Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38915602

ABSTRACT

The vestibular short-latency evoked potential (VsEP) reflects the activity of irregular vestibular afferents and their target neurons in the brain stem. Attenuation of trial-averaged VsEP waveforms is widely accepted as an indicator of vestibular dysfunction, however, more quantitative analyses of VsEP waveforms could reveal underlying neural properties of VsEP waveforms. Here, we present a time-frequency analysis of the VsEP with a wavelet transform on a single-trial basis, which allows us to examine trial-by-trial variability in the strength of VsEP waves as well as their temporal coherence across trials. Using this method, we examined changes in the VsEP following 110 dB SPL noise exposure in rats. We found detectability of head jerks based on the power of wavelet transform coefficients was significantly reduced 1 day after noise exposure but recovered nearly to pre-exposure level in 3 - 7 days and completely by 28 days after exposure. Temporal coherence of VsEP waves across trials was also significantly reduced on 1 day after exposure but recovered with a similar time course. Additionally, we found a significant reduction in the number of calretinin-positive calyces in the sacculi collected 28 days after noise exposure. Furthermore, the number of calretinin-positive calyces was significantly correlated with the degree of reduction in temporal coherence and/or signal detectability of the smallest-amplitude jerks. This new analysis of the VsEP provides more quantitative descriptions of noise-induced changes as well as new insights into potential mechanisms underlying noise-induced vestibular dysfunction. Significance Statement: Our study presents a new method of VsEP quantification using wavelet transform on a single-trial basis. It also describes a novel approach to determine the stimulus threshold of the VsEP based on signal-detection theory and Rayleigh statistics. The present analysis could also be applied to analysis of auditory brain stem response (ABR). Thus, it has the potential to provide new insights into the physiological properties that underlie peripheral vestibular and auditory dysfunction.

2.
Hear Res ; 424: 108601, 2022 10.
Article in English | MEDLINE | ID: mdl-36126618

ABSTRACT

Many factors contribute to hearing loss commonly found in older adults. There can be natural aging of cellular elements, hearing loss previously induced by environmental factors such as noise or ototoxic drugs as well as genetic and epigenetic influences. Even when noise overstimulation does not immediately cause permanent hearing loss it has recently been shown to increase later age-related hearing loss (ARHL). The present study further investigated this condition in the UMHET4 mouse model by comparing a small arms fire (SAF)-like impulse noise exposure that has the greatest immediate effect in more apical cochlear regions to a broadband noise (BBN) exposure that has the greatest immediate effect in more basal cochlear regions. Both noise exposures were given at levels that only induced temporary auditory brainstem response (ABR) threshold shifts (TS). Mice were noise exposed at 5 months of age followed by ABR assessment at 6, 12, 18, 21, and 24 months of age. Mice that received the SAF-like impulse noise had accelerated age-related TS at 4 kHz that appeared at 12 months of age (significantly increased compared to no-noise controls). This increased TS at 4 kHz continued at 18 and 21 months but was no longer significantly greater at 24 months of age. The SAF-like impulse noise also induced a significantly greater mean TS at 48 kHz, first appearing at 18 months of age and continuing to be significantly greater than controls at 21 and 24 months. The BBN induced a different pace and pattern of enhanced age-related ABR TS. The mean TS for the BBN group first became significantly greater than controls at 18 months of age and only at 48 kHz. It remained significantly greater than controls at 21 months but was no longer significantly greater at 24 months of age. Results, therefore, show different influences on ARHL for the two different noise exposure conditions. Noise-induced enhancement appears to provide more an acceleration than overall total increase in ARHL.


Subject(s)
Hearing Loss, Noise-Induced , Presbycusis , Animals , Auditory Threshold/physiology , Cochlea , Evoked Potentials, Auditory, Brain Stem/physiology , Hearing Loss, Noise-Induced/genetics , Mice , Noise/adverse effects , Presbycusis/genetics
3.
Tissue Cell ; 73: 101599, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34371293

ABSTRACT

Slc44a2 is reported to interact with tetraspanins CD9 and CD81. To investigate how Slc44a2 affects adhesion protein expression, cells from wild-type (WT) Slc44a2+/+, heterozygous (HET) Slc44a2+/-, and knockout (KO) Slc44a2-/- mice were cultured from lung tissue. The cultured cells expressed vimentin, N-cadherin, p120 catenin, beta-catenin, actin, CD9, and CD81, but not E-cadherin. Vimentin expression with lack of E-cadherin indicated that the cultured cells were of mesenchymal origin. Slc44a2 KO cells and HET cells demonstrated lower adherence and faster proliferation than the WT cells. All three groups displayed dramatically altered intracellular distribution of N-cadherin, CD9, and CD81. The CD9 membrane foci observed in WT cell membranes were less frequent and diminished in size in HET cells and KO cells. N-cadherin was dispersed throughout both the cytoplasm and membrane in WT cells, with similar yet weaker distribution in HET cells; however, in KO cells, N-cadherin was densely aggregated in the perinuclear cytoplasm. CD81 had a distribution pattern in WT, HET, and KO cells similar to that of N-cadherin with dense cytoplasmic clusters in the cells. KO cells also exhibited reduced filamentous actin as compared to WT cells. These results suggest that Slc44a2 is necessary for proper cellular localization of adhesion proteins and growth regulation that may be related to altered adhesion signals.


Subject(s)
Cadherins/metabolism , Gene Deletion , Lung/cytology , Membrane Transport Proteins/genetics , Mesoderm/cytology , Tetraspanins/metabolism , Animals , Catenins/metabolism , Cell Adhesion , Cell Proliferation , Cells, Cultured , Genotype , Heterozygote , Membrane Transport Proteins/metabolism , Mice, Knockout , beta Catenin/metabolism , Delta Catenin
4.
Front Cell Neurosci ; 15: 658972, 2021.
Article in English | MEDLINE | ID: mdl-33897373

ABSTRACT

Our previous study demonstrated rapamycin added to diet at 4 months of age had significantly less age-related outer hair cell loss in the basal half of the cochlea at 22 months of age compared to mice without rapamycin. The present study tested adding rapamycin to diet later in life, at 14 months of age, and added a longitudinal assessment of auditory brain stem response (ABR). The present study used UMHET4 mice, a 4 way cross in which all grandparental strains lack the Cdh23753A allele that predisposes to early onset, progressive hearing loss. UMHET4 mice typically have normal hearing until 16-17 months, then exhibit threshold shifts at low frequencies/apical cochlea and later in more basal high frequency regions. ABR thresholds at 4, 12, 24, and 48 kHz were assessed at 12, 18, and 24 months of age and compared to baseline ABR thresholds acquired at 5 months of age to determine threshold shifts (TS). There was no TS at 12 months of age at any frequency tested. At 18 months of age mice with rapamycin added to diet at 14 months had a significantly lower mean TS at 4 and 12 kHz compared to mice on control diet with no significant difference at 24 and 48 kHz. At 24 months of age, the mean 4 kHz TS in rapamycin diet group was no longer significantly lower than the control diet group, while the 12 kHz mean remained significantly lower. Mean TS at 24 and 48 kHz in the rapamycin diet group became significantly lower than in the control diet group at 24 months. Hair cell counts at 24 months showed large loss in the apical half of most rapamycin and control diet mice cochleae with no significant difference between groups. There was only mild outer hair cell loss in the basal half of rapamycin and control diet mice cochleae with no significant difference between groups. The results show that a later life addition of rapamycin can decrease age-related hearing loss in the mouse model, however, it also suggests that this decrease is a delay/deceleration rather than a complete prevention.

5.
Mil Med ; 185(Suppl 1): 454-461, 2020 01 07.
Article in English | MEDLINE | ID: mdl-32074366

ABSTRACT

INTRODUCTION: The vestibular system is essential for normal postural control and balance. Because of their proximity to the cochlea, the otolith organs are vulnerable to noise. We previously showed that head jerks that evoke vestibular nerve activity were no longer capable of inducing a response after noise overstimulation. The present study adds a greater range of jerk intensities to determine if the response was abolished or required more intense stimulation (threshold shift). MATERIALS AND METHODS: Vestibular short-latency evoked potential (VsEP) measurements were taken before noise exposure and compared to repeated measurements taken at specific time points for 28 days after noise exposure. Calretinin was used to identify changes in calyx-only afferents in the sacculus. RESULTS: Results showed that more intense jerk stimuli could generate a VsEP, although it was severely attenuated relative to prenoise values. When the VsEP was evaluated 4 weeks after noise exposure, partial recovery was observed. CONCLUSION: These data suggest that noise overstimulation, such as can occur in the military, could introduce an increased risk of imbalance that should be evaluated before returning a subject to situations that require normal agility and motion. Moreover, although there is recovery with time, some dysfunction persists for extended periods.


Subject(s)
Bilateral Vestibulopathy/etiology , Noise/adverse effects , Animals , Bilateral Vestibulopathy/pathology , Disease Models, Animal , Environmental Exposure/adverse effects , Evoked Potentials, Auditory/physiology , Evoked Potentials, Auditory, Brain Stem , Rats, Inbred LEC/injuries
6.
Neuroscience ; 407: 32-40, 2019 05 21.
Article in English | MEDLINE | ID: mdl-30053484

ABSTRACT

A noise-induced loss of inner hair cell (IHC) - auditory nerve synaptic connections has been suggested as a factor that can trigger the progression of maladaptive plastic changes leading to noise-induced tinnitus. The present study used a military relevant small arms fire (SAF)-like noise (50 biphasic impulses over 2.5 min at 152 dB SPL given unilaterally to the right ear) to induce loss (∼1/3) of IHC synaptic ribbons (associated with synapse loss) in rat cochleae with only minor (less than 10%) loss of outer hair cells. Approximately half of the noise-exposed rats showed poorer Gap Detection post-noise, a behavioral indication suggesting the presence of tinnitus. There was significantly greater loss of IHC ribbons in noise-exposed rats with reduced Gap Detection compared to noise-exposed rats retaining normal Gap Detection. We have previously shown systemic administration of piribedil, memantine, and/or ACEMg significantly reduced loss of IHC ribbons induced by a 3 h 4 kHz octave band 117 dB (SPL) noise. The present study examined if this treatment would also reduce ribbon loss from the SAF-like noise exposure and if this would prevent the reduced Gap Detection. As in the previous study, piribedil, memantine, and ACEMg treatment significantly reduced the noise-induced loss of ribbons, such that it was no longer significantly different from normal. However, it did not prevent development of the reduced Gap Detection indication of tinnitus in all treated noise-exposed rats, reducing the incidence but not reaching significance.


Subject(s)
Auditory Threshold/physiology , Deafness/physiopathology , Hair Cells, Auditory, Outer/physiology , Hearing Loss, Noise-Induced/physiopathology , Animals , Evoked Potentials, Auditory, Brain Stem/physiology , Male , Noise , Rats, Sprague-Dawley
7.
J Neurophysiol ; 119(2): 662-667, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29118200

ABSTRACT

The vestibular system plays a critical role in detection of head movements and is essential for normal postural control. Because of their anatomical proximity to the cochlea, the otolith organs are selectively exposed to sound pressure and are at risk for noise overstimulation. Clinical reports suggest a link between noise exposure and balance problems, but the structural and physiological basis for this linkage is not well understood. The goal of this study was to determine the effects of low-frequency noise (LFN) on the otolith organs by correlating changes in vestibular short-latency evoked potentials (VsEPs) with changes in saccular afferent endings following noise exposure. LFN exposure transiently abolished the VsEP and reduced the number of stained calyces within the sacculus. Although some recovery of the VsEP waveform could be observed within 3 days after noise, at 3 wk recovery was only partial in most animals, consistent with a reduced number of afferents with calyceal endings. These data show that a single intense noise exposure is capable of causing a vestibular deficit that appears to mirror the synaptic deficit associated with hidden hearing loss after noise-induced cochlear injury. NEW & NOTEWORTHY This is the first study to explore the effects of low-frequency high-intensity noise on vestibular short-latency evoked potential (VsEP) responses, which shows a linkage between attenuated noise-induced VsEPs and pathological changes to otolith organ afferents. This finding suggests a potential limitation of the VsEP for evaluation of vestibular dysfunction, since the VsEP measurement may assess the activity of a specific class rather than all afferents.


Subject(s)
Noise/adverse effects , Saccule and Utricle/radiation effects , Vestibular Evoked Myogenic Potentials , Animals , Male , Rats , Rats, Sprague-Dawley , Reaction Time , Saccule and Utricle/physiology
8.
Neuroscience ; 332: 242-57, 2016 09 22.
Article in English | MEDLINE | ID: mdl-27403879

ABSTRACT

In experimental animal models of auditory hair cell (HC) loss, insults such as noise or ototoxic drugs often lead to secondary changes or degeneration in non-sensory cells and neural components, including reduced density of spiral ganglion neurons, demyelination of auditory nerve fibers and altered cell numbers and innervation patterns in the cochlear nucleus (CN). However, it is not clear whether loss of HCs alone leads to secondary degeneration in these neural components of the auditory pathway. To elucidate this issue, we investigated changes of central components after cochlear insults specific to HCs using diphtheria toxin receptor (DTR) mice expressing DTR only in HCs and exhibiting complete HC loss when injected with diphtheria toxin (DT). We showed that DT-induced HC ablation has no significant impacts on the survival of auditory neurons, central synaptic terminals, and myelin, despite complete HC loss and profound deafness. In contrast, noise exposure induced significant changes in synapses, myelin and CN organization even without loss of inner HCs. We observed a decrease of neuronal size in the auditory pathway, including peripheral axons, spiral ganglion neurons, and CN neurons, likely due to loss of input from the cochlea. Taken together, selective HC ablation and noise exposure showed different patterns of pathology in the auditory pathway and the presence of HCs is not essential for the maintenance of central synaptic connectivity and myelination.


Subject(s)
Auditory Pathways/pathology , Cochlea/pathology , Cochlear Nucleus/pathology , Hair Cells, Auditory/pathology , Hearing Loss, Noise-Induced/pathology , Noise/adverse effects , Animals , Auditory Pathways/metabolism , Cell Size , Cell Survival , Cochlea/metabolism , Cochlear Nucleus/metabolism , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem , Female , Hair Cells, Auditory/metabolism , Hearing Loss, Noise-Induced/metabolism , Immunohistochemistry , Male , Mice, Transgenic , Microscopy, Electron, Transmission , Receptors, AMPA/metabolism , Vesicular Glutamate Transport Protein 1/metabolism
9.
J Assoc Res Otolaryngol ; 16(6): 695-712, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26463873

ABSTRACT

SLC44A2 (solute carrier 44a2), also known as CTL2 (choline transporter-like protein 2), is expressed in many supporting cell types in the cochlea and is implicated in hair cell survival and antibody-induced hearing loss. In mice with the mixed C57BL/6-129 background, homozygous deletion of Slc44a2 exons 3­10 (Slc44a2(Δ/Δ)resulted in high-frequency hearing loss and hair cell death. To reduce effects associated with age-related hearing loss (ARHL) in these strains, mice carrying the Slc44a2Δ allele were backcrossed to the ARHL-resistant FVB/NJ strain and evaluated after backcross seven(N7) (99 % FVB). Slc44a2(Δ/Δ) mice produced abnormally spliced Slc44a2 transcripts that contain a frame shift and premature stop codons. Neither full-length SLC44A2 nor a putative truncated protein could be detected in Slc44a2(Δ/Δ) mice, suggesting a likely null allele. Auditory brain stem responses (ABRs) of mice carrying the Slc44a2Δ allele on an FVB/NJ genetic background were tested longitudinally between the ages of 2 and 10 months. By 6 months of age,Slc44a2(Δ/Δ) mice exhibited hearing loss at 32 kHz,but at 12 and 24 kHz had sound thresholds similar to those of wild-type Slc44a2(+/+) and heterozygous +/Slc44a2Δ mice. After 6 months of age, Slc44a2(Δ/Δ) mutants exhibited progressive hearing loss at all frequencies and +/Slc44a2(Δ) mice exhibited moderate threshold elevations at high frequency. Histologic evaluation of Slc44a2(Δ/Δ) mice revealed extensive hair cell and spiral ganglion cell loss, especially in the basal turn of the cochlea. We conclude that Slc44a2 function is required for long-term hair cell survival and maintenance of hearing.


Subject(s)
Hair Cells, Auditory/pathology , Hearing Loss, Sensorineural/genetics , Membrane Transport Proteins/genetics , Spiral Ganglion/pathology , Amino Acid Sequence , Animals , Female , Gene Deletion , Hearing Loss, Sensorineural/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data
10.
Development ; 139(24): 4666-74, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23172918

ABSTRACT

This study is the first to demonstrate that macrophage migration inhibitory factor (MIF), an immune system 'inflammatory' cytokine that is released by the developing otocyst, plays a role in regulating early innervation of the mouse and chick inner ear. We demonstrate that MIF is a major bioactive component of the previously uncharacterized otocyst-derived factor, which directs initial neurite outgrowth from the statoacoustic ganglion (SAG) to the developing inner ear. Recombinant MIF acts as a neurotrophin in promoting both SAG directional neurite outgrowth and neuronal survival and is expressed in both the developing and mature inner ear of chick and mouse. A MIF receptor, CD74, is found on both embryonic SAG neurons and adult mouse spiral ganglion neurons. Mif knockout mice are hearing impaired and demonstrate altered innervation to the organ of Corti, as well as fewer sensory hair cells. Furthermore, mouse embryonic stem cells become neuron-like when exposed to picomolar levels of MIF, suggesting the general importance of this cytokine in neural development.


Subject(s)
Ear, Inner/embryology , Intramolecular Oxidoreductases/physiology , Macrophage Migration-Inhibitory Factors/physiology , Nerve Growth Factors/physiology , Animals , Animals, Newborn , Cell Survival/drug effects , Cells, Cultured , Chick Embryo , Ear, Inner/drug effects , Ear, Inner/growth & development , Ear, Inner/metabolism , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Intramolecular Oxidoreductases/pharmacology , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/metabolism , Macrophage Migration-Inhibitory Factors/pharmacology , Mice , Mice, Knockout , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nerve Growth Factors/pharmacology , Neurites/drug effects , Neurites/physiology , Neurons/cytology , Neurons/drug effects , Neurons/physiology , Organ of Corti/embryology , Organ of Corti/growth & development , Organ of Corti/metabolism , Spiral Ganglion/embryology , Spiral Ganglion/growth & development , Spiral Ganglion/metabolism
11.
PLoS One ; 7(12): e53280, 2012.
Article in English | MEDLINE | ID: mdl-23285273

ABSTRACT

Cyclodextrins are sugar compounds that are increasingly finding medicinal uses due to their ability to complex with hydrophobic molecules. One cyclodextrin in particular, 2-hydroxypropyl-ß-cyclodextrin (HPßCD), is used as a carrier to solubilize lipophilic drugs and is itself being considered as a therapeutic agent for treatment of Niemann-Pick Type C disease, due to its ability to mobilize cholesterol. Results from toxicological studies suggest that HPßCD is generally safe, but a recent study has found that it causes hearing loss in cats. Whether the hearing loss occurred via death of cochlear hair cells, rendering it permanent, was unexplored. In the present study, we examined peripheral auditory function and cochlear histology in mice after subcutaneous injection of HPßCD to test for hearing loss and correlate any observed auditory deficits with histological findings. On average, auditory brainstem response thresholds were elevated at 4, 16, and 32 kHz in mice one week after treatment with 8,000 mg/kg. In severely affected mice all outer hair cells were missing in the basal half of the cochlea. In many cases, surviving hair cells in the cochlear apex exhibited abnormal punctate distribution of the motor protein prestin, suggesting long term changes to membrane composition and integrity. Mice given a lower dose of 4,000 mg/kg exhibited hearing loss only after repeated doses, but these threshold shifts were temporary. Therefore, cyclodextrin-induced hearing loss was complex, involving cell death and other more subtle influences on cochlear physiology.


Subject(s)
Anticholesteremic Agents/adverse effects , Hair Cells, Auditory/drug effects , Hearing Loss/chemically induced , beta-Cyclodextrins/adverse effects , 2-Hydroxypropyl-beta-cyclodextrin , Animals , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/pharmacology , Cell Death/drug effects , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Evaluation, Preclinical , Hair Cells, Auditory/physiology , Hearing Loss/pathology , Infusions, Parenteral , Mice , beta-Cyclodextrins/administration & dosage , beta-Cyclodextrins/pharmacology
12.
Mol Cell Neurosci ; 49(2): 104-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22122823

ABSTRACT

The auditory sensory epithelium in non-mammalian vertebrates can replace lost hair cells by transdifferentiation of supporting cells, but this regenerative ability is lost in the mammalian cochlea. Future cell-based treatment of hearing loss may depend on stem cell transplantation or on transdifferentiation of endogenous cells in the cochlea. For both approaches, identification of cells with stem cell features within the mature cochlea may be useful. Here we use a Nestin-ß-gal mouse to examine the presence of Nestin positive cells in the mature auditory epithelium, and determine how overstimulation of the ear impacts these cells. Nestin positive cells were found in the apical turn of the cochlea lateral to the outer hair cell area. This pattern of expression persisted into mature age. The area of Nestin positive cells was increased after the noise lesion. This increase in area coincided with an increase in expression of the Nestin mRNA. The data suggest that cells with potential stem cell features remain in the mature mammalian cochlea, restricted to the apical turn, and that an additional set of signals is necessary to trigger their contribution to cell replacement therapy in the ear. As such, this population of cells could serve to generate cochlear stem cells for research and potential therapy, and may be a target for treatments based on induced transdifferentiation of endogenous cochlear cells.


Subject(s)
Cell Differentiation , Cell Transdifferentiation/physiology , Cochlea/cytology , Intermediate Filament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Organ of Corti/metabolism , Stem Cells/metabolism , Animals , Cell Proliferation , Cochlea/metabolism , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Mice , Nestin , Noise , Organ of Corti/cytology , Rats
13.
Neurobiol Aging ; 33(5): 1010.e15-6, 2012 May.
Article in English | MEDLINE | ID: mdl-22154190

ABSTRACT

Oxidative stress has been linked to noise- and drug-induced as well as age-related hearing loss. Antioxidants can attenuate the decline of cochlear structure and function after exposure to noise or drugs, but it is debated as to whether they can protect from age-related hearing loss. In a long-term longitudinal study, 10-month-old female CBA/J mice were placed on either a control or antioxidant-enriched diet and monitored through 24 months of age. Supplementation with vitamins A, C, and E, L-carnitine, and α-lipoic acid significantly increased the antioxidant capacity of inner ear tissues. However, by 24 months of age, the magnitude of hearing loss was equal between the two groups. Likewise, there were no significant differences in hair cell loss or degeneration of spiral ganglion cells. We conclude that dietary manipulations can alter cochlear antioxidant capacity but do not ameliorate age-related sensorineural hearing loss in the CBA/J mouse.


Subject(s)
Antioxidants/pharmacology , Dietary Supplements , Oxidative Stress/drug effects , Presbycusis/diet therapy , Presbycusis/prevention & control , Animals , Disease Models, Animal , Disease Progression , Female , Longitudinal Studies , Mice , Mice, Inbred CBA , Oxidative Stress/physiology
14.
Hear Res ; 243(1-2): 87-94, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18573325

ABSTRACT

Commercially obtained aged male CBA/J mice presented a complex pattern of hearing loss and morphological changes. A significant threshold shift in auditory brainstem responses (ABR) occurred at 3 months of age at 4 kHz without apparent loss of hair cells, rising slowly at later ages accompanied by loss of apical hair cells. A delayed high-frequency deficit started at 24 kHz around the age of 12 months. At 20-26 months, threshold shifts at 12 and 24 kHz and the accompanying hair cell loss at the base of the cochlea were highly variable with some animals appearing almost normal and others showing large deficits. Spiral ganglion cells degenerated by 18 months in all regions of the cochlea, with cell density reduced by approximately 25%. There was no degeneration of the stria vascularis and the endocochlear potential remained stable from 3 to 25 months of age regardless of whether the animals had normal or highly elevated ABR thresholds. The slow high-frequency hearing loss combined with a modest reduction of ganglion cell density and an unchanged endocochlear potential suggest sensorineural presbycusis. The superimposed early hearing loss at low frequencies, which is not seen in animals bred in-house, may complicate the use of these animals as a presbycusis model.


Subject(s)
Aging/pathology , Presbycusis/pathology , Animals , Auditory Threshold , Cochlea/pathology , Cochlear Microphonic Potentials , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem , Hair Cells, Auditory/pathology , Hearing Loss, High-Frequency/etiology , Hearing Loss, High-Frequency/pathology , Hearing Loss, High-Frequency/physiopathology , Male , Mice , Mice, Inbred CBA , Presbycusis/etiology , Presbycusis/physiopathology , Spiral Ganglion/pathology , Stria Vascularis/pathology
15.
Brain Res Mol Brain Res ; 130(1-2): 134-48, 2004 Nov 04.
Article in English | MEDLINE | ID: mdl-15519684

ABSTRACT

In mammals, exposure to intense noise produces a permanent hearing loss called permanent threshold shift (PTS), whereas a moderate noise produces only a temporary threshold shift (TTS). Little is known about the molecular responses to such high intensity noise exposures. In this study we used gene arrays to examine the early response to acoustic overstimulation in the rat cochlea. We compared cochlear RNA from noise-exposed rats with RNA from unexposed controls. The intense PTS noise induced several immediate early genes encoding both transcription factors (c-FOS, EGR1, NUR77/TR3) and cytokines (PC3/BTG2, LIF and IP10). In contrast, the TTS noise down-regulated the gene for growth hormone. The response of these genes to different noise intensities was examined by quantitative RT-PCR 2.5 h after the 90-min noise exposure. For most genes, the extent of induction correlates with the intensity of the noise exposure. Three proteins (EGR1, NUR77/TR3, and IP10) were detected in many regions of the unexposed cochlea. After exposure to 120 dB noise, these proteins were present at higher levels or showed extended expression in additional regions of the cochlea. LIF was undetectable in the cochlea of unexposed rats, but could be seen in the organ of Corti and spiral ganglion neurons following noise. NUR77/TR3 was a nuclear protein before noise, but following noise translocated to the cytoplasm. These studies provide new insights into the molecular response to noise overstimulation in the mammalian cochlea.


Subject(s)
Acoustic Stimulation , Cochlea/radiation effects , Gene Expression/radiation effects , Genes, Immediate-Early/physiology , Immediate-Early Proteins/metabolism , Noise , Animals , Autoradiography/methods , Cochlea/anatomy & histology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels , Gene Expression Profiling/methods , Gene Expression Regulation/radiation effects , Immediate-Early Proteins/genetics , Immunohistochemistry/methods , Leukemia Inhibitory Factor Receptor alpha Subunit , Male , Nuclear Receptor Subfamily 4, Group A, Member 1 , Oligonucleotide Array Sequence Analysis/methods , Potassium Channels/metabolism , Potassium Channels, Voltage-Gated , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Receptors, Cytoplasmic and Nuclear , Receptors, OSM-LIF , Receptors, Steroid , Reverse Transcriptase Polymerase Chain Reaction/methods , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Hear Res ; 188(1-2): 1-11, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14759565

ABSTRACT

The genes for heat shock proteins (Hsps) can be upregulated in response to cellular trauma, resulting in enhanced cell survival and protection. Hsp32, also known as heme oxygenase 1, catalyzes the degradation of heme to produce carbon monoxide and bilirubin, which play a variety of cytoprotective functions at physiological concentrations, and iron, which is rapidly sequestered by the iron-binding protein ferritin. In the present study we examined the expression and localization of Hsp32 in the rat cochlea after heat shock using semi-quantitative reverse transcription polymerase chain reaction (RT-PCR), Western blot, and immunocytochemistry. Low levels of constitutive Hsp32 expression were observed in the normal rat cochlea by RT-PCR and Western blot. Hsp32 mRNA (messenger RNA) was present at higher levels in a subfraction containing sensorineural epithelium and lateral wall than in a subfraction containing modiolus. Western blot revealed that Hsp32 protein levels increase in the rat cochlea following heat shock. Immunocytochemistry showed scattered staining of outer hair cells in the organ of Corti of normal untreated rats. Following heat shock Hsp32 is upregulated in outer hair cells and the cells of the stria vascularis. These results suggest a potential role for Hsp32 as a component of the oxidative stress response pathway in the rat cochlea.


Subject(s)
Cochlea/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Hot Temperature/adverse effects , Oxygenases/genetics , Oxygenases/metabolism , Animals , Blotting, Western , DNA, Complementary/analysis , Hair Cells, Auditory/metabolism , Heme Oxygenase (Decyclizing) , Hyperthermia, Induced , Immunohistochemistry , Organ of Corti/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Stria Vascularis/metabolism
17.
Hear Res ; 173(1-2): 109-18, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12372640

ABSTRACT

Activation of heat shock factors (Hsfs) is one of the potential mechanisms for regulating the transcription of the heat shock proteins (Hsps) and certain other stress-responsive genes. Reverse transcription polymerase chain reaction (RT-PCR), Western blot and immunocytochemistry were used to examine the expression and localization of Hsf1, the stress-responsive member of the Hsf family, in the rat and mouse cochlea. Cerebellum was used as a positive control. Semi-quantitative RT-PCR of cochlear RNA revealed that Hsf1 was more highly expressed in a subfraction containing sensorineural epithelium and lateral wall than in a subfraction containing modiolus, with the alpha splice form predominant over the beta in both subfractions. Immunocytochemistry showed selective staining in the rodent cochlea. Hsf1 immunostaining was found in the nuclei of inner and outer hair cells in the organ of Corti, spiral ganglion cells in the modiolus, and cells in the marginal and intermediate layers of the stria vascularis. This is largely consistent with where Hsp70 induction is reported. Hsf1 activation following heat shock was examined by Western blot. Hyperthermia resulted in stress-induced Hsf1 hyperphosphorylation in cochlea as well as cerebellum. This hyperphosphorylation as well as the correlation of its localization with Hsp70 induction supports a role for Hsf1 in the cochlear stress response.


Subject(s)
Cochlea/metabolism , DNA-Binding Proteins/metabolism , Mice/metabolism , Rats/metabolism , Animals , Blotting, Western , Heat Shock Transcription Factors , Immunohistochemistry , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...