Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.639
Filtrer
1.
Hear Res ; 452: 109106, 2024 10.
Article de Anglais | MEDLINE | ID: mdl-39181061

RÉSUMÉ

Several studies suggest that hearing loss results in changes in the balance between inhibition and excitation in the inferior colliculus (IC). The IC is an integral nucleus within the auditory brainstem. The majority of ascending pathways from the lateral lemniscus (LL), superior olivary complex (SOC), and cochlear nucleus (CN) synapse in the IC before projecting to the thalamus and cortex. Many of these ascending projections provide inhibitory innervation to neurons within the IC. However, the nature and the distribution of this inhibitory input have only been partially elucidated in the rat. The inhibitory neurotransmitter, gamma aminobutyric acid (GABA), from the ventral nucleus of the lateral lemniscus (VNLL), provides the primary inhibitory input to the IC of the rat with GABA from other lemniscal and SOC nuclei providing lesser, but prominent innervation. There is evidence that hearing related conditions can result in dysfunction of IC neurons. These changes may be mediated in part by changes in GABA inputs to IC neurons. We have previously used gene micro-arrays in a study of deafness-related changes in gene expression in the IC and found significant changes in GAD as well as the GABA transporters and GABA receptors (Holt 2005). This is consistent with reports of age and trauma related changes in GABA (Bledsoe et al., 1995; Mossop et al., 2000; Salvi et al., 2000). Ototoxic lesions of the cochlea produced a permanent threshold shift. The number, intensity, and density of GABA positive axon terminals in the IC were compared in normal hearing and deafened rats. While the number of GABA immunolabeled puncta was only minimally different between groups, the intensity of labeling was significantly reduced. The ultrastructural localization and distribution of labeling was also examined. In deafened animals, the number of immuno gold particles was reduced by 78 % in axodendritic and 82 % in axosomatic GABAergic puncta. The affected puncta were primarily associated with small IC neurons. These results suggest that reduced inhibition to IC neurons contribute to the increased neuronal excitability observed in the IC following noise or drug induced hearing loss. Whether these deafness diminished inhibitory inputs originate from intrinsic or extrinsic CNIC sources awaits further study.


Sujet(s)
Colliculus inférieurs , Rat Sprague-Dawley , Acide gamma-amino-butyrique , Animaux , Colliculus inférieurs/métabolisme , Colliculus inférieurs/anatomopathologie , Acide gamma-amino-butyrique/métabolisme , Surdité due au bruit/métabolisme , Surdité due au bruit/physiopathologie , Surdité due au bruit/anatomopathologie , Ototoxicité/métabolisme , Ototoxicité/étiologie , Mâle , Voies auditives/métabolisme , Voies auditives/anatomopathologie , Voies auditives/physiopathologie , Modèles animaux de maladie humaine , Immunohistochimie , Rats , Glutamate decarboxylase/métabolisme , Neurones/métabolisme , Neurones/anatomopathologie , Inhibition nerveuse
2.
Mil Med ; 189(Supplement_3): 407-415, 2024 Aug 19.
Article de Anglais | MEDLINE | ID: mdl-39160824

RÉSUMÉ

INTRODUCTION: Auditory injuries induced by repeated exposures to blasts reduce the operational performance capability and the life quality of military personnel. The treatment for blast-induced progressive hearing damage is lacking. We have recently investigated the therapeutic function of liraglutide, a glucagon-like peptide-1 receptor agonist, to mitigate blast-induced hearing damage in the animal model of chinchilla, under different blast intensities, wearing earplugs (EPs) or not during blasts, and drug-treatment plan. The goal of this study was to investigate the therapeutical function of liraglutide by comparing the results obtained under different conditions. MATERIALS AND METHODS: Previous studies on chinchillas from two under-blast ear conditions (EP/open), two blast plans (G1: 6 blasts at 3-5 psi or G2:3 blasts at 15-25 psi), and three treatment plans (blast control, pre-blast drug treatment, and post-blast drug treatment) were summarized. The auditory brainstem response (ABR), distortion product otoacoustic emission (DPOAE), and middle latency response (MLR) recorded within 14 days after the blasts were used. Statistical analysis was performed to evaluate the effect of liraglutide under different conditions. RESULTS: ABR threshold shifts indicated that the conditions of the EP and open ears were substantially different. Results from EP chinchillas indicated that the pre-blast treatment reduced the acute ABR threshold elevation on the day of blasts, and the significance of such an effect increased with the blast level. Liraglutide-treated open chinchillas showed lower ABR threshold shifts at the later stage of the experiment regardless of the blast levels. The DPOAE was less damaged after G2 blasts compared to G1 when pre-blast liraglutide was administrated. Lower post-blast MLR amplitudes were observed in the pre-blast treatment groups. CONCLUSIONS: This study indicated that the liraglutide mitigated the blast-induced auditory injuries. In EP ears, the pre-blast administration of liraglutide reduced the severity of blast-induced acute damage in ears with EP protection, especially under G2. In animals with open ears, the effect of liraglutide on the restoration of hearing increased with time. The liraglutide potentially benefits post-blast hearing through multiple approaches with different mechanics.


Sujet(s)
Traumatismes par explosion , Chinchilla , Modèles animaux de maladie humaine , Liraglutide , Animaux , Liraglutide/pharmacologie , Liraglutide/usage thérapeutique , Traumatismes par explosion/complications , Traumatismes par explosion/traitement médicamenteux , Traumatismes par explosion/physiopathologie , Surdité due au bruit/traitement médicamenteux , Surdité due au bruit/physiopathologie , Potentiels évoqués auditifs du tronc cérébral/effets des médicaments et des substances chimiques , Potentiels évoqués auditifs du tronc cérébral/physiologie , Émissions otoacoustiques spontanées/effets des médicaments et des substances chimiques , Émissions otoacoustiques spontanées/physiologie
3.
Neurobiol Dis ; 200: 106619, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39079581

RÉSUMÉ

It is well established that hearing loss can lead to widespread plasticity within the central auditory pathway, which is thought to contribute to the pathophysiology of audiological conditions such as tinnitus and hyperacusis. Emerging evidence suggests that hearing loss can also result in plasticity within brain regions involved in higher-level cognitive functioning like the prefrontal cortex; findings which may underlie the association between hearing loss and cognitive impairment documented in epidemiological studies. Using the 40-Hz auditory steady state response to assess sound-evoked gamma oscillations, we previously showed that noise-induced hearing loss results in impaired gamma phase coherence within the prefrontal but not the auditory cortex. To determine whether region-specific structural or molecular changes accompany this differential plasticity following hearing loss, in the present study we utilized Golgi-Cox staining to assess dendritic organization and synaptic density, as well as Western blotting to measure changes in synaptic signaling proteins in these cortical regions. We show that following noise exposure, impaired gamma phase coherence within the prefrontal cortex is accompanied by alterations in pyramidal cell dendritic morphology and decreased expression of proteins involved in GABAergic (GAD65) and glutamatergic (NR2B) neurotransmission; findings that were not observed in the auditory cortex, where gamma phase coherence remained unchanged post-noise exposure. In contrast to the noise-induced effects we observed in the prefrontal cortex, plasticity in the auditory cortex was characterized by an increase in NR2B suggesting increased excitability, as well as increases in the synaptic proteins PSD95 and synaptophysin within the auditory cortex. Overall, our results highlight the disparate effect of noise-induced hearing loss on auditory and higher-level brain regions as well as potential structural and molecular mechanisms by which hearing loss may contribute to impaired cognitive and sensory functions mediated by the prefrontal and auditory cortices.


Sujet(s)
Cortex auditif , Surdité due au bruit , Cortex préfrontal , Surdité due au bruit/physiopathologie , Surdité due au bruit/anatomopathologie , Surdité due au bruit/métabolisme , Cortex auditif/métabolisme , Cortex auditif/physiopathologie , Cortex auditif/anatomopathologie , Cortex préfrontal/métabolisme , Cortex préfrontal/anatomopathologie , Animaux , Mâle , Plasticité neuronale/physiologie , Glutamate decarboxylase/métabolisme , Récepteurs du N-méthyl-D-aspartate/métabolisme , Homologue-4 de la protéine Disks Large/métabolisme , Dendrites/anatomopathologie , Dendrites/métabolisme , Rythme gamma/physiologie , Cellules pyramidales/métabolisme , Cellules pyramidales/anatomopathologie , Rats
4.
Hear Res ; 451: 109077, 2024 09 15.
Article de Anglais | MEDLINE | ID: mdl-39084132

RÉSUMÉ

Musicians are at risk of hearing loss and tinnitus due to regular exposure to high levels of noise. This level of risk may have been underestimated previously since damage to the auditory system, such as cochlear synaptopathy, may not be easily detectable using standard clinical measures. Most previous research investigating hearing loss in musicians has involved cross-sectional study designs that may capture only a snapshot of hearing health in relation to noise exposure. The aim of this study was to investigate the effects of cumulative noise exposure on behavioural, electrophysiological, and self-report indices of hearing damage in early-career musicians and non-musicians with normal hearing over a 2-year period. Participants completed an annual test battery consisting of pure tone audiometry, extended high-frequency hearing thresholds, distortion product otoacoustic emissions (DPOAEs), speech perception in noise, auditory brainstem responses, and self-report measures of tinnitus, hyperacusis, and hearing in background noise. Participants also completed the Noise Exposure Structured Interview to estimate cumulative noise exposure across the study period. Linear mixed models assessed changes over time. The longitudinal analysis comprised 64 early-career musicians (female n = 34; age range at T0 = 18-26 years) and 30 non-musicians (female n = 20; age range at T0 = 18-27 years). There were few longitudinal changes as a result of musicianship. Small improvements over time in some measures may be attributable to a practice/test-retest effect. Some measures (e.g., DPOAE indices of outer hair cell function) were associated with noise exposure at each time point, but did not show a significant change over time. A small proportion of participants reported a worsening of their tinnitus symptoms, which participants attributed to noise exposure, or not using hearing protection. Future longitudinal studies should attempt to capture the effects of noise exposure over a longer period, taken at several time points, for a precise measure of how hearing changes over time. Hearing conservation programmes for "at risk" individuals should closely monitor DPOAEs to detect early signs of noise-induced hearing loss when audiometric thresholds are clinically normal.


Sujet(s)
Audiométrie tonale , Seuil auditif , Potentiels évoqués auditifs du tronc cérébral , Surdité due au bruit , Ouïe , Musique , Exposition professionnelle , Émissions otoacoustiques spontanées , Autorapport , Acouphène , Humains , Femelle , Surdité due au bruit/physiopathologie , Surdité due au bruit/diagnostic , Surdité due au bruit/étiologie , Adulte , Acouphène/diagnostic , Acouphène/physiopathologie , Acouphène/étiologie , Études longitudinales , Mâle , Jeune adulte , Adolescent , Exposition professionnelle/effets indésirables , Perception de la parole , Hyperacousie/physiopathologie , Hyperacousie/diagnostic , Hyperacousie/étiologie , Bruit/effets indésirables , Facteurs temps , Bruit au travail/effets indésirables , Maladies professionnelles/diagnostic , Maladies professionnelles/physiopathologie , Maladies professionnelles/étiologie , Facteurs de risque , Stimulation acoustique , Modèles linéaires
5.
Sci Rep ; 14(1): 15655, 2024 07 08.
Article de Anglais | MEDLINE | ID: mdl-38977724

RÉSUMÉ

Fear and anxiety among patients are sometimes evoked in dental clinics due to the sound of dental drills. This study aimed to explore the impact of age-related hearing loss in the extended high frequency (EHF) range above 8 kHz on individuals' subjective discomfort towards dental drill noise. After measuring pure-tone audiometric thresholds at both conventional and extended high frequencies, we used a psychoacoustic approach to evaluate subjective impressions of four dental drill sound stimuli, which featured varying frequency components, in 62 participants (aged 12-67 years). We found a significant decrease in hearing sensitivity within the EHF range as age increased, with notable differences in hearing thresholds at 14 kHz between teenage and older adults exceeding 65 dB. Furthermore, significant differences were observed between younger and older (above 40 years) participants in the subjective impressions of dental drill noise, emphasizing age as a critical factor in the perception of high frequency components. Consequently, age may influence the unpleasantness of dental drilling noise. Compared to older individuals, young participants may exhibit increased fear of dental procedures owing to physiological factors. These results underscore the need for age-appropriate noise control strategies in dental clinics to mitigate anxiety and improve patient comfort.


Sujet(s)
Bruit , Humains , Adulte , Adulte d'âge moyen , Sujet âgé , Mâle , Femelle , Adolescent , Jeune adulte , Enfant , Bruit/effets indésirables , Seuil auditif/physiologie , Facteurs âges , Surdité due au bruit/physiopathologie , Surdité due au bruit/étiologie , Audiométrie tonale
6.
Hear Res ; 450: 109071, 2024 09 01.
Article de Anglais | MEDLINE | ID: mdl-38941694

RÉSUMÉ

Following adult-onset hearing impairment, crossmodal plasticity can occur within various sensory cortices, often characterized by increased neural responses to visual stimulation in not only the auditory cortex, but also in the visual and audiovisual cortices. In the present study, we used an established model of loud noise exposure in rats to examine, for the first time, whether the crossmodal plasticity in the audiovisual cortex that occurs following a relatively mild degree of hearing loss emerges solely from altered intracortical processing or if thalamocortical changes also contribute to the crossmodal effects. Using a combination of an established pharmacological 'cortical silencing' protocol and current source density analysis of the laminar activity recorded across the layers of the audiovisual cortex (i.e., the lateral extrastriate visual cortex, V2L), we observed layer-specific changes post-silencing in the strength of the residual visual, but not auditory, input in the noise exposed rats with mild hearing loss compared to rats with normal hearing. Furthermore, based on a comparison of the laminar profiles pre- versus post-silencing in both groups, we can conclude that noise exposure caused a re-allocation of the strength of visual inputs across the layers of the V2L cortex, including enhanced visual-evoked activity in the granular layer; findings consistent with thalamocortical plasticity. Finally, we confirmed that audiovisual integration within the V2L cortex depends on intact processing within intracortical circuits, and that this form of multisensory processing is vulnerable to disruption by noise-induced hearing loss. Ultimately, the present study furthers our understanding of the contribution of intracortical and thalamocortical processing to crossmodal plasticity as well as to audiovisual integration under both normal and mildly-impaired hearing conditions.


Sujet(s)
Stimulation acoustique , Cortex auditif , Modèles animaux de maladie humaine , Potentiels évoqués visuels , Plasticité neuronale , Stimulation lumineuse , Cortex visuel , Animaux , Cortex visuel/physiopathologie , Cortex auditif/physiopathologie , Mâle , Surdité due au bruit/physiopathologie , Perception visuelle , Perception auditive , Bruit/effets indésirables , Potentiels évoqués auditifs , Rats , Ouïe , Rat Sprague-Dawley
7.
Hear Res ; 447: 109021, 2024 06.
Article de Anglais | MEDLINE | ID: mdl-38703432

RÉSUMÉ

Understanding the complex pathologies associated with hearing loss is a significant motivation for conducting inner ear research. Lifelong exposure to loud noise, ototoxic drugs, genetic diversity, sex, and aging collectively contribute to human hearing loss. Replicating this pathology in research animals is challenging because hearing impairment has varied causes and different manifestations. A central aspect, however, is the loss of sensory hair cells and the inability of the mammalian cochlea to replace them. Researching therapeutic strategies to rekindle regenerative cochlear capacity, therefore, requires the generation of animal models in which cochlear hair cells are eliminated. This review discusses different approaches to ablate cochlear hair cells in adult mice. We inventoried the cochlear cyto- and histo-pathology caused by acoustic overstimulation, systemic and locally applied drugs, and various genetic tools. The focus is not to prescribe a perfect damage model but to highlight the limitations and advantages of existing approaches and identify areas for further refinement of damage models for use in regenerative studies.


Sujet(s)
Cochlée , Modèles animaux de maladie humaine , Cellules ciliées auditives , Régénération , Animaux , Cellules ciliées auditives/anatomopathologie , Cellules ciliées auditives/métabolisme , Souris , Cochlée/anatomopathologie , Cochlée/physiopathologie , Humains , Ouïe , Surdité due au bruit/physiopathologie , Surdité due au bruit/anatomopathologie , Perte d'audition/anatomopathologie , Perte d'audition/physiopathologie , Stimulation acoustique
8.
Hear Res ; 447: 109010, 2024 06.
Article de Anglais | MEDLINE | ID: mdl-38744019

RÉSUMÉ

Auditory nerve (AN) function has been hypothesized to deteriorate with age and noise exposure. Here, we perform a systematic review of published studies and find that the evidence for age-related deficits in AN function is largely consistent across the literature, but there are inconsistent findings among studies of noise exposure history. Further, evidence from animal studies suggests that the greatest deficits in AN response amplitudes are found in noise-exposed aged mice, but a test of the interaction between effects of age and noise exposure on AN function has not been conducted in humans. We report a study of our own examining differences in the response amplitude of the compound action potential N1 (CAP N1) between younger and older adults with and without a self-reported history of noise exposure in a large sample of human participants (63 younger adults 18-30 years of age, 103 older adults 50-86 years of age). CAP N1 response amplitudes were smaller in older than younger adults. Noise exposure history did not appear to predict CAP N1 response amplitudes, nor did the effect of noise exposure history interact with age. We then incorporated our results into two meta-analyses of published studies of age and noise exposure history effects on AN response amplitudes in neurotypical human samples. The meta-analyses found that age effects across studies are robust (r = -0.407), but noise exposure effects are weak (r = -0.152). We conclude that noise exposure effects may be highly variable depending on sample characteristics, study design, and statistical approach, and researchers should be cautious when interpreting results. The underlying pathology of age-related and noise-induced changes in AN function are difficult to determine in living humans, creating a need for longitudinal studies of changes in AN function across the lifespan and histological examination of the AN from temporal bones collected post-mortem.


Sujet(s)
Stimulation acoustique , Nerf cochléaire , Bruit , Humains , Bruit/effets indésirables , Sujet âgé , Nerf cochléaire/physiopathologie , Adulte d'âge moyen , Adulte , Sujet âgé de 80 ans ou plus , Facteurs âges , Jeune adulte , Adolescent , Vieillissement/physiologie , Potentiels évoqués auditifs , Surdité due au bruit/physiopathologie , Femelle , Mâle , Animaux , Potentiels d'action
9.
J Acoust Soc Am ; 155(5): 3254-3266, 2024 05 01.
Article de Anglais | MEDLINE | ID: mdl-38742964

RÉSUMÉ

Testudines are a highly threatened group facing an array of stressors, including alteration of their sensory environment. Underwater noise pollution has the potential to induce hearing loss and disrupt detection of biologically important acoustic cues and signals. To examine the conditions that induce temporary threshold shifts (TTS) in hearing in the freshwater Eastern painted turtle (Chrysemys picta picta), three individuals were exposed to band limited continuous white noise (50-1000 Hz) of varying durations and amplitudes (sound exposure levels ranged from 151 to 171 dB re 1 µPa2 s). Control and post-exposure auditory thresholds were measured and compared at 400 and 600 Hz using auditory evoked potential methods. TTS occurred in all individuals at both test frequencies, with shifts of 6.1-41.4 dB. While the numbers of TTS occurrences were equal between frequencies, greater shifts were observed at 600 Hz, a frequency of higher auditory sensitivity, compared to 400 Hz. The onset of TTS occurred at 154 dB re 1 µPa2 s for 600 Hz, compared to 158 dB re 1 µPa2 s at 400 Hz. The 400-Hz onset and patterns of TTS growth and recovery were similar to those observed in previously studied Trachemys scripta elegans, suggesting TTS may be comparable across Emydidae species.


Sujet(s)
Stimulation acoustique , Seuil auditif , Tortues , Animaux , Tortues/physiologie , Facteurs temps , Bruit/effets indésirables , Potentiels évoqués auditifs/physiologie , Surdité due au bruit/physiopathologie , Surdité due au bruit/étiologie , Mâle , Femelle , Ouïe/physiologie
10.
Trends Hear ; 28: 23312165241239541, 2024.
Article de Anglais | MEDLINE | ID: mdl-38738337

RÉSUMÉ

Cochlear synaptopathy, a form of cochlear deafferentation, has been demonstrated in a number of animal species, including non-human primates. Both age and noise exposure contribute to synaptopathy in animal models, indicating that it may be a common type of auditory dysfunction in humans. Temporal bone and auditory physiological data suggest that age and occupational/military noise exposure also lead to synaptopathy in humans. The predicted perceptual consequences of synaptopathy include tinnitus, hyperacusis, and difficulty with speech-in-noise perception. However, confirming the perceptual impacts of this form of cochlear deafferentation presents a particular challenge because synaptopathy can only be confirmed through post-mortem temporal bone analysis and auditory perception is difficult to evaluate in animals. Animal data suggest that deafferentation leads to increased central gain, signs of tinnitus and abnormal loudness perception, and deficits in temporal processing and signal-in-noise detection. If equivalent changes occur in humans following deafferentation, this would be expected to increase the likelihood of developing tinnitus, hyperacusis, and difficulty with speech-in-noise perception. Physiological data from humans is consistent with the hypothesis that deafferentation is associated with increased central gain and a greater likelihood of tinnitus perception, while human data on the relationship between deafferentation and hyperacusis is extremely limited. Many human studies have investigated the relationship between physiological correlates of deafferentation and difficulty with speech-in-noise perception, with mixed findings. A non-linear relationship between deafferentation and speech perception may have contributed to the mixed results. When differences in sample characteristics and study measurements are considered, the findings may be more consistent.


Sujet(s)
Cochlée , Perception de la parole , Acouphène , Humains , Cochlée/physiopathologie , Acouphène/physiopathologie , Acouphène/diagnostic , Animaux , Perception de la parole/physiologie , Hyperacousie/physiopathologie , Bruit/effets indésirables , Perception auditive/physiologie , Synapses/physiologie , Surdité due au bruit/physiopathologie , Surdité due au bruit/diagnostic , Perception sonore
11.
Sci Rep ; 14(1): 10689, 2024 05 09.
Article de Anglais | MEDLINE | ID: mdl-38724641

RÉSUMÉ

Homeostatic plasticity, the ability of neurons to maintain their averaged activity constant around a set point value, is thought to account for the central hyperactivity after hearing loss. Here, we investigated the putative role of GABAergic neurotransmission in this mechanism after a noise-induced hearing loss larger than 50 dB in high frequencies in guinea pigs. The effect of GABAergic inhibition is linked to the normal functioning of K + -Cl- co-transporter isoform 2 (KCC2) which maintains a low intracellular concentration of chloride. The expression of membrane KCC2 were investigated before and after noise trauma in the ventral and dorsal cochlear nucleus (VCN and DCN, respectively) and in the inferior colliculus (IC). Moreover, the effect of gabazine (GBZ), a GABA antagonist, was also studied on the neural activity in IC. We show that KCC2 is downregulated in VCN, DCN and IC 3 days after noise trauma, and in DCN and IC 30 days after the trauma. As expected, GBZ application in the IC of control animals resulted in an increase of spontaneous and stimulus-evoked activity. In the noise exposed animals, on the other hand, GBZ application decreased the stimulus-evoked activity in IC neurons. The functional implications of these central changes are discussed.


Sujet(s)
Surdité due au bruit , , Symporteurs , Acide gamma-amino-butyrique , Animaux , Symporteurs/métabolisme , Symporteurs/antagonistes et inhibiteurs , Cochons d'Inde , Surdité due au bruit/métabolisme , Surdité due au bruit/physiopathologie , Acide gamma-amino-butyrique/métabolisme , Mâle , Noyau cochléaire/métabolisme , Pyridazines/pharmacologie , Neurones/métabolisme
12.
Article de Chinois | MEDLINE | ID: mdl-38677986

RÉSUMÉ

Objective: To explore the mechanism of noise-induced hidden hearing loss by proteomics. Methods: In October 2022, 64 SPF male C57BL/6J mice were divided into control group and noise exposure group with 32 mice in each group according to random sampling method. The noise exposure group was exposed to 100 dB sound pressure level, 2000-16000 Hz broadband noise for 2 h, and the mouse hidden hearing loss model was established. Auditory brainstem response (ABR) was used to test the change of hearing threshold of mice on the 7th day after noise exposure, the damage of basal membrane hair cells was observed by immunofluorescence, and the differentially expressed proteins in the inner ear of mice in each group were identified and analyzed by 4D-Label-free quantitative proteomics, and verified by Western blotting. The results were statistically analyzed by ANOVA and t test. Results: On the 7th day after noise exposure, there was no significant difference in hearing threshold between the control group and the noise exposure group at click and 8000 Hz acoustic stimulation (P>0.05) . The hearing threshold in the noise exposure group was significantly higher than that in the control group under 16000 Hz acoustic stimulation (P<0.05) . Confocal immunofluorescence showed that the basal membrane hair cells of cochlear tissue in noise exposure group were arranged neatly, but the relative expression of C-terminal binding protein 2 antibody of presynaptic membrane in middle gyrus and basal gyrus was significantly lower than that in control group (P<0.05) . GO enrichment analysis showed that the functions of differentially expressed proteins were mainly concentrated in membrane potential regulation, ligand-gated channel activity, and ligand-gated ion channel activity. KEGG pathway enrichment analysis showed that differentially expressed proteins were significantly enriched in phosphatidylinositol 3 kinase-protein kinase B (PI3K-Akt) signaling pathway, NOD-like receptor signaling pathway, calcium signaling pathway, etc. Western blotting showed that the expression of inositol 1, 4, 5-trisphosphate receptor 3 (Itpr3) was increased and the expression of solute carrier family 38 member 2 (Slc38a2) was decreased in the noise exposure group (P<0.05) . Conclusion: Through proteomic analysis, screening and verification of the differential expression proteins Itpr3 and Slc38a2 in the constructed mouse noise-induced hidden hearing loss model, the glutaminergic synaptic related pathways represented by Itpr3 and Slc38a2 may be involved in the occurrence of hidden hearing loss.


Sujet(s)
Potentiels évoqués auditifs du tronc cérébral , Surdité due au bruit , Souris de lignée C57BL , Bruit , Protéomique , Animaux , Souris , Surdité due au bruit/métabolisme , Surdité due au bruit/physiopathologie , Mâle , Bruit/effets indésirables , Modèles animaux de maladie humaine , Seuil auditif , Oreille interne/métabolisme ,
13.
Brain Behav ; 14(4): e3479, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38648388

RÉSUMÉ

OBJECTIVE: To explore the changes in the cerebral microstructure of patients with noise-induced hearing loss (NIHL) using diffusion tensor imaging (DTI). METHOD: Overall, 122 patients with NIHL (mild [MP, n = 79], relatively severe patients [including moderate and severe; RSP, n = 32], and undetermined [lost to follow-up, n = 11]) and 84 healthy controls (HCs) were enrolled. All clinical data, including age, education level, hearing threshold, occupation type, noise exposure time, and some scale scores (including the Mini-Mental State Examination [MMSE], tinnitus handicap inventory [THI], and Hamilton Anxiety Scale [HAMA]), were collected and analyzed. All participants underwent T1WI3DFSPGR and DTI, and tract-based spatial statistics and region of interest (ROI) analysis were used for assessment. RESULTS: The final sample included 71 MP, 28 RSP, and 75 HCs. The HAMA scores of the three groups were significantly different (p < .05). The noise exposure times, hearing thresholds, and HAMA scores of the MP and RSP were significantly different (p < .05). The noise exposure time was positively correlated with the hearing threshold and negatively correlated with the HAMA scores (p < .05), whereas the THI scores were positively correlated with the hearing threshold (p < .05). DTI analysis showed that all DTI parameters (fractional anisotropy [FA], axial diffusivity [AD], mean diffusivity [MD], and radial diffusivity [RD]) were significantly different in the left inferior longitudinal fasciculus (ILF) and left inferior fronto-occipital fasciculus (IFOF) for the three groups (p < .05). In addition, the FA values were significantly lower in the bilateral corticospinal tract (CST), right fronto-pontine tract (FPT), right forceps major, left superior longitudinal fasciculus (temporal part) (SLF), and left cingulum (hippocampus) (C-H) of the MP and RSP than in those of the HCs (p < .05); the AD values showed diverse changes in the bilateral CST, left IFOF, right anterior thalamic radiation, right external capsule (EC), right SLF, and right superior cerebellar peduncle (SCP) of the MP and RSP relative to those of the HC (p < .05). However, there were no significant differences among the bilateral auditory cortex ROIs of the three groups (p > .05). There was a significant negative correlation between the FA and HAMA scores for the left IFOF/ILF, right FPT, left SLF, and left C-H for the three groups (p < .05). There was a significant positive correlation between the AD and HAMA scores for the left IFOF/ILF and right EC of the three groups (p < .05). There were significantly positive correlations between the RD/MD and HAMA scores in the left IFOF/ILF of the three groups (p < .05). There was a significant negative correlation between the AD in the right SCP and noise exposure time of the MP and RSP groups (p < .05). The AD, MD, and RD in the left ROI were significantly positively correlated with hearing threshold in the MP and RSP groups (p < .05), whereas FA in the right ROI was significantly positively correlated with the HAMA scores for the three groups (p < .05). CONCLUSION: The changes in the white matter (WM) microstructure may be related to hearing loss caused by noise exposure, and the WM structural abnormalities in patients with NIHL were mainly located in the syndesmotic fibers of the temporooccipital region, which affected the auditory and language pathways. This confirmed that the auditory pathways have abnormal structural connectivity in patients with NIHL.


Sujet(s)
Imagerie par tenseur de diffusion , Surdité due au bruit , Humains , Mâle , Femelle , Adulte , Adulte d'âge moyen , Surdité due au bruit/anatomopathologie , Surdité due au bruit/imagerie diagnostique , Surdité due au bruit/physiopathologie , Substance blanche/imagerie diagnostique , Substance blanche/anatomopathologie , Substance blanche/physiopathologie , Encéphale/imagerie diagnostique , Encéphale/anatomopathologie , Encéphale/physiopathologie
14.
J Neurophysiol ; 127(2): 452-462, 2022 02 01.
Article de Anglais | MEDLINE | ID: mdl-35020518

RÉSUMÉ

Noise-induced hearing deficits are important health problems in the industrialized world. As the underlying physiological dysfunctions are not well understood, research in suitable animal models is urgently needed. Three rodent species (Mongolian gerbil, rat, and mouse) were studied to compare the temporal dynamics of noise-induced hearing loss after identical procedures of noise exposure. Auditory brainstem responses (ABRs) were measured before, during, and up to 8 wk after noise exposure for threshold determination and ABR waveform analysis. Trauma induction with stepwise increasing sound pressure level was interrupted by five interspersed ABR measurements. Comparing short- and long-term dynamics underlying the following noise-induced hearing loss revealed diverging time courses between the three species. Hearing loss occurred early on during noise exposure in all three rodent species at or above trauma frequency. Initial noise level (105 dB SPL) was most effective in rats whereas the delayed level increase to 115 dB SPL affected mice much stronger. Induced temporary threshold shifts in rats and mice were larger in animals with lower pretrauma ABR thresholds. The increase in activity (gain) along the auditory pathway was derived by comparing the amplitudes of short- and long-latency ABR waveform components. Directly after trauma, significant effects were found for rats (decreasing gain) and mice (increasing gain) whereas gerbils revealed high individual variability in gain changes. Taken together, our comparative study revealed pronounced species-specific differences in the development of noise-induced hearing loss and the related processing along the auditory pathway.NEW & NOTEWORTHY We compared deficits after noise trauma in different rodents that are typically used in hearing research (Mongolian gerbil, rat, and mouse). We observed noise-induced threshold changes and alterations in the activity of processing auditory information along the ascending auditory pathway. Our results reveal pronounced differences in the characteristics of trauma-induced damage in these different rodent groups.


Sujet(s)
Voies auditives/physiopathologie , Potentiels évoqués auditifs du tronc cérébral/physiologie , Surdité due au bruit/physiopathologie , Animaux , Seuil auditif/physiologie , Comportement animal , Modèles animaux de maladie humaine , Gerbillinae , Souris , Bruit , Rats , Spécificité d'espèce
15.
J Neurophysiol ; 127(1): 313-327, 2022 01 01.
Article de Anglais | MEDLINE | ID: mdl-34907797

RÉSUMÉ

It is critical for hearing that the descending cochlear efferent system provides a negative feedback to hair cells to regulate hearing sensitivity and protect hearing from noise. The medial olivocochlear (MOC) efferent nerves project to outer hair cells (OHCs) to regulate OHC electromotility, which is an active cochlear amplifier and can increase hearing sensitivity. Here, we report that the MOC efferent nerves also could innervate supporting cells (SCs) in the vicinity of OHCs to regulate hearing sensitivity. MOC nerve fibers are cholinergic, and acetylcholine (ACh) is a primary neurotransmitter. Immunofluorescent staining showed that MOC nerve endings, presynaptic vesicular acetylcholine transporters (VAChTs), and postsynaptic ACh receptors were visible at SCs and in the SC area. Application of ACh in SCs could evoke a typical inward current and reduce gap junctions (GJs) between them, which consequently enhanced the direct effect of ACh on OHCs to shift but not eliminate OHC electromotility. This indirect, GJ-mediated inhibition had a long-lasting influence. In vivo experiments further demonstrated that deficiency of this GJ-mediated efferent pathway decreased the regulation of active cochlear amplification and compromised the protection against noise. In particular, distortion product otoacoustic emission (DPOAE) showed a delayed reduction after noise exposure. Our findings reveal a new pathway for the MOC efferent system via innervating SCs to control active cochlear amplification and hearing sensitivity. These data also suggest that this SC GJ-mediated efferent pathway may play a critical role in long-term efferent inhibition and is required for protection of hearing from noise trauma.NEW & NOTEWORTHY The cochlear efferent system provides a negative feedback to control hair cell activity and hearing sensitivity and plays a critical role in noise protection. We reveal a new efferent control pathway in which medial olivocochlear efferent fibers have innervations with cochlear supporting cells to control their gap junctions, therefore regulating outer hair cell electromotility and hearing sensitivity. This supporting cell gap junction-mediated efferent control pathway is required for the protection of hearing from noise.


Sujet(s)
Nerf cochléaire/physiopathologie , Cellules ciliées auditives externes/physiologie , Surdité due au bruit/physiopathologie , Neurones efférents/physiologie , Animaux , Voies efférentes/physiopathologie , Femelle , Cochons d'Inde , Mâle
16.
J Neurophysiol ; 127(1): 239-254, 2022 01 01.
Article de Anglais | MEDLINE | ID: mdl-34936500

RÉSUMÉ

In the auditory modality, noise trauma has often been used to investigate cortical plasticity as it causes cochlear hearing loss. One limitation of these past studies, however, is that the effects of noise trauma have been mostly documented at the granular layer, which is the main cortical recipient of thalamic inputs. Importantly, the cortex is composed of six different layers each having its own pattern of connectivity and specific role in sensory processing. The present study aims at investigating the effects of acute and chronic noise trauma on the laminar pattern of spontaneous activity (SA) in primary auditory cortex (A1) of the anesthetized guinea pig. We show that spontaneous activity is dramatically altered across cortical layers after acute and chronic noise-induced hearing loss. First, spontaneous activity was globally enhanced across cortical layers, both in terms of firing rate and amplitude of spike-triggered average of local field potentials. Second, current source density on (spontaneous) spike-triggered average of local field potentials indicates that current sinks develop in the supra- and infragranular layers. These latter results suggest that supragranular layers become a major input recipient and the propagation of spontaneous activity over a cortical column is greatly enhanced after acute and chronic noise-induced hearing loss. We discuss the possible mechanisms and functional implications of these changes.NEW & NOTEWORTHY The present study investigates the effects of acute and chronic noise trauma on the laminar pattern of spontaneous activity in the primary auditory cortex. Our study is first to report that noise trauma alters the sequence of cortical column activation during ongoing activity. In particular, we show that the supragranular layer becomes a major input recipient and the synaptic activity in the infragranular layers is enhanced.


Sujet(s)
Cortex auditif/physiopathologie , Phénomènes électrophysiologiques/physiologie , Surdité due au bruit/physiopathologie , Plasticité neuronale/physiologie , Animaux , Cortex auditif/cytologie , Cochons d'Inde
17.
Int J Mol Sci ; 22(22)2021 Nov 11.
Article de Anglais | MEDLINE | ID: mdl-34830090

RÉSUMÉ

Recent studies have identified sex-differences in auditory physiology and in the susceptibility to noise-induced hearing loss (NIHL). We hypothesize that 17ß-estradiol (E2), a known modulator of auditory physiology, may underpin sex-differences in the response to noise trauma. Here, we gonadectomized B6CBAF1/J mice and used a combination of electrophysiological and histological techniques to study the effects of estrogen replacement on peripheral auditory physiology in the absence of noise exposure and on protection from NIHL. Functional analysis of auditory physiology in gonadectomized female mice revealed that E2-treatment modulated the peripheral response to sound in the absence of changes to the endocochlear potential compared to vehicle-treatment. E2-replacement in gonadectomized female mice protected against hearing loss following permanent threshold shift (PTS)- and temporary threshold shift (TTS)-inducing noise exposures. Histological analysis of the cochlear tissue revealed that E2-replacement mitigated outer hair cell loss and cochlear synaptopathy following noise exposure compared to vehicle-treatment. Lastly, using fluorescent in situ hybridization, we demonstrate co-localization of estrogen receptor-2 with type-1C, high threshold spiral ganglion neurons, suggesting that the observed protection from cochlear synaptopathy may occur through E2-mediated preservation of these neurons. Taken together, these data indicate the estrogen signaling pathways may be harnessed for the prevention and treatment of NIHL.


Sujet(s)
Cochlée , Oestradiol/pharmacologie , Potentiels évoqués auditifs/effets des médicaments et des substances chimiques , Surdité due au bruit , Animaux , Cochlée/métabolisme , Cochlée/anatomopathologie , Cochlée/physiopathologie , Femelle , Surdité due au bruit/métabolisme , Surdité due au bruit/anatomopathologie , Surdité due au bruit/physiopathologie , Surdité due au bruit/prévention et contrôle , Souris , Ovariectomie
18.
J Neurophysiol ; 126(6): 2027-2038, 2021 12 01.
Article de Anglais | MEDLINE | ID: mdl-34788179

RÉSUMÉ

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


Sujet(s)
Maladies cochléaires/physiopathologie , Nerf cochléaire/physiopathologie , Surdité due au bruit/physiopathologie , Ganglion spiral/physiopathologie , Synapses/anatomopathologie , Animaux , Modèles animaux de maladie humaine , Mâle , Souris , Souris de lignée CBA
19.
Biol Open ; 10(10)2021 10 15.
Article de Anglais | MEDLINE | ID: mdl-34668520

RÉSUMÉ

Over 1.1 billion individuals are at risk for noise induced hearing loss yet there is no accepted therapy. A long history of research has demonstrated that excessive noise exposure will kill outer hair cells (OHCs). Such observations have fueled the notion that dead OHCs underlie hearing loss. Therefore, previous and current therapeutic approaches are based on preventing the loss of OHCs. However, the relationship between OHC loss and hearing loss is at best a modest correlation. This suggests that in addition to the death of OHCs, other mechanisms may regulate the type and degree of hearing loss. In the current study, we tested the hypothesis that permanent noise-induced-hearing loss is consequent to additional mechanisms beyond the noise dose and the death of OHCs. Hooded male rats were randomly divided into noise and control groups. Morphological and physiological assessments were conducted on both groups. The combined results suggest that beyond OHC loss, the surviving cochlear elements shape sensorineural outcomes, which can be nondeterministic. These findings provide the basis for individualized ototherapeutics that manipulate surviving cellular elements in order to bias cochlear function towards normal hearing even in the presence of dead OHCs.


Sujet(s)
Cochlée/effets des radiations , Cellules ciliées auditives externes/effets des radiations , Surdité due au bruit/physiopathologie , Bruit/effets indésirables , Animaux , Modèles animaux de maladie humaine , Mâle , Rats
20.
Cell Rep ; 36(13): 109758, 2021 09 28.
Article de Anglais | MEDLINE | ID: mdl-34592158

RÉSUMÉ

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


Sujet(s)
Oreille interne/métabolisme , Cellules ciliées auditives/métabolisme , Surdité due au bruit/métabolisme , Surdité due au bruit/physiopathologie , Ganglion spiral/métabolisme , Animaux , Cochlée/métabolisme , Cochlée/physiopathologie , Oreille interne/physiopathologie , Potentiels évoqués auditifs du tronc cérébral/physiologie , Surdité due au bruit/génétique , Souris , Neurones/métabolisme , Bruit , Ganglion spiral/cytologie , Ganglion spiral/physiopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE