induced model of hearing loss and hyperacusis, as high doses of sodium salicylate reliably induce temporary hearing loss, neural hyperactivity, and auditory perceptual disruptions like tinnitus and hyperacusis. Salicylate induced parallel changes to loudness growth and evoked response-intensity functions consistent with temporary hearing loss and hyperacusis. Most importantly, we found that salicylate-mediated changes in loudness growth and sound-evoked activity were correlated within individual animals. These results provide strong support for the central gain model of hyperacusis and demonstrate the utility of using an experimental design that allows for within-subject comparison of behavioral and electrophysiological measures, thereby making inter-subject variability a strength rather than a limitation.


Assuntos
Perda Auditiva/fisiopatologia , Hiperacusia/fisiopatologia , Percepção Sonora/fisiologia , Salicilato de Sódio/farmacologia , Estimulação Acústica/métodos , Animais , Córtex Auditivo/efeitos dos fármacos , Córtex Auditivo/fisiopatologia , Potenciais Evocados Auditivos/efeitos dos fármacos , Feminino , Audição/efeitos dos fármacos , Audição/fisiologia , Perda Auditiva/tratamento farmacológico , Colículos Inferiores/efeitos dos fármacos , Colículos Inferiores/fisiopatologia , Percepção Sonora/efeitos dos fármacos , Masculino , Ratos Sprague-Dawley , Roedores
13.
Epilepsy Behav ; 88: 227-234, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30316149

RESUMO

It is known that audiogenic seizure (AGS) expression is based on the activation of the midbrain structures such as the inferior colliculus (IC). It was demonstrated that excessive sound exposure during the postnatal developments of the IC in rats led to AGS susceptibility in adulthood, which correlated with underdevelopment of the IC. In adult rodents, noise overstimulation induced apoptosis in the IC. The purpose of this study was to investigate postnatal development of the IC in rats genetically prone to AGS and to check if audiogenic kindling would activate apoptosis and/or proliferation in the IC. In our study, we used inbred audiogenic Krushinsky-Molodkina (KM) rats, which are characterized by age-dependent seizure expression. Analysis of postnatal development showed the increased number of proliferating cells in the IC central nucleus of KM rats on the 14th postnatal day (P14) in comparison with those of Wistar rats. Moreover, we also observed increased apoptosis level and decreased general cell population in the IC central nucleus. These data pointed towards a delayed development of the IC in KM rats. Analysis of the IC central nucleus of KM rat after audiogenic kindling for a week, with one AGS per day, demonstrated dramatically increased cell death, which was accompanied with a reduction of general cell population. Audiogenic kindling also decreased proliferation in the IC central nucleus. However, a week after the last AGS, the number of proliferating cells was increased, which supposes a certain compensatory mechanism to prevent cell loss.


Assuntos
Apoptose/fisiologia , Proliferação de Células/fisiologia , Epilepsia Reflexa/fisiopatologia , Colículos Inferiores/fisiopatologia , Estimulação Acústica/efeitos adversos , Animais , Epilepsia Reflexa/patologia , Feminino , Colículos Inferiores/crescimento & desenvolvimento , Colículos Inferiores/patologia , Excitação Neurológica/fisiologia , Masculino , Ratos , Ratos Endogâmicos , Ratos Wistar
14.
Hear Res ; 367: 207-212, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29954642

RESUMO

Neural plasticity is a characteristic of the brain that helps it adapt to changes in sensory input. We hypothesize that auditory deafferentation may induce plastic changes in the cytoskeleton of the neurons in the inferior colliculus (IC). In this study, we evaluated the dynamic status of neurofilament (NF) phosphorylation in the IC after hearing loss. We induced auditory deafferentation via unilateral or bilateral cochlear ablation in rats, aged 4 weeks. To evaluate cytoskeletal changes in neurons, we evaluated mRNA fold changes in NF heavy chain expression, non-phosphorylated NF protein fold changes using SMI-32 antibody, and the ratio of SMI-32 immunoreactive (SMI-32-ir) neurons to the total neuronal population in the IC at 4 and 12 weeks after deafness. In the bilateral deafness (BD) group, the ratios of SMI-32-ir neurons significantly increased at 4 weeks after ablation in the right and left IC (6.1 ±â€¯4.4%, 5.0 ±â€¯3.4%, respectively), compared with age-matched controls (P < 0.01, P < 0.01). At 12 weeks after ablation, the ratio of SMI-32 positive neurons was higher (right, 3.4 ±â€¯2.0%; left, 3.2 ±â€¯2.3%) than that in the age-matched control group, albeit not significant in the right and left side (P = 0.38, P = 0.24, respectively). Consistent with the results of the ratio of SMI-32-ir neurons, SMI-32-ir protein expression was increased at 4 weeks after BD, and the changes at 12 weeks after bilateral ablation were not significant in the right or left IC. The age-matched control fold changes of NF mRNA expression after bilateral deafness were not significant at 4 and 12 weeks after deafness in right and left IC. Unilateral deafness did not induce significant change of NF mRNA expression, SMI-32-ir protein expression, and the ratio of SMI-32-ir neurons in the IC at 4 and 12 weeks after hearing loss. Bilateral auditory deafferentation induces structural changes in the neuronal cytoskeleton within the IC, which is prominent at 4 weeks after BD. The structural remodeling of neurons stabilized at 12 weeks after BD. Unlike BD, unilateral auditory deafferentation did not affect the dynamic status of NFs in the IC.


Assuntos
Cóclea/inervação , Citoesqueleto/patologia , Surdez/patologia , Denervação/métodos , Colículos Inferiores/patologia , Plasticidade Neuronal , Neurônios/patologia , Animais , Citoesqueleto/metabolismo , Surdez/genética , Surdez/metabolismo , Surdez/fisiopatologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Colículos Inferiores/metabolismo , Colículos Inferiores/fisiopatologia , Masculino , Proteínas de Neurofilamentos/genética , Proteínas de Neurofilamentos/metabolismo , Neurônios/metabolismo , Fosforilação , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Fatores de Tempo
15.
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
16.
Alcohol ; 66: 21-26, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29277284

RESUMO

We previously reported increased current density through P-type voltage-gated Ca2+ channels in inferior colliculus (IC) neurons during alcohol withdrawal. However, the molecular correlate of this increased P-type channel current is currently unknown. Here, we probe changes in mRNA and protein expression of the pore-forming CaV2.1-α1 (P/Q-type) subunits in IC neurons during the course of alcohol withdrawal-induced seizures (AWSs). Rats received three daily doses of ethanol or the vehicle every 8 h for 4 consecutive days. The IC was dissected at various time intervals following alcohol withdrawal, and the mRNA and protein levels of the CaV2.1-α1 subunits were measured. In separate experiments, rats were tested for acoustically evoked seizure susceptibility 3, 24, and 48 h after alcohol withdrawal. AWSs were observed 24 h after withdrawal; no seizures were observed at 3 or 48 h or in the control-treated rats. Compared to control-treated rats, the mRNA levels of the CaV2.1-α1 subunit were increased 1.9-fold and 2.1-fold at 3 and 24 h, respectively; change in mRNA expression was nonsignificant at 48 h following alcohol withdrawal. Western blot analyses revealed that protein levels of the CaV2.1-α1 subunits were not altered in IC neurons following alcohol withdrawal. We conclude that expression of the Cacna1a mRNA increased before the onset of AWS susceptibility, suggesting that altered CaV2.1 channel expression may play a role in AWS pathogenesis.


Assuntos
Convulsões por Abstinência de Álcool/metabolismo , Canais de Cálcio Tipo N/metabolismo , Etanol , Colículos Inferiores/metabolismo , Neurônios/metabolismo , RNA Mensageiro/metabolismo , Convulsões por Abstinência de Álcool/genética , Convulsões por Abstinência de Álcool/fisiopatologia , Animais , Canais de Cálcio Tipo N/genética , Modelos Animais de Doenças , Colículos Inferiores/fisiopatologia , Masculino , Potenciais da Membrana , RNA Mensageiro/genética , Ratos Sprague-Dawley , Fatores de Tempo , Regulação para Cima
17.
J Neurosci Methods ; 291: 227-237, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28864083

RESUMO

BACKGROUND: Subjective tinnitus is a hearing disorder in which a person perceives sound when no external sound is present. It can be acute or chronic. Because our current understanding of its pathology is incomplete, no effective cures have yet been established. Mouse models are useful for studying the pathophysiology of tinnitus as well as for developing therapeutic treatments. NEW METHOD: We have developed a new method for determining acute and chronic tinnitus in mice, called sound-based avoidance detection (SBAD). The SBAD method utilizes one paradigm to detect tinnitus and another paradigm to monitor possible confounding factors, such as motor impairment, loss of motivation, and deficits in learning and memory. RESULTS: The SBAD method has succeeded in monitoring both acute and chronic tinnitus in mice. Its detection ability is further validated by functional studies demonstrating an abnormal increase in neuronal activity in the inferior colliculus of mice that had previously been identified as having tinnitus by the SBAD method. COMPARISON WITH EXISTING METHODS: The SBAD method provides a new means by which investigators can detect tinnitus in a single mouse accurately and with more control over potential confounding factors than existing methods. CONCLUSION: This work establishes a new behavioral method for detecting tinnitus in mice. The detection outcome is consistent with functional validation. One key advantage of mouse models is they provide researchers the opportunity to utilize an extensive array of genetic tools. This new method could lead to a deeper understanding of the molecular pathways underlying tinnitus pathology.


Assuntos
Condicionamento Operante , Modelos Animais de Doenças , Zumbido/diagnóstico , Estimulação Acústica , Análise de Variância , Animais , Aprendizagem da Esquiva , Eletrochoque , Desenho de Equipamento , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Feminino , Colículos Inferiores/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Atividade Motora , Neurônios/fisiologia , Emissões Otoacústicas Espontâneas/fisiologia , Salicilato de Sódio , Zumbido/fisiopatologia , Técnicas de Cultura de Tecidos , Imagens com Corantes Sensíveis à Voltagem
18.
J Neurosci ; 37(26): 6314-6330, 2017 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-28583912

RESUMO

Hearing loss leads to a host of cellular and synaptic changes in auditory brain areas that are thought to give rise to auditory perception deficits such as temporal processing impairments, hyperacusis, and tinnitus. However, little is known about possible changes in synaptic circuit connectivity that may underlie these hearing deficits. Here, we show that mild hearing loss as a result of brief noise exposure leads to a pronounced reorganization of local excitatory and inhibitory circuits in the mouse inferior colliculus. The exact nature of these reorganizations correlated with the presence or absence of the animals' impairments in detecting brief sound gaps, a commonly used behavioral sign for tinnitus in animal models. Mice with gap detection deficits (GDDs) showed a shift in the balance of synaptic excitation and inhibition that was present in both glutamatergic and GABAergic neurons, whereas mice without GDDs showed stable excitation-inhibition balances. Acoustic enrichment (AE) with moderate intensity, pulsed white noise immediately after noise trauma prevented both circuit reorganization and GDDs, raising the possibility of using AE immediately after cochlear damage to prevent or alleviate the emergence of central auditory processing deficits.SIGNIFICANCE STATEMENT Noise overexposure is a major cause of central auditory processing disorders, including tinnitus, yet the changes in synaptic connectivity underlying these disorders remain poorly understood. Here, we find that brief noise overexposure leads to distinct reorganizations of excitatory and inhibitory synaptic inputs onto glutamatergic and GABAergic neurons and that the nature of these reorganizations correlates with animals' impairments in detecting brief sound gaps, which is often considered a sign of tinnitus. Acoustic enrichment immediately after noise trauma prevents circuit reorganizations and gap detection deficits, highlighting the potential for using sound therapy soon after cochlear damage to prevent the development of central processing deficits.


Assuntos
Estimulação Acústica/métodos , Percepção Auditiva , Colículos Inferiores/fisiopatologia , Inibição Neural , Reflexo de Sobressalto , Zumbido/fisiopatologia , Adaptação Fisiológica , Animais , Potenciais Pós-Sinápticos Excitadores , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Rede Nervosa/fisiopatologia , Ruído/efeitos adversos , Estatística como Assunto , Zumbido/etiologia
19.
Hear Res ; 350: 32-42, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28431308

RESUMO

Chronic tinnitus and hyperacusis often develop with age-related hearing loss presumably due to aberrant neural activity in the central auditory system (CAS) induced by cochlear pathologies. However, the full spectrum of physiological changes that occur in the CAS as a result age-related hearing loss are still poorly understood. To address this issue, neurophysiological measures were obtained from the cochlea and the inferior colliculus (IC) of 2, 6 and 12 month old C57BL/6J mice, a mouse model for early age-related hearing loss. Thresholds of the compound action potentials (CAP) in 6 and 12 month old mice were significantly higher than in 2 month old mice. The sound driven and spontaneous firing rates of IC neurons, recorded with 16 channel electrodes, revealed mean IC thresholds of 22.8 ± 6.5 dB (n = 167) at 2 months, 37.9 ± 6.2 dB (n = 132) at 6 months and 47.1 ± 15.3 dB (n = 151) at 12 months of age consistent with the rise in CAP thresholds. The characteristic frequencies (CF) of IC neurons ranged from 3 to 32 kHz in 2 month old mice; the upper CF ranged decreased to 26 kHz and 16 kHz in 6 and 12 month old mice respectively. The percentage of IC neurons with CFs between 8 and 12 kHz increased from 36.5% in 2 month old mice, to 48.8% and 76.2% in 6 and 12 month old mice, respectively, suggesting a downshift of IC CFs due to the high-frequency hearing loss. The average spontaneous firing rate (SFRs) of all recorded neurons in 2 month old mice was 3.2 ± 2.5 Hz (n = 167). For 6 and 12 month old mice, the SFRs of low CF neurons (<8 kHz) was maintained at 3-6 spikes/s; whereas SFRs of IC neurons with CFs > 8 kHz increased to 13.0 ± 15.4 (n = 68) Hz at 6 months of age and then declined to 4.8 ± 7.4 (n = 110) spikes/s at 12 months of age. In addition, sound-evoked activity at suprathreshold levels at 6 months of age was much higher than at 2 and 12 months of age. To evaluate the behavioral consequences of sound evoked hyperactivity in the IC, the amplitude of the acoustic startle reflex was measured at 4, 8 and 16 kHz using narrow band noise bursts. Acoustic startle reflex amplitudes in 6 and 12 month old mice (n = 4) were significantly larger than 2 month old mice (n = 4) at 4 and 8 kHz, but not 16 kHz. The enhanced reflex amplitudes suggest that high-intensity, low-frequency sounds are perceived as louder than normal in 6 and 12 month old mice compared to 2 month olds. The increased spontaneous activity, particularly at 6 months, may be related to tinnitus whereas the increase in sound-evoked activity and startle reflex amplitudes may be related to hyperacusis.


Assuntos
Cóclea/fisiopatologia , Colículos Inferiores/fisiopatologia , Presbiacusia/fisiopatologia , Reflexo de Sobressalto , Estimulação Acústica , Fatores Etários , Envelhecimento , Animais , Limiar Auditivo , Modelos Animais de Doenças , Potenciais Evocados Auditivos , Audição , Hiperacusia/fisiopatologia , Hiperacusia/psicologia , Percepção Sonora , Camundongos Endogâmicos C57BL , Presbiacusia/psicologia , Zumbido/fisiopatologia , Zumbido/psicologia
20.
Neuroscience ; 347: 48-56, 2017 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-28188855

RESUMO

Epilepsy is a neurological disease related to the occurrence of pathological oscillatory activity, but the basic physiological mechanisms of seizure remain to be understood. Our working hypothesis is that specific sensory processing circuits may present abnormally enhanced predisposition for coordinated firing in the dysfunctional brain. Such facilitated entrainment could share a similar mechanistic process as those expediting the propagation of epileptiform activity throughout the brain. To test this hypothesis, we employed the Wistar audiogenic rat (WAR) reflex animal model, which is characterized by having seizures triggered reliably by sound. Sound stimulation was modulated in amplitude to produce an auditory steady-state-evoked response (ASSR; -53.71Hz) that covers bottom-up and top-down processing in a time scale compatible with the dynamics of the epileptic condition. Data from inferior colliculus (IC) c-Fos immunohistochemistry and electrographic recordings were gathered for both the control Wistar group and WARs. Under 85-dB SLP auditory stimulation, compared to controls, the WARs presented higher number of Fos-positive cells (at IC and auditory temporal lobe) and a significant increase in ASSR-normalized energy. Similarly, the 110-dB SLP sound stimulation also statistically increased ASSR-normalized energy during ictal and post-ictal periods. However, at the transition from the physiological to pathological state (pre-ictal period), the WAR ASSR analysis demonstrated a decline in normalized energy and a significant increase in circular variance values compared to that of controls. These results indicate an enhanced coordinated firing state for WARs, except immediately before seizure onset (suggesting pre-ictal neuronal desynchronization with external sensory drive). These results suggest a competing myriad of interferences among different networks that after seizure onset converge to a massive oscillatory circuit.


Assuntos
Córtex Auditivo/fisiopatologia , Potenciais Evocados Auditivos , Colículos Inferiores/fisiopatologia , Convulsões/fisiopatologia , Estimulação Acústica , Animais , Córtex Auditivo/metabolismo , Sincronização Cortical , Modelos Animais de Doenças , Eletroencefalografia , Colículos Inferiores/metabolismo , Vias Neurais/fisiopatologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...