Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Hear Res ; 374: 1-4, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30669034

ABSTRACT

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


Subject(s)
Auditory Threshold/physiology , Mutation , NAV1.6 Voltage-Gated Sodium Channel/deficiency , NAV1.6 Voltage-Gated Sodium Channel/genetics , Acoustic Stimulation , Animals , Audiometry, Pure-Tone , Cochlear Nucleus/physiopathology , Hearing Tests , Homozygote , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Mutant Strains , NAV1.6 Voltage-Gated Sodium Channel/physiology , Neural Conduction/genetics , Neural Conduction/physiology , Ranvier's Nodes/physiology , Time Factors
2.
Hear Res ; 343: 14-33, 2017 01.
Article in English | MEDLINE | ID: mdl-27473502

ABSTRACT

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


Subject(s)
Cochlear Nerve/ultrastructure , Cochlear Nucleus/ultrastructure , Hearing Loss/pathology , Hearing , Synapses/ultrastructure , Acoustic Stimulation , Age Factors , Animals , Auditory Threshold , Behavior, Animal , Cochlear Nerve/physiopathology , Cochlear Nucleus/physiopathology , Disease Models, Animal , Disease Progression , Evoked Potentials, Auditory, Brain Stem , Female , Genetic Predisposition to Disease , Hearing/genetics , Hearing Loss/genetics , Hearing Loss/physiopathology , Hearing Loss/psychology , Male , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred DBA , Mice, Knockout , Microscopy, Electron, Transmission , Myosins/deficiency , Myosins/genetics , Phenotype , Severity of Illness Index , Time Factors
3.
Med Hypotheses ; 98: 2-4, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28012597

ABSTRACT

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


Subject(s)
Cervical Vertebrae/pathology , Tinnitus/complications , Tinnitus/physiopathology , Acoustic Stimulation , Animals , Cochlear Nucleus/physiopathology , Hemodynamics , Humans , Models, Neurological , Models, Theoretical , Nerve Net , Neuronal Plasticity/physiology , Neurons/metabolism , Positron-Emission Tomography , Serotonin/metabolism , Signal Transduction , Synaptic Transmission , Trigeminal Nerve/physiopathology
4.
J Neurophysiol ; 112(12): 3197-208, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25231612

ABSTRACT

Atypical medial olivocochlear (MOC) feedback from brain stem to cochlea has been proposed to play a role in tinnitus, but even well-constructed tests of this idea have yielded inconsistent results. In the present study, it was hypothesized that low sound tolerance (mild to moderate hyperacusis), which can accompany tinnitus or occur on its own, might contribute to the inconsistency. Sound-level tolerance (SLT) was assessed in subjects (all men) with clinically normal or near-normal thresholds to form threshold-, age-, and sex-matched groups: 1) no tinnitus/high SLT, 2) no tinnitus/low SLT, 3) tinnitus/high SLT, and 4) tinnitus/low SLT. MOC function was measured from the ear canal as the change in magnitude of distortion-product otoacoustic emissions (DPOAE) elicited by broadband noise presented to the contralateral ear. The noise reduced DPOAE magnitude in all groups ("contralateral suppression"), but significantly more reduction occurred in groups with tinnitus and/or low SLT, indicating hyperresponsiveness of the MOC system compared with the group with no tinnitus/high SLT. The results suggest hyperresponsiveness of the interneurons of the MOC system residing in the cochlear nucleus and/or MOC neurons themselves. The present data, combined with previous human and animal data, indicate that neural pathways involving every major division of the cochlear nucleus manifest hyperactivity and/or hyperresponsiveness in tinnitus and/or low SLT. The overactivation may develop in each pathway separately. However, a more parsimonious hypothesis is that top-down neuromodulation is the driving force behind ubiquitous overactivation of the auditory brain stem and may correspond to attentional spotlighting on the auditory domain in tinnitus and hyperacusis.


Subject(s)
Auditory Perception/physiology , Cochlear Nucleus/physiopathology , Hearing/physiology , Hyperacusis/physiopathology , Superior Olivary Complex/physiopathology , Tinnitus/physiopathology , Acoustic Stimulation , Adult , Humans , Male , Middle Aged , Noise , Reflex , Stapedius/physiology
5.
Hear Res ; 295: 124-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22349094

ABSTRACT

Hyperactivity in the form of increased spontaneous firing rates of single neurons develops in the guinea pig inferior colliculus (IC) after unilateral loud sound exposures that result in behavioural signs of tinnitus. The hyperactivity is found in those parts of the topographic frequency map in the IC where neurons possess characteristic frequencies (CFs) closely related to the region in the cochlea where lasting sensitivity changes occur as a result of the loud sound exposure. The observed hyperactivity could be endogenous to the IC, or it could be driven by hyperactivity at lower stages of the auditory pathway. In addition to the dorsal cochlear nucleus (DCN) hyperactivity reported by others, specific cell types in the ventral cochlear nucleus (VCN) also show hyperactivity in this animal model suggesting that increased drive from several regions of the lower brainstem could contribute to the observed hyperactivity in the midbrain. In addition, spontaneous afferent drive from the cochlea itself is necessary for the maintenance of hyperactivity up to about 8 weeks post cochlear trauma. After 8 weeks however, IC hyperactivity becomes less dependent on cochlear input, suggesting that central neurons transition from a state of hyperexcitability to a state in which they generate their own endogenous firing. The results suggest that there might be a "therapeutic window" for early-onset tinnitus, using treatments that reduce cochlear afferent firing.


Subject(s)
Auditory Pathways/physiopathology , Mesencephalon/physiopathology , Animals , Auditory Cortex/physiopathology , Cochlea/physiopathology , Cochlear Nucleus/physiopathology , Electric Stimulation Therapy , Electrophysiological Phenomena , Guinea Pigs , Hearing Loss, Noise-Induced/physiopathology , Inferior Colliculi/physiopathology , Models, Neurological , Olivary Nucleus/physiopathology , Sensory Gating , Tinnitus/etiology , Tinnitus/physiopathology , Tinnitus/therapy
6.
Neurosci Lett ; 522(1): 16-20, 2012 Jul 26.
Article in English | MEDLINE | ID: mdl-22683504

ABSTRACT

Previous studies indicate that the dorsal cochlear nucleus (DCN) may serve as a generator and/or modulator of noise-induced tinnitus. This prompted an interest to investigate the modulatory role of the DCN in tinnitus suppression. In this study, we chronically implanted the DCN of rats with behavioral evidence of intense tone-induced tinnitus. Behavioral evidence of tinnitus was measured using a gap detection acoustic startle reflex paradigm. Our results demonstrated that electrical stimulation of the DCN suppressed behavioral evidence of tinnitus, especially at high frequencies. The data suggest that the DCN may be used as a target to suppress tinnitus through a bottom-up neuromodulation approach. The underlying mechanism of DCN-stimulation-induced tinnitus suppression was discussed by comparing it with other stimulation modalities.


Subject(s)
Cochlear Nucleus/physiopathology , Tinnitus/physiopathology , Acoustic Stimulation , Animals , Electric Stimulation , Male , Rats , Rats, Long-Evans , Reflex, Startle , Tinnitus/psychology
7.
Hear Res ; 287(1-2): 57-66, 2012 May.
Article in English | MEDLINE | ID: mdl-22726617

ABSTRACT

The purpose of this study was to investigate the effects of early hearing loss on the anatomy of the central auditory system, specifically, the ascending projections to the inferior colliculus (IC). We compared normal animals with animals deafened during early development by administration of amikacin, an ototoxic antibiotic that is known to destroy the hair cells in the inner ear. The amikacin was injected subcutaneously every day from postnatal days P7 to P16. A retrograde tract tracer, Fluoro-Gold (FG), was then injected unilaterally directly into the IC at either 4 weeks of age or 12 weeks of age. After axonal transport the animals were sacrificed and their brains were prepared for histology. The FG labeled neurons in the cochlear nucleus (CN) and the dorsal nucleus of lateral lemniscus (DNLL) were counted for each of the animals in the two age groups. For deaf animals sacrificed at 4 weeks of age there was a significant reduction in the number of FG labeled neurons that was limited to the ventral CN ipsilateral to the tracer injection. For deaf animals sacrificed at 12 weeks of age, however, there was a significant decrease in the number of labeled cells in both dorsal and ventral CN on both sides of the brain. In DNLL there was no change in the number or pattern of labeled neurons. The results show that neonatal deafness reduces the number of labeled neurons projecting from the CN to the IC with the effect being more evident during later stages of deafness. In contrast, there are no significant changes in the projections from DNLL to IC.


Subject(s)
Cochlear Nucleus/pathology , Hearing Loss/pathology , Inferior Colliculi/pathology , Acoustic Stimulation , Age Factors , Amikacin , Animals , Animals, Newborn , Auditory Pathways/pathology , Auditory Pathways/physiopathology , Auditory Threshold , Cochlear Nucleus/physiopathology , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem , Female , Fluorescent Dyes , Hearing Loss/chemically induced , Hearing Loss/physiopathology , Inferior Colliculi/physiopathology , Male , Neuroanatomical Tract-Tracing Techniques , Neuronal Tract-Tracers , Rats , Rats, Sprague-Dawley , Stilbamidines
8.
Eur J Neurosci ; 31(9): 1616-28, 2010 May.
Article in English | MEDLINE | ID: mdl-20525074

ABSTRACT

Hearing loss from acoustic trauma is a risk factor for tinnitus. Animal models using acoustic trauma have demonstrated hyperactivity in central auditory pathways, which has been suggested as a substrate for tinnitus. We used a guinea-pig model of unilateral acoustic trauma. Within the same animals, measurements of peripheral hearing loss, spontaneous activity of single neurons in the inferior colliculus and gene expression in cochlear nucleus and inferior colliculus were combined, acutely and after recovery from acoustic trauma. Genes investigated related to inhibitory (GABA-A receptor subunit alpha 1; glycine receptor subunit alpha 1) and excitatory neurotransmission (glutamate decarboxylase 1; glutamate receptor AMPA subunit alpha 2; glutamate receptor NMDA subunit 1), regulation of transmitter release (member of RAB family of small GTPase; RAB3 GTPase activating protein subunit 1) and neuronal excitability (potassium channel subfamily K member 15). Acoustic trauma resulted in unilateral hearing loss and hyperactivity bilaterally in inferior colliculus. Changes in expression of different mRNAs were observed in ipsilateral cochlear nucleus and in ipsi- and contralateral inferior colliculus, immediately after acoustic trauma, and after 2 and 4 weeks' recovery. Gene expression was generally reduced immediately after trauma, followed by a return to near normal levels or over-expression as recovery time increased. Different mechanisms appear to underlie the spontaneous hyperactivity observed. There is evidence of down-regulation of genes associated with neuronal inhibition in the contralateral inferior colliculus, whereas in ipsilateral cochlear nucleus, competing actions of inhibitory and excitatory systems seem to play a major role in determining overall excitability.


Subject(s)
Cochlear Nucleus/physiopathology , Gene Expression , Hearing Loss, Noise-Induced/genetics , Hearing Loss, Noise-Induced/physiopathology , Inferior Colliculi/physiopathology , Neurons/physiology , Acoustic Stimulation/adverse effects , Action Potentials , Animals , Brain Stem/physiopathology , Disease Models, Animal , Functional Laterality , Guinea Pigs , Hearing Tests , RNA, Messenger/metabolism , Random Allocation , Time Factors
9.
Am J Audiol ; 19(1): 17-25, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20308289

ABSTRACT

PURPOSE: A hallmark listening problem of individuals presenting with auditory processing disorder (APD) is their poor recognition of speech in noise. The underlying perceptual problem of the listening difficulties in unfavorable listening conditions is unknown. The objective of this article was to demonstrate theoretically how to determine whether the speech recognition problems are related to an auditory dysfunction, a language-based dysfunction, or a combination of both. METHOD: Tests such as the Speech Perception in Noise (SPIN) test allow the exploration of the auditory and language-based functions involved in speech perception in noise, which is not possible with most other speech-in-noise tests. Psychometric functions illustrating results from hypothetical groups of individuals with APD on the SPIN test are presented. This approach makes it possible to postulate about the origin of the speech perception problems in noise. CONCLUSION: APD is a complex and heterogeneous disorder for which the underlying deficit is currently unclear. Because of their design, SPIN-like tests can potentially be used to identify the nature of the deficits underlying problems with speech perception in noise for this population. A better understanding of the difficulties with speech perception in noise experienced by many listeners with APD should lead to more efficient intervention programs.


Subject(s)
Auditory Perceptual Disorders/diagnosis , Perceptual Masking , Speech Reception Threshold Test , Acoustic Stimulation , Adult , Auditory Perceptual Disorders/physiopathology , Auditory Perceptual Disorders/rehabilitation , Child , Child, Preschool , Cochlear Nucleus/physiopathology , Corpus Callosum/physiopathology , Diagnosis, Differential , Female , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/physiopathology , Hearing Loss, Sensorineural/rehabilitation , Hearing Tests/statistics & numerical data , Humans , Male , Psycholinguistics , Psychometrics , Semantics , Sound Localization/physiology , Sound Spectrography , Speech Acoustics , Speech Discrimination Tests , Speech Perception/physiology , Speech Reception Threshold Test/statistics & numerical data
10.
Neuroscience ; 164(2): 747-59, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19699270

ABSTRACT

Fifteen percent to 35% of the United States population experiences tinnitus, a subjective "ringing in the ears". Up to 10% of those afflicted report severe and disabling symptoms. Tinnitus was induced in rats using unilateral, 1 h, 17 kHz-centered octave-band noise (116 dB SPL) and assessed using a gap-startle method. The dorsal cochlear nucleus (DCN) is thought to undergo plastic changes suggestive of altered inhibitory function during tinnitus development. Exposed rats showed near pre-exposure auditory brainstem response (ABR) thresholds for clicks and all tested frequencies 16 weeks post-exposure. Sound-exposed rats showed significantly worse gap detection at 24 and 32 kHz 16 weeks following sound exposure, suggesting the development of chronic, high frequency tinnitus. Message and protein levels of alpha(1-3,) and beta glycine receptor subunits (GlyRs), and the anchoring protein, gephyrin, were measured in DCN fusiform cells 4 months following sound exposure. Rats with evidence of tinnitus showed significant GlyR alpha(1) protein decreases in the middle and high frequency regions of the DCN while alpha(1) message levels were paradoxically increased. Gephyrin levels showed significant tinnitus-related increases in sound-exposed rats suggesting intracellular receptor trafficking changes following sound exposure. Consistent with decreased alpha(1) subunit protein levels, strychnine binding studies showed significant tinnitus-related decreases in the number of GlyR binding sites, supporting tinnitus-related changes in the number and/or composition of GlyRs. Collectively, these findings suggest the development of tinnitus is likely associated with functional GlyR changes in DCN fusiform cells consistent with previously described behavioral and neurophysiologic changes. Tinnitus related GlyR changes could provide a unique receptor target for tinnitus pharmacotherapy or blockade of tinnitus initiation.


Subject(s)
Cochlear Nucleus/physiopathology , Glycine/metabolism , Neuronal Plasticity/physiology , Neurons/physiology , Synapses/physiology , Tinnitus/physiopathology , Acoustic Stimulation , Animals , Carrier Proteins/metabolism , Chronic Disease , Evoked Potentials, Auditory, Brain Stem , Male , Membrane Proteins/metabolism , Psychoacoustics , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains , Receptors, Glycine/metabolism , Time Factors
11.
Neuroscience ; 163(4): 1264-76, 2009 Nov 10.
Article in English | MEDLINE | ID: mdl-19646510

ABSTRACT

Neurons restore their function in response to external or internal perturbations and maintain neuronal or network stability through a homeostatic scaling mechanism. Homeostatic responses at synapses along the auditory system would be important for adaptation to normal and abnormal fluctuations in the sensory environment. We investigated at the electron microscopic level and after postembedding immunogold labeling whether projection neurons in the cochlear nucleus responded to modifications of auditory nerve activity. After unilaterally reducing the level of auditory inputs by approximately 20 dB by monaural earplugging, auditory nerve synapses on bushy cells somata and basal dendrites of fusiform cells of the ventral and dorsal cochlear nucleus, respectively, upregulated GluR3 AMPA receptor subunit, while inhibitory synapses decreased the expression of GlyRalpha1 subunit. These changes in expression levels were fully reversible once the earplug was removed, indicating that activity affects the trafficking of receptors at synapses. Excitatory synapses on apical dendrites of fusiform cells (parallel fibers) with different synaptic AMPA receptor subunit composition, were not affected by sound attenuation, as the expression levels of AMPA receptor subunits were the same as in normal hearing littermates. GlyRalpha1 subunit expression at inhibitory synapses on apical dendrites of fusiform cells was also found unaffected. Furthermore, fusiform and bushy cells of the contralateral side to the earplugging upregulated the GluR3 subunit at auditory nerve synapses. These results show that cochlear nucleus neurons innervated by the auditory nerve, are able to respond to small changes in sound levels by redistributing specific AMPA and glycine receptor subunits.


Subject(s)
Cochlear Nucleus/physiopathology , Hearing Loss, Conductive/physiopathology , Neurons/physiology , Receptors, AMPA/metabolism , Receptors, Glycine/metabolism , Synapses/physiology , Acoustic Stimulation , Animals , Cochlear Nerve/physiopathology , Cochlear Nucleus/ultrastructure , Dendrites/physiology , Dendrites/ultrastructure , Immunohistochemistry , Microscopy, Electron , Neuronal Plasticity/physiology , Neurons/ultrastructure , Rats , Rats, Sprague-Dawley , Sensory Deprivation/physiology , Synapses/ultrastructure
12.
Am J Audiol ; 17(2): S193-209, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19056923

ABSTRACT

PURPOSE: This review outlines the anatomical and functional bases of somatosensory influences on auditory processing in the normal brainstem and midbrain. It then explores how interactions between the auditory and somatosensory system are modified through deafness, and their impact on tinnitus is discussed. METHOD: Literature review, tract tracing, immunohistochemistry, and in vivo electrophysiological recordings were used. RESULTS: Somatosensory input originates in the dorsal root ganglia and trigeminal ganglia, and is transmitted directly and indirectly through 2nd-order nuclei to the ventral cochlear nucleus, dorsal cochlear nucleus (DCN), and inferior colliculus. The glutamatergic somatosensory afferents can be segregated from auditory nerve inputs by the type of vesicular glutamate transporters present in their terminals. Electrical stimulation of the somatosensory input results in a complex combination of excitation and inhibition, and alters the rate and timing of responses to acoustic stimulation. Deafness increases the spontaneous rates of those neurons that receive excitatory somatosensory input and results in a greater sensitivity of DCN neurons to trigeminal stimulation. CONCLUSIONS: Auditory-somatosensory bimodal integration is already present in 1st-order auditory nuclei. The balance of excitation and inhibition elicited by somatosensory input is altered following deafness. The increase in somatosensory influence on auditory neurons when their auditory input is diminished could be due to cross-modal reinnervation or increased synaptic strength, and may contribute to mechanisms underlying somatic tinnitus.


Subject(s)
Attention/physiology , Auditory Perception/physiology , Brain Stem/physiopathology , Deafness/physiopathology , Mesencephalon/physiopathology , Sensation/physiology , Tinnitus/physiopathology , Acoustic Stimulation , Afferent Pathways/physiopathology , Animals , Auditory Pathways/physiopathology , Brain Mapping , Cochlear Nucleus/physiopathology , Electric Stimulation , Ganglia, Spinal/physiopathology , Humans , Inferior Colliculi/physiopathology , Neural Inhibition/physiology , Neuronal Plasticity/physiology , Neurons/physiology , Sensory Thresholds/physiology , Synaptic Transmission/physiology , Trigeminal Ganglion/physiopathology
13.
J Commun Disord ; 41(1): 70-83, 2008.
Article in English | MEDLINE | ID: mdl-17585930

ABSTRACT

UNLABELLED: To investigate the transient evoked otoacoustic emissions (TEOAE) contralateral suppression in neonates at risk for hearing loss, 55 neonates at risk for hearing loss (risk group) and 72 full-term neonates not at such risk (control group) were bilaterally tested. In all neonates, the TEOAE were recorded in two stimulation modes (linear and nonlinear clicks), with and without contralateral acoustic stimulation. Findings revealed significant contralateral suppression of otoacoustic emissions in both groups, but the amount of TEOAE contralateral suppression was reduced for at risk group (p=0.001), supporting the hypothesis that medial olivocochlear bundle function is reduced in neonates at risk for hearing loss. The combination of contralateral acoustic stimulation and TEOAE enables easy and noninvasive study of auditory efferent function. However it should be emphasized that the reduction in TEOAE contralateral suppression in the risk group, statistically identified as a group effect, might not be detectable in individual cases. Further studies are needed in order to determine whether the lower amount of TEOAE contralateral suppression in neonates at risk for hearing loss represents a risk for developing auditory processing disorders. LEARNING OUTCOMES: The reader will be introduced to the study using auditory efferent pathway activation by contralateral acoustic stimulation (CAS), resulting in the TEOAE suppression effect. The characteristics of TEOAE suppression in the neonatal population, in which it provides evidence of the reduced medial olivocochlear system function in those at risk for hearing loss, will also be addressed.


Subject(s)
Audiometry, Evoked Response/methods , Dominance, Cerebral/physiology , Hearing Disorders/congenital , Infant, Premature, Diseases/diagnosis , Otoacoustic Emissions, Spontaneous/physiology , Signal Processing, Computer-Assisted , Acoustic Stimulation , Cochlear Nucleus/physiopathology , Efferent Pathways/physiopathology , Female , Hearing Disorders/diagnosis , Hearing Disorders/physiopathology , Humans , Infant, Newborn , Infant, Premature, Diseases/physiopathology , Male , Olivary Nucleus/physiopathology , Reference Values , Risk Factors
14.
J Am Acad Audiol ; 19(7): 542-7, 2008.
Article in English | MEDLINE | ID: mdl-19248730

ABSTRACT

BACKGROUND: It has been reported that up to 40% of patients over age 60 fail to generate a vestibular evoked myogenic potential (VEMP; Su et al, 2004). When this occurs it is difficult to determine whether the absent VEMP represents evidence of bilateral impairment of the vestibulocollic reflex pathway or a normal age-related variant (i.e., idiopathic absence). PURPOSE: The purpose of the present investigation was to determine whether both VEMPs and PAMs could be recorded reliably in a sample of neurologically and otologically intact young adults. If both could be obtained with high reliability in normal subjects, then the bilateral presence of PAM in the bilateral absence of VEMP, at least in younger patients, could be used to support the contention that the absent VEMP represented evidence of bilateral impairment. RESEARCH DESIGN: A descriptive study. STUDY SAMPLE: Attempts were made to record both the VEMP and a second sonomotor response, the postauricular muscle potential (PAM) from 20 young adults. RESULTS: Results showed both the VEMP and the PAM were present in 90% of the ears. Both the VEMP and PAM responses were bilaterally absent for one subject. Also, the VEMP and PAM were unilaterally absent for two subjects. Subjects who generated VEMPs also generated a PAM in at least one ear. CONCLUSIONS: The present investigation represents an initial step in the determination of whether the presence of PAMs in the absence of VEMPs can be used as a method of validating the presence of an impairment affecting the vestibulocollic reflex pathway.


Subject(s)
Acoustic Stimulation , Cochlear Nucleus/physiopathology , Electromyography/statistics & numerical data , Evoked Potentials, Motor/physiology , Neck Muscles/innervation , Reflex, Acoustic/physiology , Vestibular Function Tests/statistics & numerical data , Vestibular Nuclei/physiopathology , Vestibulocochlear Nerve/physiopathology , Auditory Pathways/physiopathology , Functional Laterality/physiology , Humans , Male , Otolithic Membrane/innervation , Reaction Time/physiology , Reference Values , Saccule and Utricle/innervation , Signal Processing, Computer-Assisted , Synaptic Transmission/physiology , Young Adult
15.
Hear Res ; 230(1-2): 88-92, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17590548

ABSTRACT

Among possible therapies after acute acoustic trauma, hyperbaric oxygenation (HBO) combined with corticoid was found effective in several animal studies. Such evidence was obtained for moderate 20-25 dB losses. The aim of this study was to further assess this therapy for noise-induced hearing losses greater than previously examined. Sixty-five ears from thirty-six adult guinea pigs were used. Acoustically evoked responses from intracranial electrodes chronically implanted bilaterally into the ventral cochlear nucleus were used to assess acoustic sensitivity alterations. Trauma sound was a third-octave noise-band around 8 kHz presented bilaterally at 115 dB SPL for 45 min. One control group received no treatment, one group was treated with HBO only and another with corticoid only both starting within one day post-trauma, two groups were treated with both HBO and corticoid starting for one group within one day post-trauma, and for the second group at 6 days post-trauma. Acoustic thresholds were measured between the 6th and the 16th days after acoustic trauma. Animals treated with HBO alone or corticoid alone did not differ from controls. Combined HBO and corticoid therapy provided significant protection from noise-induced loss of auditory thresholds, especially when started one day post-exposure. Hearing loss reduction induced by HBO combined with corticoid was of similar magnitude (about 10-15 dB) as in previous studies although the induced hearing loss was considerably greater (about 40 dB instead of 20-25 dB).


Subject(s)
Cochlear Nucleus/drug effects , Glucocorticoids/pharmacology , Hearing Loss, Noise-Induced/therapy , Hyperbaric Oxygenation , Methylprednisolone Hemisuccinate/pharmacology , Animals , Auditory Threshold/drug effects , Cochlear Nucleus/physiopathology , Combined Modality Therapy , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/drug effects , Glucocorticoids/therapeutic use , Guinea Pigs , Hearing Loss, Noise-Induced/drug therapy , Hearing Loss, Noise-Induced/etiology , Hearing Loss, Noise-Induced/physiopathology , Methylprednisolone Hemisuccinate/therapeutic use , Noise/adverse effects , Time Factors
16.
Auris Nasus Larynx ; 34(4): 453-8, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17467218

ABSTRACT

OBJECTIVE: The purpose of this study is to evaluate whether the middle latency responses (MLR) can be used for an objective differentiation of patients with presbycusis having relatively good (Group I) and relatively poor speech recognition scores (Group II). METHODS: All the participants of these groups had high frequency down-sloping hearing loss with an average of 26-60 dB HL. Data were collected from two described study groups and a control group, using pure tone audiometry, monosyllabic phonetically balanced word and synthetic sentence identification, as well as MLR. The study groups were compared with the control group. RESULTS: When patients in Group I were compared with the control group, only ipsilateral Na latency of middle latency evoked response was statistically significant in the right ear whereas ipsilateral Na latency in the right ear, ipsilateral and contralateral Na latency in the left ear of the patients in Group II were statistically significant. CONCLUSION: Thus, as an objective complementary tool for the evaluation of the speech perception ability of the patients with presbycusis, Na latency of MLR may be used in combination with the speech discrimination tests.


Subject(s)
Audiometry, Speech/methods , Evoked Potentials, Auditory, Brain Stem/physiology , Presbycusis/physiopathology , Reaction Time/physiology , Acoustic Stimulation , Aged , Audiometry, Pure-Tone , Auditory Threshold/physiology , Cochlear Nucleus/physiopathology , Dominance, Cerebral/physiology , Female , Humans , Male , Middle Aged , Phonetics , Presbycusis/diagnosis , Prognosis
17.
Ear Hear ; 28(3): 381-93, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17485987

ABSTRACT

OBJECTIVE: The effect on speech perception of using higher stimulation rates than the 14.4 kHz available in the Nucleus 24 cochlear implant system was investigated. The study used the Nucleus Research Platform 8 (RP8) system, comprising the CI24RE receiver-stimulator with the Contour electrode array, the L34SP body-worn research speech processor, and the Nucleus Programming Environment (NPE) fitting and Neural Response Telemetry (NRT) software. This system enabled clinical investigation of higher stimulation rates before an implementation in the Freedom cochlear implant system commercially released by Cochlear Limited. DESIGN: Use of higher stimulation rates in the ACE coding strategy was assessed in 15 adult subjects. An ABAB experimental design was used to control for order effects. Program A used a total stimulation rate of between 12 kHz and 14.4 kHz. This program was used for at least the first 3 mo after initial device activation. After evaluation with this program, each subject was provided with two different higher stimulation rate programs: one with a total stimulation rate of 24 kHz and the other with a total stimulation rate of 32 kHz. After a 6-week period of familiarization, each subject identified his/her preferred higher rate program (program B), and this was used for the evaluation. Subjects then repeated their use of program A for 3 wk, then program B for 3 wk, before the second evaluation with each. Speech perception was evaluated by using CNC open-set monosyllabic words presented in quiet and CUNY open-set sentences presented in noise. Preference for stimulation rate program was assessed via a subjective questionnaire. Threshold (T)- and Comfortable (C)-levels, as well as subjective reports of tinnitus, were monitored for each subject throughout the study to determine whether there were any changes that might be associated with the use of higher stimulation rates. RESULTS: No significant mean differences in speech perception results were found for the group between the two programs for tests in either quiet or noise. Analysis of individual subject data showed that five subjects had significant benefit from use of program B for tests administered in quiet and for tests administered in noise. However, only two of these subjects showed benefit in both test conditions. One subject showed significant benefit from use of program A when tested in quiet, whereas another showed benefit with this program in noise. Each subject's preferred program varied. Five subjects reported a preference for program A, eight subjects reported a preference for program B and two reported no overall preference. Preference between the different stimulation rates provided within program B also varied, with 10 subjects preferring 24 kHz and five preferring 32 kHz total stimulation rates. A significant increase in T-levels from baseline measures was observed after three weeks of initial experience with program B, however there was no difference between the baseline levels and those obtained after five weeks of use. No significant change in C-levels was found over the monitoring period. No long-term changes in tinnitus that could be associated with the use of the higher stimulation rates were reported by any of the subjects. CONCLUSIONS: The use of higher stimulation rates may provide benefit to some but not all cochlear implant recipients. It is important to optimize the stimulation rate for an individual to ensure maximal benefit. The absence of any changes in T- and C-levels or in tinnitus suggests that higher stimulation rates are safe for clinical use.


Subject(s)
Acoustic Stimulation/instrumentation , Cochlear Implants , Cochlear Nucleus/physiopathology , Speech Perception , Adult , Aged , Auditory Threshold/physiology , Electrodes , Female , Hearing Loss, Sensorineural/epidemiology , Hearing Loss, Sensorineural/physiopathology , Hearing Loss, Sensorineural/therapy , Humans , Male , Middle Aged , Noise , Prosthesis Design , Psychophysics , Severity of Illness Index , Software , Surveys and Questionnaires , Tinnitus/epidemiology , Vocabulary
18.
B-ENT ; 3 Suppl 7: 23-5, 2007.
Article in English | MEDLINE | ID: mdl-18225605

ABSTRACT

Tinnitus, or the phantom perception of sound, is one of the great unsolved problems of otology. It is present in all patients with hearing loss and, in approximately 5-10% of individuals, it has a significant impact on quality of life. Progress in the treatment of tinnitus has been limited by a lack of animal models that can be used to study the neurophysiology of tinnitus and to examine prospective treatment. In the last ten years, several physiological and behavioural animal models of tinnitus have been developed that have significantly increased our understanding. The next ten years will see the application of these models to drug development and electrical stimulus approaches to curing tinnitus.


Subject(s)
Cochlear Nucleus/physiopathology , Electric Stimulation Therapy/methods , GABA Agonists/therapeutic use , Tinnitus/therapy , Animals , Tinnitus/physiopathology
19.
Acta Otolaryngol Suppl ; (556): 20-6, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17114138

ABSTRACT

Evidence has accumulated in the last decade that the dorsal cochlear nucleus (DCN) may be an important site in the etiology of tinnitus. This evidence comes from a combination of studies conducted in animals and humans. This paper will review the key findings, as follows. 1) Direct electrical stimulation of the DCN leads to changes in the loudness of tinnitus. This suggests that the loudness of tinnitus may be linked to changes in the level of neural activity in the DCN. 2) Exposure to tinnitus inducers, such as intense sound or cisplatin, causes neural activity in the DCN to become chronically elevated, a condition known as neuronal hyperactivity. 3) This hyperactivity is very similar to the activity that is evoked in the DCN by sound stimulation, suggesting that the hyperactivity represents a code that signals the presence of sound, even when there is no longer any sound stimulus. 4) Noise-induced hyperactivity in the DCN is correlated with tinnitus. Behavioral studies have demonstrated that animals exposed to the same intense sound that causes hyperactivity in the DCN develop tinnitus-like percepts. The correlation between the level of hyperactivity and the behavioral index of tinnitus was found to be statistically significant. 5) The DCN is a polysensory integration center, and electrophysiological studies have shown that both spontaneous activity and hyperactivity of neurons in the DCN can be modulated by stimulation of certain ipsilateral cranial nerves, such as the sensory branch of the trigeminal nerve. This ipsilateral modulation of DCN activity offers a plausible explanation of how tinnitus, when perceived on one side, can be modulated by certain manipulations of the head and neck on the side ipsilateral to the tinnitus, but rarely on the contralateral side. 6) The DCN exhibits various forms of neuronal plasticity that parallel the various forms of plasticity that characterize tinnitus. These findings collectively strengthen the view that the DCN may be a key structure that should be included as a target of anti-tinnitus treatment.


Subject(s)
Cochlear Nucleus/physiopathology , Tinnitus/etiology , Tinnitus/physiopathology , Acoustic Stimulation/adverse effects , Animals , Cricetinae , Electric Stimulation/instrumentation , Electrodes, Implanted , Evoked Potentials, Auditory/physiology , Neuronal Plasticity/physiology , Severity of Illness Index , Time Factors , Tinnitus/diagnosis
20.
Hear Res ; 216-217: 146-53, 2006.
Article in English | MEDLINE | ID: mdl-16650703

ABSTRACT

Two-pore domain potassium channels (K(2PD)+) play an important role in setting resting membrane potential by regulating background leakage of potassium ions, which in turn controls neuronal excitability. To determine whether these channels contribute to activity-dependent plasticity following deafness, we used quantitative real-time PCR to examine the expression of 10 K(2PD)+ subunits in the rat cochlear nucleus at 3 days, 3 weeks and 3 months after bilateral cochlear ablation. There was a large sustained decrease in the expression of TASK-5, a subunit that is predominantly expressed in auditory brain stem neurons, and in the TASK-1 subunit which is highly expressed in several types of cochlear nucleus neurons. TWIK-1 and THIK-2 also showed significant decreases in expression that were maintained across all time points. TWIK-2, TREK-1 and TREK-2 showed no significant change in expression at 3 days but showed large decreases at 3 weeks and 3 months following deafness. TRAAK and TASK-3 subunits showed significant decreases at 3 days and 3 weeks following deafness, but these differences were no longer significant at 3 months. Dramatic changes in expression of K(2PD)+ subunits suggest these channels may play a role in deafness-associated changes in the excitability of cochlear nucleus neurons.


Subject(s)
Cochlear Nucleus/physiopathology , Deafness/physiopathology , Neuronal Plasticity/physiology , Potassium Channels, Tandem Pore Domain/metabolism , Animals , Cochlear Nucleus/cytology , DNA, Complementary/chemistry , Deafness/pathology , Evoked Potentials, Auditory, Brain Stem , Male , Potassium Channels, Tandem Pore Domain/chemistry , Potassium Channels, Tandem Pore Domain/genetics , Potassium Channels, Tandem Pore Domain/physiology , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , Random Allocation , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL