Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 546
1.
Cereb Cortex ; 34(5)2024 May 02.
Article En | MEDLINE | ID: mdl-38715406

Presbycusis has been reported as related to cognitive decline, but its underlying neurophysiological mechanism is still unclear. This study aimed to investigate the relationship between metabolite levels, cognitive function, and node characteristics in presbycusis based on graph theory methods. Eighty-four elderly individuals with presbycusis and 63 age-matched normal hearing controls underwent magnetic resonance spectroscopy, functional magnetic resonance imaging scans, audiological assessment, and cognitive assessment. Compared with the normal hearing group, presbycusis patients exhibited reduced gamma-aminobutyric acid and glutamate levels in the auditory region, increased nodal characteristics in the temporal lobe and precuneus, as well as decreased nodal characteristics in the superior occipital gyrus and medial orbital. The right gamma-aminobutyric acid levels were negatively correlated with the degree centrality in the right precuneus and the executive function. Degree centrality in the right precuneus exhibited significant correlations with information processing speed and executive function, while degree centrality in the left medial orbital demonstrated a negative association with speech recognition ability. The degree centrality and node efficiency in the superior occipital gyrus exhibited a negative association with hearing loss and speech recognition ability, respectively. These observed changes indicate alterations in metabolite levels and reorganization patterns at the brain network level after auditory deprivation.


Cognitive Dysfunction , Magnetic Resonance Imaging , Presbycusis , Humans , Male , Female , Presbycusis/diagnostic imaging , Presbycusis/metabolism , Presbycusis/physiopathology , Aged , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Magnetic Resonance Spectroscopy , Glutamic Acid/metabolism , gamma-Aminobutyric Acid/metabolism , Middle Aged , Brain/diagnostic imaging , Brain/metabolism
2.
Hear Res ; 446: 109006, 2024 May.
Article En | MEDLINE | ID: mdl-38583350

Hair cells in the cochlear sensory epithelia serve as mechanosensory receptors, converting sound into neuronal signals. The basal sensory epithelia are responsible for transducing high-frequency sounds, while the apex handles low-frequency sounds. Age-related hearing loss predominantly affects hearing at high frequencies and is indicative of damage to the basal sensory epithelia. However, the precise mechanism underlying this site-selective injury remains unclear. In this study, we employed a microscale proteomics approach to examine and compare protein expression in different regions of the cochlear sensory epithelia (upper half and lower half) in 1.5-month-old (normal hearing) and 6-month-old (severe high-frequency hearing loss without hair cell loss) C57BL/6J mice. A total of 2,386 proteins were detected, and no significant differences in protein expression were detected in the upper half of the cochlear sensory epithelia between the two age groups. The expression of 20 proteins in the lower half of the cochlear sensory epithelia significantly differed between the two age groups (e.g., MATN1, MATN4, and AQP1). Moreover, there were 311 and 226 differentially expressed proteins between the upper and lower halves of the cochlear sensory epithelia in 1.5-month-old and 6-month-old mice, respectively. The expression levels of selected proteins were validated by Western blotting. These findings suggest that the spatial differences in protein expression within the cochlear sensory epithelia may play a role in determining the susceptibility of cells at different sites of the cochlea to age-related damage.


Cochlea , Mice, Inbred C57BL , Presbycusis , Proteomics , Animals , Cochlea/metabolism , Cochlea/pathology , Presbycusis/metabolism , Presbycusis/pathology , Presbycusis/physiopathology , Presbycusis/genetics , Age Factors , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Aging/metabolism , Aging/pathology , Disease Models, Animal , Hearing , Epithelium/metabolism , Male , Mice
3.
Hear Res ; 446: 109005, 2024 May.
Article En | MEDLINE | ID: mdl-38598943

Auditory nerve (AN) fibers that innervate inner hair cells in the cochlea degenerate with advancing age. It has been proposed that age-related reductions in brainstem frequency-following responses (FFR) to the carrier of low-frequency, high-intensity pure tones may partially reflect this neural loss in the cochlea (Märcher-Rørsted et al., 2022). If the loss of AN fibers is the primary factor contributing to age-related changes in the brainstem FFR, then the FFR could serve as an indicator of cochlear neural degeneration. In this study, we employed electrocochleography (ECochG) to investigate the effects of age on frequency-following neurophonic potentials, i.e., neural responses phase-locked to the carrier frequency of the tone stimulus. We compared these findings to the brainstem-generated FFRs obtained simultaneously using the same stimulation. We conducted recordings in young and older individuals with normal hearing. Responses to pure tones (250 ms, 516 and 1086 Hz, 85 dB SPL) and clicks were recorded using both ECochG at the tympanic membrane and traditional scalp electroencephalographic (EEG) recordings of the FFR. Distortion product otoacoustic emissions (DPOAE) were also collected. In the ECochG recordings, sustained AN neurophonic (ANN) responses to tonal stimulation, as well as the click-evoked compound action potential (CAP) of the AN, were significantly reduced in the older listeners compared to young controls, despite normal audiometric thresholds. In the EEG recordings, brainstem FFRs to the same tone stimulation were also diminished in the older participants. Unlike the reduced AN CAP response, the transient-evoked wave-V remained unaffected. These findings could indicate that a decreased number of AN fibers contributes to the response in the older participants. The results suggest that the scalp-recorded FFR, as opposed to the clinical standard wave-V of the auditory brainstem response, may serve as a more reliable indicator of age-related cochlear neural degeneration.


Acoustic Stimulation , Aging , Audiometry, Evoked Response , Cochlea , Cochlear Nerve , Evoked Potentials, Auditory, Brain Stem , Nerve Degeneration , Humans , Female , Cochlea/physiopathology , Cochlea/innervation , Adult , Aged , Male , Middle Aged , Young Adult , Age Factors , Cochlear Nerve/physiopathology , Aging/physiology , Electroencephalography , Audiometry, Pure-Tone , Auditory Threshold , Presbycusis/physiopathology , Presbycusis/diagnosis , Predictive Value of Tests , Time Factors
4.
Hear Res ; 447: 109008, 2024 Jun.
Article En | MEDLINE | ID: mdl-38636186

The auditory cortex is the source of descending connections providing contextual feedback for auditory signal processing at almost all levels of the lemniscal auditory pathway. Such feedback is essential for cognitive processing. It is likely that corticofugal pathways are degraded with aging, becoming important players in age-related hearing loss and, by extension, in cognitive decline. We are testing the hypothesis that surface, epidural stimulation of the auditory cortex during aging may regulate the activity of corticofugal pathways, resulting in modulation of central and peripheral traits of auditory aging. Increased auditory thresholds during ongoing age-related hearing loss in the rat are attenuated after two weeks of epidural stimulation with direct current applied to the surface of the auditory cortex for two weeks in alternate days (Fernández del Campo et al., 2024). Here we report that the same cortical electrical stimulation protocol induces structural and cytochemical changes in the aging cochlea and auditory brainstem, which may underlie recovery of age-degraded auditory sensitivity. Specifically, we found that in 18 month-old rats after two weeks of cortical electrical stimulation there is, relative to age-matched non-stimulated rats: a) a larger number of choline acetyltransferase immunoreactive neuronal cell body profiles in the ventral nucleus of the trapezoid body, originating the medial olivocochlear system.; b) a reduction of age-related dystrophic changes in the stria vascularis; c) diminished immunoreactivity for the pro-inflammatory cytokine TNFα in the stria vascularis and spiral ligament. d) diminished immunoreactivity for Iba1 and changes in the morphology of Iba1 immunoreactive cells in the lateral wall, suggesting reduced activation of macrophage/microglia; d) Increased immunoreactivity levels for calretinin in spiral ganglion neurons, suggesting excitability modulation by corticofugal stimulation. Altogether, these findings support that non-invasive neuromodulation of the auditory cortex during aging preserves the cochlear efferent system and ameliorates cochlear aging traits, including stria vascularis dystrophy, dysregulated inflammation and altered excitability in primary auditory neurons.


Aging , Auditory Cortex , Auditory Pathways , Cochlea , Electric Stimulation , Presbycusis , Animals , Auditory Cortex/metabolism , Auditory Cortex/physiopathology , Cochlea/innervation , Cochlea/metabolism , Cochlea/physiopathology , Cochlea/pathology , Presbycusis/physiopathology , Presbycusis/metabolism , Presbycusis/pathology , Auditory Pathways/physiopathology , Auditory Pathways/metabolism , Male , Aging/pathology , Aging/metabolism , Disease Models, Animal , Age Factors , Neurons, Efferent/metabolism , Microglia/metabolism , Microglia/pathology , Auditory Threshold , Choline O-Acetyltransferase/metabolism , Olivary Nucleus/metabolism , Evoked Potentials, Auditory, Brain Stem , Hearing , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism , Calcium-Binding Proteins , Microfilament Proteins
5.
Hear Res ; 446: 109004, 2024 May.
Article En | MEDLINE | ID: mdl-38608332

The naturally occurring amino acid, l-ergothioneine (EGT), has immense potential as a therapeutic, having shown promise in the treatment of other disease models, including neurological disorders. EGT is naturally uptaken into cells via its specific receptor, OCTN1, to be utilized by cells as an antioxidant and anti-inflammatory. In our current study, EGT was administered over a period of 6 months to 25-26-month-old CBA/CaJ mice as a possible treatment for age-related hearing loss (ARHL), since presbycusis has been linked to higher levels of cochlear oxidative stress, apoptosis, and chronic inflammation. Results from the current study indicate that EGT can prevent aging declines of some key features of ARHL. However, we found a distinct sex difference for the response to the treatments, for hearing - Auditory Brainstem Responses (ABRs) and Distortion Product Otoacoustic Emissions (DPOAEs). Males exhibited lower threshold declines in both low dose (LD) and high dose (HD) test groups throughout the testing period and did not display some of the characteristic aging declines in hearing seen in Control animals. In contrast, female mice did not show any therapeutic effects with either treatment dose. Further confirming this sex difference, EGT levels in whole blood sampling throughout the testing period showed greater uptake of EGT in males compared to females. Additionally, RT-PCR results from three tissue types of the inner ear confirmed EGT activity in the cochlea in both males and females. Males and females exhibited significant differences in biomarkers related to apoptosis (Cas-3), inflammation (TNF-a), oxidative stress (SOD2), and mitochondrial health (PGC1a).These changes were more prominent in males as compared to females, especially in stria vascularis tissue. Taken together, these findings suggest that EGT has the potential to be a naturally derived therapeutic for slowing down the progression of ARHL, and possibly other neurodegenerative diseases. EGT, while effective in the treatment of some features of presbycusis in aging males, could also be modified into a general prophylaxis for other age-related disorders where treatment protocols would include eating a larger proportion of EGT-rich foods or supplements. Lastly, the sex difference discovered here, needs further investigation to see if therapeutic conditions can be developed where aging females show better responsiveness to EGT.


Aging , Antioxidants , Cochlea , Disease Models, Animal , Disease Progression , Ergothioneine , Evoked Potentials, Auditory, Brain Stem , Mice, Inbred CBA , Oxidative Stress , Presbycusis , Animals , Ergothioneine/pharmacology , Female , Evoked Potentials, Auditory, Brain Stem/drug effects , Male , Presbycusis/physiopathology , Presbycusis/pathology , Presbycusis/drug therapy , Presbycusis/metabolism , Presbycusis/prevention & control , Oxidative Stress/drug effects , Aging/drug effects , Aging/pathology , Antioxidants/pharmacology , Sex Factors , Cochlea/drug effects , Cochlea/metabolism , Cochlea/physiopathology , Cochlea/pathology , Age Factors , Apoptosis/drug effects , Otoacoustic Emissions, Spontaneous/drug effects , Superoxide Dismutase/metabolism , Auditory Threshold/drug effects , Hearing/drug effects , Mice , Anti-Inflammatory Agents/pharmacology
6.
Neurobiol Aging ; 111: 1-13, 2022 03.
Article En | MEDLINE | ID: mdl-34915240

Age-related sensorineural hearing loss (HL) leads to localized brain changes in the primary auditory cortex, long-range functional alterations, and is considered a risk factor for dementia. Nonhuman studies have repeatedly highlighted cross-modal brain plasticity in sensorial brain networks other than those primarily involved in the peripheral damage, thus in this study, the possible cortical alterations associated with HL have been analyzed using a whole-brain multimodal connectomic approach. Fifty-two HL and 30 normal hearing participants were examined in a 3T MRI study along with audiological and neurological assessments. Between-regions functional connectivity and whole-brain probabilistic tractography were calculated in a connectome-based manner and graph theory was used to obtain low-dimensional features for the analysis of brain connectivity at global and local levels. The HL condition was associated with a different functional organization of the visual subnetwork as revealed by a significant increase in global efficiency, density, and clustering coefficient. These functional effects were mirrored by similar (but more subtle) structural effects suggesting that a functional repurposing of visual cortical centers occurs to compensate for age-related loss of hearing abilities.


Connectome/methods , Neuronal Plasticity , Presbycusis/diagnosis , Presbycusis/physiopathology , Aged , Auditory Cortex/pathology , Auditory Cortex/physiopathology , Brain/diagnostic imaging , Brain/physiopathology , Diffusion Tensor Imaging , Female , Hearing , Humans , Magnetic Resonance Imaging/methods , Male , Middle Aged , Neural Pathways/physiopathology , Visual Cortex/physiopathology
7.
Neurobiol Aging ; 110: 61-72, 2022 02.
Article En | MEDLINE | ID: mdl-34861480

Recent evidence suggests that modulation of the large-conductance, calcium-activated potassium (BK) channel regulates auditory processing in the brain. Because ion channel expression often changes during aging, this could be a factor in age-related hearing loss. The current study explored how the novel BK channel modulator LS3 shapes central auditory processing in young and old adult mice. In vivo extracellular recordings in the auditory midbrain demonstrated that LS3 differentially modulates neural processing along the tonotopic axis. Though sound-evoked activity was reduced in the mid and ventral tonotopic regions, LS3 enhanced excitatory drive and sound-evoked responses for some neurons in the dorsal, low-frequency region. Behavioral assessment using acoustic reflex modification audiometry indicated improved tone salience following systemic LS3 administration. Moderation of these responses with aging correlated with an age-related decline in BK channel expression. These findings suggest that targeting the BK channel enhances responsivity to tonal sounds, providing the potential to improve hearing acuity and treat hearing loss.


Aging/physiology , Auditory Perception/physiology , Behavior, Animal/physiology , Intercellular Signaling Peptides and Proteins/administration & dosage , Intercellular Signaling Peptides and Proteins/pharmacology , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Mesencephalon/physiology , Presbycusis/etiology , Aging/metabolism , Animals , Evoked Potentials, Auditory/drug effects , Gene Expression/drug effects , Hearing/drug effects , Large-Conductance Calcium-Activated Potassium Channels/genetics , Large-Conductance Calcium-Activated Potassium Channels/physiology , Mice , Molecular Targeted Therapy , Neurons/physiology , Presbycusis/physiopathology , Presbycusis/therapy , Reflex, Acoustic/physiology
8.
Neurobiol Aging ; 110: 47-60, 2022 02.
Article En | MEDLINE | ID: mdl-34852306

Impaired temporal resolution of the central auditory system has long been suggested to contribute to speech understanding deficits in the elderly. However, it has been difficult to differentiate between direct age-related central deficits and indirect effects of confounding peripheral age-related hearing loss on temporal resolution. To differentiate this, we measured temporal acuity in the inferior colliculus (IC) of aged CBA/J and C57BL/6 mice, as a model of aging with and without concomitant hearing loss. We used two common measures of auditory temporal processing: gap detection as a measure of temporal fine structure and amplitude-modulated noise as a measure of envelope sensitivity. Importantly, auditory temporal acuity remained precise in the IC of old CBA/J mice when no or only minimal age-related hearing loss was present. In contrast, temporal acuity was only indirectly reduced by the presence of age-related hearing loss in aged C57BL/6 mice, not by affecting the brainstem precision, but by affecting the signal-to-noise ratio of the neuronal activity in the IC. This demonstrates that indirect effects of age-related peripheral hearing loss likely remain an important factor for temporal processing in aging in comparison to 'pure' central auditory decline itself. It also draws attention to the issue that the threshold difference between 'nearly normal' or 'clinically normal' hearing aging subjects in comparison to normal hearing young subjects still can have indirect effects on central auditory neural representations of temporal processing.


Aging/physiology , Auditory Perception/physiology , Inferior Colliculi/physiology , Presbycusis/physiopathology , Time Perception/physiology , Animals , Mice, Inbred C57BL , Mice, Inbred CBA , Neurons/physiology , Presbycusis/etiology , Signal-To-Noise Ratio
9.
J Neurosci ; 41(50): 10293-10304, 2021 12 15.
Article En | MEDLINE | ID: mdl-34753738

A common complaint of older adults is difficulty understanding speech, particularly in challenging listening conditions. Accumulating evidence suggests that these difficulties may reflect a loss and/or dysfunction of auditory nerve (AN) fibers. We used a novel approach to study age-related changes in AN structure and several measures of AN function, including neural synchrony, in 58 older adults and 42 younger adults. AN activity was measured in response to an auditory click (compound action potential; CAP), presented at stimulus levels ranging from 70 to 110 dB pSPL. Poorer AN function was observed for older than younger adults across CAP measures at higher but not lower stimulus levels. Associations across metrics and stimulus levels were consistent with age-related AN disengagement and AN dyssynchrony. High-resolution T2-weighted structural imaging revealed age-related differences in the density of cranial nerve VIII, with lower density in older adults with poorer neural synchrony. Individual differences in neural synchrony were the strongest predictor of speech recognition, such that poorer synchrony predicted poorer recognition of time-compressed speech and poorer speech recognition in noise for both younger and older adults. These results have broad clinical implications and are consistent with an interpretation that age-related atrophy at the level of the AN contributes to poorer neural synchrony and may explain some of the perceptual difficulties of older adults.SIGNIFICANCE STATEMENT Differences in auditory nerve (AN) pathophysiology may contribute to the large variations in hearing and communication abilities of older adults. However, current diagnostics focus largely on the increase in detection thresholds, which is likely because of the absence of indirect measures of AN function in standard clinical test batteries. Using novel metrics of AN function, combined with estimates of AN structure and auditory function, we identified age-related differences across measures that we interpret to represent age-related reductions in AN engagement and poorer neural synchrony. Structure-function associations are consistent with an explanation of AN deficits that arise from age-related atrophy of the AN. Associations between neural synchrony and speech recognition suggest that individual and age-related deficits in neural synchrony contribute to speech recognition deficits.


Cochlear Nerve/physiopathology , Presbycusis/physiopathology , Age Factors , Aged , Aged, 80 and over , Audiometry , Auditory Threshold/physiology , Electroencephalography , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged
10.
J Neurosci ; 41(46): 9650-9668, 2021 11 17.
Article En | MEDLINE | ID: mdl-34611028

Age-related hearing loss (presbycusis) is a chronic health condition that affects one-third of the world population. One hallmark of presbycusis is a difficulty hearing in noisy environments. Presbycusis can be separated into two components: alterations of peripheral mechanotransduction of sound in the cochlea and central alterations of auditory processing areas of the brain. Although the effects of the aging cochlea in hearing loss have been well studied, the role of the aging brain in hearing loss is less well understood. Therefore, to examine how age-related central processing changes affect hearing in noisy environments, we used a mouse model (Thy1-GCaMP6s X CBA) that has excellent peripheral hearing in old age. We used in vivo two-photon Ca2+ imaging to measure the responses of neuronal populations in auditory cortex (ACtx) of adult (2-6 months, nine male, six female, 4180 neurons) and aging mice (15-17 months, six male, three female, 1055 neurons) while listening to tones in noisy backgrounds. We found that ACtx neurons in aging mice showed larger responses to tones and have less suppressed responses consistent with reduced inhibition. Aging neurons also showed less sensitivity to temporal changes. Population analysis showed that neurons in aging mice showed higher pairwise activity correlations and showed a reduced diversity in responses to sound stimuli. Using neural decoding techniques, we show a loss of information in neuronal populations in the aging brain. Thus, aging not only affects the responses of single neurons but also affects how these neurons jointly represent stimuli.SIGNIFICANCE STATEMENT Aging results in hearing deficits particularly under challenging listening conditions. We show that auditory cortex contains distinct subpopulations of excitatory neurons that preferentially encode different stimulus features and that aging selectively reduces certain subpopulations. We also show that aging increases correlated activity between neurons and thereby reduces the response diversity in auditory cortex. The loss of population response diversity leads to a decrease of stimulus information and deficits in sound encoding, especially in noisy backgrounds. Future work determining the identities of circuits affected by aging could provide new targets for therapeutic strategies.


Aging/pathology , Auditory Cortex/physiopathology , Neurons/pathology , Presbycusis/physiopathology , Animals , Female , Male , Mice , Mice, Inbred CBA
11.
Neurobiol Aging ; 108: 133-145, 2021 12.
Article En | MEDLINE | ID: mdl-34601244

Loss of inner hair cell-auditory nerve fiber synapses is considered to be an important early stage of neural presbyacusis. Mass potentials, recorded at the cochlear round window, can be used to derive the neural index (NI), a sensitive measure for pharmacologically-induced synapse loss. Here, we investigate the applicability of the NI for measuring age-related auditory synapse loss in young-adult, middle-aged, and old Mongolian gerbils. Synapse loss, which was progressively evident in the 2 aged groups, correlated weakly with NI when measured at a fixed sound level of 60 dB SPL. However, the NI was confounded by decreases in single-unit firing rates at 60 dB SPL. NI at 30 dB above threshold, when firing rates were similar between age groups, did not correlate with synapse loss. Our results show that synapse loss is poorly reflected in the NI of aged gerbils, particularly if further peripheral pathologies are present. The NI may therefore not be a reliable clinical tool to assess synapse loss in aged humans with peripheral hearing loss.


Aging/pathology , Hair Cells, Auditory, Inner/pathology , Presbycusis/pathology , Synapses/pathology , Acoustic Stimulation , Animals , Auditory Threshold , Gerbillinae , Presbycusis/physiopathology
12.
Biomed Pharmacother ; 143: 112149, 2021 Nov.
Article En | MEDLINE | ID: mdl-34507120

Age-related hearing loss (AHL) is the most common sensory disorder of aged population. Currently, one of the most important sources of experimental medicine for AHL is medicinal plants. This study performed the first investigation of the effect of thymoquinone (TQ), a potent antioxidant, on AHL. Here, we used inbred C57BL/6J mice (B6 mice) as a successful experimental model of the early onset of AHL. The behavioral assessment of hearing revealed that the injection of a high dose of TQ (40 mg/kg; TQ40) significantly improved the auditory sensitivity of B6 mice at all tested frequencies (8, 16 and 22 kHz). Histological sections of cochlea from B6 mice injected with a low dose (20 mg/kg; TQ20) and high dose showed relatively less degenerative signs in the modiolus, hair cells and spiral ligaments, the main constituents of the cochlea. In addition, TQ40 completely restored the normal pattern of hair cells in B6 mice, as shown in scanning electron micrographs. Our data indicated that TQ20 and TQ40 reduced levels of Bak1-mediated apoptosis in the cochlea of B6 mice. Interestingly, the level of Sirt1, a positive regulator of autophagy, was significantly increased in B6 mice administered TQ40. In conclusion, TQ relieves the symptoms of AHL by downregulating Bak1 and activating Sirt1 in the cochlea of B6 mice.


Antioxidants/pharmacology , Benzoquinones/pharmacology , Cochlea/drug effects , Hearing/drug effects , Presbycusis/drug therapy , Sirtuin 1/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , Animals , Apoptosis/drug effects , Auditory Threshold/drug effects , Autophagy/drug effects , Cochlea/metabolism , Cochlea/physiopathology , Cochlea/ultrastructure , Disease Models, Animal , Female , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/ultrastructure , Mice, Inbred C57BL , Presbycusis/metabolism , Presbycusis/pathology , Presbycusis/physiopathology , Signal Transduction , Sirtuin 1/genetics , bcl-2 Homologous Antagonist-Killer Protein/genetics
13.
Int J Mol Sci ; 22(15)2021 Jul 27.
Article En | MEDLINE | ID: mdl-34360766

Age-related hearing loss (ARHL) is the most common sensory disorder among older people, and yet, the treatment options are limited to medical devices such as hearing aids and cochlear implants. The high prevalence of ARHL mandates the development of treatment strategies that can prevent or rescue age-related cochlear degeneration. In this study, we investigated a novel pharmacological strategy based on inhibition of the adenosine A2A receptor (A2AR) in middle aged C57BL/6 mice prone to early onset ARHL. C57BL/6J mice were treated with weekly istradefylline (A2AR antagonist; 1 mg/kg) injections from 6 to 12 months of age. Auditory function was assessed using auditory brainstem responses (ABR) to tone pips (4-32 kHz). ABR thresholds and suprathreshold responses (wave I amplitudes and latencies) were evaluated at 6, 9, and 12 months of age. Functional outcomes were correlated with quantitative histological assessments of sensory hair cells. Cognitive function was assessed using the Morris water maze and the novel object recognition test, and the zero maze test was used to assess anxiety-like behaviour. Weekly injections of istradefylline attenuated ABR threshold shifts by approximately 20 dB at mid to high frequencies (16-32 kHz) but did not improve ABR suprathreshold responses. Istradefylline treatment improved hair cell survival in a turn-dependent manner, whilst the cognitive function was unaffected by istradefylline treatment. This study presents the first evidence for the rescue potential of istradefylline in ARHL and highlights the role of A2AR in development of age-related cochlear degeneration.


Aging , Auditory Threshold/drug effects , Evoked Potentials, Auditory, Brain Stem/drug effects , Presbycusis , Purines/pharmacology , Animals , Male , Mice , Presbycusis/drug therapy , Presbycusis/pathology , Presbycusis/physiopathology
14.
Biomed Res Int ; 2021: 6667531, 2021.
Article En | MEDLINE | ID: mdl-34409106

The results of recent animal studies have suggested that cochlear synaptopathy may be an important factor involved in presbycusis. Therefore, here, we aimed to examine whether cochlear synaptopathy frequently exists in patients with presbycusis and to describe the effect of cochlear synaptopathy on speech recognition in noise. Based on the medical history and an audiological examination, 94 elderly patients with bilateral, symmetrical, sensorineural hearing loss were diagnosed as presbycusis. An electrocochleogram, auditory brainstem responses, auditory cortical evoked potentials, and speech audiometry were recorded to access the function of the auditory pathway. First, 65 ears with hearing levels of 41-50 dB HL were grouped based on the summating potential/action potential (SP/AP) ratio, and the amplitudes of AP and SP were compared between the two resulting groups. Second, 188 ears were divided into two groups: the normal SP/AP and abnormal SP/AP groups. The speech recognition abilities in the two groups were compared. Finally, the relationship between abnormal electrocochleogram and poor speech recognition (signal-to-noise ratio loss ≥7 dB) was analyzed in 188 ears. The results of the present study showed: (1) a remarkable reduction in the action potential amplitude was observed in patients with abnormal SP/AP ratios; this suggests that cochlear synaptopathy was involved in presbycusis. (2) There was a large proportion of patients with poor speech recognition in the abnormal SP/AP group. Furthermore, a larger number of cases with abnormal SP/AP ratios were confirmed among patients with presbycusis and poor speech recognition. We concluded that cochlear synaptopathy is not uncommon among elderly individuals who have hearing ability deficits, and it may have a more pronounced effect on ears with declining auditory performance in noisy environments.


Audiometry/methods , Cochlea/physiopathology , Presbycusis/physiopathology , Aged , Aged, 80 and over , Audiometry, Speech , Auditory Threshold , Evoked Potentials, Auditory, Brain Stem , Female , Humans , Male , Middle Aged , Speech Perception
15.
Acta Otolaryngol ; 141(6): 545-550, 2021 Jun.
Article En | MEDLINE | ID: mdl-33827362

BACKGROUND: Vestibulo-ocular reflex (VOR) function is expected to be normal in patients with presbycusis during sudden head rotations. AIM: This study aimed to determine whether presbycusis was accompanied by vestibular system pathologies. In addition, it was examined whether there was a difference existed between the patients with and without presbycusis in terms of normative data. MATERIALS AND METHODS: A total of 40 individuals were included in the study: 20 in the presbycusis group and 20 in the control group. The vestibular systems of both groups were evaluated using the video head impulse test and videonystagmography. RESULTS: The right and left lateral VOR gain values were decreased in the group with presbycusis compared to the control group. The difference between the two groups in the mean VOR gains in the right lateral canal and left lateral canal were statistically significant (p = .040 and p = .050, respectively). The air caloric tests of all individuals were found to be normal. CONCLUSIONS: This result suggests that the loss of vestibular hair cells and vestibular nerve degeneration in the lateral semicircular canal may be more severe in presbycusis than in the same age group with normal hearing.


Hair Cells, Vestibular/pathology , Presbycusis/physiopathology , Reflex, Vestibulo-Ocular , Semicircular Canals/physiology , Vestibular Diseases/complications , Vestibular Nerve/physiology , Aged , Case-Control Studies , Female , Head Impulse Test , Humans , Male , Middle Aged , Nystagmus, Pathologic/complications , Presbycusis/complications , Presbycusis/pathology , Vestibular Function Tests , Vestibular Nerve/physiopathology , Vestibule, Labyrinth
16.
BMC Biol ; 19(1): 18, 2021 02 01.
Article En | MEDLINE | ID: mdl-33526032

BACKGROUND: Age-related hearing loss (ARHL), also known as presbycusis, is the most common sensory impairment seen in elderly people. However, the cochlear aging process does not affect people uniformly, suggesting that both genetic and environmental (e.g., noise, ototoxic drugs) factors and their interaction may influence the onset and severity of ARHL. Considering the potential links between thyroid hormone, mitochondrial activity, and hearing, here, we probed the role of p43, a N-terminally truncated and ligand-binding form of the nuclear receptor TRα1, in hearing function and in the maintenance of hearing during aging in p43-/- mice through complementary approaches, including in vivo electrophysiological recording, ultrastructural assessments, biochemistry, and molecular biology. RESULTS: We found that the p43-/- mice exhibit no obvious hearing loss in juvenile stages, but that these mice developed a premature, and more severe, ARHL resulting from the loss of cochlear sensory outer and inner hair cells and degeneration of spiral ganglion neurons. Exacerbated ARHL in p43-/- mice was associated with the early occurrence of a drastic fall of SIRT1 expression, together with an imbalance between pro-apoptotic Bax, p53 expression, and anti-apoptotic Bcl2 expression, as well as an increase in mitochondrial dysfunction, oxidative stress, and inflammatory process. Finally, p43-/- mice were also more vulnerable to noise-induced hearing loss. CONCLUSIONS: These results demonstrate for the first time a requirement for p43 in the maintenance of hearing during aging and highlight the need to probe the potential link between human THRA gene polymorphisms and/or mutations and accelerated age-related deafness or some adult-onset syndromic deafness.


Aging , Presbycusis/genetics , Receptors, Thyroid Hormone/genetics , Animals , Male , Mice , Presbycusis/physiopathology , Receptors, Thyroid Hormone/metabolism
17.
Ann Otol Rhinol Laryngol ; 130(9): 1093-1099, 2021 Sep.
Article En | MEDLINE | ID: mdl-33615825

OBJECTIVE: The effect of hearing aid use on the evolution of presbycusis has not been well described in the literature, with only a handful of publications addressing this topic. This paper aims to evaluate the long-term use of amplification and its effect on pure-tone thresholds and word recognition scores. METHOD: Monaurally fitted patients were followed with serial audiograms. Data was collected from hearing aid centers. Seventy-seven patients with presbycusis met the inclusion criteria and participated in the present study. The progression of hearing loss in both pure tone thresholds and word recognition scores were compared between the hearing aid ears (HA), and the non-hearing aid ears (NHA). Pure tone thresholds were analyzed by comparing the pure tone average at the initial and last audiograms. Word Recognition Scores (WRS) were analyzed using the model of Thornton and Raffin (1978), and by comparing the change in the absolute values of WRS from the initial to the last audiogram between the HA ear and the NHA ear. RESULTS: No significant difference in pure-tone thresholds between the HA ear and NHA ear was found at the last audiogram (P = .696), even after dividing the patients into groups based on the duration of amplification. Both methods of analysis of patients' WRS showed a statistically significant worsening in NHA (P < .05). CONCLUSION: The present study supports the previously defined auditory deprivation effect on non-fitted ears, which showed worsening of word recognition over time and no effect on pure tone average. It provides an additional argument for the counseling of patients with presbycusis considering amplification, and highlights the importance of bilateral amplification in preserving the residual hearing of hearing impaired patients.


Correction of Hearing Impairment/methods , Hearing Aids , Presbycusis/rehabilitation , Speech Perception , Aged , Aged, 80 and over , Audiometry, Pure-Tone , Disease Progression , Female , Humans , Male , Middle Aged , Presbycusis/physiopathology , Time Factors
18.
Biosci Rep ; 41(2)2021 02 26.
Article En | MEDLINE | ID: mdl-33506259

The present study examined auditory function across age in the dark agouti (DA) rat strain. Auditory brainstem responses (ABRs) were measured for frequencies 8, 16, and 32 kHz in male and female DA rats from 3 to 18 months of age. Hearing thresholds and absolute and interpeak latencies (IPLs) were analyzed. Male hearing thresholds remained stable for the first year of life and then significantly increased at 18 months across all frequencies; female hearing remained stable at all tested ages out to 18 months. At 12 months, male DA rats showed significantly longer absolute latencies by age (i.e., compared with 3-month-old males) and sex (compared with 12-month-old females), with no differences in IPLs. At 18 months, female DA rats showed significantly longer absolute latencies with age (compared with 3-month-old females) and sex (compared with 18-month-old males), particularly for the later waves. Female IPLs were also significantly longer with age and by sex for the later waves. This report supports the feasibility of using male DA rats in studies to investigate age-related hearing loss (ARHL; presbycusis).


Brain Stem/physiology , Evoked Potentials, Auditory, Brain Stem , Presbycusis/physiopathology , Animals , Auditory Threshold , Cochlea/anatomy & histology , Cochlea/pathology , Cross-Sectional Studies , Disease Models, Animal , Female , Male , Rats , Reaction Time
19.
Aging Cell ; 19(12): e13275, 2020 12.
Article En | MEDLINE | ID: mdl-33222382

Aging of the auditory system is associated with the incremental production of reactive oxygen species (ROS) and the accumulation of oxidative damage in macromolecules, which contributes to cellular malfunction, compromises cell viability, and, ultimately, leads to functional decline. Cellular detoxification relies in part on the production of NADPH, which is an important cofactor for major cellular antioxidant systems. NADPH is produced principally by the housekeeping enzyme glucose-6-phosphate dehydrogenase (G6PD), which catalyzes the rate-limiting step in the pentose phosphate pathway. We show here that G6PD transgenic mice (G6PD-Tg), which show enhanced constitutive G6PD activity and NADPH production along life, have lower auditory thresholds than wild-type mice during aging, together with preserved inner hair cell (IHC) and outer hair cell (OHC), OHC innervation, and a conserved number of synapses per IHC. Gene expression of antioxidant enzymes was higher in 3-month-old G6PD-Tg mice than in wild-type counterparts, whereas the levels of pro-apoptotic proteins were lower. Consequently, nitration of proteins, mitochondrial damage, and TUNEL+ apoptotic cells were all lower in 9-month-old G6PD-Tg than in wild-type counterparts. Unexpectedly, G6PD overexpression triggered low-grade inflammation that was effectively resolved in young mice, as shown by the absence of cochlear cellular damage and macrophage infiltration. Our results lead us to propose that NADPH overproduction from an early stage is an efficient mechanism to maintain the balance between the production of ROS and cellular detoxification power along aging and thus prevents hearing loss progression.


Aging/metabolism , Glucosephosphate Dehydrogenase/genetics , Glucosephosphate Dehydrogenase/metabolism , Presbycusis/enzymology , Presbycusis/prevention & control , Aging/physiology , Animals , Apoptosis , Auditory Threshold/physiology , Cochlea/metabolism , Cochlea/pathology , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NADP/biosynthesis , Oxidative Stress , Presbycusis/physiopathology , Reactive Oxygen Species/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Up-Regulation
20.
Int J Mol Sci ; 21(19)2020 Sep 29.
Article En | MEDLINE | ID: mdl-33003463

Aging is associated with functional and morphological changes in the sensory organs, including the auditory system. Mitophagy, a process that regulates the turnover of dysfunctional mitochondria, is impaired with aging. This study aimed to investigate the effect of aging on mitophagy in the central auditory system using an age-related hearing loss mouse model. C57BL/6J mice were divided into the following four groups based on age: 1-, 6-, 12-, and 18-month groups. The hearing ability was evaluated by measuring the auditory brainstem response (ABR) thresholds. The mitochondrial DNA damage level and the expression of mitophagy-related genes, and proteins were investigated by real-time polymerase chain reaction and Western blot analyses. The colocalization of mitophagosomes and lysosomes in the mouse auditory cortex and inferior colliculus was analyzed by immunofluorescence analysis. The expression of genes involved in mitophagy, such as PINK1, Parkin, and BNIP3 in the mouse auditory cortex and inferior colliculus, was investigated by immunohistochemical staining. The ABR threshold increased with aging. In addition to the mitochondrial DNA integrity, the mRNA levels of PINK1, Parkin, NIX, and BNIP3, as well as the protein levels of PINK1, Parkin, BNIP3, COX4, LC3B, mitochondrial oxidative phosphorylation (OXPHOS) subunits I-IV in the mouse auditory cortex significantly decreased with aging. The immunofluorescence analysis revealed that the colocalization of mitophagosomes and lysosomes in the mouse auditory cortex and inferior colliculus decreased with aging. The immunohistochemical analysis revealed that the expression of PINK1, Parkin, and BNIP3 decreased in the mouse auditory cortex and inferior colliculus with aging. These findings indicate that aging-associated impaired mitophagy may contribute to the cellular changes observed in an aged central auditory system, which result in age-related hearing loss. Thus, the induction of mitophagy can be a potential therapeutic strategy for age-related hearing loss.


Aging/genetics , Mitochondria/genetics , Mitophagy/genetics , Presbycusis/genetics , Aging/pathology , Animals , Auditory Diseases, Central/genetics , Auditory Diseases, Central/physiopathology , DNA, Mitochondrial/genetics , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/genetics , Evoked Potentials, Auditory, Brain Stem/physiology , Lysosomes/genetics , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Oxidative Phosphorylation , Presbycusis/physiopathology
...