Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Mil Med ; 188(Suppl 6): 176-184, 2023 11 08.
Article in English | MEDLINE | ID: mdl-37948248

ABSTRACT

INTRODUCTION: Although existing auditory injury prevention standards benefit warfighters, the Department of Defense could do more to understand and address auditory injuries (e.g., hearing loss, tinnitus, and central processing deficits) among service members. The Blast Injury Prevention Standards Recommendation (BIPSR) Process is designed to address the needs of all the Military Services for biomedically valid Military Health System (MHS) Blast Injury Prevention Standards. MATERIALS AND METHODS: Through the BIPSR Process, stakeholders provided their intended uses and requested functionalities for an MHS Blast Injury Prevention Standard. The BIPSR Process established a broad-based, non-advocacy panel of auditory injury Subject Matter Expert (SME) Panel with members drawn from industry, academia, and government. The SME Panel selected evaluation factors, weighted priorities, and then evaluated the resulting candidate MHS Auditory Blast Injury Prevention Standards against the evaluation criteria. The SME Panel members provided rationales for their decisions, documented discussions, and used iterative rounds of feedback to promote consensus building among members. The BIPSR Process used multi-attribute utility theory to combine members' evaluations and compare the candidate standards. RESULTS: The SME Panel identified and collated information about existing auditory injury datasets to identify gaps and promote data sharing and comprehensive evaluations of standards for preventing auditory blast injury. The panel evaluated the candidate standards and developed recommendations for an MHS Blast Injury Prevention Standard. CONCLUSIONS: The BIPSR Process illuminated important characteristics, capabilities, and limitations of candidate standards and existing datasets (e.g., limited human exposure data to evaluate the validity of injury prediction) for auditory blast injury prevention. The evaluation resulted in the recommendation to use the 8-hour Equivalent Level (LAeq8hr) as the interim MHS Auditory Blast Injury Prevention Standard while the community performs additional research to fill critical knowledge gaps.


Subject(s)
Blast Injuries , Hearing Loss , Military Health Services , Military Personnel , Tinnitus , Humans , Blast Injuries/prevention & control , Explosions , Tinnitus/prevention & control
2.
Elife ; 112022 09 16.
Article in English | MEDLINE | ID: mdl-36111669

ABSTRACT

Neurons in sensory cortex exhibit a remarkable capacity to maintain stable firing rates despite large fluctuations in afferent activity levels. However, sudden peripheral deafferentation in adulthood can trigger an excessive, non-homeostatic cortical compensatory response that may underlie perceptual disorders including sensory hypersensitivity, phantom limb pain, and tinnitus. Here, we show that mice with noise-induced damage of the high-frequency cochlear base were behaviorally hypersensitive to spared mid-frequency tones and to direct optogenetic stimulation of auditory thalamocortical neurons. Chronic two-photon calcium imaging from ACtx pyramidal neurons (PyrNs) revealed an initial stage of spatially diffuse hyperactivity, hyper-correlation, and auditory hyperresponsivity that consolidated around deafferented map regions three or more days after acoustic trauma. Deafferented PyrN ensembles also displayed hypersensitive decoding of spared mid-frequency tones that mirrored behavioral hypersensitivity, suggesting that non-homeostatic regulation of cortical sound intensity coding following sensorineural loss may be an underlying source of auditory hypersensitivity. Excess cortical response gain after acoustic trauma was expressed heterogeneously among individual PyrNs, yet 40% of this variability could be accounted for by each cell's baseline response properties prior to acoustic trauma. PyrNs with initially high spontaneous activity and gradual monotonic intensity growth functions were more likely to exhibit non-homeostatic excess gain after acoustic trauma. This suggests that while cortical gain changes are triggered by reduced bottom-up afferent input, their subsequent stabilization is also shaped by their local circuit milieu, where indicators of reduced inhibition can presage pathological hyperactivity following sensorineural hearing loss.


Subject(s)
Auditory Cortex , Hearing Loss, Noise-Induced , Tinnitus , Acoustic Stimulation , Animals , Calcium , Cochlea , Mice , Noise
3.
Front Cell Neurosci ; 15: 699978, 2021.
Article in English | MEDLINE | ID: mdl-34385909

ABSTRACT

Previous work in animals with recovered hearing thresholds but permanent inner hair cell synapse loss after noise have suggested initial vulnerability of low spontaneous rate (SR) auditory nerve fibers (ANF). As these fibers have properties of response that facilitate robust sound coding in continuous noise backgrounds, their targeted loss would have important implications for function. To address the issue of relative ANF vulnerabilities after noise, we assessed cochlear physiologic and histologic consequences of temporary threshold shift-producing sound over-exposure in the gerbil, a species with well-characterized distributions of auditory neurons by SR category. The noise exposure targeted a cochlear region with distributed innervation (low-, medium- and high-SR neurons). It produced moderate elevations in outer hair cell-based distortion-product otoacoustic emission and whole nerve compound action potential thresholds in this region, with accompanying reductions in suprathreshold response amplitudes, quantified at 24 h. These parameters of response recovered well with post-exposure time. Chronic synapse loss was maximum in the frequency region initially targeted by the noise. Cochlear round window recorded mass potentials (spontaneous neural noise and sound-driven peri-stimulus time responses, PSTR) reflected parameters of the loss not detected by the conventional assays. Spontaneous activity was acutely reduced. Steady-state (PSTR plateau) activity was correlated with synapse loss in frequency regions with high concentrations of low-SR neurons, whereas the PSTR onset peak and spontaneous round window noise, both dominated by high-SR fiber activity, were relatively unaltered across frequency in chronic ears. Together, results suggest that acute targets of noise were of mixed SR subtypes, but chronic targets were predominantly low-SR neurons. PSTRs captured key properties of the auditory nerve response and vulnerability to injury that should yield important diagnostic information in hearing loss etiologies producing cochlear synaptic and neural loss.

4.
Noise Health ; 23(108): 42-49, 2021.
Article in English | MEDLINE | ID: mdl-33753680

ABSTRACT

CONTEXT: Sound levels in fitness classes often exceed safe levels despite studies that show many participants find high sound levels stressful. AIMS: The objective is to determine if lower sound levels in spinning classes significantly impact exercise intensity and to determine if class participants prefer the music played at lower levels. SETTINGS AND DESIGN: Observational study of 1-hour group spin classes. METHODS AND MATERIALS: Sound levels were measured in 18 spin classes over two weeks. No adjustments were made in week-1 and sound levels were decreased by 3 dB in week-2. Participant preferences and data on post-class hearing changes were collected via post-class questionnaires (n = 213) and divided into three terciles based on the total sound exposure of corresponding classes. STATISTICAL ANALYSIS USED: Unweighted survey generalized linear models are used to sort the causal relationships between different variables simultaneously and participant responses. The Chi-square test is used to reveal statistically significant relationships between two or more categorical variables. RESULTS: When mean sound levels exceeded 98.4 dBC, respondents were 23 times more likely to report the music as too loud than too quiet (P < 0.05), and four times more likely to prefer a decrease, rather than an increase, in sound level (P < 0.05). There was no significant difference in respondents reporting high exercise intensity between the middle (95.7-98.1 dBC) and upper (98.4-101.0 dBC) terciles, 67.1% and 71.8%, respectively (P = 0.53). Overall, 25.9% of respondents reported auditory symptoms following classes. Analysis in the context of dBA and dBC produced congruent conclusions and interpretations. CONCLUSIONS: Sound levels in many fitness classes remain dangerously high. However, music level can be lowered without a significant impact on perceived exercise intensity and many participants prefer lower sound levels than current levels.


Subject(s)
Bicycling/psychology , Exercise/psychology , Hearing Loss, Noise-Induced/psychology , Music/psychology , Noise/adverse effects , Adult , Auditory Threshold , Female , Hearing Loss, Noise-Induced/epidemiology , Hearing Loss, Noise-Induced/etiology , Humans , Male , Perception , Sound
5.
JCI Insight ; 6(3)2021 02 08.
Article in English | MEDLINE | ID: mdl-33373328

ABSTRACT

TrkB agonist drugs are shown here to have a significant effect on the regeneration of afferent cochlear synapses after noise-induced synaptopathy. The effects were consistent with regeneration of cochlear synapses that we observed in vitro after synaptic loss due to kainic acid-induced glutamate toxicity and were elicited by administration of TrkB agonists, amitriptyline, and 7,8-dihydroxyflavone, directly into the cochlea via the posterior semicircular canal 48 hours after exposure to noise. Synaptic counts at the inner hair cell and wave 1 amplitudes in the auditory brainstem response (ABR) were partially restored 2 weeks after drug treatment. Effects of amitriptyline on wave 1 amplitude and afferent auditory synapse numbers in noise-exposed ears after systemic (as opposed to local) delivery were profound and long-lasting; synapses in the treated animals remained intact 1 year after the treatment. However, the effect of systemically delivered amitriptyline on synaptic rescue was dependent on dose and the time window of administration: it was only effective when given before noise exposure at the highest injected dose. The long-lasting effect and the efficacy of postexposure treatment indicate a potential broad application for the treatment of synaptopathy, which often goes undetected until well after the original damaging exposures.


Subject(s)
Hearing Loss, Noise-Induced/drug therapy , Membrane Glycoproteins/agonists , Amitriptyline/administration & dosage , Amitriptyline/pharmacology , Animals , Auditory Threshold/drug effects , Auditory Threshold/physiology , Cochlea/drug effects , Cochlea/physiopathology , Cochlear Nerve/drug effects , Cochlear Nerve/physiopathology , Coculture Techniques , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/drug effects , Evoked Potentials, Auditory, Brain Stem/physiology , Flavones/administration & dosage , Flavones/pharmacology , Hair Cells, Auditory, Inner/drug effects , Hair Cells, Auditory, Inner/physiology , Hearing Loss, Noise-Induced/physiopathology , Membrane Glycoproteins/physiology , Mice , Mice, Inbred CBA , Protein-Tyrosine Kinases/physiology , Regeneration/drug effects , Regeneration/physiology , Synapses/drug effects , Synapses/physiology
7.
Neuroscience ; 427: 43-57, 2020 02 10.
Article in English | MEDLINE | ID: mdl-31887361

ABSTRACT

Prior work has provided extensive documentation of threshold sensitivity and sensory hair cell losses after noise exposure. It is now clear, however, that cochlear synaptic loss precedes such losses, at least at low-moderate noise doses, silencing affected neurons. To address questions of whether, and how, cochlear synaptopathy and underlying mechanisms change as noise dose is varied, we assessed cochlear physiologic and histologic consequences of a range of exposures varied in duration from 15 min to 8 h and in level from 85 to 112 dB SPL. Exposures delivered to adult CBA/CaJ mice produced acute elevations in hair cell- and neural-based response thresholds ranging from trivial (∼5 dB) to large (∼50 dB), followed by varying degrees of recovery. Males appeared more noise vulnerable for some conditions of exposure. There was little to no inner hair cell (IHC) loss, but outer hair cell (OHC) loss could be substantial at highest frequencies for highest noise doses. Synapse loss was an early manifestation of noise injury and did not scale directly with either temporary or permanent threshold shift. With increasing noise dose, synapse loss grew to ∼50%, then declined for exposures yielding permanent hair cell injury/loss. All synaptopathic, but no non-synaptopathic exposures produced persistent neural response amplitude declines; those additionally yielding permanent OHC injury/loss also produced persistent reductions in OHC-based responses and exaggerated neural amplitude declines. Findings show that widespread cochlear synaptopathy can be present with and without noise-induced sensory cell loss and that differing patterns of cellular injury influence synaptopathic outcomes.


Subject(s)
Cochlea/pathology , Hair Cells, Auditory, Outer/pathology , Hair Cells, Auditory, Outer/physiology , Synapses/pathology , Animals , Auditory Threshold/physiology , Hair Cells, Auditory, Inner/pathology , Hearing Loss, Noise-Induced/pathology , Hearing Loss, Noise-Induced/physiopathology , Mice , Mice, Inbred CBA , Noise/adverse effects , Sex Factors
8.
Hear Res ; 377: 44-52, 2019 06.
Article in English | MEDLINE | ID: mdl-30903954

ABSTRACT

Acquired sensorineural hearing loss is one of the most prevalent chronic diseases, and aging and acoustic overexposure are common contributors. Decades of study in animals and humans have clarified the cellular targets and perceptual consequences of these forms of hearing loss, and preclinical studies have led to the development of therapeutics designed to slow, prevent or reverse them. Here, we review the histopathological changes underlying age-related and noise-induced hearing loss and the functional consequences of these pathologies. Based on these relations, we consider the ambiguities that arise in diagnosing underlying pathology from minimally invasive tests of auditory function, and how those ambiguities present challenges in the design and interpretation of clinical trials.


Subject(s)
Cochlea/physiopathology , Hearing Loss, Noise-Induced/therapy , Presbycusis/therapy , Translational Research, Biomedical , Age Factors , Aging , Animals , Auditory Perception , Cochlea/pathology , Disease Models, Animal , Hearing , Hearing Loss, Noise-Induced/etiology , Hearing Loss, Noise-Induced/pathology , Hearing Loss, Noise-Induced/physiopathology , Humans , Noise , Presbycusis/pathology , Presbycusis/physiopathology , Species Specificity
9.
Neuroscience ; 407: 21-31, 2019 05 21.
Article in English | MEDLINE | ID: mdl-30553793

ABSTRACT

Aging listeners often experience difficulties in perceiving temporally complex acoustic cues in noisy environments. These difficulties likely have neurophysiological contributors from various levels of auditory processing. Cochlear synapses between inner hair cells and auditory nerve fibers exhibit a progressive decline with age which is not reflected in the threshold audiogram. The functional consequences of this loss for the coding of suprathreshold sound remain poorly understood. Recent studies suggest that cochlear synaptopathy results in degraded representations of temporal envelope cues at the earliest levels of the auditory pathway. Central nuclei downstream of the auditory nerve exhibit a compensatory plasticity in response to this deafferentation, in the form of altered gain. This results in a modulation frequency selective increase in the representation of envelope cues at the level of the auditory midbrain and cortex. These changes may be shaped by mechanisms such as decreased inhibitory neurotransmission occurring with age across various central auditory nuclei. Altered representations of the differing temporal components of speech due to these interactions between multiple levels of the auditory pathway may contribute to the age-related difficulties hearing speech in noisy environments.


Subject(s)
Auditory Perception/physiology , Auditory Threshold/physiology , Cochlear Nerve/physiology , Hearing/physiology , Aging , Animals , Evoked Potentials, Auditory, Brain Stem/physiology , Hearing Loss, Noise-Induced/physiopathology , Humans
10.
Hear Res ; 370: 113-119, 2018 12.
Article in English | MEDLINE | ID: mdl-30366194

ABSTRACT

Cochlear synaptopathy, the loss of synaptic connections between inner hair cells (IHCs) and auditory nerve fibers, has been documented in animal models of aging, noise, and ototoxic drug exposure, three common causes of acquired sensorineural hearing loss in humans. In each of these models, synaptopathy begins prior to changes in threshold sensitivity or loss of hair cells; thus, this underlying injury can be hidden behind a normal threshold audiogram. Since cochlear synaptic loss cannot be directly confirmed in living humans, non-invasive assays will be required for diagnosis. In animals with normal auditory thresholds, the amplitude of wave 1 of the auditory brainstem response (ABR) is highly correlated with synapse counts. However, synaptopathy can also co-occur with threshold elevation, complicating the use of the ABR alone as a diagnostic measure. Using an age-graded series of mice and a partial least squares regression approach to model structure-function relationships, this study shows that the combination of a small number of ABR and distortion product otoacoustic emission (DPOAE) measurements can predict synaptic ribbon counts at various cochlear frequencies to within 1-2 synapses per IHC of their true value. In contrast, the model, trained using the age-graded series of mice, overpredicted synapse counts in a small sample of young noise-exposed mice, perhaps due to differences in the underlying pattern of damage between aging and noise-exposed mice. These results provide partial validation of a noninvasive approach to identify synaptic/neuronal loss in humans using ABRs and DPOAEs.


Subject(s)
Cochlea/pathology , Cochlear Diseases/diagnosis , Evoked Potentials, Auditory, Brain Stem , Hearing Loss, Noise-Induced/diagnosis , Synapses/pathology , Acoustic Stimulation , Age Factors , Alcohol Oxidoreductases , Animals , Auditory Threshold , Biomarkers/metabolism , Co-Repressor Proteins , Cochlea/metabolism , Cochlear Diseases/metabolism , Cochlear Diseases/pathology , Cochlear Diseases/physiopathology , DNA-Binding Proteins/metabolism , Disease Models, Animal , Hearing Loss, Noise-Induced/metabolism , Hearing Loss, Noise-Induced/pathology , Hearing Loss, Noise-Induced/physiopathology , Immunohistochemistry , Least-Squares Analysis , Machine Learning , Male , Mice, Inbred CBA , Noise , Phosphoproteins/metabolism , Predictive Value of Tests , Receptors, AMPA/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Synapses/metabolism
11.
Cell ; 174(5): 1229-1246.e17, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30078709

ABSTRACT

In the auditory system, type I spiral ganglion neurons (SGNs) convey complex acoustic information from inner hair cells (IHCs) to the brainstem. Although SGNs exhibit variation in physiological and anatomical properties, it is unclear which features are endogenous and which reflect input from synaptic partners. Using single-cell RNA sequencing, we derived a molecular classification of mouse type I SGNs comprising three subtypes that express unique combinations of Ca2+ binding proteins, ion channel regulators, guidance molecules, and transcription factors. Based on connectivity and susceptibility to age-related loss, these subtypes correspond to those defined physiologically. Additional intrinsic differences among subtypes and across the tonotopic axis highlight an unexpectedly active role for SGNs in auditory processing. SGN identities emerge postnatally and are disrupted in a mouse model of deafness that lacks IHC-driven activity. These results elucidate the range, nature, and origins of SGN diversity, with implications for treatment of congenital deafness.


Subject(s)
Ear, Inner/physiology , Hair Cells, Auditory, Inner/physiology , Sensory Receptor Cells/physiology , Amino Acid Transport Systems, Acidic/genetics , Animals , Calbindin 2/genetics , Cochlea/physiology , Deafness/genetics , Female , Male , Mice , Mice, Inbred C57BL , Sequence Analysis, RNA , Spiral Ganglion/physiology , Synaptic Transmission , Transgenes
12.
J Neurosci ; 38(32): 7108-7119, 2018 08 08.
Article in English | MEDLINE | ID: mdl-29976623

ABSTRACT

Aging listeners, even in the absence of overt hearing loss measured as changes in hearing thresholds, often experience impairments processing temporally complex sounds such as speech in noise. Recent evidence has shown that normal aging is accompanied by a progressive loss of synapses between inner hair cells and auditory nerve fibers. The role of this cochlear synaptopathy in degraded temporal processing with age is not yet understood. Here, we used population envelope following responses, along with other hair cell- and neural-based measures from an age-graded series of male and female CBA/CaJ mice to study changes in encoding stimulus envelopes. By comparing responses obtained before and after the application of the neurotoxin ouabain to the inner ear, we demonstrate that we can study changes in temporal processing on either side of the cochlear synapse. Results show that deficits in neural coding with age emerge at the earliest neural stages of auditory processing and are correlated with the degree of cochlear synaptopathy. These changes are seen before losses in neural thresholds and particularly affect the suprathreshold processing of sound. Responses obtained from more central sources show smaller differences with age, suggesting compensatory gain. These results show that progressive cochlear synaptopathy is accompanied by deficits in temporal coding at the earliest neural generators and contribute to the suprathreshold sound processing deficits observed with age.SIGNIFICANCE STATEMENT Aging listeners often experience difficulty hearing and understanding speech in noisy conditions. The results described here suggest that age-related loss of cochlear synapses may be a significant contributor to those performance declines. We observed aberrant neural coding of sounds in the early auditory pathway, which was accompanied by and correlated with an age-progressive loss of synapses between the inner hair cells and the auditory nerve. Deficits first appeared before changes in hearing thresholds and were largest at higher sound levels relevant to real world communication. The noninvasive tests described here may be adapted to detect cochlear synaptopathy in the clinical setting.


Subject(s)
Aging/physiology , Cochlea/physiopathology , Evoked Potentials, Auditory/physiology , Hearing Loss, Sensorineural/physiopathology , Synapses/pathology , Acoustic Stimulation , Afferent Pathways , Animals , Auditory Threshold , Cochlea/growth & development , Cochlea/pathology , Cochlear Nerve/physiopathology , Evoked Potentials, Auditory, Brain Stem/physiology , Female , Hair Cells, Auditory, Inner/drug effects , Hair Cells, Auditory, Inner/physiology , Hearing Loss, Sensorineural/pathology , Male , Mice , Mice, Inbred CBA , Otoacoustic Emissions, Spontaneous/physiology , Ouabain/toxicity , Time Factors
13.
Hear Res ; 349: 138-147, 2017 06.
Article in English | MEDLINE | ID: mdl-28087419

ABSTRACT

Common causes of hearing loss in humans - exposure to loud noise or ototoxic drugs and aging - often damage sensory hair cells, reflected as elevated thresholds on the clinical audiogram. Recent studies in animal models suggest, however, that well before this overt hearing loss can be seen, a more insidious, but likely more common, process is taking place that permanently interrupts synaptic communication between sensory inner hair cells and subsets of cochlear nerve fibers. The silencing of affected neurons alters auditory information processing, whether accompanied by threshold elevations or not, and is a likely contributor to a variety of perceptual abnormalities, including speech-in-noise difficulties, tinnitus and hyperacusis. Work described here will review structural and functional manifestations of this cochlear synaptopathy and will consider possible mechanisms underlying its appearance and progression in ears with and without traditional 'hearing loss' arising from several common causes in humans.


Subject(s)
Cochlear Nerve/pathology , Hair Cells, Auditory, Inner/pathology , Hearing Loss, Noise-Induced/pathology , Hearing Loss, Sensorineural/pathology , Hearing , Synapses/pathology , Animals , Auditory Perception , Auditory Threshold , Cochlear Nerve/metabolism , Cochlear Nerve/physiopathology , Glutamic Acid/metabolism , Hair Cells, Auditory, Inner/metabolism , Hearing Loss, Noise-Induced/metabolism , Hearing Loss, Noise-Induced/physiopathology , Hearing Loss, Noise-Induced/psychology , Hearing Loss, Sensorineural/metabolism , Hearing Loss, Sensorineural/physiopathology , Hearing Loss, Sensorineural/psychology , Humans , Nerve Degeneration , Noise/adverse effects , Risk Factors , Synapses/metabolism , Synaptic Transmission
14.
Otol Neurotol ; 37(8): e271-5, 2016 09.
Article in English | MEDLINE | ID: mdl-27518135

ABSTRACT

OBJECTIVE: To review basic and clinical findings relevant to defining temporary (TTS) and permanent (PTS) threshold shifts and their sequelae. DATA SOURCES: Relevant scientific literature and government definitions were broadly reviewed. DATA SYNTHESIS: The definitions and characteristics of TTS and PTS were assessed and recent advances that expand our knowledge of the extent, nature, and consequences of noise-induced hearing loss were reviewed. CONCLUSION: Exposure to intense sound can produce TTS, acute changes in hearing sensitivity that recover over time, or PTS, a loss that does not recover to preexposure levels. In general, a threshold shift ≥10 dB at 2, 3, and 4 kHz is required for reporting purposes in human studies. The high-frequency regions of the cochlea are most sensitive to noise damage. Resonance of the ear canal also results in a frequency region of high-noise sensitivity at 4 to 6 kHz. A primary noise target is the cochlear hair cell. Although the mechanisms that underlie such hair cell damage remain unclear, there is evidence to support a role for reactive oxygen species, stress pathway signaling, and apoptosis. Another target is the synapse between the hair cell and the primary afferent neurons. Large numbers of these synapses and their neurons can be lost after noise, even though hearing thresholds may return to normal. This affects auditory processing and detection of signals in noise. The consequences of TTS and PTS include significant deficits in communication that can impact performance of military duties or obtaining/retaining civilian employment. Tinnitus and exacerbation of posttraumatic stress disorder are also potential sequelae.


Subject(s)
Auditory Threshold/physiology , Hearing Loss, Noise-Induced/physiopathology , Animals , Humans , Noise
15.
Ear Hear ; 37(5): 560-71, 2016.
Article in English | MEDLINE | ID: mdl-27050773

ABSTRACT

OBJECTIVE: The objective is to develop methods to utilize newborn reflectance measures for the identification of middle-ear transient conditions (e.g., middle-ear fluid) during the newborn period and ultimately during the first few months of life. Transient middle-ear conditions are a suspected source of failure to pass a newborn hearing screening. The ability to identify a conductive loss during the screening procedure could enable the referred ear to be either (1) cleared of a middle-ear condition and recommended for more extensive hearing assessment as soon as possible, or (2) suspected of a transient middle-ear condition, and if desired, be rescreened before more extensive hearing assessment. DESIGN: Reflectance measurements are reported from full-term, healthy, newborn babies in which one ear referred and one ear passed an initial auditory brainstem response newborn hearing screening and a subsequent distortion product otoacoustic emission screening on the same day. These same subjects returned for a detailed follow-up evaluation at age 1 month (range 14 to 35 days). In total, measurements were made on 30 subjects who had a unilateral refer near birth (during their first 2 days of life) and bilateral normal hearing at follow-up (about 1 month old). Three specific comparisons were made: (1) Association of ear's state with power reflectance near birth (referred versus passed ear), (2) Changes in power reflectance of normal ears between newborn and 1 month old (maturation effects), and (3) Association of ear's newborn state (referred versus passed) with ear's power reflectance at 1 month. In addition to these measurements, a set of preliminary data selection criteria were developed to ensure that analyzed data were not corrupted by acoustic leaks and other measurement problems. RESULTS: Within 2 days of birth, the power reflectance measured in newborn ears with transient middle-ear conditions (referred newborn hearing screening and passed hearing assessment at age 1 month) was significantly greater than power reflectance on newborn ears that passed the newborn hearing screening across all frequencies (500 to 6000 Hz). Changes in power reflectance in normal ears from newborn to 1 month appear in approximately the 2000 to 5000 Hz range but are not present at other frequencies. The power reflectance at age 1 month does not depend significantly on the ear's state near birth (refer or pass hearing screening) for frequencies above 700 Hz; there might be small differences at lower frequencies. CONCLUSIONS: Power reflectance measurements are significantly different for ears that pass newborn hearing screening and ears that refer with middle-ear transient conditions. At age 1 month, about 90% of ears that referred at birth passed an auditory brainstem response hearing evaluation; within these ears the power reflectance at 1 month did not differ between the ear that initially referred at birth and the ear that passed the hearing screening at birth for frequencies above 700 Hz. This study also proposes a preliminary set of criteria for determining when reflectance measures on young babies are corrupted by acoustic leaks, probes against the ear canal, or other measurement problems. Specifically proposed are "data selection criteria" that depend on the power reflectance, impedance magnitude, and impedance angle. Additional data collected in the future are needed to improve and test these proposed criteria.


Subject(s)
Ear, Middle/physiopathology , Evoked Potentials, Auditory, Brain Stem/physiology , Hearing Loss, Conductive/physiopathology , Otoacoustic Emissions, Spontaneous/physiology , Female , Healthy Volunteers , Hearing Loss, Conductive/diagnosis , Humans , Infant, Newborn , Male , Neonatal Screening
16.
Lab Chip ; 16(5): 829-46, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26778829

ABSTRACT

The anatomical and pharmacological inaccessibility of the inner ear is a major challenge in drug-based treatment of auditory disorders. This also makes pharmacokinetic characterization of new drugs with systemic delivery challenging, because efficacy is coupled with how efficiently a drug can reach its target. Direct delivery of drugs to cochlear fluids bypasses pharmacokinetic barriers and helps to minimize systemic toxicity, but anatomical barriers make administration of multiple doses difficult without an automated delivery system. Such a system may be required for hair-cell regeneration treatments, which will likely require timed delivery of several drugs. To address these challenges, we have developed a micropump for controlled, automated inner-ear drug delivery with the ultimate goal of producing a long-term implantable/wearable delivery system. The current pump is designed to be used with a head mount for guinea pigs in preclinical drug characterization experiments. In this system, we have addressed several microfluidic challenges, including maintaining controlled delivery at safe, low flow rates and delivering drug without increasing the volume of fluid in the cochlea. By integrating a drug reservoir and all fluidic components into the microfluidic structure of the pump, we have made the drug delivery system robust compared to previous systems that utilized separate, tubing-connected components. In this study, we characterized the pump's unique infuse-withdraw and on-demand dosing capabilities on the bench and in guinea pig animal models. For the animal experiments, we used DNQX, a glutamate receptor antagonist, as a physiological indicator of drug delivery. DNQX suppresses compound action potentials (CAPs), so we were able to infer the distribution and spreading of the DNQX over time by measuring the changes in CAPs in response to stimuli at several characteristic frequencies.


Subject(s)
Cochlea , Drug Delivery Systems , Infusion Pumps , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques , Animals , Drug Delivery Systems/instrumentation , Drug Delivery Systems/methods , Guinea Pigs , Humans , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods
17.
J Neurosci ; 35(19): 7509-20, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25972177

ABSTRACT

Cochlear synaptic loss, rather than hair cell death, is the earliest sign of damage in both noise- and age-related hearing impairment (Kujawa and Liberman, 2009; Sergeyenko et al., 2013). Here, we compare cochlear aging after two types of noise exposure: one producing permanent synaptic damage without hair cell loss and another producing neither synaptopathy nor hair cell loss. Adult mice were exposed (8-16 kHz, 100 or 91 dB SPL for 2 h) and then evaluated from 1 h to ∼ 20 months after exposure. Cochlear function was assessed via distortion product otoacoustic emissions and auditory brainstem responses (ABRs). Cochlear whole mounts and plastic sections were studied to quantify hair cells, cochlear neurons, and the synapses connecting them. The synaptopathic noise (100 dB) caused 35-50 dB threshold shifts at 24 h. By 2 weeks, thresholds had recovered, but synaptic counts and ABR amplitudes at high frequencies were reduced by up to ∼ 45%. As exposed animals aged, synaptopathy was exacerbated compared with controls and spread to lower frequencies. Proportional ganglion cell losses followed. Threshold shifts first appeared >1 year after exposure and, by ∼ 20 months, were up to 18 dB greater in the synaptopathic noise group. Outer hair cell losses were exacerbated in the same time frame (∼ 10% at 32 kHz). In contrast, the 91 dB exposure, producing transient threshold shift without acute synaptopathy, showed no acceleration of synaptic loss or cochlear dysfunction as animals aged, at least to ∼ 1 year after exposure. Therefore, interactions between noise and aging may require an acute synaptopathy, but a single synaptopathic exposure can accelerate cochlear aging.


Subject(s)
Aging/physiology , Cochlea/cytology , Evoked Potentials, Auditory, Brain Stem/physiology , Otoacoustic Emissions, Spontaneous/physiology , Synapses/pathology , Acoustic Stimulation , Alcohol Oxidoreductases , Animals , Auditory Threshold , Cell Death , Co-Repressor Proteins , DNA-Binding Proteins/metabolism , Hair Cells, Auditory/pathology , Hearing Loss, Noise-Induced/physiopathology , Mice , Mice, Inbred CBA , Microscopy, Electron, Transmission , Noise/adverse effects , Phosphoproteins/metabolism , Receptors, AMPA/metabolism , Spiral Ganglion/pathology , Synapses/ultrastructure , Time Factors , Wakefulness/physiology
18.
Hear Res ; 330(Pt B): 191-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25769437

ABSTRACT

The classic view of sensorineural hearing loss (SNHL) is that the "primary" targets are hair cells, and that cochlear-nerve loss is "secondary" to hair cell degeneration. Our recent work in mouse and guinea pig has challenged that view. In noise-induced hearing loss, exposures causing only reversible threshold shifts (and no hair cell loss) nevertheless cause permanent loss of >50% of cochlear-nerve/hair-cell synapses. Similarly, in age-related hearing loss, degeneration of cochlear synapses precedes both hair cell loss and threshold elevation. This primary neural degeneration has remained hidden for three reasons: 1) the spiral ganglion cells, the cochlear neural elements commonly assessed in studies of SNHL, survive for years despite loss of synaptic connection with hair cells, 2) the synaptic terminals of cochlear nerve fibers are unmyelinated and difficult to see in the light microscope, and 3) the degeneration is selective for cochlear-nerve fibers with high thresholds. Although not required for threshold detection in quiet (e.g. threshold audiometry or auditory brainstem response threshold), these high-threshold fibers are critical for hearing in noisy environments. Our research suggests that 1) primary neural degeneration is an important contributor to the perceptual handicap in SNHL, and 2) in cases where the hair cells survive, neurotrophin therapies can elicit neurite outgrowth from spiral ganglion neurons and re-establishment of their peripheral synapses. This article is part of a Special Issue entitled .


Subject(s)
Cochlea/innervation , Cochlear Nerve/pathology , Hearing Loss, Noise-Induced/pathology , Hearing Loss, Sensorineural/pathology , Hearing , Nerve Degeneration , Noise/adverse effects , Synaptic Transmission , Acoustic Stimulation , Animals , Auditory Pathways/pathology , Auditory Pathways/physiopathology , Auditory Perception , Auditory Threshold , Cochlear Nerve/physiopathology , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem , Guinea Pigs , Hair Cells, Auditory/pathology , Hearing Loss, Noise-Induced/physiopathology , Hearing Loss, Sensorineural/physiopathology , Hearing Loss, Sensorineural/psychology , Humans , Mice
19.
Biomed Microdevices ; 17(2): 37, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25686902

ABSTRACT

One of the major challenges in treatment of auditory disorders is that many therapeutic compounds are toxic when delivered systemically. Local intracochlear delivery methods are becoming critical in emerging treatments and in drug discovery. Direct infusion via cochleostomy, in particular, is attractive from a pharmacokinetics standpoint, as there is potential for the kinetics of delivery to be well-controlled. Direct infusion is compatible with a large number of drug types, including large, complex molecules such as proteins and unstable molecules such as siRNA. In addition, hair-cell regeneration therapy will likely require long-term delivery of a timed series of agents. This presents unknown risks associated with increasing the volume of fluid within the cochlea and mechanical damage caused during delivery. There are three key requirements for an intracochlear drug delivery system: (1) a high degree of miniaturization (2) a method for pumping precise and small volumes of fluid into the cochlea in a highly controlled manner, and (3) a method for removing excess fluid from the limited cochlear fluid space. To that end, our group is developing a head-mounted microfluidics-based system for long-term intracochlear drug delivery. We utilize guinea pig animal models for development and demonstration of the device. Central to the system is an infuse-withdraw micropump component that, unlike previous micropump-based systems, has fully integrated drug and fluid storage compartments. Here we characterize the infuse-withdraw capabilities of our micropump, and show experimental results that demonstrate direct drug infusion via cochleostomy in animal models. We utilized DNQX, a glutamate receptor antagonist that suppresses CAPs, as a test drug. We monitored the frequency-dependent changes in auditory nerve CAPs during drug infusion, and observed CAP suppression consistent with the expected drug transport path based on the geometry and tonotopic organization of the cochlea.


Subject(s)
Cochlea , Drug Delivery Systems/instrumentation , Infusion Pumps , Microfluidics/instrumentation , Animals , Cochlea/drug effects , Drug Administration Routes , Drug Delivery Systems/methods , Equipment Design , Guinea Pigs , Male , Microtechnology , Miniaturization , Quinoxalines/administration & dosage
20.
Otol Neurotol ; 35(10): 1825-34, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25393974

ABSTRACT

OBJECTIVE: Otologic trauma was the most common physical injury sustained after the April 15, 2013, Boston Marathon bombings. The goal of this study is to describe the resultant otologic morbidity and to report on early outcomes. STUDY DESIGN: Multi-institutional prospective cohort study. METHODS: Children and adults seen for otologic complaints related to the Boston Marathon bombings comprised the study population. Participants completed symptom assessments, quality-of-life questionnaires, and audiograms at initial and 6-month visits. Otologic evaluation and treatment, including tympanoplasty results, were reviewed. RESULTS: More than 100 patients from eight medical campuses have been evaluated for blast-related otologic injuries; 94 have enrolled. Only 7% had any otologic symptoms before the blasts. Ninety percent of hospitalized patients sustained tympanic membrane perforation. Proximity to blast (RR = 2.7, p < 0.01) and significant nonotologic injury (RR = 2.7, p < 0.01) were positive predictors of perforation. Spontaneous healing occurred in 38% of patients, and tympanoplasty success was 86%. After oral steroid therapy in eight patients, improvement in hearing at 2 and 4 kHz was seen, although changes did not reach statistical significance. Hearing loss, tinnitus, hyperacusis, and difficulty hearing in noise remain persistent and, in some cases, progressive complaints for patients. Otologic-specific quality of life was impaired in this population. CONCLUSION: Blast-related otologic injuries constitute a major source of ongoing morbidity after the Boston Marathon bombings. Continued follow-up and care of this patient population are warranted.


Subject(s)
Blast Injuries/complications , Explosions , Hearing Loss/etiology , Hyperacusis/etiology , Tinnitus/etiology , Tympanic Membrane Perforation/etiology , Adrenal Cortex Hormones/therapeutic use , Adult , Blast Injuries/psychology , Blast Injuries/therapy , Child , Cohort Studies , Female , Hearing Loss/psychology , Hearing Tests , Humans , Hyperacusis/psychology , Male , Prospective Studies , Quality of Life/psychology , Surveys and Questionnaires , Tinnitus/psychology , Tympanic Membrane Perforation/psychology , Tympanic Membrane Perforation/therapy , Tympanoplasty
SELECTION OF CITATIONS
SEARCH DETAIL
...