Next Article in Journal
Effects of Radiofrequency Diathermy Plus Therapeutic Exercises on Pain and Functionality of Patients with Patellofemoral Pain Syndrome: A Randomized Controlled Trial
Next Article in Special Issue
Comparative Transcriptomic Analysis of Archival Human Vestibular Schwannoma Tissue from Patients with and without Tinnitus
Previous Article in Journal
Circulating ANGPTL8 as a Potential Protector of Metabolic Complications in Patients with Psoriasis
Previous Article in Special Issue
Associations of Tinnitus Incidence with Use of Tumor Necrosis Factor-Alpha Inhibitors among Patients with Autoimmune Conditions
 
 
Correction published on 7 February 2024, see J. Clin. Med. 2024, 13(4), 944.
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Noise-Induced Hearing Loss

by
Nirvikalpa Natarajan
1,
Shelley Batts
1 and
Konstantina M. Stankovic
1,2,3,*
1
Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA 94304, USA
2
Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA 94304, USA
3
Wu Tsai Neuroscience Institute, Stanford University, Stanford, CA 94305, USA
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2023, 12(6), 2347; https://doi.org/10.3390/jcm12062347
Submission received: 7 February 2023 / Revised: 10 March 2023 / Accepted: 14 March 2023 / Published: 17 March 2023 / Corrected: 7 February 2024

Abstract

:
Noise-induced hearing loss (NIHL) is the second most common cause of sensorineural hearing loss, after age-related hearing loss, and affects approximately 5% of the world’s population. NIHL is associated with substantial physical, mental, social, and economic impacts at the patient and societal levels. Stress and social isolation in patients’ workplace and personal lives contribute to quality-of-life decrements which may often go undetected. The pathophysiology of NIHL is multifactorial and complex, encompassing genetic and environmental factors with substantial occupational contributions. The diagnosis and screening of NIHL are conducted by reviewing a patient’s history of noise exposure, audiograms, speech-in-noise test results, and measurements of distortion product otoacoustic emissions and auditory brainstem response. Essential aspects of decreasing the burden of NIHL are prevention and early detection, such as implementation of educational and screening programs in routine primary care and specialty clinics. Additionally, current research on the pharmacological treatment of NIHL includes anti-inflammatory, antioxidant, anti-excitatory, and anti-apoptotic agents. Although there have been substantial advances in understanding the pathophysiology of NIHL, there remain low levels of evidence for effective pharmacotherapeutic interventions. Future directions should include personalized prevention and targeted treatment strategies based on a holistic view of an individual’s occupation, genetics, and pathology.

1. Introduction

Noise-induced hearing loss (NIHL) is a consequence of multifactorial damage to auditory structures following exposure to occupational, environmental, or recreational sources of loud sound. Noise has been recognized as a factor contributing to hearing loss long before rigorous data collection, sophisticated analyses, and careful experimental design became the norm. Although earplugs were patented in 1864, hearing protection devices are mentioned in ancient Greek mythology [1]. NIHL was formally acknowledged as a medical condition in the United States (US) during the Industrial Revolution, first named ‘boilermaker’s disease’ as a reference to the hearing loss suffered by workers building engines that powered transportation and production [2]. Historical data on US women who worked in the factories during World War I and II reveal devastating health effects, including hearing loss, although disorders caused by exposure to chemicals received more attention than those attributable to noise [3]. Noted physician and Nobel Prize winner Robert Koch predicted in 1910 that “one day man will have to fight noise as fiercely as cholera and pest” [4]. Despite this prediction and the long-standing knowledge of the adverse effects of noise on hearing and extensive research in the modern era, hearing loss continues to rank among the most common work-related illnesses both in the US and the world [5].
NIHL may be unilateral (affecting one ear) or bilateral (affecting both ears), and the hearing deficits may be transient or permanent [6]. The duration and severity of NIHL depends on the extent and location of cellular damage, which correlates with intensity and duration of the sound stimulus. Because the mammalian auditory sensory epithelium—the organ of Corti—does not spontaneously regenerate when sensory cells are lost, noise-induced hair cell or neural degeneration can result in permanent hearing loss particularly in the setting of repeated exposure [6,7]. Furthermore, NIHL is frequently irreversible and can have a profoundly negative impact on an individual’s quality of life and on the economy and society at large. However, NIHL is largely a preventable condition when appropriate precautions, such as the use of hearing protection, can be taken. Therefore, implementing measures to detect and attenuate causative factors, raising awareness of the condition and implementing protective strategies, and developing therapies that protect against or mitigate damage from noise exposure can aid in the prevention of this common condition. In this review, we describe the epidemiology, anatomy, pathophysiology, diagnosis, and prevention strategies of NIHL and protective pharmacological agents which have demonstrated some efficacy in humans, and we close with an outlook on emerging therapies.

2. Epidemiology

The most recent Global Burden of Disease report (2019) estimated that 1.57 billion people, or 20.3% of the world population, are affected by any kind of hearing loss, with 62% over the age of 50 years [8]. As NIHL is the second most common cause of hearing loss after presbycusis (age-related hearing loss) [9,10], it imposes an enormous burden on individuals and health systems. Globally, NIHL is estimated to affect approximately 5% of the population and is generally more common among adult men [11,12]. However, this may be an underestimate as the prevalence of NIHL varies widely across populations and age groups. For example, greater exposure to occupational and urban noise in developing nations increases the risk of NIHL, and limited access to healthcare and screening tests may leave much of the burden undetected [13,14,15]. Further, developing nations may lack governmental guidance or legislation to limit noise exposure or lack public education measures to encourage the use of hearing protection. This can be observed in data on the prevalence of occupational NIHL across nations within the same geographic region. Approximately 16% of disabling hearing loss in adults globally is attributed to occupational noise, ranging from 7 to 21% across various geographic regions [16]. Both the highest and lowest rates are in the West Pacific region (as defined by the World Health Organization (WHO)): rates are lowest among developed nations including Australia, Japan, New Zealand, and Singapore, and the highest in the region include Cambodia, Laos, Malaysia, Philippines, and Vietnam [16].
Due to lack of public records and funding for research on NIHL, the true burden of NIHL in the developing world is unknown, although several studies in India and Africa have highlighted it [17,18]. For example, a survey of the effects of urban noise on traffic police in Hyderabad, India, reported that 76% had NIHL, and high rates of NIHL were observed in workers in textile, mining, and heavy engineering industries [17]. Similarly, a cross-sectional study reported that the prevalence of NIHL among Tanzanian iron/steel workers was significantly higher than among schoolteachers (48% vs. 31%) but was high in both groups [19].
NIHL is also common among developed nations. In the US, interviews and hearing tests conducted by the Centers for Disease Control and Prevention (CDC) National Health and Nutrition Examination Survey (NHANES) led to estimates that at least 6% and up to 24% of adults have hearing loss in one or both ears due to noise exposure, with higher rates among males [20,21]. An estimated 17% of US youths (aged 12–19 years) have hearing tests indicative of NIHL, with higher rates among females, primarily attributable to unsafe recreational noise levels [22]. In the European Union, noise is a main cause of disabling hearing loss, affecting over 34.4 million people in 2019 and contributing to over 185 billion euros in annual costs related to reduced quality of life and lost productivity [23]. NIHL is also reported to be more prevalent in eastern and central Europe compared to western Europe [24]. In the United Kingdom (UK), approximately 180,000 people aged 35–64 years were estimated to have severe hearing difficulties attributable to noise at work in 2002 [25]. NIHL and occupational deafness have been declining in the UK but are still estimated to impact over 11,000 workers annually [26].

3. Pathophysiology

3.1. Harmful Noise Levels

Sound frequency is measured in Hertz (Hz), and intensity (loudness) is measured by sound pressure level on a logarithmic decibel (dB) scale, which ranges from safe to unsafe exposure levels (Figure 1). Normal human sound discrimination typically begins at 0 dB within the frequency ranges of 20 Hz to 20,000 kHz [27]. In comparison, a normal conversation is approximately 60 dB, traffic is 80 dB, very loud music at a rock concert or nightclub is 120 dB, and a jet engine is 140 dB. Broad categories of noise exposure include continuous noise, which is sustained over time, and impulsive noise, which occurs rapidly (i.e., a gunshot or explosion) and is generally at a higher sound pressure level [28]. A sound pressure level above 110 dB is considered discomfort threshold, and above 130 dB is the pain threshold [3]. Sustained noise above 70 dB can result in cumulative hearing loss while noise above 120 dB can cause immediate hearing loss [29].

3.2. Auditory Structures and Functions Impacted by Noise

3.2.1. Sound Transmission from the Outer to the Middle and Inner Ear

The damage associated with NIHL begins when harmful sound is channeled to auditory structures from the outer ear via the auditory canal (Figure 2a). The unique shape of the auditory canal, where it is closed at one end by the tympanic membrane and open at the lateral end, serves as a quarter-wave resonator tube funneling sound to the middle ear [32]. Sound waves vibrate the tympanic membrane which transmits information about the frequency and amplitude of sound through the air-filled middle ear cavity via the tiny ossicles (malleus, incus, and finally to the stapes) [33]. Amplification of sound pressure is a key feature of the transmission of sound from the middle to inner ear, allowing the stapes push against the higher resistance fluid in the inner ear behind the oval window [34,35,36]. As a result of the larger surface area of the tympanic membrane compared to the cochlea’s oval window (a ratio of 20:1), the sound pressure ultimately received by the oval window is approximately 20 times greater than the original sound stimulus [37]. Resonance in the ear canal produces amplification of acoustic frequencies whose wavelengths are approximately four times the length of the canal, which, in humans, results in enhancement of frequencies around 4 kHz [38].
The mammalian cochlea is a spiral-shaped structure divided into three fluid-filled chambers—the scala tympani, the scala vestibuli, and the scala media (i.e., the cochlear duct) (Figure 2b). In humans, the cochlea has a conical central canal (modiolus) around which the cochlear duct makes two- and three-quarter turns. The footplate of the stapes rests on the cochlea’s oval window and moves in a piston-like manner to transmit sound vibrations to the cochlear duct commensurate with the loudness and intensity of the sound stimulus. Accordingly, vibrations from displacement of the oval window spiral up the cochlea to cause physical movement of fluids and of the flexible basilar membrane. The basilar membrane does not vibrate as a whole; rather, specific areas vibrate in response to different sound frequencies. This is achieved by variations in stiffness and width along its length: it is thin and stiff at the base and broader and more flexible toward the apex. Therefore, lower frequencies vibrate the basilar membrane closer to the apex of the cochlea while higher frequencies produce vibrations closer to the base, near the oval window. This arrangement is known as a tonotopic organization [39,40]. Thus, our ability to differentiate sounds of varying loudness and pitch depends on the ability of the cochlea to respond appropriately to transmitted sound amplitudes and frequencies.

3.2.2. Auditory Transduction in the Organ of Corti

The cochlea’s scala media contains the organ of Corti, the cellular apparatus for transduction of sound vibration into neural signals that can be interpreted by the brain. The organ of Corti is a mosaic of sensory hair cells and non-sensory supporting cells which, in mammals, does not have the capacity to replace lost cells once damaged (Figure 2c). There are two types of sensory hair cells: a single row of inner hair cells (IHC; ~3500 in humans) and three to five rows of outer hair cells (OHC; ~12,000) [41]. Both types of hair cells have 3–4 rows of actin-rich “hairs” called stereocilia, which increase in height along the basilar membrane from base to apex. The roles of the supporting cells are to generally provide structural, metabolic, and immune support to the organ of Corti and are essential for hair cell survival. These include pillar cells, separating IHC and OHC to form the tunnel of Corti, as well as Deiters’, Hensen’s, and Claudius cells [42].
While the scala vestibuli communicates with the scala tympani at the apex of the cochlea (helicotrema), the scala media is separated from both chambers by Reissner’s membrane and the basilar membrane, respectively. This separation is crucial for the maintenance of the unique ionic composition of the extracellular fluids in these chambers, generating the endocochlear potential which is essential for auditory transduction. Specifically, the scala vestibuli and scala tympani contain perilymph which is K+-poor and Na+-rich. Conversely, the scala media is filled with K+-rich and Na+-poor endolymph which is secreted by a specialized epithelium called the stria vascularis. The tips of the hair cells’ stereocilia are embedded in the flexible tectorial membrane and move in response to sound vibrations. The stereocilia bundles are also connected to each other (within the same hair cell) via extracellular tip links formed in part by cadherin 23 (CDH23) and protocadherin 15 (PCDH15) proteins [43]. These links allow the stereocilia to move together and cause non-selective mechanically gated ion channels to open, conducting inward currents of cations (K+ and Ca2+) from endolymph [44].
While the endocochlear potential of endolymph in the scala media is +80 mV compared to perilymph [45], hair cells have a potential of −55 mV to up to −150 to mV [46]. Thus, the influx of cations depolarizes the hair cell, resulting in different effects in OHCs and IHCs. OHCs physically elongate and contract, activating the “cochlear amplifier” to narrow the area of excitation and give the traveling sound waves more sensitivity [47,48]. This aids in frequency discrimination and in the detection of quiet sounds. Meanwhile, IHCs release the neurotransmitter glutamate to synaptic boutons of type I (myelinated) spiral ganglion neurites (SGNs) which form the auditory nerve. Glutamate activates the AMPA glutamate receptors in the afferent nerve fibers which in turn transmit the neural signal to the cochlear nucleus and higher auditory processing regions. A single IHC synapses with approximately 10 boutons from 10 different spiral ganglion cells, but the number varies tonotopically (can be up to 20 boutons in the most sensitive frequency regions). IHCs have synaptic ribbons which sustain high rates of glutamate release and maintain synchronous auditory signaling [49,50,51,52]. The temporal change in neuronal firing is rapid in the ear, to the effect of 2 to 5 kHz, and distinctions between these various frequencies facilitates sound discrimination [51,53].

3.3. Mechanisms of Damage in NIHL

The mechanisms by which loud noise induces hearing loss include mechanical damage of cochlear structures, reduction in blood flow, sterile inflammation, and oxidative stress and excitotoxicity due to overstimulation of hair cells and nerves (Figure 3). The loss of hair cells via apoptosis is ultimately the most severe injury and contributes to permanent hearing loss [54,55]. Noise at specific frequencies can cause discrete areas of hair cell damage which manifest as frequency-specific hearing deficits [56,57].
The variables impacting the severity of damage include factors attributable to the sound stimulus (i.e., intensity, spectral energy of noise exposure, and duration) and the physical, mechanical, and chemical characteristics of the outer, middle, and inner ear. The damage caused by noise can cause temporary or permanent damage and associated hearing loss. Prolonged duration of exposure to hazardous noise levels or a one-time exposure to high-intensity sound levels can both cause permanent threshold shifts (PTS) [6,38,58]. PTS occurs when the standard threshold is stabilized at an elevated level due to the destruction of the cochlear hair cells, mechanosensory hair bundles, or nerve fibers [34,59]. Hearing loss persisting at 14 days after noise exposure, with the upper recovery limit being 30 days, is indicative of PTS [58]. Transient attenuation of hearing with recovery within 24–48 h is called a temporary threshold shift (TTS) and results from more moderate noise damage [60]. TTS and PTS show distinct histopathological patterns, as described below.

3.3.1. Mechanical Damage

Because the hair cells sit atop the basilar membrane and because their stereocilia are embedded in the tectorial membrane, they are subject to mechanical shearing forces in response to sound vibrations. This continuous mechanical stress causes damage over time and is a feature of typical age-related hearing loss. However, in the context of intense or persistent noise, the shearing forces can cause stereocilia core breakage, destruction of tip links, and ultimately premature hair cell death [61]. At low frequencies of 2 kHz and below, the middle ear muscle reflexes may contract on exposure to noise and provide a degree of protection against mechanical shearing forces [62]. Non-linearities in these frequencies, auditory responses, and the mechanical response differences in the basilar membrane explain the finding that low-frequency hearing thresholds are less vulnerable to the loss of apical OHCs [63,64].
Loss of stereocilia tip links due to excessive force, uncoupling the mechanoelectrical transduction (MET) channel, precludes mechanotransduction by the hair cell [65]. However, the tip links have some capacity to repair via replacement of PCDH15 and CDH23 proteins. In mammals, tip links broken in vitro have been observed to repair within 24 h, followed by restoration of mechanotransduction, although the MET current remains impaired for 36 h [66,67]. This phenomenon is suggested to underlie TTS after noise exposure, although PTS may occur if the tip links are damaged too extensively to repair. Furthermore, noise may damage the F-actin core of the stereocilia itself. Mechanical overstimulation can decrease the stiffness of the core, leaving stereocilia with a “floppy” appearance resulting from actin depolymerization, loss of actin cross-links, or fusion with other stereocilia [68]. As the stereocilia have some capacity to repair actin, this can result in TTS or PTS depending on the extent of the damage to the stereocilia core [61,68].
Supporting cells may also be physically damaged by noise. Pillar cell damage has been observed both after high-level impulse (160 dB) and continuous noise (100–120 dB SPL) [69,70]. Dieters’ and Hensen’s cells also have a protective effect on OHCs, and acoustic trauma may displace them toward the center of the cochlear turn, resulting in loss of hearing sensitivity [71]. Buckling of the supporting cells, particularly the pillar cells, has been shown to result in an uncoupling of the OHC stereocilia from the tectorial membrane in a chinchilla model of NIHL [72]. This decreases the hair cell stimulation and results in a TTS. Moderate noise exposure can also result in microchemical changes, decreasing pillar rigidity [72].

3.3.2. Oxidative Stress and Reduced Blood Flow

Apart from their function in the Krebs cycle, the mitochondria play an essential role in apoptosis and oxidative stress. Following infection, trauma, and in this context, noise-induced trauma, cellular damage or degradation of cell connections may lead to the activation of apoptotic pathways by hair cells [73]. Reactive oxygen species (ROS, e.g., superoxide, hydroxyl ion, hydrogen peroxide) may acquire an unstable number of electrons which also triggers pro-apoptotic pathways [73,74]. On receiving apoptotic signals, the balance between pro-apoptotic proteins, such as BAX or BAD, and anti-apoptotic proteins, such as those in the BCL family, is tilted [75]. Outer mitochondrial membrane permeability increases, leading to the transport of cytochrome c from the inner mitochondrial membrane to the cytoplasm [76]. Caspase enzymes in the cytoplasm are activated, resulting in cell degradation [76]. Yamane et al. demonstrated the stagnation of blood flow and the disturbance of strial circulation following acoustic trauma [77] and the appearance of free radicals such as the superoxide ion in the luminal surface of the stria vascularis marginal cells [78]. Stress and ischemia activate MAP3 kinases which in turn activate the JNK pathway, and phosphorylated JNK enters the nucleus and activates pro-apoptotic pathways such as c-Jun and Fos [79]. The downstream products lead to the expression of transcription factors and cellular effectors of apoptosis [76].
Peroxynitrite (ONOO–), a highly reactive nitrogen species (RNS) derived from NO and superoxide, is one of the most harmful free radicals to hair cells [80]. Noise-induced generation of RNS and ROS continues as an active biochemical process, not just in the immediate aftermath of exposure, but progresses over more than ten days post-exposure and can cause subsequent damage to cochlear structures [81]. Antioxidant enzymes such as superoxide dismutase play an important role in countering free radicals by catalyzing the dismutation of superoxide to hydrogen peroxide [82]. Other enzymes such as glutathione, glutathione peroxide, and catalase then balance with Sod1 to provide cellular defense, and the loss of Sod1 in knockout mice increased the susceptibility to noise-induced PTS [83].
The stria vascularis forms the lateral wall of the cochlear duct and comprises three cell layers: marginal, intermediate, and basal. It is responsible for maintaining the endocochlear potential and has a rich blood supply [84,85]. Hazardous noise exposure can cause damage, particularly to the intermediate cells, leading to temporary or permanent changes in the endocochlear potential and impaired hair cell mechanotransduction [86,87]. Furthermore, disruption of blood flow may induce cell hypoxia or alterations of the ionic equilibrium in the inner ear. The rise in the K+ levels in the endolymph and the Na+ levels in the perilymph result in cellular edema and structural damage [88].
Additionally, free radical formation in response to high-intensity noise can lead to vasoconstriction and reperfusion of cochlear cells, with subsequent cell death [81,89,90]. The calcium–magnesium ratio plays a vital role in controlling membrane permeability, voltage-dependent calcium and potassium channels, and polarization. Magnesium is also a potent vasodilator, co-operatively binds with potassium, and has a calcium channel blocker mimetic effect. A decrease in magnesium results in increased membrane permeability, an influx of calcium and sodium into the hair cell, and an efflux of potassium via passive diffusion [91]. In animal models, magnesium deficiency has been reported to exacerbate cochlear trauma in response to noise [92]. A sustained rise in intracellular calcium can deplete cell energy and can also lead to eventual cell death [93,94,95]. Additionally, low magnesium states have been associated with increased catecholamines and prostaglandins inducing vasoconstriction [93,96,97,98,99]. Thus, a decrease in magnesium and an increase in calcium-magnesium ratio can increase blood viscosity, reduce cochlear blood flow, and exacerbate the vasoconstrictor effects of acoustic trauma.

3.3.3. Inflammation

Neuroinflammation, a critical component of maintaining homeostasis in the central and peripheral nervous system, has been implicated in a wide range of pathological processes, including NIHL. There is extensive evidence showing that pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), interleukins, and chemokines (i.e., CCL2) are induced in the mammalian cochlea after noise trauma [100,101,102,103,104]. Additionally, noise exposure results in the recruitment of inflammatory cells such as macrophages to the cochlea [105,106,107,108]. It is currently unclear whether these inflammatory processes cause or exacerbate the threshold shifts associated with NIHL, although some of these molecules have demonstrated ototoxicity. For example, perfusion of TNF-α into the cochlea of guinea pigs resulted in synaptic degeneration and reduced auditory nerve compound action potentials, which could be protected against by blocking TNF-α [109].

3.3.4. Excitotoxicity and Synaptopathy

Louder sounds cause more hair cells to depolarize, causing greater release of glutamate by IHC ribbon synapses and increased motility of OHC. Noise exposure causes a rapid reduction in the number of IHC ribbon synapses, which is irreversible after neuropathic noise exposure [34,110,111]. Furthermore, excessive release of glutamate leads to glutamate excitotoxicity, characterized by an increased influx of ions in the postsynaptic cochlear nerve terminals leading to swelling of the postsynaptic cell bodies and dendrites [112]. This overstimulation can cause the loss of spiral ganglion cells, even after a delay of several months, and the damage can progress over years [113]. The long-term consequences of primary neuronal loss are progressive and present despite the recovery of threshold sensitivity [34]. Indeed, long-term noise exposure, even beneath the levels to induce cochlea trauma, can result in increased spontaneous firing rates and reorganization of cortical tonotopic maps in mammals [114,115,116,117]. The most common cause of cochlear damage leading to deafferentation is environmental noise overexposure [118,119,120].
Ribbon synapses and the peripheral dendrites of afferent neurons can be damaged by noise levels that are not as high as those required to effectuate threshold shifts [34,121,122]. This deafferentation of the IHC (no contact between them and the dendrites) is termed “synaptopathy” and may not result in detectable threshold shifts. However, as fewer neurons connect to the hair cells, the amplitude growth of neural excitation is reduced, and hearing may be impacted. This phenomenon occurs at noise levels previously thought not to be very harmful. The injury is permanent and not typically detected on an audiogram, hence the term “hidden hearing loss” [34,121,122]. IHCs have two orientation axes: pillar-modiolar and the habenular cuticular. The auditory neurons have high spontaneous rates and lower thresholds on the pillar face, while the modiolar facing neurons have low spontaneous rates and higher thresholds [123,124]. Neurons on the modiolar side of hair cells have smaller receptor patches and more synapses that are more sensitive to noise-induced degeneration compared to the pillar side [125]. In addition, these neurons show a reversible downregulation of protective glutamate receptor expression in peripheral terminals [122]. This spatial distribution of loss of high threshold neurons compared to low threshold neurons might explain the absence of threshold shifts even in the presence of synaptopathy [122].

3.4. Additional Negative Effects of Noise on the Inner Ear

3.4.1. Tinnitus

Tinnitus is the conscious perception of sound without an external auditory stimulus, often experienced as ringing or buzzing [126]. Tinnitus is typically self-reported and is primarily subjective [126]. Nevertheless, chronic tinnitus with or without hearing loss has been associated with low-level DPOAEs [127] and low-level transiently evoked otoacoustic emissions [128] compared with people without tinnitus and the same hearing level. Exposure to loud occupational, leisure-time, and firearm noise has been associated with higher prevalence of tinnitus and more frequent symptoms [118,120,129]. Although the precise mechanism of tinnitus remains an area of active research, cochlear deafferentation is thought to play a role. The most common cause of cochlear damage leading to deafferentation is environmental noise overexposure [118,119,120]. Similar to NIHL, neuroinflammation in the auditory cortex may also contribute to tinnitus, with elevated pro-inflammatory cytokines and microglial activation demonstrated in murine models [130,131].
Hearing loss is present in approximately 60% of people with tinnitus [132,133], suggestive of similar pathological processes after harmful noise exposure. The variability between tinnitus and hearing loss in noise-exposed subjects can be due to the differential vulnerability of cochlear and central components to the duration and intensity of noise exposure. A retrospective study of 531 patients with chronic tinnitus found that 83% had a high-frequency hearing loss corresponding to NIHL, and the degree of hearing loss was positively correlated with tinnitus loudness [7]. Similarly, several studies have reported that high proportions (i.e., up to 80%) of military personnel with NIHL also have tinnitus [7,134]. Additionally, the degree of hearing loss positively correlated with the two subscales (“intrusiveness” and “auditory perceptional difficulties”) of the Tinnitus Questionnaire [7]. A study of acute acoustic trauma in the Finnish armed forces found that 47% of personnel reported hearing impairment, and 94% reported tinnitus immediately following acute acoustic trauma, which persisted until discharge in 45% of cases [135]. In addition, otoacoustic emissions were better predictors of tinnitus persistence in a cohort of French military personnel than hearing thresholds alone as early as 24 h after an acute acoustic trauma [136].

3.4.2. Vestibular Dysfunction

The vestibular system consists of the utricle and saccule, which detect gravitational forces and horizontal and vertical plane movements, respectively, as well as three perpendicular semicircular canals [137]. The primary function of the vestibular system is the maintenance of gaze and postural stability, informing head position and spatial orientation, which are crucial to balance [137]. Movement of the endolymph in the semicircular canals during head rotation corresponds to the plane of rotation. Endolymph flows into the ampulla, an expansion of the semicircular canal containing mechanotransducing hair cells, causing deflection of the stereocilia and the release of neurotransmitters that send information about the plane of movement to the brain.
The vestibular labyrinth Is in proximity to and interconnected with the auditory system, and the fluids in the vestibular system have a degree of patency with the cochlea [138]. The cochlea and vestibular hair cells have similar ultrastructure; the balance and auditory receptors share the membranous labyrinth and a common arterial blood supply. These factors increase the likelihood of vestibular trauma in concurrence with NIHL [138]. Similar to the cochlea, vestibular end organ damage by noise can occur via direct mechanical destruction, metabolic decompensation with sensory degeneration, excitotoxicity, and free radical damage [28,139]. Indeed, neurophysiological studies have shown that, similar to the cochlea, the vestibular organs, particularly the saccule and utricle, are vulnerable to noise [140,141,142]. Accordingly, multiple studies have reported associations between NIHL and vestibular dysfunction or balance disorders such as vertigo, oscillopsia, postural instability, and/or motion intolerance [138,143,144,145,146,147].
Neurophysiological studies have measured the effects of noise on the vestibular system via vestibular evoked myogenic potentials (VEMPs) [28,148,149]. Vestibular afferents give rise to VEMPs, which are short-latency myogenic potentials in response to air-conducted sound or bone-conducted vibration. Cervical VEMPs are a measure of the sacculocollic pathway, and ocular VEMPs are a measure of utricular/superior vestibular nerve function [28,148,149]. A cross-sectional observational study of 43 military personnel with bilateral asymmetric hearing loss found that the severity of NIHL was associated with cervical VEMP, suggesting that the sacculocollic pathway is susceptible to noise damage [150]. Indeed, there are saccular neurons responsive to sound [151]. Another measure of vestibular function, particularly of otolith function, is the vestibular short-latency evoked potential [149]. Jerk amplitudes are used to elicit far-field potential responses generated by irregular primary vestibular afferent discharges [152,153]. Studies have shown that vestibular damage can occur due to brief exposure to elevated sound levels and sustained exposure to low-frequency continuous sound at more moderate levels [154,155,156,157]. A study of 258 military personnel found that vestibular damage from intense noise exposure was corelated with asymmetric NIHL [138]. Additionally, symptoms of vestibular dysfunction were observed in 11.2% of individuals in the study with symmetrical hearing loss compared to 21% with asymmetrical hearing loss, which could be attributed to compensation by the vestibular system to symmetrical progressive injury [138]. Additionally, individuals with higher levels of hearing loss (pure tone audiometry [PTA] >40 dB) had more abnormal vestibular test results and worse dual-task performance than those without NIHL [158]. However, participants’ reports of imbalance intensity via the Visual Analog Scale were similar, highlighting the need for vestibular evaluation in patients with hearing loss [158].

4. Screening and Diagnosis of NIHL

4.1. Screening

Screening of NIHL is typically performed via taking a patient’s history of noise exposure and performing audiograms, and clinicians will distinguish sensorineural from conductive hearing loss. However, it is often difficult to precisely quantify the noise exposure of individuals, and the type of exposure may vary (i.e., intense blast noise, cumulative noise, single tones, or wide-spectrum noise), leading to different patterns of damage. Thus, other tests may include the measurement of distortion product otoacoustic emissions (DPOAE), speech-in-noise testing, and auditory brainstem response (ABR), as detailed below. Each of these methods have notable strengths and limitations. Clinical assessments of signs of tinnitus and vestibular dysfunction may also be included if appropriate.

4.1.1. Audiograms

PTA, which measures the lowest intensity at which sound can still be heard across a range of audible frequencies (hearing threshold), is performed as a baseline evaluation test for hearing loss [159]. Air and bone conduction tests are typically performed over a range of frequencies from 125 to 8000 Hz [160]. NIHL produces characteristics audiometric signatures, where hearing is normal from low to mid frequencies, and there is a sudden drop past 3000 Hz, most pronounced at 4000 Hz, and a slight recovery in higher frequencies (Figure 4) [161]. Multiple factors contribute to this characteristic pattern, including the length and volume of the outer ear canal and the outer ear resonant frequency [60,161,162]. As the duration of noise exposure increases, the notch deepens to involve both higher and lower frequencies.
A 10 dB confirmed threshold shift from the baseline PTA at 2000, 3000, and 4000 Hz is the metric used by the US Occupational Safety and Health Administration (OSHA) to determine a standard threshold shift in cases of occupational hearing loss [163]. While not necessarily indicating hearing impairment, this change can be a warning sign of permanent hearing loss [88]. A limitation of using audiograms for NIHL screening is the need to consider natural age-related declines in hearing sensitivity (i.e., age-associated hearing levels). Thus, age correction of audiograms, which take averages of a population, is recommended for accurate comparisons with a non-noise exposed population [164,165]. However, there is considerable variability in the audiometric profiles among professions. Military personnel are exposed to high impulse sounds and show a pattern of hearing loss more significantly at 6 kHz than at 4 kHz [166].

4.1.2. Speech-in-Noise Testing

While PTA testing helps identify hearing loss, it does not account for the ability to discriminate sound in the presence of background noise, a primary presenting complaint of patients with NIHL [167]. PTA is limited in its ability to effectively predict speech perception because it indicates the patient’s access to sound rather than their functional hearing ability [168]. The speech-in-noise test simulates real-world situations by adding background noise or a competing signal in an isolated sound chamber and better represents the processing of speech [169]. This test measures a patient’s speech recognition threshold and responses to suprathreshold speech (i.e., stimuli presented above the detection threshold) [170]. Signal-to-noise ratio (SNR) loss refers to the increase in SNR required by a listener to repeat words, sentences, or words in sentences with 50% accuracy compared to typical performance [171]. Commonly performed tests are Hearing in Noise Tests, Speech Perception in Noise Test-Revised (SPIN-R), the Bamford–Kowal–Bench Speech-In-Noise (BKB-SIN) test, and QuickSIN [172,173]. In the rapid (~2 min) QuickSIN, test sentences are played at a high SNR of +25 dB to a low SNR of 0 dB, and the patient repeats the sentences back to the audiologist [171]. The number of words that are repeated inaccurately is subtracted from the accurate number.
Speech-in-noise testing gives a useful baseline estimation of hearing impairment, helps to infer the degree of benefit from different assistive hearing devices, and allows the quantification of improvement [174]. Speech-in-noise testing can be used to determine the potential utility of hearing aids for a patient because hearing aids are very effective at improving the ability to understand speech in quiet settings but are less effective in noisy settings [175]. This can result in better alignment with patient treatment priorities, and a 2021 systematic review found that patients who underwent speech-in-noise testing had higher measures of hearing aid satisfaction [176]. Further, this test can potentially detect hearing impairments that may not be evident in PTA testing. For example, when comparing ten noise-exposed Royal Air Force aircrew pilots with ten age/sex-matched unexposed administrators, the groups had comparable audiograms, but the exposed group had relatively elevated SIN thresholds, potentially reflecting abnormal retrocochlear processing [177].

4.1.3. Distortion Product Otoacoustic Emissions (DPOAE) Measurement

The OHCs amplify the intensity and sharpen the peak of the traveling sound wave in the cochlea via prestin-mediated elongation and contraction [178]. The resulting nonlinear electromechanical distortion of the sound wave by OHC motility can be measured via the DPAOE testing [179,180]. In addition to the distortion mechanism, otoacoustic emissions are also attributed to the reflection caused by the random scatter of the incoming traveling wave [180,181]. Therefore, the presence of DPOAEs is a marker of normal cochlear function and, specifically, of OHC health [182].
DPOAEs are evoked via a probe placed into the ear canal which emits two sounds at designated intensity levels (L1 and L2) and frequencies (f1 and f2), while a microphone detects the otoacoustic emission [182]. Cochlear stimulation with two pure tones, f1 and f2, results in a family of distortion products mathematically related to the input tones. Of these, the cubic distortion product 2f1–f2 is most commonly used in clinical and research settings [182]. The test is easy to implement and provides quick results [183].
Decreased DPOAE amplitudes are typically detected in older patients, those exposed to noise, and those with cochlear pathologies [184,185,186]. Thus, the DPOAE testing has been used for myriad clinical applications including newborn hearing screening, diagnostic audiological assessment, ototoxicity monitoring, and the study of cochlear mechanics [187]. DPOAE testing may be superior to PTA testing for military-related NIHL screening, as a study identified it as a quicker, easier tool still capable of detecting early cochlear injury from noise [183].

4.1.4. ABR Measurement

ABR, or auditory brainstem evoked potentials, are electrical signals produced by the nervous system within the first 10–15 ms following a transient acoustic stimulus [188]. A typical ABR consists of a sequence of five vertex positive waves with negative throughs [189]. Regarding the origins of component ABR waves, there are slight differences between humans and rodent models. In humans, wave I and II are believed to originate from the distal and proximal auditory nerve fibers, respectively; wave III is generated by the cochlear nucleus; IV reflects activity in the superior olivary complex; and wave V is associated with the lateral lemniscus [190]. In mice, wave II is believed to be generated by the posterior ventral cochlear nucleus; wave III is believed to be generated by the anterior ventral cochlear nuclear and trapezoid body; wave IV is believed to be generated by the superior olivary complex; and wave V is believed to be generated by the lateral lemniscus and inferior colliculus [191,192,193]. There is also some variation between mammalian models, as reported for rats [192,194,195], guinea pigs [196], and cats [197]. The sound intensity is directly proportional to the waves’ amplitude and inversely proportional to latency.
ABR is not routinely used in clinical screening of NIHL but does have expanded applications for neurodiagnostic testing, intraoperative monitoring, hearing screening/audiometry, and neurophysiological research [193]. In animal models, noise-induced synaptopathy results in a permanent decrease in the supra-threshold growth of ABR wave I amplitude despite full recovery of otoacoustic emission amplitudes and ABR thresholds [110]. A similar pattern of reduced suprathreshold ABR wave I amplitude has been observed in humans exposed to noise, although synaptopathy has not yet been confirmed [198] and is termed “hidden hearing loss” because these functional deficits are hidden by otherwise normal ABR thresholds [63,110]. However, some studies have shown a lack of sensitivity of ABR to noise exposure, which makes wave 1 amplitude a less ideal measure of cochlear synaptopathy for individuals with NIHL [56,170]. These findings underscore the importance of incorporating suprathreshold audiological testing when screening for NIHL to accurately understand a patient’s functional hearing ability.

4.2. Diagnosis

The UK utilizes Coles, Lutman, and Buffin diagnostic guidelines, which identify NIHL by a notch or downward bulge in the frequency range 3–6 kHz during PTA testing [199]. Additional requirements for diagnosing NIHL per these guidelines are high-frequency hearing impairment and a potentially hazardous amount of noise exposure. Four modifying factors which also need consideration include (1) the clinical picture (i.e., the mode, nature, and age of onset, symptom progression, and use of hearing amplification devices), (2) compatibility with age and noise exposure, (3) Robinson’s criteria for other causation [200], and (4) complications such as symptom asymmetry, mixed disorder, and conductive hearing impairment. Modifications have been proposed to account for age-related hearing loss and to delineate the effect of age from NIHL [199]. The US utilizes similar criteria put forth by the American Medical Association in 1979 [201], which were validated in 2011 among 1001 patients via audiometric testing and questionnaires [202]. Modified diagnostic criteria have been proposed to quantify hearing loss thresholds of military-related NIHL by comparing individuals’ hearing thresholds to those of non-noise-exposed individuals [166,171].

5. Prevention and Management of NIHL

5.1. Prevention

NIHL is mostly preventable, and tangible steps to reduce the burden of the disorder can be taken via the implementation of educational programs, regulation, and legislation to raise awareness and pre-emptively mitigate the damage caused by noise. In the US, the 1972 Noise Control Act established federal noise emission standards for commercial products and required that the public be provided information about noise emission levels and ways of reducing them [203]. Two US governmental departments—OSHA and the National Institute for Occupational Safety & Health (NIOSH)—have made recommendations for the permissible noise limit (PEL) of workplace noise exposure based on the average time a worker is exposed [204,205]. Daily noise dose is expressed as a percentage, per occupational standards, taking duration, sound exposure level, and course of exposure into account. For example, reaching 100% of a worker’s daily noise dose could be expressed as 85 dBA per NIOSH and 90 dBA per OSHA over a shift of 8 h. The course of exposure is cut when there is an increase in noise levels [206] (Table 1). Additionally, OSHA regulates that employers must provide hearing protection if employees are exposed to noise over the permissible exposure limit of 90 dB over an eight-hour time-weighted average [207]. Arenas et al. compared the occupational noise exposure levels in Latin America, the US, and Canada and found that 81% of the countries have a PEL of 85 dBA and that the majority of the countries limit impulsive noise exposure to a peak unweighted sound pressure level of 140 dB [208]. However, there were no established regulations in 27% of the countries, potentially exposing millions of people to NIHL.
Environmental noise level is conventionally measured via a sound level meter. However, they are expensive for small businesses and require maintenance and calibration, thereby limiting their widespread implementation. However, smartphone applications for this purpose are now available and provide inexpensive alternatives to specialist calibrated sound meters. An evaluation of the reliability of nine applications found the NIOShH Sound Level Meter to be the most accurate [209]. A free, accessible, and reliable app may help increase compliance with legislation and easy monitoring of environmental noise levels [210]. Additionally, in 2009, the European Commission mandated that output levels in new personal audio devices should be set to a standard of 85 dB, allowing users to increase the volume to a maximum of 100 dB. When users raise the volume to maximum level, a message was required to pop up that warns of the potential for hearing loss.
Hearing protective devices (HPDs) such as earmuffs and earplugs play an important role in protection against noise exposure. Plugs need to be inserted to ensure coverage of the entire ear canal’s circumference to provide protection and minimize irritation. Noise Reduction Ratings (NRRs) are calibrations to assess the protection range of HPDs in a single attenuation value (in dB). Because NRRs are derived via laboratory-based testing, they may overestimate the actual protection provided in non-experimental environments. Therefore, the NRR is derated by 50% on a dB scale before estimating exposure protection [211]. A Cochrane systematic review (2017) of interventions to prevent occupational NIHL found evidence that training on the proper insertion of ear plugs significantly reduced short-term noise exposure but called for more studies on the effectiveness of stricter legislation or better use of hearing protection devices [212]. Additionally, a randomized controlled trial showed that effective training of earplug usage led to significant improvement in the efficacy of HPDs in comparison to the usage of a device with higher NRR [213]. HPD fit-testing systems provide a customized fit for increased attenuation; however, they often require special facilities for testing such as a quiet room or audiometric booth or specialized equipment. Compliance in wearing HPDs is a barrier, especially in the military, as they are thought to decrease auditory situational awareness (e.g., sound detection, sound localization, and speech perception) [211,214]. However, novel fit test techniques (i.e., via a smartphone application) may enable better training and monitoring compliance [211].
Finally, clinicians can actively counsel patients at risk of NIHL on hearing protection strategies and the hazards of noise exposure in the workplace or recreationally (i.e., concerts, sports events, gun ranges, etc.). Simple strategies outlined by the US Centers for Disease Control and Prevention include avoiding exposure to excessive noise, turning down the volume on music, moving away from sources of noise, and using HPDs to reduce exposure to safe levels [215].

5.2. Clinical Management

There is no cure for NIHL and, to date, no approved pharmacological treatment indicated for its treatment. Although there are currently no clinical practice guidelines specifically for NIHL management, such guidelines exist for sensorineural hearing loss in adults (i.e., from the American Academy of Otolaryngology-Head and Neck Surgery (AAOHNS) [216], American Academy of Audiology [217], and the UK National Institute for Health and Care Excellence [218]) and are applicable. Relevant recommendations include exclusion of conductive hearing loss, audiometric confirmation of sensorineural hearing loss (SNHL) characteristic of noise trauma, and exclusion of retrocochlear pathology in case of asymmetric SNHL based on contrast-enhanced brain MRI or ABR testing. NIHL is clinically managed with hearing aids and/or use of hearing protection during exposure, although if hearing loss worsens, patients may be eligible for cochlear implants [56]. In some cases of acute noise-induced TTS, clinicians may consider the use of intratympanic steroids such as dexamethasone [219,220], although high-quality, long-term efficacy evidence in humans is lacking, and it is not considered for chronic occupational noise exposure. Additionally, the WHO classifies hearing loss into mild, moderate, severe, and profound, listing the typical signs and various recommendations for each level of disability (Box 1). Clinicians can use this classification to educate patients regarding the natural history of NIHL while counseling them on protective measures and the benefits of auditory rehabilitation (i.e., hearing aids or other assistive devices). However, the WHO severity levels are arbitrarily defined, and the system does not address patients with <25 dB hearing level and unilateral hearing loss [221]. Additionally, the speech-in-noise test, which is an effective test for measuring noise interference on speech perception skills, is not considered by the WHO system [222].
Box 1. WHO grades of hearing loss. WHO grades from the 1991 working group on prevention of deafness and hearing impairment [223]. Additional comments on the classifications from Olusanya et al. [221]. Abbreviations: dB, decibel; ISO, International Organization for Standardization; WHO, World Health Organization.
Grade of impairmentAudiometric ISO valuePerformanceRecommendationComments added to the prior classification
0-None≥25 dBNo or very slight hearing problem.
Can hear whispers.
None20 dB also recommended. People with 15–20 dB levels
may have hearing problems. People with unilateral hearing
loss may have problems even if the better ear is normal.
1-Slight26–40 dBCan hear and repeat words spoken
in a normal voice at 1 m
Counseling. Hearing aids
may be needed
Some difficulty in hearing but can usually hear
normal level of conversation
2-Moderate41–60 dBCan hear and repeat words spoken
in raised voice at 1 m
Hearing aids are
usually recommended
None
3-Severe61–80 dBCan hear some words when shouted
into better ear
Hearing aids needed. Otherwise
lip-reading and signing should
be taught
Discrepancies between pure-tone thresholds and speech
discrimination score should be noted
4-Profound≥81 dBUnable to hear and understand even
a shouted voice
Hearing aids may help understanding
words. Additional rehabilitation needed.
Lip-reading and sometimes signing are
essential.
Speech is distorted, the degree depending on the age at
which hearing was lost

6. Risk Factors

There are myriad risk factors associated with NIHL, as listed in Box 2 [224,225,226,227,228].
Box 2. Common risk factors for NIHL. Abbreviation: NIHL, noise-induced hearing loss.
  • Older age, although all ages are at risk
  • Repeated occupational noise exposure (construction, machine shop/factory, landscaping, mining, agriculture, musician, etc.)
  • Repeated recreational noise exposure (loud music at concerts, loud volume via earphones/earbuds)
  • Intense blast or explosion exposure
  • Shooting firearms (recreational or military)
  • Hypertension, smoking
  • Lack of hearing protection
  • Exposure to organic solvents, heavy metals, pesticides, asphyxiants

6.1. Occupational

Occupational NIHL is the most prevalent occupational disease globally [229], with approximately 7.0 million years lived with disability attributed to occupational noise exposure in 2019 [8]. People employed in construction, manufacturing, mining, agriculture, utility, and transportation industries, military personnel, and musicians are at high risk for NIHL [230]. As discussed above, OSHA considers US workers at increased risk of NIHL if noise exposure is at or above 85 dB, averaged over eight working hours or an eight-hour time-weighted average [231]. According to a report from the NHANES (1999–2004), 22 million US workers (17%) reported exposure to hazardous workplace noise, with 34% of these workers reporting no use of hearing protection devices [232]. Their report identified the highest weighted prevalence of workplace noise exposure for mining (76%) followed by lumber/wood product manufacturing (55%). Other high-risk occupations included repair and maintenance, motor vehicle operators, and construction trades. The position statement from the American College of Occupational and Environmental Medicine (ACOEM) states that occupational and environmental medicine physicians should understand a worker’s history of noise exposure and become proficient in the early detection and prevention of NIHL [233].

6.2. Military

Hearing loss is common in the military and is particularly disabling as it relates to both safety of the personnel and that of the nation. Personnel are often exposed to continuous as well as intermittent hazardous levels of noise, including gun fire and blast exposure, and increased hearing loss is associated with solvent exposure and longer service [234,235,236,237]. Combining acoustic and pressure wave energies can result in rupture of the organ of Corti, separation from the basilar membrane, and fracture and displacement of stereocilia. Indeed, as of 2021, tinnitus is the most prevalent service-connected disability for US veterans while hearing loss is the third-most prevalent, affecting 2.5 and 1.4 million veterans, respectively [238]. Individuals with medically disqualifying audiograms or hearing loss at application for service in the US Army and Marine Corp were eight and four times more likely, respectively, to have a hearing loss disability evaluation compared with matched controls [239]. Thus, early identification of enlistees at risk of hearing loss and counseling on hearing protection measures may reduce the burden of NIHL in the military. Currently, the post-exposure protocol for acute acoustic trauma in the US military includes removing the individual from the noise hazard and maintaining them in an effectively quiet environment (ambient levels of ≤70 dB) for 21 days [88,240].

6.3. Environmental and Recreational

Environmental factors, including ambient urban noise, also play an important role in the development of NIHL. For example, an observational study of environmental noise in Bangkok, Thailand, found that urban populations experienced greater mean hearing loss than suburban populations and found that among the occupational population in the urban monitoring sites, drivers had the highest risk of traffic-related NIHL [241]. Per a 2011 WHO report, approximately 1.5 million healthy life years are lost due to traffic-related noise in Western Europe alone [242]. Further, recreational noise exposure can contribute to NIHL. A recent systematic review reported that over 1 billion young adults are at risk of permanent, avoidable hearing loss primarily due to unsafe listening practices (i.e., amplified music) [243]. According to the 2021 National Firearms Survey, 81.4 million US adults (31.9 %) own firearms, and recreational shooting is a major cause of non-occupational NIHL [244,245]. Other non-occupational noise sources include chain saws and other power tools, toys, and recreational vehicles such as snowmobiles and motorcycles [9,245].

6.4. Genetic

Multiple studies have found that certain genetic and epigenetic characteristics increase the susceptibility to acoustic trauma in animal models of hearing loss [70,246,247,248]. Transgenic mice expressing genes modeling age-related hearing loss, such as Ahl1 (Cdh23735A>G) in C57BL/6J mice, are more susceptible to further hearing deterioration caused by noise exposure [247]. Conversely, mice that lack Ahl1 (i.e., 129/SvEv, Cast/Ei, and MOLF/Ei) are less vulnerable to acoustic injury [249,250,251,252]. Studies of knockout mice have detected pathways involving cochlear structures, oxidative stress, potassium recycling (vital for sensory transduction [85]), and heat shock proteins (HSPs) which increase the susceptibility of the inner ear to NIHL [253]. Some of the mouse genes implicated include Cdh23 [254], Pmca2 [255], Sod1 [83], Gpx1 [256], Trpv4 [257], Vasp [258], Hsf1 [259], and mdx [249].
Evidence of genetic risk factors in human NIHL is still emerging, although studies have suggested heritability of hearing loss [248,260,261,262]. For example, a study of noise sensitivity among 573 twin pairs in a Finnish cohort demonstrated heritability of 36% overall and of 40% after exclusion of hearing-impaired individuals [248]. Genes in pathways involving oxidative stress, potassium recycling, and HSPs have also been associated with NIHL in humans. Among the oxidative stress pathway genes, polymorphisms of GSTM1, PON2, SOD2, and CAT have been associated with NIHL, and CAT was independently validated in Swedish and Polish populations [263,264,265]. GJB1, GJB2, GJB3, GJB4, GJB6, KCNJ10, KCNQ1, KCNQ4, KCNE1, and SLC12A2 are ten genes in the potassium recycling pathway with known or suspected association with syndromic and non-syndromic hearing loss [266]. An analysis of 35 small nuclear polymorphisms in these genes from a Swedish population showed associations between KCNE1, KCNQ1, and KCNQ4 with NIHL [266], and a Polish study replicating the study found associations between polymorphisms in GJB1, GJB2, GJB4, KCNJ10, and KCNQ1 with NIHL [267].
HSPs are involved in the synthesis, folding, assembly, and intracellular transport of many other proteins. HPS expression increases during oxidative or other stress, including that caused by noise exposure, which may have a protective effect against cochlear damage via stabilization of stereocilia or enzyme regulation [268,269]. Three polymorphisms in HSP70 genes have been associated with susceptibility to NIHL: HSP70-1, HSP70-2, and HSP70-hom [268,270,271]. An association study with 53 candidate genes in two independent noise-exposed populations identified associations between NIHL and two monogenic genes, the deafness genes protocadherin 15 (PCDH15) and myosin 14 (MYH14) [272]. Cadherins are responsible for mechanical transduction and form the tip links between hair cells, while myosin regulates cytokinesis, cell motility, and cell polarity in hair cells [43,268]. In addition, noise exposure can result in epigenetic effects (i.e., alternations in gene transcription) via DNA methylation changes [273,274], histone modifications [275,276,277,278], and alterations in non-coding microRNAs (miRNAs) [279,280,281,282].
Finally, the inner ear has many glucocorticoid receptors (GR), which are affected by acoustic trauma or restraint stress at a transcriptional level [283,284,285]. In animal models, acoustic trauma increased endogenous corticosterone and decreased cochlear GR mRNA expression [283,284]. This led to a reduction in nuclear translocation of GR in the spiral ganglion neurons and increased activity of NF-κB. Pretreatment with a glucocorticoid agonist (dexamethasone) resulted in a decreased hearing threshold. Conversely, pretreatment with a glucocorticoid synthesis inhibitor (metyrapone) in combination with a GR antagonist (RU 486) exacerbated auditory threshold shifts (25–60 dB) after acoustic trauma [283].

7. Emerging Therapies

7.1. Completed and Ongoing Clinical Trials

The development of novel treatments for NIHL is an area of active research, encompassing interventions with anti-inflammatory, antioxidant, anti-excitatory, anti-apoptotic, or other effects. A search of ClinicalTrials.gov on 20 January 2023 identified 21 registered trials of interventional therapies inclusive of NIHL: one active and not recruiting (NCT05086276 (FX-322)), one not yet recruiting (NCT05511753 (acupuncture)), one recruiting (NCT04766853 (steroids)), two enrolling by invitation only (NCT04768569 and NCT04774250 (zonisamide)), one terminated (NCT02903355 (D-methionine)), one withdrawn (NCT02049073 (zonisamide, steroids)), three of unknown status (NCT02779192 (SPI-1005), NCT04482998 (steroids, hyperbaric oxygen therapy [HBOT]), and NCT01727492 (antioxidants)), and 13 completed (NCT02257983 (EPI-743), NCT01444846 (ebselen), NCT00808470 (micronutrients), NCT00552786 (antioxidant), NCT02951715 (zinc), NCT04120116, NCT04601909, and NCT04629664 (FX-322), NCT02259595 (HPN-07, N-acetylcysteine), NCT03834714 (near infrared light), NCT00802425 (AM-111), and NCT03878875 (sound conditioning)) (Table S1). Of the 12 completed/terminated clinical trials, just five have reported efficacy results via ClinicalTrials.gov or in publications, and all have focused on agents with antioxidant or anti-inflammatory capacity: micronutrients (i.e., vitamins/minerals), D-methionine, N-acetylcysteine, and ebselen (see details in Table 2 and described below). Additional findings have been published from clinical trials conducted outside of the US registry system, with therapies primarily aimed at preventing or attenuating TTS due to noise exposure (Table 3).

7.2. Antioxidant Therapy

Beta carotene, vitamins B, C, and E, zinc, and magnesium have antioxidant properties—particularly when combined—and have been shown to reduce vasoconstriction, cochlear cell death, and hearing loss in animal models when administered prior to noise exposure [89,286,287,288,289]. In humans, a 2021 systematic review concluded that certain micronutrients demonstrated protective effects for NIHL but that the results were not consistent [290]. A randomized Phase 2 trial (NCT00808470) of micronutrients (beta carotene, vitamins C and E, and magnesium) administered to young adults prior to exposure to 4 h of loud music reported that it was not superior to placebo in preventing TTS at any frequency level [291]. However, oral magnesium significantly reduced the magnitude of noise-induced TTS and PTS compared with placebo among an Israeli military personnel [292,293]. Similarly, separate prospective randomized trials demonstrated that vitamin B12 [294] and vitamin E (plus inhaled carbogen) [295] significantly attenuated noise-induced TTS compared with placebo. However, in an open-label study of oral zinc (NCT02951715) among adults with NIHL-related tinnitus, treatment improved tinnitus symptoms but not hearing thresholds from baseline after 2 months [296].
N-acetylcysteine is a thiol which supplies cysteine for intracellular glutathione synthesis. It acts as a free radical scavenger of hydrogen peroxide and hydrogen radicals to promote detoxification of ROS and RNS [297,298,299]. A randomized Phase 2 crossover trial (NCT00552786) of N-acetylcysteine administered to male Taiwanese workers exposed to loud occupational noise reported that it significantly reduced the magnitude of TTS from baseline (measured with PTA) compared with placebo among some workers [300]. Specifically, the benefit was limited to those without polymorphisms in the genes GSTM1 or GSTT1, which code for glutathione S-transferases (mu and theta, respectively) involved in cellular detoxification and nullification of carcinogens. Thus, antioxidant therapy with N-acetylcysteine may help to reduce the magnitude of noise-induced TTS among certain populations, although individuals’ genetic profiles influence the effect. D-methionine is the active form of methionine, an essential amino acid needed for tissue repair and selenium/zinc absorption. A randomized, double-blind Phase 3 trial (NCT02903355) of D-methionine administered prior to and during firearms training among US drill sergeant trainees (18 total days) reported in ClinicalTrials.gov that it was not superior to placebo in preventing hearing threshold shift. A separate Phase II-like trial of N-acetylcysteine conducted among a similar US Marines Corp population prior to weapons training also failed to demonstrate superiority of treatment to placebo in preventing noise-induced TTS [301].
Sound conditioning, or exposure to low-level noise prior to intense noise, is also aimed at increasing antioxidant activity in the inner ear [302]. This benign stimulus is hypothesized to “prime” the stria vascularis and organ of Corti to produce enzymes (i.e., glutathione reductase, gamma-glutamyl cysteine synthetase, and catalase) protective from noise-induced free radical damage [303]. A completed prospective clinical trial (NCT03878875) examined whether sound conditioning could attenuate TTS and tinnitus among young adults in the UK, although no results have been posted or published. To date, only studies in animal models have demonstrated any benefit from sound conditioning [304,305].

7.3. Anti-Inflammatory Therapy

Ebselen is a synthetic selenium-containing molecule that has anti-inflammatory properties and has been assessed as a treatment for various forms of SNHL [306]. It mimics glutathione peroxidase, the primary antioxidant enzyme in the cochlea and which decreases in activity following noise or ototoxic injury, and activates the Keap1-Nrf2 cytoprotective pathway [307,308]. A Phase 2 randomized clinical trial (NCT01444846) among 83 normal-hearing adults compared a placebo with 200, 400, or 600 mg ebselen administered 2 days prior to and 2 days following a calibrated noise exposure challenge [309]. The results indicated that the mean TTS at 4 kHz was significantly reduced (by 68%) in the group receiving 400 mg ebselen compared with placebo.
Table 2. Completed clinical trials of interventions for NIHL registered at ClinicalTrials.gov with reported results.
Table 2. Completed clinical trials of interventions for NIHL registered at ClinicalTrials.gov with reported results.
ClinicalTrial.gov
ID/Name
InterventionStudy DesignPopulation, NActive Arm, NComparator Arm, NPrimary OutcomeSecondary OutcomesResultsPublicationSponsor
NCT02903355: Phase 3 Clinical Trial: D-methionine to Reduce Noise-Induced Hearing Loss (NIHL)D-methionine to prevent NIHL or tinnitus in a military population Phase 3 RCT US drill sergeant instructor trainees (21–45 y) receiving weapons training, N = 266 18 days of oral D-methionine, n = 124Placebo, n = 142 ASHA shift from baseline to day 29–36 post-drug (day 15–22 post-noise exposure) Change in DOEHRS-HC, EWS STS, THI D- methionine was not superior to placebo on any outcome in the interim analysisNoneMetamor, Inc., USA [discontinued development]
NCT00808470: Micronutrients to Prevent Noise-induced Hearing LossMicronutrients to reduce magnitude of TTS from 4 h of loud music in young adultsPhase 2 RCTAdults (18–35 y) with normal hearing, N = 704 days of oral beta carotene, vitamins C and E, magnesium, n = 35Placebo, n = 35Mean threshold shift at 4 kHz in both ears 15 min post-music vs. baselineThreshold shift at 0.25, 0.5, 1, 2, 3, 6, and 8 kHz post-music; change in DPOAE amplitude, PMTF thresholds, and tinnitus measuresMicronutrients were not superior to placebo on any outcome in the final analysisLePrell et al., 2016 [291]University of Michigan, USA
NCT00552786: Antioxidation Medication for Noise-induced Hearing LossNAC to prevent TTS in workers exposed to noise in TaiwanRandomized Phase 2 crossoverMales (25–65 y) with known work noise exposure, N = 5314 days of 1200 mg NAC Glucose capsuleThreshold shift from baseline measured with PTA, four time-spaced assessmentsThreshold shift from baseline measured with DPOAE, four time-spaced assessmentsNAC significantly reduced TTS among men without any GSTIM1 or GSTT1 polymorphisms Lin et al. (2010) [300]National Taiwan University Hospital, Taiwan
NCT02951715: Improvement of Tinnitus After Oral Zinc on Patients With Noise-induced Hearing LossZine to improve NIHL and tinnitus symptomsOpen label, single armAdults with confirmed NIHL-related tinnitus, N = 202 months of 40 mg oral zinc dailyNoneTHI change from baselineThreshold shift, speech discrimination, DPOAE, tinnitus pitch/loudness from baselineZinc did not significantly improve hearing threshold but did improve THI score from baselineYeh et al. (2019) [296]Chang Gung Memorial Hospital, China
NCT01444846: Otoprotection With SPI-1005 for Prevention of Temporary Auditory Threshold Shift Ebselen to prevent TTSPhase 2 RCTAdults (18–31 y) with normal hearing, N = 834 days of oral 200, 400, or 600 mg ebselen, total n = 63Placebo capsule, n = 20Reduction in TTS from baseline (15 min post-sound exposure) at 4 kHzN/ASignificant reduction in TTS at 4 kHz in 400 mg ebselen vs. placebo groupsKil et al. (2017) [309]Sound Pharmaceuticals, Inc., USA
Abbreviations: ASHA, American Speech-Language-Hearing Association; dB, decibel; DOEHRS-HC, Defense Occupational and Environmental Health Readiness System-Hearing Conservation; DPOAE, distortion product otoacoustic emissions; EWS, early warning shift; h, hour; kHz, kilohertz; min, minutes; NAC, N-acetylcysteine; NIHL, noise-induced hearing loss; PMTF, psycho-acoustical modulation transfer function; RCT, randomized controlled trial; STS, standard threshold shift; THI, Tinnitus Handicap Inventory; TTS, temporary threshold shift; US, United States; y, year.
Clinical trials have been conducted to assess HBOT and corticosteroids as treatments for NIHL, primarily focusing on acute acoustic trauma (AAT) (Table 3). Treatment with corticosteroids has been proposed to reduce the inflammation in the inner ear associated with AAT and with intratympanic administration—clinically achieved via laser-assisted myringotomy—resulting in higher perilymph concentrations than systemic administration in animal models [240]. However, the therapeutic benefits are largely predicated on very rapid treatment after NIHL onset. A randomized trial of intratympanic plus systemic or just systemic corticosteroids within 3 days of NIHL onset reported that Chinese patients who received intratympanic delivery had significantly better improvement in audiological outcomes and word recognition than the systemic group, although individuals’ outcomes varied widely [219]. A prospective trial assessed the timing of intravenous corticosteroids and piracetam—a GABA derivative sold outside the US as a nootropic or possible anti-convulsive—as early treatment for AAT among Greek military personnel [310]. The results indicated that patients who were treated within 1 h post-noise exposure achieved the best audiological recovery compared with patients treated 16 or 24+ h later, although there was no control group. However, a case-control study of oral corticosteroids after AAT reported that Israeli miliary personnel treated with <24 h of exposure had significantly better hearing outcomes than those who received no treatment [311].
Hyperbaric oxygen has been previously used to counteract cochlear hypoxia in sudden idiopathic hearing loss [312], including in concert with intratympanic steroids [313]. In the context of NIHL, several retrospective studies have noted that HBOT with or without steroid therapy improved hearing recovery after AAT, although the benefits have been limited and inconsistent [314,315,316,317]. Additionally, a prospective randomized clinical trial examined three regimens of HBOT and/or corticosteroids and piracetam (i.e., medical therapy), following AAT among Belgian soldiers: (1) 10 days oral medical therapy, (2) intensive (twice daily for 3 days then once daily for 7 days) HBOT plus 5 days intravenous then oral medical therapy, or (3) 10 days once-daily HBOT plus oral medical therapy [318]. The results indicated that the audiological outcomes (10 days post-therapy) were superior in the groups that received HBOT versus oral medical therapy alone, although it was not clear if one HBOT regimen was superior to the other. Similar to corticosteroid therapy, the delivery of HBOT treatment very soon after AAT appears crucial to realize benefits [319].
Table 3. Additional studies of therapies to prevent or attenuate NIHL (conducted outside of the US).
Table 3. Additional studies of therapies to prevent or attenuate NIHL (conducted outside of the US).
Study NationAuthor, YearInterventionStudy DesignPopulation, NActive Arm, NComparator arm, NPrimary OutcomeResults
Following AAT (attenuation)
GreecePsillas et al. (2008) [310]Prednisone and piracetamRandomized cohort studyMale soldiers with firearms-related AAT, N = 52Therapy in <1 h, n = 20Therapy in >1–<16 h, n = 17
Therapy in 24+ h, n = 15
Complete or partial recovery in 1 month Significantly higher recovery rate (65%) and better final threshold shifts if treatment at <1 h vs. with delayed treatment (13–24% recovery)
BelgiumLafère et al. (2010) [318]HBOT with or without methyl-prednisolone and piracetam (medical therapy)Cohort studyBelgian soldiers with firearms-related AAT, N = 68HBOT + oral medical therapy, n = 17
HBOT + IV then oral medical therapy, n = 32
10 days oral medical therapy, N = 17Average hearing gain and average residual hearing loss 10 days post-treatment vs. baseline pre-AATBoth regimens of HBOT + medical therapy was superior to medical therapy alone
ChinaZhou et al. (2013) [219]Intratympanic steroid (methyl-prednisolone) with or without oral steroid Prospective RCTAdults recently diagnosed with NIHL, N = 52Intratympanic + oral steroid 3 days after NIHL onset, n = 27Intratympanic placebo + oral steroid 3 days after NIHL onset, n = 26PTA and speech discrimination score change from baselineSignificantly more of the intratympanic group had ≥15 dB improvement in PTA and ≥15% speech discrimination vs. placebo + oral steroid only
Before noise exposure (prophylaxis)
IsraelAttias et al, (2004) [293]Mg Prospective RCTMales (16–37 y) with normal hearing, N = 2010 days 122 mg oral Mg, n = 10Placebo, n = 10TTS immediately following noise exposure
(90 dB for 10 min)
12% of Mg-treated patients experienced TTS ≥ 20 dB, vs. 28% in placebo and no-intake groups; reduced magnitude of TTS was significant for all frequencies between 2 and 8 kHz
IsraelAttias et al, (1994) [292] MgProspective RCTMale military recruits exposed to noise during 8 weeks of weapons training, N = 255167 mg oral Mg twice daily during training, n = 125Placebo, n = 130PTS 7–10 days post-exposure vs. baselinePTS was significantly more common and more severe in the placebo vs. Mg-treated group, and negatively correlated with red blood cell Mg level
ItalyQuaranta et al. (2004) [294]Vitamin B12 Prospective RCTAdults (20–30 y) with normal hearing, N = 201 mg B12 daily for 7 days, then 5 mg on day 8, n = 10Placebo, n = 10TTS following
112 dB 3 kHz noise for 10 min measured 8 days post-treatment
Significant reduction in TTS at 3 at 4 kHz in B12 group vs. control
IndiaKapoor et al. (2011) [295]Vitamin E + inhaled carbogen (5% CO2 +95% oxygen)Prospective RCTMale industrial workers with exposure to intense occupational noise, N = 40 6 days carbogen, vitamin E only, or combination, total n = 30 (10 each)Placebo, n = 10TTS from baseline following 5 h noise exposure
(90–113.5 dB)
Combined carbogen + vitamin E reduced TTS by 1.6–5.1 dB across frequencies; vitamin E groups had reduced serum markers of oxidative stress
Abbreviations: AAT, acute acoustic trauma; dB, decibel; h, hour; HBOT, hyperbaric oxygen therapy; IV, intravenous; kHz, kilohertz; Mg, magnesium; min, minutes; NAC, N-acetylcysteine; NIHL, noise-induced hearing loss; PTA, pure tone audiometry; PTS, permanent threshold shift; RCT, randomized controlled trial; TTS, temporary threshold shift; y, year.

8. Conclusions

This review of the literature described the etiology of and risk factors for NIHL, as well as approaches for screening, prevention, and treatment of affected patients. We also summarized the evidence from publications of experimental therapies which, although promising for prophylaxis or AAT that can be treated very quickly, have yet to address the needs of patients with long-standing or cumulative NIHL. The impact of NIHL is not limited to decreased quality of life of affected individuals but can extend to lower productivity of the workforce and impaired performance of military personnel. Further, NIHL is associated with a substantial economic burden to governments and society at large, expected to increase as the global population ages and experiences increased costs of NIHL-related healthcare and disability. Thus, the prevention of noise-induced cochlear injury, via the use of hearing protection in loud occupational and recreational settings, remains the cornerstone of reducing the enormous burden of NIHL. Governmental regulations and safety-focused organizations play essential roles in advising the public and industry on safe noise exposure levels and in implementing educational programs. For example, a cost-effectiveness analysis of a military hearing conservation program estimated a lifetime savings of USD $10,657 per person in service-related compensation on implementation of the program [320]. Additional harm reduction may also result from better alignment of occupational noise exposure limits recommended by NIOSH and OSHA. It is encouraging that there is increased awareness of the need for NIHL prevention compared to decades ago, although there remains a large unmet medical need for therapies beyond prophylaxis or AAT.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/2077-0383/12/6/2347/s1. Table S1. Clinical trials of interventions for NIHL registered at ClinicalTrials.gov without reported results.

Funding

This research was funded by the Remondi Foundation, Rich Robbins, Larry Bowman, and the Bertarelli Foundation Endowed Professorship (KMS).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors thank Samuel Early for early help with the literature review.

Conflicts of Interest

The authors attest they have no conflicts of interest.

References

  1. Acton, W.I. History and development of hearing protection devices. J. Acoust. Soc. Am. 1987, 81, S4. [Google Scholar] [CrossRef]
  2. Thurston, F.E. The worker’s ear: A history of noise-induced hearing loss. Am. J. Ind. Med. 2013, 56, 367–377. [Google Scholar] [CrossRef] [PubMed]
  3. Institute for Quality and Efficiency in Health Care (IQWiG). Hearing Loss and Deafness: Normal Hearing and Impaired Hearing. Available online: https://www.ncbi.nlm.nih.gov/books/NBK390300/ (accessed on 25 January 2023).
  4. Münzel, T.; Gori, T.; Babisch, W.; Basner, M. Cardiovascular effects of environmental noise exposure. Eur. Heart J. 2014, 35, 829–836. [Google Scholar] [CrossRef] [PubMed]
  5. World Health Organization. Deafness and Hearing Loss. Available online: https://www.who.int/news-room/fact-sheets/detail/deafness-and-hearing-loss (accessed on 10 January 2022).
  6. Liberman, M.C. Noise-induced hearing loss: Permanent versus temporary threshold shifts and the effects of hair cell versus neuronal degeneration. Adv. Exp. Med. Biol. 2016, 875, 1–7. [Google Scholar] [PubMed]
  7. Mazurek, B.; Olze, H.; Haupt, H.; Szczepek, A.J. The more the worse: The grade of noise-induced hearing loss associates with the severity of tinnitus. Int. J. Environ. Res. Public. Health 2010, 7, 3071–3079. [Google Scholar] [CrossRef]
  8. GBD 2019 Hearing Loss Collaborators. Hearing loss prevalence and years lived with disability, 1990–2019: Findings from the Global Burden of Disease Study 2019. Lancet 2021, 397, 996–1009. [Google Scholar] [CrossRef]
  9. Rabinowitz, P.M. Noise-induced hearing loss. Am. Fam. Physician 2000, 61, 2749–2756, 2759–2760. [Google Scholar]
  10. Goines, L.; Hagler, L. Noise pollution: A modem plague. South. Med. J. 2007, 100, 287–294. [Google Scholar] [CrossRef]
  11. Oishi, N.; Schacht, J. Emerging treatments for noise-induced hearing loss. Expert. Opin. Emerg. Drugs 2011, 16, 235–245. [Google Scholar] [CrossRef]
  12. Daniel, E. Noise and hearing loss: A review. J. Sch. Health 2007, 77, 225–231. [Google Scholar] [CrossRef] [PubMed]
  13. Schwela, D. Environmental noise challenges and policies in low- and middle- income countries. SFJH 2021, 2, 26–45. [Google Scholar] [CrossRef]
  14. Kalisa, E.; Irankunda, E.; Rugengamanzi, E.; Amani, M. Noise levels associated with urban land use types in Kigali, Rwanda. Heliyon 2022, 8, e10653. [Google Scholar] [CrossRef] [PubMed]
  15. King, E.A. Here, there, and everywhere: How the SDGs must include noise pollution in their development challenges. Environ. Sci. Policy 2022, 64, 17–32. [Google Scholar] [CrossRef]
  16. Nelson, D.I.; Nelson, R.Y.; Concha-Barrientos, M.; Fingerhut, M. The global burden of occupational noise-induced hearing loss. Am. J. Indust Med. 2005, 48, 446–458. [Google Scholar] [CrossRef] [PubMed]
  17. Nandi, S.S.; Dhatrak, S.V. Occupational noise-induced hearing loss in India. Indian. J. Occup. Environ. Med. 2008, 12, 53–56. [Google Scholar] [CrossRef]
  18. Khoza-Shangase, K.; Moroe, N.F.; Edwards, A. Occupational hearing loss in Africa: An interdisciplinary view of the current status. S. Afr. J. Commun. Disord. 2020, 67, e1–e3. [Google Scholar] [CrossRef]
  19. Nyarubeli, I.P.; Tungu, A.M.; Moen, B.E.; Bråtveit, M. Prevalence of noise-induced hearing loss among Tanzanian iron and steel workers: A cross-sectional study. Int. J. Environ. Res. Public. Health 2019, 16, 1367. [Google Scholar] [CrossRef]
  20. US Centers for Disease Control and Prevention. Noise-induced Hearing Loss. Available online: https://www.nidcd.nih.gov/health/noise-induced-hearing-loss (accessed on 28 November 2022).
  21. Carroll, Y.I.; Eichwald, J.; Scinicariello, F.; Hoffman, H.J.; Deitchman, S.; Radke, M.S.; Themann, C.L.; Breysse, P. Vital signs: Noise-induced hearing loss among adults—United States 2011–2012. MMWR Morb. Mortal. Wkly. Rep. 2017, 66, 139–144. [Google Scholar] [CrossRef]
  22. Henderson, E.; Testa, M.A.; Hartnick, C. Prevalence of noise-induced hearing-threshold shifts and hearing loss among US youths. Pediatrics 2011, 127, e39–e46. [Google Scholar] [CrossRef]
  23. Shield, B.; Hearing Loss: Numbers and Costs. Evaluation of the Social and Economic Costs of Hearing Impairment. A Report for Hear-It AISBL. Available online: https://www.hear-it.org/sites/default/files/2021-01/Hear-it%20Report%20Hearing%20Loss%20Numbers%20and%20Costs%202019.pdf (accessed on 28 November 2022).
  24. Pawlaczyk-Luszczynska, M.; Dudarewicz, A.; Zaborowski, K.; Zamojska, M.; Sliwinska-Kowalska, M. Noise induced hearing loss: Research in Central, Eastern and South-Eastern Europe and newly independent states. Noise Health 2013, 15, 55–66. [Google Scholar] [CrossRef]
  25. Palmer, K.T.; Griffin, M.J.; Syddall, H.E.; Davis, A.; Pannett, B.; Coggon, D. Occupational exposure to noise and the attributable burden of hearing difficulties in Great Britain. Occup. Environ. Med. 2002, 59, 634–639. [Google Scholar] [CrossRef] [PubMed]
  26. Health and Safety Executive. Noise Induced Hearing Loss in Great Britain. Available online: https://www.hse.gov.uk/statistics/causdis/deafness/index.htm (accessed on 28 November 2022).
  27. Purves, D.; Augustine, G.; Fitzpatrick, D. (Eds.) The Audible Spectrum. In Neuroscience, 2nd ed.; Sinauer Associates: Sunderland, MA, USA, 2001. [Google Scholar]
  28. Stewart, C.E.; Holt, A.G.; Altschuler, R.A.; Cacace, A.T.; Hall, C.D.; Murnane, O.D.; King, W.M.; Akin, F.W. Effects of noise exposure on the vestibular system: A systematic review. Front. Neurol. 2020, 11, 593919. [Google Scholar] [CrossRef]
  29. US Centers for Disease Control and Prevention. What Noises Cause Hearing Loss? Available online: https://www.cdc.gov/nceh/hearing_loss/what_noises_cause_hearing_loss.html (accessed on 11 January 2023).
  30. US National Institute for Occupational Safety and Health (NIOSH). Noise & Hearing Loss Prevention. Available online: https://www.cdc.gov/niosh/topics/noise/default.html (accessed on 28 November 2022).
  31. Hearing Health Foundation. Decibel Levels. Available online: https://hearinghealthfoundation.org/decibel-levels (accessed on 28 November 2022).
  32. Sinyor, A.; Laszlo, C.A. Acoustic behavior of the outer ear of the guinea pig and the influence of the middle ear. J. Acoust. Soc. Am. 1973, 54, 916–921. [Google Scholar] [CrossRef] [PubMed]
  33. Luers, J.C.; Hüttenbrink, K.B. Surgical anatomy and pathology of the middle ear. J. Anat. 2016, 228, 338–353. [Google Scholar] [CrossRef]
  34. Kujawa, S.G.; Liberman, M.C. Adding insult to injury: Cochlear nerve degeneration after “temporary” noise-induced hearing loss. J. Neurosci. 2009, 29, 14077–14085. [Google Scholar] [CrossRef] [PubMed]
  35. Skrzat, J.; Kozerska, M.; Wroński, S.; Tarasiu, J.; Walocha, J. Volume rendering of the tympanic cavity from micro-CT data. Folia Med. Cracov. 2015, 55, 81–89. [Google Scholar] [PubMed]
  36. Zdilla, M.J.; Skrzat, J.; Kozerska, M.; Leszczyński, B.; Tarasiuk, J.; Wroński, S. Oval window size and shape: A micro-CT anatomical study with considerations for stapes surgery. Otol. Neurotol. 2018, 39, 558–564. [Google Scholar] [CrossRef] [PubMed]
  37. Békésy, G.V. The sound pressure difference between the round and the oval windows and the artificial window of labyrinthine fenestration. Acta Otolaryngol. 1947, 35, 301–315. [Google Scholar] [CrossRef]
  38. Ryan, A.F.; Kujawa, S.G.; Hammill, T.; Le Prell, C.; Kil, J. Temporary and permanent noise-induced threshold shifts: A review of basic and clinical observations. Otol. Neurotol. 2016, 37, e271–e275. [Google Scholar] [CrossRef]
  39. Costanzo, L. Neurophysiology. In Physiology, 6th ed.; Costanzo, L., Ed.; Elsevier: Philadelphia, PA, USA, 2018. [Google Scholar]
  40. Raftery AT, J.S. Head and neck. In Applied Basic Science for Basic Surgical Training, 2nd ed.; Raftery, A., Ed.; Churchill Livingstone: Edinburgh, Scotland, 2008. [Google Scholar]
  41. Pujol, R.; Nouvian, R.; Lenoir, M.; Irving, S. Hair Cells: Overview. Available online: http://www.cochlea.eu/en/hair-cells (accessed on 28 November 2022).
  42. Zetes, D.E.; Tolomeo, J.A.; Holley, M.C. Structure and mechanics of supporting cells in the guinea pig organ of Corti. PLoS ONE 2012, 7, e49338. [Google Scholar] [CrossRef]
  43. Sakaguchi, H.; Tokita, J.; Müller, U.; Kachar, B. Tip links in hair cells: Molecular composition and role in hearing loss. Curr. Opin. Otolaryngol. Head. Neck Surg. 2009, 17, 388–393. [Google Scholar] [CrossRef] [PubMed]
  44. Hakizimana, P.; Fridberger, A. Inner hair cell stereocilia are embedded in the tectorial membrane. Nat. Commun. 2021, 12, 2604. [Google Scholar] [CrossRef] [PubMed]
  45. Von Bekesy, G. Resting potentials inside the cochlear partition of the guinea pig. Nature 1952, 169, 241–242. [Google Scholar] [CrossRef]
  46. Kurbel, S.; Borzan, V.; Golem, H.; Dinjar, K. Cochlear potential difference between endolymph fluid and the hair cell’s interior: A retold interpretation based on the Goldman equation. Med. Glas. 2017, 14, 8–15. [Google Scholar]
  47. Liberman, M.C.; Gao, J.; He, D.Z.Z.; Wu, X.; Jia, S.; Zuo, J. Prestin is required for electromotility of the outer hair cell and for the cochlear amplifier. Nature 2002, 419, 300–304. [Google Scholar] [CrossRef] [PubMed]
  48. Gale, J.E.; Ashmore, J.F. An intrinsic frequency limit to the cochlear amplifier. Nature 1997, 389, 63–66. [Google Scholar] [CrossRef]
  49. Khimich, D.; Nouvian, R.; Pujol, R.; Tom Dieck, S.; Egner, A.; Gundelfinger, E.D.; Moser, T. Hair cell synaptic ribbons are essential for synchronous auditory signalling. Nature 2005, 434, 889–894. [Google Scholar] [CrossRef] [PubMed]
  50. Fuchs, P.A.; Glowatzki, E.; Moser, T. The afferent synapse of cochlear hair cells. Curr. Opin. Neurobiol. 2003, 13, 452–458. [Google Scholar] [CrossRef]
  51. Sterling, P.; Matthews, G. Structure and function of ribbon synapses. Trends Neurosci. 2005, 28, 20–29. [Google Scholar] [CrossRef]
  52. Lagnado, L. Ribbon synapses. Curr. Biol. 2003, 13, R631. [Google Scholar] [CrossRef]
  53. Johnson, D.H. The relationship between spike rate and synchrony in responses of auditory-nerve fibers to single tones. J. Acoust. Soc. Am. 1980, 68, 1115–1122. [Google Scholar] [CrossRef]
  54. Furness, D.N. Molecular basis of hair cell loss. Cell Tissue Res. 2015, 361, 387–399. [Google Scholar] [CrossRef]
  55. Salvi, R.; Sun, W.; Ding, D.; Chen, G.-D.; Lobarinas, E.; Wang, J.; Radziwon, K.; Auerbach, B.D. Inner hair cell loss disrupts hearing and cochlear function leading to sensory deprivation and enhanced central auditory gain. Front. Neurosci. 2017, 10, 621. [Google Scholar] [CrossRef] [PubMed]
  56. Le, T.N.; Straatman, L.V.; Lea, J.; Westerberg, B. Current insights in noise-induced hearing loss: A literature review of the underlying mechanism, pathophysiology, asymmetry, and management options. J. Otolaryngol. Head Neck Surg. 2017, 46, 41. [Google Scholar] [CrossRef]
  57. Wu, P.Z.; O’Malley, J.T.; de Gruttola, V.; Liberman, M.C. Primary neural degeneration in noise-exposed human cochleas: Correlations with outer hair cell loss and word-discrimination scores. J. Neurosci. 2021, 41, 4439–4447. [Google Scholar] [CrossRef] [PubMed]
  58. Campbell, K.; Hammill, T.; Hoffer, M.; Kil, J.; Le Prell, C. Guidelines for auditory threshold measurement for significant threshold shift. Otol. Neurotol. 2016, 37, e263–e270. [Google Scholar] [CrossRef]
  59. Liberman, M.C.; Dodds, L.W. Single-neuron labeling and chronic cochlear pathology. III. Stereocilia damage and alterations of threshold tuning curves. Hear. Res. 1984, 16, 55–74. [Google Scholar] [CrossRef] [PubMed]
  60. Gerhardt, K.J.; Rodriguez, G.P.; Hepler, E.L.; Moul, M.L. Ear canal volume and variability in the patterns of temporary threshold shifts. Ear Hear. 1987, 8, 316–321. [Google Scholar] [CrossRef]
  61. Wagner, E.L.; Shin, J.B. Mechanisms of hair cell damage and repair. Trends Neurosci. 2019, 42, 414–424. [Google Scholar] [CrossRef]
  62. Kim, J.; Koo, M. Mass and stiffness impact on the middle ear and the cochlear partition. J. Audiol. Otol. 2015, 19, 1–6. [Google Scholar] [CrossRef]
  63. Robles, L.; Ruggero, M.A. Mechanics of the mammalian cochlea. Physiol. Rev. 2001, 81, 1305–1352. [Google Scholar] [CrossRef]
  64. Prosen, C.A.; Moody, D.B.; Stebbins, W.C.; Smith, D.W.; Sommers, M.S.; Brown, J.N.; Altschuler, R.A.; Hawkins, J.E. Apical hair cells and hearing. Hear. Res. 1990, 44, 179–193. [Google Scholar] [CrossRef]
  65. Assad, J.A.; Shepherd, G.M.; Corey, D.P. Tip-link integrity and mechanical transduction in vertebrate hair cells. Neuron 1991, 7, 985–994. [Google Scholar] [CrossRef] [PubMed]
  66. Jia, S.; Yang, S.; Guo, W.; He, D.Z. Fate of mammalian cochlear hair cells and stereocilia after loss of the stereocilia. J. Neurosci. 2009, 29, 15277–15285. [Google Scholar] [CrossRef] [PubMed]
  67. Indzhykulian, A.A.; Stepanyan, R.; Nelina, A.; Spinelli, K.J.; Ahmed, Z.M.; Belyantseva, I.A.; Friedman, T.B.; Barr-Gillespie, P.G.; Frolenkov, G.I. Molecular remodeling of tip links underlies mechanosensory regeneration in auditory hair cells. PLoS Biol. 2013, 11, e1001583. [Google Scholar] [CrossRef]
  68. Liberman, M.C. Chronic ultrastructural changes in acoustic trauma: Serial-section reconstruction of stereocilia and cuticular plates. Hear. Res. 1987, 26, 65–88. [Google Scholar] [CrossRef]
  69. Roberto, M.; Zito, F. Scar formation following impulse noise-induced mechanical damage to the organ of Corti. J. Laryngol. Otol. 1988, 102, 2–9. [Google Scholar] [CrossRef]
  70. Ou, H.C.; Bohne, B.A.; Harding, G.W. Noise damage in the C57BL/CBA mouse cochlea. Hear. Res. 2000, 145, 111–122. [Google Scholar] [CrossRef]
  71. Flock, A.; Flock, B.; Fridberger, A.; Scarfone, E.; Ulfendahl, M. Supporting cells contribute to control of hearing sensitivity. J. Neurosci. 1999, 19, 4498–4507. [Google Scholar] [CrossRef] [PubMed]
  72. Nordmann, A.S.; Bohne, B.A.; Harding, G.W. Histopathological differences between temporary and permanent threshold shift. Hear. Res. 2000, 139, 13–30. [Google Scholar] [CrossRef]
  73. Fetoni, A.R.; Paciello, F.; Rolesi, R.; Paludetti, G.; Troiani, D. Targeting dysregulation of redox homeostasis in noise-induced hearing loss: Oxidative stress and ROS signaling. Free. Radic. Biol. Med. 2019, 135, 46–59. [Google Scholar] [CrossRef]
  74. Giorgio, M.; Migliaccio, E.; Orsini, F.; Paolucci, D.; Moroni, M.; Contursi, C.; Pelliccia, G.; Luzi, L.; Minucci, S.; Marcaccio, M.; et al. Electron transfer between cytochrome C and p66Shc generates reactive oxygen species that trigger mitochondrial apoptosis. Cell 2005, 122, 221–233. [Google Scholar] [CrossRef]
  75. Yamashita, D.; Minami, S.B.; Kanzaki, S.; Ogawa, K.; Miller, J.M. Bcl-2 genes regulate noise-induced hearing loss. J. Neurosci. Res. 2008, 86, 920–928. [Google Scholar] [CrossRef]
  76. Kurabi, A.; Keithley, E.M.; Housley, G.D.; Ryan, A.F.; Wong, A.C.Y. Cellular mechanisms of noise-induced hearing loss. Hear. Res. 2017, 349, 129–137. [Google Scholar] [CrossRef]
  77. Yamane, H.; Nakai, Y.; Konishi, K.; Sakamoto, H.; Matsuda, Y.; Iguchi, H. Strial circulation impairment due to acoustic trauma. Acta Otolaryngol. 1991, 111, 85–93. [Google Scholar] [CrossRef] [PubMed]
  78. Yamane, H.; Nakai, Y.; Takayama, M.; Iguchi, H.; Nakagawa, T.; Kojima, A. Appearance of free radicals in the guinea pig inner ear after noise-induced acoustic trauma. Eur. Arch. Otorhinolaryngol. 1995, 252, 504–508. [Google Scholar] [CrossRef] [PubMed]
  79. Chen, W.; White, M.A.; Cobb, M.H. Stimulus-specific requirements for MAP3 kinases in activating the JNK pathway. J. Biolog. Chem. 2002, 277, 49105–49110. [Google Scholar] [CrossRef] [PubMed]
  80. Ahmad, R.; Hussain, A.; Ahsan, H. Peroxynitrite: Cellular pathology and implications in autoimmunity. J. Immunoass. Immunochem. 2019, 40, 123–138. [Google Scholar] [CrossRef]
  81. Yamashita, D.; Jiang, H.Y.; Schacht, J.; Miller, J.M. Delayed production of free radicals following noise exposure. Brain Res. 2004, 1019, 201–209. [Google Scholar] [CrossRef]
  82. Mruk, D.D.; Silvestrini, B.; Mo, M.-y.; Cheng, C.Y. Antioxidant superoxide dismutase—a review: Its function, regulation in the testis, and role in male fertility. Contraception 2002, 65, 305–311. [Google Scholar] [CrossRef]
  83. Ohlemiller, K.K.; McFadden, S.L.; Ding, D.L.; Flood, D.G.; Reaume, A.G.; Hoffman, E.K.; Scott, R.W.; Wright, J.S.; Putcha, G.V.; Salvi, R.J. Targeted deletion of the cytosolic Cu/Zn-superoxide dismutase gene (Sod1) increases susceptibility to noise-induced hearing loss. Audiol. Neurootol. 1999, 4, 237–246. [Google Scholar] [CrossRef]
  84. Castaldo, A.; Linthicum, F.H. Stria vascularis hearing loss. Otol. Neurotol. 2006, 27, 285–286. [Google Scholar] [CrossRef]
  85. Wangemann, P. K(+) cycling and its regulation in the cochlea and the vestibular labyrinth. Audiol. Neurootol. 2002, 7, 199–205. [Google Scholar] [CrossRef]
  86. Hirose, K.; Liberman, M.C. Lateral wall histopathology and endocochlear potential in the noise-damaged mouse cochlea. J. Assoc. Res. Otolaryngol. 2003, 4, 339–352. [Google Scholar] [CrossRef]
  87. Ide, M.; Morimitsu, T. Long term effects of intense sound on endocochlear DC potential. Auris Nasus Larynx 1990, 17, 1–10. [Google Scholar] [CrossRef] [PubMed]
  88. Kuokkanen, J.; Aarnisalo, A.A.; Ylikoski, J. Efficiency of hyperbaric oxygen therapy in experimental acute acoustic trauma from firearms. Acta Otolaryngol. Suppl. 2000, 543, 132–134. [Google Scholar] [CrossRef] [PubMed]
  89. Le Prell, C.G.; Yamashita, D.; Minami, S.B.; Yamasoba, T.; Miller, J.M. Mechanisms of noise-induced hearing loss indicate multiple methods of prevention. Hear Res. 2007, 226, 22–43. [Google Scholar] [CrossRef] [PubMed]
  90. Sha, S.-H.; Taylor, R.; Forge, A.; Schacht, J. Differential vulnerability of basal and apical hair cells is based on intrinsic susceptibility to free radicals. Hear Res. 2001, 155, 1–8. [Google Scholar] [CrossRef]
  91. Cevette, M.J.; Vormann, J.; Franz, K. Magnesium and hearing. J. Am. Acad. Audiol. 2003, 14, 202–212. [Google Scholar] [CrossRef] [PubMed]
  92. Ising, H.; Handrock, M.; Günther, T.; Fischer, R.; Dombrowski, M. Increased noise trauma in guinea pigs through magnesium deficiency. Arch. Otorhinolaryngol. 1982, 236, 139–146. [Google Scholar] [CrossRef] [PubMed]
  93. Nageris, B.I.; Ulanovski, D.; Attias, J. Magnesium treatment for sudden hearing loss. Ann. Otol. Rhinol. Laryngol. 2004, 113, 672–675. [Google Scholar] [CrossRef] [PubMed]
  94. White, R.E.; Hartzell, H.C. Effects of intracellular free magnesium on calcium current in isolated cardiac myocytes. Science 1988, 239, 778–780. [Google Scholar] [CrossRef]
  95. Schanne, F.A.; Kane, A.B.; Young, E.E.; Farber, J.L. Calcium dependence of toxic cell death: A final common pathway. Science 1979, 206, 700–702. [Google Scholar] [CrossRef]
  96. Houston, M. The role of magnesium in hypertension and cardiovascular disease. J. Clin. Hypertens. 2011, 13, 843–847. [Google Scholar] [CrossRef] [PubMed]
  97. Das, U.N. Essential fatty acids: Biochemistry, physiology and pathology. Biotechnol. J. 2006, 1, 420–439. [Google Scholar] [CrossRef] [PubMed]
  98. Das, U.N. Nutrients, essential fatty acids and prostaglandins interact to augment immune responses and prevent genetic damage and cancer. Nutrition 1989, 5, 106–110. [Google Scholar]
  99. Das, U.N. Delta6 desaturase as the target of the beneficial actions of magnesium. Med. Sci. Monit. 2010, 16, LE11–LE12. [Google Scholar]
  100. Satoh, H.; Firestein, G.S.; Billings, P.B.; Harris, J.P.; Keithley, E.M. Tumor necrosis factor-alpha, an initiator, and etanercept, an inhibitor of cochlear inflammation. Laryngoscope 2002, 112, 1627–1634. [Google Scholar] [CrossRef]
  101. Wakabayashi, K.; Fujioka, M.; Kanzaki, S.; Okano, H.J.; Shibata, S.; Yamashita, D.; Masuda, M.; Mihara, M.; Ohsugi, Y.; Ogawa, K.; et al. Blockade of interleukin-6 signaling suppressed cochlear inflammatory response and improved hearing impairment in noise-damaged mice cochlea. Neurosci. Res. 2010, 66, 345–352. [Google Scholar] [CrossRef]
  102. Seist, R.; Landegger, L.D.; Robertson, N.G.; Vasilijic, S.; Morton, C.C.; Stankovic, K.M. Cochlin deficiency protects against noise-induced hearing loss. Front. Mol. Neurosci. 2021, 14, 670013. [Google Scholar] [CrossRef]
  103. Landegger, L.D.; Vasilijic, S.; Fujita, T.; Soares, V.Y.; Seist, R.; Xu, L.; Stankovic, K.M. Cytokine levels in inner ear fluid of young and aged mice as molecular biomarkers of noise-induced hearing loss. Front. Neurol. 2019, 10, 977. [Google Scholar] [CrossRef]
  104. Ren, Y.; Stankovic, K.M. The role of tumor necrosis factor alpha (TNFα) in hearing loss and vestibular schwannomas. Curr. Otorhinolaryngol. Rep. 2018, 6, 15–23. [Google Scholar] [CrossRef]
  105. Hirose, K.; Discolo, C.M.; Keasler, J.R.; Ransohoff, R. Mononuclear phagocytes migrate into the murine cochlea after acoustic trauma. J. Comp. Neurol. 2005, 489, 180–194. [Google Scholar] [CrossRef]
  106. Yang, W.; Vethanayagam, R.R.; Dong, Y.; Cai, Q.; Hu, B.H. Activation of the antigen presentation function of mononuclear phagocyte populations associated with the basilar membrane of the cochlea after acoustic overstimulation. Neuroscience 2015, 303, 1–15. [Google Scholar] [CrossRef]
  107. Frye, M.D.; Zhang, C.; Hu, B.H. Lower level noise exposure that produces only TTS modulates the immune homeostasis of cochlear macrophages. J. Neuroimmunol. 2018, 323, 152–166. [Google Scholar] [CrossRef] [PubMed]
  108. Hough, K.; Verschuur, C.A.; Cunningham, C.; Newman, T.A. Macrophages in the cochlea: An immunological link between risk factors and progressive hearing loss. Glia 2022, 70, 219–238. [Google Scholar] [CrossRef] [PubMed]
  109. Katsumi, S.; Sahin, M.I.; Lewis, R.M.; Iyer, J.S.; Landegger, L.D.; Stankovic, K.M. Intracochlear perfusion of tumor necrosis factor-alpha induces sensorineural hearing loss and synaptic degeneration in guinea pigs. Front. Neurol. 2020, 10, 1353. [Google Scholar] [CrossRef] [PubMed]
  110. Kujawa, S.G.; Liberman, M.C. Synaptopathy in the noise-exposed and aging cochlea: Primary neural degeneration in acquired sensorineural hearing loss. Hear. Res. 2015, 330, 191–199. [Google Scholar] [CrossRef]
  111. Jensen, J.B.; Lysaght, A.C.; Liberman, M.C.; Qvortrup, K.; Stankovic, K.M. Immediate and delayed cochlear neuropathy after noise exposure in pubescent mice. PLoS ONE 2015, 10, e0125160. [Google Scholar] [CrossRef]
  112. Pujol, R.; Rebillard, G.; Puel, J.L.; Lenoir, M.; Eybalin, M.; Recasens, M. Glutamate neurotoxicity in the cochlea: A possible consequence of ischaemic or anoxic conditions occurring in ageing. Acta Otolaryngol. Suppl. 1990, 476, 32–36. [Google Scholar] [CrossRef]
  113. Kujawa, S.G.; Liberman, M.C. Acceleration of age-related hearing loss by early noise exposure: Evidence of a misspent youth. J. Neurosci. 2006, 26, 2115–2123. [Google Scholar] [CrossRef] [PubMed]
  114. Eggermont, J.J. Effects of long-term non-traumatic noise exposure on the adult central auditory system. Hearing problems without hearing loss. Hear. Res. 2017, 352, 12–22. [Google Scholar] [CrossRef]
  115. Pienkowski, M.; Eggermont, J.J. Intermittent exposure with moderate-level sound impairs central auditory function of mature animals without concomitant hearing loss. Hear. Res. 2010, 261, 30–35. [Google Scholar] [CrossRef] [PubMed]
  116. Takacs, J.D.; Forrest, T.J.; Basura, G.J. Noise exposure alters long-term neural firing rates and synchrony in primary auditory and rostral belt cortices following bimodal stimulation. Hear. Res. 2017, 356, 1–15. [Google Scholar] [CrossRef] [PubMed]
  117. Seki, S.; Eggermont, J.J. Changes in spontaneous firing rate and neural synchrony in cat primary auditory cortex after localized tone-induced hearing loss. Hear Res 2003, 180, 28–38. [Google Scholar] [CrossRef]
  118. Agrawal, Y.; Platz, E.A.; Niparko, J.K. Prevalence of hearing loss and differences by demographic characteristics among US adults: Data from the National Health and Nutrition Examination Survey, 1999–2004. Arch. Intern. Med. 2008, 168, 1522–1530. [Google Scholar] [CrossRef]
  119. Shore, S.E.; Wu, C. Mechanisms of noise-induced tinnitus: Insights from cellular studies. Neuron 2019, 103, 8–20. [Google Scholar] [CrossRef] [PubMed]
  120. Agrawal, Y.; Platz, E.A.; Niparko, J.K. Risk factors for hearing loss in US adults: Data from the National Health and Nutrition Examination Survey, 1999 to 2002. Otol. Neurotol. 2009, 30, 139–145. [Google Scholar] [CrossRef] [PubMed]
  121. Moser, T.; Starr, A. Auditory neuropathy—Neural and synaptic mechanisms. Nat. Rev. Neurol. 2016, 12, 135–149. [Google Scholar] [CrossRef] [PubMed]
  122. Liberman, L.D.; Sukuzi, J.; Liberman, M.C. Dynamics of cochlear synaptopathy after acoustic overexposure. J. Assoc. Res. Otolaryngol. 2015, 16, 205–219. [Google Scholar] [CrossRef] [PubMed]
  123. Liberman, M.C. Morphological differences among radial afferent fibers in the cat cochlea: An electron-microscopic study of serial sections. Hear. Res. 1980, 3, 45–63. [Google Scholar] [CrossRef]
  124. Liberman, M.C. Single-neuron labeling in the cat auditory nerve. Science 1982, 216, 1239–1241. [Google Scholar] [CrossRef] [PubMed]
  125. Yin, Y.; Liberman, L.D.; Maison, S.F.; Liberman, M.C. Olivocochlear innervation maintains the normal modiolar-pillar and habenular-cuticular gradients in cochlear synaptic morphology. J. Assoc. Res. Otolaryngol. 2014, 15, 571–583. [Google Scholar] [CrossRef] [PubMed]
  126. Esmaili, A.A.; Renton, J. A review of tinnitus. Aust. J. Gen. Pract. 2018, 47, 205–208. [Google Scholar] [CrossRef]
  127. Shiomi, Y.; Tsuji, J.; Naito, Y.; Fujiki, N.; Yamamoto, N. Characteristics of DPOAE audiogram in tinnitus patients. Hear. Res. 1997, 108, 83–88. [Google Scholar] [CrossRef]
  128. Ceranic, B.J.; Prasher, D.K.; Raglan, E.; Luxon, L.M. Tinnitus after head injury: Evidence from otoacoustic emissions. J. Neurol. Neurosurg. Psychiatry 1998, 65, 523–529. [Google Scholar] [CrossRef]
  129. Shargorodsky, J.; Curhan, G.C.; Farwell, W.R. Prevalence and characteristics of tinnitus among US adults. Am. J. Med. 2010, 123, 711–718. [Google Scholar] [CrossRef]
  130. Shulman, A.; Wang, W.; Luo, H.; Bao, S.; Searchfield, G.; Zhang, J. Neuroinflammation and tinnitus. Curr. Top. Behav. Neurosci. 2021, 51, 161–174. [Google Scholar]
  131. Jang, D.I.; Lee, A.H.; Shin, H.Y.; Song, H.R.; Park, J.H.; Kang, T.B.; Lee, S.R.; Yang, S.H. The role of tumor necrosis factor alpha (TNF-α) in autoimmune disease and current TNF-α inhibitors in therapeutics. Int. J. Mol. Sci. 2021, 22, 2719. [Google Scholar] [CrossRef] [PubMed]
  132. Savastano, M. Tinnitus with or without hearing loss: Are its characteristics different? Eur. Arch. Otorhinolaryngol. 2008, 265, 1295–1300. [Google Scholar] [CrossRef]
  133. Martines, F.; Bentivegna, D.; Martines, E.; Sciacca, V.; Martinciglio, G. Assessing audiological, pathophysiological and psychological variables in tinnitus patients with or without hearing loss. Eur. Arch. Otorhinolaryngol. 2010, 267, 1685–1693. [Google Scholar] [CrossRef] [PubMed]
  134. Yankaskas, K. Prelude: Noise-induced tinnitus and hearing loss in the military. Hear. Res. 2013, 295, 3–8. [Google Scholar] [CrossRef]
  135. Mrena, R.; Savolainen, S.; Pirvola, U.; Ylikoski, J. Characteristics of acute acoustical trauma in the Finnish Defence Forces. Int. J. Audiol. 2004, 43, 177–181. [Google Scholar] [CrossRef]
  136. Nottet, J.B.; Moulin, A.; Brossard, N.; Suc, B.; Job, A. Otoacoustic emissions and persistent tinnitus after acute acoustic trauma. Laryngoscope 2006, 116, 970–975. [Google Scholar] [CrossRef] [PubMed]
  137. Day, B.L.; Fitzpatrick, R.C. The vestibular system. Curr. Biol. 2005, 15, R583–R586. [Google Scholar] [CrossRef]
  138. Golz, A.; Westerman, S.T.; Westerman, L.M.; Goldenberg, D.; Netzer, A.; Wiedmyer, T.; Fradis, M.; Joachims, H.Z. The effects of noise on the vestibular system. Am. J. Otolaryngol. 2001, 22, 190–196. [Google Scholar] [CrossRef]
  139. Oosterveld, W.J.; Polman, A.R.; Schoonheyt, J. Vestibular implications of noise-induced hearing loss. Br. J. Audiol. 1982, 16, 227–232. [Google Scholar] [CrossRef] [PubMed]
  140. McCue, M.P.; Guinan, J.J. Acoustically responsive fibers in the vestibular nerve of the cat. J. Neurosci. 1994, 14, 6058–6070. [Google Scholar] [CrossRef]
  141. Zhu, H.; Tang, X.; Wei, W.; Mustain, W.; Xu, Y.; Zhou, W. Click-evoked responses in vestibular afferents in rats. J. Neurophysiol. 2011, 106, 754–763. [Google Scholar] [CrossRef]
  142. Murofushi, T.; Curthoys, I.S.; Topple, A.N.; Colebatch, J.G.; Halmagyi, G.M. Responses of guinea pig primary vestibular neurons to clicks. Exp. Brain Res. 1995, 103, 174–178. [Google Scholar] [CrossRef]
  143. Ylikoski, J.; Juntunen, J.; Matikainen, E.; Ylikoski, M.; Ojala, M. Subclinical vestibular pathology in patients with noise-induced hearing loss from intense impulse noise. Acta Otolaryngol. 1988, 105, 558–563. [Google Scholar] [CrossRef] [PubMed]
  144. Manabe, Y.; Kurokawa, T.; Saito, T.; Saito, H. Vestibular dysfunction in noise induced hearing loss. Acta Otolaryngol. Suppl. 1995, 519, 262–264. [Google Scholar] [CrossRef] [PubMed]
  145. Ylikoski, J. Delayed endolymphatic hydrops syndrome after heavy exposure to impulse noise. Am. J. Otol. 1988, 9, 282–285. [Google Scholar]
  146. Shupak, A.; Bar-El, E.; Podoshin, L.; Spitzer, O.; Gordon, C.R.; Ben-David, J. Vestibular findings associated with chronic noise induced hearing impairment. Acta Otolaryngol. 1994, 114, 579–585. [Google Scholar] [CrossRef]
  147. Pyykkö, I.; Aalto, H.; Ylikoski, J. Does impulse noise induce vestibular disturbances? Acta Otolaryngol. Suppl. 1989, 468, 211–216. [Google Scholar] [CrossRef]
  148. Curthoys, I.S.; Vulovic, V.; Sokolic, L.; Pogson, J.; Burgess, A.M. Irregular primary otolith afferents from the guinea pig utricular and saccular maculae respond to both bone conducted vibration and to air conducted sound. Brain Res. Bull. 2012, 89, 16–21. [Google Scholar] [CrossRef]
  149. Dlugaiczyk, J. Ocular vestibular evoked myogenic potentials: Where are we now? Otol. Neurotol. 2017, 38, e513–e521. [Google Scholar] [CrossRef]
  150. Akin, F.W.; Murnane, O.D.; Tampas, J.W.; Clinard, C.; Byrd, S.; Kelly, J.K. The effect of noise exposure on the cervical vestibular evoked myogenic potential. Ear Hear. 2012, 33, 458–465. [Google Scholar] [CrossRef] [PubMed]
  151. McCue, M.P.; Guinan, J.J., Jr. Sound-evoked activity in primary afferent neurons of a mammalian vestibular system. Am. J. Otol. 1997, 18, 355–360. [Google Scholar]
  152. Stewart, C.E.; Kanicki, A.C.; Bauer, D.S.; Altschuler, R.A.; King, W.M. Exposure to intense noise causes vestibular loss. Mil. Med. 2020, 185, 454–461. [Google Scholar] [CrossRef]
  153. Jones, T.A.; Jones, S.M.; Vijayakumar, S.; Brugeaud, A.; Bothwell, M.; Chabbert, C. The adequate stimulus for mammalian linear vestibular evoked potentials (VsEPs). Hear. Res. 2011, 280, 133–140. [Google Scholar] [CrossRef]
  154. Tamura, H.; Ohgami, N.; Yajima, I.; Iida, M.; Ohgami, K.; Fujii, N.; Itabe, H.; Kusudo, T.; Yamashita, H.; Kato, M. Chronic exposure to low frequency noise at moderate levels causes impaired balance in mice. PLoS ONE 2012, 7, e39807. [Google Scholar] [CrossRef] [PubMed]
  155. McCabe, B.F.; Lawrence, M. The effects of intense sound on the non-auditory labyrinth. Acta Otolaryngol. 1958, 49, 147–157. [Google Scholar] [CrossRef] [PubMed]
  156. Kühl, A.; Dixon, A.; Hali, M.; Apawu, A.K.; Muca, A.; Sinan, M.; Warila, J.; Braun, R.D.; Berkowitz, B.A.; Holt, A.G. Novel QUEST MRI in vivo measurement of noise-induced oxidative stress in the cochlea. Sci. Rep. 2019, 9, 16265. [Google Scholar] [CrossRef]
  157. Fetoni, A.R.; Ferraresi, A.; Picciotti, P.; Gaetani, E.; Paludetti, G.; Troiani, D. Noise induced hearing loss and vestibular dysfunction in the guinea pig. Int. J. Audiol. 2009, 48, 804–810. [Google Scholar] [CrossRef] [PubMed]
  158. Soylemez, E.; Mujdeci, B. Dual-task performance and vestibular functions in individuals with noise induced hearing loss. Am. J. Otolaryngol. 2020, 41, 102665. [Google Scholar] [CrossRef]
  159. Musiek, F.E.; Shinn, J.; Chermak, G.D.; Bamiou, D.E. Perspectives on the pure-tone audiogram. J. Am. Acad. Audiol. 2017, 28, 655–671. [Google Scholar] [CrossRef]
  160. Kileny, P.R.; Zwolan, T.A.; Slager, H.K. Diagnostic audiology and electrophysiologic assessment of hearing. In Cummings Otolaryngology: Head and Neck Surgery; Flint, P., Ed.; Elsevier: Amsterdam, The Netherlands, 2021; pp. 2021–2041. [Google Scholar]
  161. Pierson, L.L.; Gerhardt, K.J.; Rodriguez, G.P.; Yanke, R.B. Relationship between outer ear resonance and permanent noise-induced hearing loss. Am. J. Otolaryngol. 1994, 15, 37–40. [Google Scholar] [CrossRef] [PubMed]
  162. Rodriguez, G.P.; Gerhardt, K.J. Influence of outer ear resonant frequency on patterns of temporary threshold shift. Ear Hear. 1991, 12, 110–114. [Google Scholar] [CrossRef] [PubMed]
  163. OSHA. 1904.10. Recording Criteria for Cases Involving Occupational Hearing Loss. Available online: https://www.osha.gov/laws-regs/regulations/standardnumber/1904/1904.10 (accessed on 12 December 2022).
  164. Kirchner, D.B.; Evenson, E.; Dobie, R.A.; Rabinowitz, P.; Crawford, J.; Kopke, R.; Hudson, T.W. Occupational noise-induced hearing loss: ACOEM Task Force on Occupational Hearing Loss. J. Occupat Environ. Med. 2012, 54, 106–108. [Google Scholar] [CrossRef]
  165. Moore, B.C.J.; Lowe, D.A.; Cox, G. Guidelines for diagnosing and quantifying noise-induced hearing loss. Trends Hear 2022, 26, 23312165221093156. [Google Scholar] [CrossRef]
  166. Moore, B.C.J. Diagnosis and quantification of military noise-induced hearing loss. J. Acoust. Soc. Am. 2020, 148, 884. [Google Scholar] [CrossRef]
  167. Moore, B.C. Cochlear hearing loss: Physiological, psychological and technical issues; John Wiley & Sons: Hoboken, NJ, USA, 2007. [Google Scholar]
  168. Liberman, M.C. Noise-induced and age-related hearing loss: New perspectives and potential therapies. F1000Res 2017, 6, 927. [Google Scholar] [CrossRef]
  169. Audiograms and Functional Auditory Testing to Assess Hearing Speech in Noise: A Review of the Clinical Evidence [Internet]. Ottawa (ON): Canadian Agency for Drugs and Technologies in Health; 2015 Aug 17. Appendix 1, Functional Tests to Assess Speech in Noise. Available online: https://www.ncbi.nlm.nih.gov/books/NBK315846/ (accessed on 13 January 2023).
  170. Le Prell, C.G.; Brungart, D.S. Speech-in-noise tests and supra-threshold auditory evoked potentials as metrics for noise damage and clinical trial outcome measures. Otol. Neurotol. 2016, 37, e295–e302. [Google Scholar] [CrossRef]
  171. Killion, M.C.; Niquette, P.A.; Gudmundsen, G.I.; Revit, L.J.; Banerjee, S. Development of a quick speech-in-noise test for measuring signal-to-noise ratio loss in normal-hearing and hearing-impaired listeners. J. Acoust. Soc. Am. 2004, 116, 2395–2405. [Google Scholar] [CrossRef]
  172. Wilson, R.H.; McArdle, R.A.; Smith, S.L. An evaluation of the BKB-SIN, HINT, QuickSIN, and WIN materials on listeners with normal hearing and listeners with hearing loss. J. Speech Lang. Hear. Res. 2007, 50, 844–856. [Google Scholar] [CrossRef]
  173. Taylor, B. Speech-in-noise tests: How and why to include them in your basic test battery. Hear. J. 2003, 56, 40, 42–46. [Google Scholar] [CrossRef]
  174. Anderson, S.; Kraus, N. Sensory-cognitive interaction in the neural encoding of speech in noise: A review. J. Am. Acad. Audiol. 2010, 21, 575–585. [Google Scholar] [CrossRef]
  175. Souza, P. Speech perception and hearing aids. In Hearing Aids; Popelka, G.R., Moore, B.C.J., Fay, R.R., Popper, A.N., Eds.; Springer Nature: Berlin, Germany, 2016; pp. 151–180. [Google Scholar]
  176. Davidson, A.; Marrone, N.; Wong, B.; Musiek, F. Predicting hearing aid satisfaction in adults: A systematic review of speech-in-noise tests and other behavioral measures. Ear Hear. 2021, 42, 1485–1498. [Google Scholar] [CrossRef]
  177. Hope, A.J.; Luxon, L.M.; Bamiou, D.E. Effects of chronic noise exposure on speech-in-noise perception in the presence of normal audiometry. J. Laryngol. Otol. 2013, 127, 233–238. [Google Scholar] [CrossRef]
  178. Dehghani-Ghahnaviyeh, S.; Zhao, Z.; Tajkhorshid, E. Lipid-mediated prestin organization in outer hair cell membranes and its implications in sound amplification. Nat. Commun. 2022, 13, 6877. [Google Scholar] [CrossRef]
  179. Brownell, W.E.; Bader, C.R.; Bertrand, D.; de Ribaupierre, Y. Evoked mechanical responses of isolated cochlear outer hair cells. Science 1985, 227, 194–196. [Google Scholar] [CrossRef]
  180. Fitzgerald, T.S.; Prieve, B.A. Detection of hearing loss using 2f2-f1 and 2f1-f2 distortion-product otoacoustic emissions. J. Speech Lang. Hear. Res. 2005, 48, 1165–1186. [Google Scholar] [CrossRef] [PubMed]
  181. Shera, C.A.; Guinan, J.J. Evoked otoacoustic emissions arise by two fundamentally different mechanisms: A taxonomy for mammalian OAEs. J. Acoust. Soc. Am. 1999, 105, 782–798. [Google Scholar] [CrossRef]
  182. Baiduc, R.R.; Dhar, S. Exploring optimal stimulus frequency ratio for measurement of the quadratic f2-f1 distortion product otoacoustic emission in humans. J. Speech Lang. Hear. Res. 2018, 61, 1794–1806. [Google Scholar] [CrossRef]
  183. Kapoor, N.; Mani, K.V.; Shukla, M. Distortion product oto-acoustic emission: A superior tool for hearing assessment than pure tone audiometry. Noise Health 2019, 21, 164–168. [Google Scholar] [PubMed]
  184. Abdala, C.; Dhar, S. Maturation and aging of the human cochlea: A view through the DPOAE looking glass. J. Assoc. Res. Otolaryngol. 2012, 13, 403–421. [Google Scholar] [CrossRef]
  185. Attias, J.; Bresloff, I.; Reshef, I.; Horowitz, G.; Furman, V. Evaluating noise induced hearing loss with distortion product otoacoustic emissions. Br. J. Audiol. 1998, 32, 39–46. [Google Scholar] [CrossRef] [PubMed]
  186. Mills, D.M. Determining the cause of hearing loss: Differential diagnosis using a comparison of audiometric and otoacoustic emission responses. Ear Hear. 2006, 27, 508–525. [Google Scholar] [CrossRef]
  187. Petersen, L.; Wilson, W.J.; Kathard, H. Towards the preferred stimulus parameters for distortion product otoacoustic emissions in adults: A preliminary study. S. Afr. J. Commun. Disord. 2018, 65, e1–e10. [Google Scholar] [CrossRef]
  188. Petrova, L.D. Brainstem auditory evoked potentials. Am. J. Electroneurodiagn. Technol. 2009, 49, 317–332. [Google Scholar] [CrossRef]
  189. Young, A.; Cornejo, J.; Spinner, A. Auditory Brainstem Response. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2022. Available online: https://www.ncbi.nlm.nih.gov/books/NBK564321/ (accessed on 19 January 2023).
  190. Møsller, A.R.; Jannetta, P.J.; Møsller, M.B. Neural generators of brainstem evoked potentials results from human intracranial recordings. Ann. Otol. Rhinol. Laryngol. 1981, 90, 591–596. [Google Scholar] [CrossRef]
  191. Parham, K.; Sun, X.-M.; Kim, D.O. Noninvasive assessment of auditory function in mice: Auditory brainstem response and distortion product otoacoustic emissions. In Handbook of Mouse Auditory Research; CRC Press: Boca Raton, FL, USA, 2001; pp. 51–72. [Google Scholar]
  192. Henry, K.R. Differential changes of auditory nerve and brain stem short latency evoked potentials in the laboratory mouse. Electroencephalogr. Clin. Neurophysiol. 1979, 46, 452–459. [Google Scholar] [CrossRef]
  193. Eggermont, J.J. Auditory brainstem response. Handb. Clin. Neurol. 2019, 160, 451–464. [Google Scholar]
  194. Alvarado, J.C.; Fuentes-Santamaría, V.; Jareño-Flores, T.; Blanco, J.L.; Juiz, J.M. Normal variations in the morphology of auditory brainstem response (ABR) waveforms: A study in Wistar rats. Neurosci. Res. 2012, 73, 302–311. [Google Scholar] [CrossRef]
  195. Chen, T.-J.; Chen, S.-S. Generator study of brainstem auditory evoked potentials by a radiofrequency lesion method in rats. Exp. Brain Res. 1991, 85, 537–542. [Google Scholar] [CrossRef]
  196. Gardi, J.N.; Berlin, C.I. Binaural interaction components. Their possible origins in guinea pig auditory brainstem response. Arch Otolaryngol 1981, 107, 164–168. [Google Scholar] [CrossRef]
  197. Melcher, J.R.; Kiang, N.Y.S. Generators of the brainstem auditory evoked potential in cat III: Identified cell populations. Hear. Res. 1996, 93, 52–71. [Google Scholar] [CrossRef]
  198. Bramhall, N.F.; Konrad-Martin, D.; McMillan, G.P.; Griest, S.E. Auditory brainstem response altered in humans with noise exposure despite normal outer hair cell function. Ear Hear. 2017, 38, e1–e12. [Google Scholar] [CrossRef]
  199. Coles, R.R.; Lutman, M.E.; Buffin, J.T. Guidelines on the diagnosis of noise-induced hearing loss for medicolegal purposes. Clin. Otolaryngol. Allied Sci. 2001, 25, 264–273. [Google Scholar] [CrossRef]
  200. Robinson, D.W. The audiogram in hearing loss due to noise: A probability test to uncover other causation. Ann. Occupat. Hyg. 1985, 29, 477–493. [Google Scholar]
  201. American Medical Association. Guides for the Evaluation of Permanent Impairment, 6th ed.; AMA: Chicago, IL, USA, 2021. [Google Scholar]
  202. Dobie, R.A. The AMA method of estimation of hearing disability: A validation study. Ear Hear. 2011, 32, 732–740. [Google Scholar] [CrossRef] [PubMed]
  203. US Environmental Protection Agency. Summary of the Noise Control Act-42 U.S.C. §4901 et seq. 1972. Available online: www.epa.gov/laws-regulations/summary-noise-control-act (accessed on 25 January 2023).
  204. OSHA. 1910.95 App A—Noise Exposure Computation. Available online: https://www.osha.gov/laws-regs/regulations/standardnumber/1910/1910.95AppA (accessed on 27 March 2023).
  205. NIOSH. Occupational Noise Exposure—Revised Criteria 1998. Available online: https://www.cdc.gov/niosh/docs/98-126/ (accessed on 27 March 2023).
  206. Centers for Disease Control and Prevention. Noise and Occupational Hearing Loss. Available online: https://www.cdc.gov/niosh/topics/noise/noise.html (accessed on 27 March 2023).
  207. OSHA. Hearing Conservation. Available online: https://www.osha.gov/sites/default/files/publications/osha3074.pdf (accessed on 19 January 2023).
  208. Arenas, J.P.; Suter, A.H. Comparison of occupational noise legislation in the Americas: An overview and analysis. Noise Health 2014, 16, 306–319. [Google Scholar] [CrossRef]
  209. NIOSH. NIOSH Sound Level Meter Application for iOS Devices. Available online: https://www.cdc.gov/niosh/topics/noise/pdfs/NIOSH-Sound-Level-Meter-Application-app-English.pdf (accessed on 19 January 2023).
  210. Crossley, E.; Biggs, T.; Brown, P.; Singh, T. The accuracy of iPhone applications to monitor environmental noise levels. Laryngoscope 2021, 131, E59–E62. [Google Scholar] [CrossRef]
  211. Smalt, C.J.; Ciccarelli, G.A.; Rodriguez, A.R.; Murphy, W.J. A deep neural-network classifier for photograph-based estimation of hearing protection attenuation and fit. J. Acoust. Soc. Am. 2021, 150, 1067. [Google Scholar] [CrossRef] [PubMed]
  212. Tikka, C.; Verbeek, J.H.; Kateman, E.; Morata, T.C.; Dreschler, W.A.; Ferrite, S. Interventions to prevent occupational noise-induced hearing loss. Cochrane Database Syst. Rev. 2017, 7, CD006396. [Google Scholar] [CrossRef] [PubMed]
  213. Salmani Nodoushan, M.; Mehrparvar, A.H.; Torab Jahromi, M.; Safaei, S.; Mollasadeghi, A. Training in using earplugs or using earplugs with a higher than necessary noise reduction rating? A randomized clinical trial. Int. J. Occup. Environ. Med. 2014, 5, 187–193. [Google Scholar]
  214. Smalt, C.J.; Calamia, P.T.; Dumas, A.P.; Perricone, J.P.; Patel, T.; Bobrow, J.; Collins, P.P.; Markey, M.L.; Quatieri, T.F. The effect of hearing-protection devices on auditory situational awareness and listening effort. Ear Hear. 2020, 41, 82–94. [Google Scholar] [CrossRef] [PubMed]
  215. Centers for Disease Control and Prevention. Preventing Noise-Induced Hearing Loss. Available online: https://www.cdc.gov/ncbddd/hearingloss/noise.html (accessed on 19 January 2023).
  216. Chandrasekhar, S.S.; Tsai Do, B.S.; Schwartz, S.R.; Bontempo, L.J.; Faucett, E.A.; Finestone, S.A.; Hollingsworth, D.B.; Kelley, D.M.; Kmucha, S.T.; Moonis, G.; et al. Clinical practice guideline: Sudden hearing loss (update). Otolaryngol. Head. Neck Surg. 2019, 161, S1–S45. [Google Scholar] [CrossRef]
  217. American Academy of Audiology. Audiology Clinical Practice Algorithms and Statements. Available online: https://www.audiology.org/practice-guideline/audiology-clinical-practice-algorithms-and-statements/ (accessed on 20 January 2023).
  218. National Guideline Centre (UK); National Institute for Health and Care Excellence (NICE). Hearing Loss in Adults: Assessment and Management. (NICE Guideline, No. 98.) 1, Guideline Summary. Available online: https://www.ncbi.nlm.nih.gov/books/NBK536565/ (accessed on 20 January 2023).
  219. Zhou, Y.; Zheng, G.; Zheng, H.; Zhou, R.; Zhu, X.; Zhang, Q. Primary observation of early transtympanic steroid injection in patients with delayed treatment of noise-induced hearing loss. Audiol. Neurotol. 2013, 18, 89–94. [Google Scholar] [CrossRef]
  220. Chang, Y.S.; Bang, K.H.; Jeong, B.; Lee, G.G. Effects of early intratympanic steroid injection in patients with acoustic trauma caused by gunshot noise. Acta Otolaryngol. 2017, 137, 716–719. [Google Scholar] [CrossRef]
  221. Olusanya, B.O.; Davis, A.C.; Hoffman, H.J. Hearing loss grades and the International classification of functioning, disability and health. Bull. World Health Organ. 2019, 97, 725–728. [Google Scholar] [CrossRef] [PubMed]
  222. Almeida, G.V.M.; Ribas, A.; Calleros, J. Free Field Word recognition test in the presence of noise in normal hearing adults. Braz. J. Otorhinolaryngol. 2017, 83, 665–669. [Google Scholar] [CrossRef] [PubMed]
  223. World Health Organization. Report of the Informal Working Group on Prevention of Deafness and Hearing Impairment Programme Planning, Geneva, 18–21 June 1991. Available online: https://apps.who.int/iris/handle/10665/58839 (accessed on 25 January 2023).
  224. Hapsari, A.A.; Kusmawan, D. Noise-Induced Hearing Loss (NIHL) Risk Factors Among Manufacturing Industry Workers: A Systematic Review [PREPRINT]. Available online: https://doi.org/10.21203/rs.3.rs-643998/v1 (accessed on 19 January 2023). [CrossRef]
  225. Toppila, E.; Pyykkö, I.I.; Starck, J.; Kaksonen, R.; Ishizaki, H. Individual risk factors in the development of noise-induced hearing loss. Noise Health 2000, 2, 59–70. [Google Scholar] [PubMed]
  226. Barone, J.A.; Peters, J.M.; Garabrant, D.H.; Bernstein, L.; Krebsbach, R. Smoking as a risk factor in noise-induced hearing loss. J. Occup. Med. 1987, 29, 741–745. [Google Scholar]
  227. Toppila, E.; Pyykkö, I.; Starck, J. Age and noise-induced hearing loss. Scand. Audiol. 2001, 30, 236–244. [Google Scholar] [CrossRef]
  228. Golmohammadi, R.; Darvishi, E. The combined effects of occupational exposure to noise and other risk factors—a systematic review. Noise Health 2019, 21, 125–141. [Google Scholar]
  229. Chen, K.H.; Su, S.B.; Chen, K.T. An overview of occupational noise-induced hearing loss among workers: Epidemiology, pathogenesis, and preventive measures. Environ. Health Prev. Med. 2020, 25, 65. [Google Scholar] [CrossRef]
  230. Masterson, E.A.; Bushnell, P.T.; Themann, C.L.; Morata, T.C. Hearing impairment among noise-exposed workers—United States, 2003–2012. MMWR Morb. Mortal. Wkly. Rep. 2016, 65, 389–394. [Google Scholar] [CrossRef]
  231. OSHA. Occupational Noise Exposure. Available online: https://www.osha.gov/noise (accessed on 25 January 2023).
  232. Tak, S.; Davis, R.R.; Calvert, G.M. Exposure to hazardous workplace noise and use of hearing protection devices among US workers--NHANES, 1999–2004. Am. J. Ind. Med. 2009, 52, 358–371. [Google Scholar] [CrossRef]
  233. Mirza, R.; Kirchner, D.B.; Dobie, R.A.; Crawford, J.; ACOEM Task Force on Occupational Hearing Loss. Occupational noise-induced hearing loss. J. Occup. Environ. Med. 2018, 60, e498–e501. [Google Scholar] [CrossRef] [PubMed]
  234. Yong, J.S.; Wang, D.Y. Impact of noise on hearing in the military. Mil. Med. Res. 2015, 2, 6. [Google Scholar] [CrossRef]
  235. Henselman, L.W.; Henderson, D.; Shadoan, J.; Subramaniam, M.; Saunders, S.; Ohlin, D. Effects of noise exposure, race, and years of service on hearing in US Army soldiers. Ear Hear. 1995, 16, 382–391. [Google Scholar] [CrossRef] [PubMed]
  236. Collee, A.; Legrand, C.; Govaerts, B.; Veken, P.; De Boodt, F.; Degrave, E. Occupational exposure to noise and the prevalence of hearing loss in a Belgian military population: A cross-sectional study. Noise Health 2011, 13, 64–70. [Google Scholar] [PubMed]
  237. Hughes, H.; Hunting, K.L. Evaluation of the effects of exposure to organic solvents and hazardous noise among US Air Force Reserve personnel. Noise Health 2013, 15, 379–387. [Google Scholar] [CrossRef]
  238. US Veteran’s Administration. 2021 Annual Benefits Report: Service-Connected Disability or Death Benefits. Available online: https://www.benefits.va.gov/REPORTS/abr/docs/2021_compensation.pdf (accessed on 30 January 2023).
  239. Gubata, M.E.; Packnett, E.R.; Feng, X.; Cowan, D.N.; Niebuhr, D.W. Pre-enlistment hearing loss and hearing loss disability among US soldiers and marines. Noise Health 2013, 15, 289–295. [Google Scholar] [CrossRef]
  240. Chandrasekhar, S.S.; Rubinstein, R.Y.; Kwartler, J.A.; Gatz, M.; Connelly, P.E.; Huang, E.; Baredes, S. Dexamethasone pharmacokinetics in the inner ear: Comparison of route of administration and use of facilitating agents. Otolaryngol. Head. Neck Surg. 2000, 122, 521–528. [Google Scholar]
  241. Leong, S.T.; Laortanakul, P. Monitoring and assessment of daily exposure of roadside workers to traffic noise levels in an Asian city: A case study of Bangkok streets. Environ. Monit. Assess. 2003, 85, 69–85. [Google Scholar] [CrossRef]
  242. World Health Organization. Regional Office for, E. Burden of Disease from Environmental Noise: Quantification of Healthy Life Years Lost in Europe; World Health Organization, Regional Office for Europe: Copenhagen, Denmark, 2011. [Google Scholar]
  243. Dillard, L.K.; Arunda, M.O.; Lopez-Perez, L.; Martinez, R.X.; Jiménez, L.; Chadha, S. Prevalence and global estimates of unsafe listening practices in adolescents and young adults: A systematic review and meta-analysis. BMJ Glob. Health 2022, 7, e010501. [Google Scholar] [CrossRef]
  244. English, W. 2021 National Firearms Survey (14 July 2021). Georgetown McDonough School of Business Research Paper No. 3887145. Available online: https://ssrn.com/abstract=3887145 (accessed on 25 January 2023).
  245. Bielefeld, E.C.; Harrison, R.T.; Riley DeBacker, J. Pharmaceutical otoprotection strategies to prevent impulse noise-induced hearing loss. J. Acoust. Soc. Am. 2019, 146, 3790. [Google Scholar] [CrossRef]
  246. Ohlemiller, K.K. Recent findings and emerging questions in cochlear noise injury. Hear. Res. 2008, 245, 5–17. [Google Scholar] [CrossRef]
  247. Watkins-Chow, D.E.; Pavan, W.J. Genomic copy number and expression variation within the C57BL/6J inbred mouse strain. Genome Res. 2008, 18, 60–66. [Google Scholar] [CrossRef]
  248. Heinonen-Guzejev, M.; Vuorinen, H.S.; Mussalo-Rauhamaa, H.; Heikkilä, K.; Koskenvuo, M.; Kaprio, J. Genetic component of noise sensitivity. Twin Res. Hum. Genet. 2005, 8, 245–249. [Google Scholar] [CrossRef]
  249. Konings, A.; Van Laer, L.; Van Camp, G. Genetic studies on noise-induced hearing loss: A review. Ear Hear. 2009, 30, 151–159. [Google Scholar] [CrossRef]
  250. Candreia, C.; Martin, G.K.; Stagner, B.B.; Lonsbury-Martin, B.L. Distortion product otoacoustic emissions show exceptional resistance to noise exposure in MOLF/Ei mice. Hear. Res. 2004, 194, 109–117. [Google Scholar] [CrossRef] [PubMed]
  251. Vázquez, A.E.; Jimenez, A.M.; Martin, G.K.; Luebke, A.E.; Lonsbury-Martin, B.L. Evaluating cochlear function and the effects of noise exposure in the B6.CAST+Ahl mouse with distortion product otoacoustic emissions. Hear. Res. 2004, 194, 87–96. [Google Scholar] [CrossRef] [PubMed]
  252. Yoshida, N.; Hequembourg, S.J.; Atencio, C.A.; Rosowski, J.J.; Liberman, M.C. Acoustic injury in mice: 129/SvEv is exceptionally resistant to noise-induced hearing loss. Hear. Res. 2000, 141, 97–106. [Google Scholar] [CrossRef] [PubMed]
  253. Sugahara, K.; Inouye, S.; Izu, H.; Katoh, Y.; Katsuki, K.; Takemoto, T.; Shimogori, H.; Yamashita, H.; Nakai, A. Heat shock transcription factor HSF1 is required for survival of sensory hair cells against acoustic overexposure. Hear. Res. 2003, 182, 88–96. [Google Scholar] [CrossRef]
  254. Tan, J.; Kaiserman, D.; O’Leary, S.J.; Bird, P.I. Increased susceptibility to acoustic trauma in a mouse model of non-syndromic sensorineural deafness, DFNB91. Eur. J. Neurosci. 2021, 53, 1638–1651. [Google Scholar] [CrossRef]
  255. Kozel, P.J.; Davis, R.R.; Krieg, E.F.; Shull, G.E.; Erway, L.C. Deficiency in plasma membrane calcium ATPase isoform 2 increases susceptibility to noise-induced hearing loss in mice. Hear. Res. 2002, 164, 231–239. [Google Scholar] [CrossRef]
  256. Ohlemiller, K.K.; McFadden, S.L.; Ding, D.L.; Lear, P.M.; Ho, Y.S. Targeted mutation of the gene for cellular glutathione peroxidase (Gpx1) increases noise-induced hearing loss in mice. J. Assoc. Res. Otolaryngol. 2000, 1, 243–254. [Google Scholar] [CrossRef]
  257. Tabuchi, K.; Suzuki, M.; Mizuno, A.; Hara, A. Hearing impairment in TRPV4 knockout mice. Neurosci. Lett. 2005, 382, 304–308. [Google Scholar] [CrossRef]
  258. Schick, B.; Praetorius, M.; Eigenthaler, M.; Jung, V.; Müller, M.; Walter, U.; Knipper, M. Increased noise sensitivity and altered inner ear MENA distribution in VASP-/- mice. Cell. Tissue Res. 2004, 318, 493–502. [Google Scholar] [CrossRef]
  259. Fairfield, D.A.; Lomax, M.I.; Dootz, G.A.; Chen, S.; Galecki, A.T.; Benjamin, I.J.; Dolan, D.F.; Altschuler, R.A. Heat shock factor 1-deficient mice exhibit decreased recovery of hearing following noise overstimulation. J. Neurosci. Res. 2005, 81, 589–596. [Google Scholar] [CrossRef] [PubMed]
  260. Duan, H.; Zhang, D.; Liang, Y.; Xu, C.; Wu, Y.; Tian, X.; Pang, Z.; Tan, Q.; Li, S.; Qiu, C. Heritability of age-related hearing loss in middle-aged and elderly chinese: A population-based twin study. Ear Hear. 2019, 40, 253–259. [Google Scholar] [CrossRef]
  261. Kvestad, E.; Czajkowski, N.; Krog, N.H.; Engdahl, B.; Tambs, K. Heritability of hearing loss. Epidemiology 2012, 23, 328–331. [Google Scholar] [CrossRef]
  262. Chen, X.-M.; Xue, X.-M.; Yu, N.; Guo, W.-W.; Yuan, S.-L.; Jiang, Q.-Q.; Yang, S.-M. The role of genetic variants in the susceptibility of noise-induced hearing loss. Front. Cell. Neurosci. 2022, 16, 946206. [Google Scholar] [CrossRef]
  263. Konings, A.; Van Laer, L.; Pawelczyk, M.; Carlsson, P.I.; Bondeson, M.L.; Rajkowska, E.; Dudarewicz, A.; Vandevelde, A.; Fransen, E.; Huyghe, J.; et al. Association between variations in CAT and noise-induced hearing loss in two independent noise-exposed populations. Hum. Mol. Genet. 2007, 16, 1872–1883. [Google Scholar] [CrossRef]
  264. Rabinowitz, P.M.; Pierce Wise, J.; Hur Mobo, B.; Antonucci, P.G.; Powell, C.; Slade, M. Antioxidant status and hearing function in noise-exposed workers. Hear. Res. 2002, 173, 164–171. [Google Scholar] [CrossRef] [PubMed]
  265. Fortunato, G.; Marciano, E.; Zarrilli, F.; Mazzaccara, C.; Intrieri, M.; Calcagno, G.; Vitale, D.F.; La Manna, P.; Saulino, C.; Marcelli, V.; et al. Paraoxonase and superoxide dismutase gene polymorphisms and noise-induced hearing loss. Clin. Chem. 2004, 50, 2012–2018. [Google Scholar] [CrossRef] [PubMed]
  266. Van Laer, L.; Carlsson, P.I.; Ottschytsch, N.; Bondeson, M.L.; Konings, A.; Vandevelde, A.; Dieltjens, N.; Fransen, E.; Snyders, D.; Borg, E.; et al. The contribution of genes involved in potassium-recycling in the inner ear to noise-induced hearing loss. Hum. Mutat. 2006, 27, 786–795. [Google Scholar] [CrossRef]
  267. Pawelczyk, M.; Van Laer, L.; Fransen, E.; Rajkowska, E.; Konings, A.; Carlsson, P.I.; Borg, E.; Van Camp, G.; Sliwinska-Kowalska, M. Analysis of gene polymorphisms associated with K ion circulation in the inner ear of patients susceptible and resistant to noise-induced hearing loss. Ann. Hum. Genet. 2009, 73, 411–421. [Google Scholar] [CrossRef]
  268. Sliwinska-Kowalska, M.; Pawelczyk, M. Contribution of genetic factors to noise-induced hearing loss: A human studies review. Mutat. Res. 2013, 752, 61–65. [Google Scholar] [CrossRef]
  269. Yoshida, N.; Kristiansen, A.; Liberman, M.C. Heat stress and protection from permanent acoustic injury in mice. J. Neurosci. 1999, 19, 10116–10124. [Google Scholar] [CrossRef] [PubMed]
  270. Yang, M.; Tan, H.; Yang, Q.; Wang, F.; Yao, H.; Wei, Q.; Tanguay, R.M.; Wu, T. Association of hsp70 polymorphisms with risk of noise-induced hearing loss in Chinese automobile workers. Cell. Stress. Chaperones 2006, 11, 233–239. [Google Scholar] [CrossRef]
  271. Konings, A.; Van Laer, L.; Michel, S.; Pawelczyk, M.; Carlsson, P.I.; Bondeson, M.L.; Rajkowska, E.; Dudarewicz, A.; Vandevelde, A.; Fransen, E.; et al. Variations in HSP70 genes associated with noise-induced hearing loss in two independent populations. Eur. J. Hum. Genet. 2009, 17, 329–335. [Google Scholar] [CrossRef]
  272. Konings, A.; Van Laer, L.; Wiktorek-Smagur, A.; Rajkowska, E.; Pawelczyk, M.; Carlsson, P.I.; Bondeson, M.L.; Dudarewicz, A.; Vandevelde, A.; Fransen, E.; et al. Candidate gene association study for noise-induced hearing loss in two independent noise-exposed populations. Ann. Hum. Genet. 2009, 73, 215–224. [Google Scholar] [CrossRef] [PubMed]
  273. Wang, Z.; Wilson, C.M.; Mendelev, N.; Ge, Y.; Galfalvy, H.; Elder, G.; Ahlers, S.; Yarnell, A.M.; LoPresti, M.L.; Kamimori, G.H.; et al. Acute and chronic molecular signatures and associated symptoms of blast exposure in military breachers. J. Neurotrauma 2020, 37, 1221–1232. [Google Scholar] [CrossRef] [PubMed]
  274. Guo, L.; Li, P.H.; Li, H.; Colicino, E.; Colicino, S.; Wen, Y.; Zhang, R.; Feng, X.; Barrow, T.M.; Cayir, A.; et al. Effects of environmental noise exposure on DNA methylation in the brain and metabolic health. Environ. Res. 2017, 153, 73–82. [Google Scholar] [CrossRef] [PubMed]
  275. Wen, L.T.; Wang, J.; Wang, Y.; Chen, F.Q. Association between histone deacetylases and the loss of cochlear hair cells: Role of the former in noise-induced hearing loss. Int. J. Mol. Med. 2015, 36, 534–540. [Google Scholar] [CrossRef]
  276. Chen, J.; Hill, K.; Sha, S.H. Inhibitors of histone deacetylases attenuate noise-induced hearing loss. J. Assoc. Res. Otolaryngol. 2016, 17, 289–302. [Google Scholar] [CrossRef]
  277. Yang, D.H.; Xie, J.; Liu, K.; Peng, Z.; Guo, J.Y.; Yu, S.K.; Wang, G.P.; Gong, S.S. The histone deacetylase inhibitor sodium butyrate protects against noise-induced hearing loss in Guinea pigs. Neurosci. Lett. 2017, 660, 140–146. [Google Scholar] [CrossRef] [PubMed]
  278. Xiong, H.; Long, H.; Pan, S.; Lai, R.; Wang, X.; Zhu, Y.; Hill, K.; Fang, Q.; Zheng, Y.; Sha, S.H. Inhibition of histone methyltransferase G9a attenuates noise-induced cochlear synaptopathy and hearing loss. J. Assoc. Res. Otolaryngol. 2019, 20, 217–232. [Google Scholar] [CrossRef]
  279. Ding, L.; Liu, J.; Shen, H.X.; Pan, L.P.; Liu, Q.D.; Zhang, H.D.; Han, L.; Shuai, L.G.; Ding, E.M.; Zhao, Q.N.; et al. Analysis of plasma microRNA expression profiles in male textile workers with noise-induced hearing loss. Hear. Res. 2016, 333, 275–282. [Google Scholar] [CrossRef]
  280. Li, Y.H.; Yang, Y.; Yan, Y.T.; Xu, L.W.; Ma, H.Y.; Shao, Y.X.; Cao, C.J.; Wu, X.; Qi, M.J.; Wu, Y.Y.; et al. Analysis of serum microRNA expression in male workers with occupational noise-induced hearing loss. Braz. J. Med. Biol. Res. 2018, 51, e6426. [Google Scholar] [CrossRef]
  281. Patel, M.; Cai, Q.; Ding, D.; Salvi, R.; Hu, Z.; Hu, B.H. The miR-183/Taok1 target pair is implicated in cochlear responses to acoustic trauma. PLoS ONE 2013, 8, e58471. [Google Scholar] [CrossRef]
  282. Leso, V.; Fontana, L.; Finiello, F.; De Cicco, L.; Luigia Ercolano, M.; Iavicoli, I. Noise induced epigenetic effects: A systematic review. Noise Health 2020, 22, 77–89. [Google Scholar]
  283. Tahera, Y.; Meltser, I.; Johansson, P.; Bian, Z.; Stierna, P.; Hansson, A.C.; Canlon, B. NF-kappaB mediated glucocorticoid response in the inner ear after acoustic trauma. J. Neurosci. Res. 2006, 83, 1066–1076. [Google Scholar] [CrossRef]
  284. Terunuma, T.; Hara, A.; Senarita, M.; Motohashi, H.; Kusakari, J. Effect of acoustic overstimulation on regulation of glucocorticoid receptor mRNA in the cochlea of the guinea pig. Hear. Res. 2001, 151, 121–124. [Google Scholar] [CrossRef] [PubMed]
  285. Curtis, L.M.; Rarey, K.E. Effect of stress on cochlear glucocorticoid protein. II. Restraint. Hear. Res. 1995, 92, 120–125. [Google Scholar] [CrossRef] [PubMed]
  286. Scheibe, F.; Haupt, H.; Vlastos, G.A. Preventive magnesium supplement reduces ischemia-induced hearing loss and blood viscosity in the guinea pig. Eur. Arch. Otorhinolaryngol. 2000, 257, 355–361. [Google Scholar] [CrossRef]
  287. Altura, B.M.; Altura, B.T.; Gebrewold, A.; Ising, H.; Günther, T. Noise-induced hypertension and magnesium in rats: Relationship to microcirculation and calcium. J. Appl. Physiol. 1992, 72, 194–202. [Google Scholar] [CrossRef]
  288. Choi, Y.H.; Miller, J.M.; Tucker, K.L.; Hu, H.; Park, S.K. Antioxidant vitamins and magnesium and the risk of hearing loss in the US general population. Am. J. Clin. Nutr. 2014, 99, 148–155. [Google Scholar] [CrossRef]
  289. Alvarado, J.C.; Fuentes-Santamaría, V.; Melgar-Rojas, P.; Gabaldón-Ull, M.C.; Cabanes-Sanchis, J.J.; Juiz, J.M. Oral antioxidant vitamins and magnesium limit noise-induced hearing loss by promoting sensory hair cell survival: Role of antioxidant enzymes and apoptosis genes. Antioxidants 2020, 9, 1177. [Google Scholar] [CrossRef]
  290. Abbasi, M.; Pourrajab, B.; Tokhi, M.O. Protective effects of vitamins/antioxidants on occupational noise-induced hearing loss: A systematic review. J. Occupat Health 2021, 63, e12217. [Google Scholar] [CrossRef]
  291. Le Prell, C.G.; Fulbright, A.; Spankovich, C.; Griffiths, S.K.; Lobarinas, E.; Campbell, K.C.; Antonelli, P.J.; Green, G.E.; Guire, K.; Miller, J.M. Dietary supplement comprised of β-carotene, vitamin C, vitamin E, and magnesium: Failure to prevent music-induced temporary threshold shift. Audiol. Neurotol. Extra 2016, 6, 20–39. [Google Scholar] [CrossRef] [PubMed]
  292. Attias, J.; Weisz, G.; Almog, S.; Shahar, A.; Wiener, M.; Joachims, Z.; Netzer, A.; Ising, H.; Rebentisch, E.; Guenther, T. Oral magnesium intake reduces permanent hearing loss induced by noise exposure. Am. J. Otolaryngol. 1994, 15, 26–32. [Google Scholar] [CrossRef] [PubMed]
  293. Attias, J.; Sapir, S.; Bresloff, I.; Reshef-Haran, I.; Ising, H. Reduction in noise-induced temporary threshold shift in humans following oral magnesium intake. Clin. Otolaryngol. Allied Sci. 2004, 29, 635–641. [Google Scholar] [CrossRef] [PubMed]
  294. Quaranta, A.; Scaringi, A.; Bartoli, R.; Margarito, M.A.; Quaranta, N. The effects of ’supra-physiological’ vitamin B12 administration on temporary threshold shift. Int. J. Audiol. 2004, 43, 162–165. [Google Scholar] [CrossRef]
  295. Kapoor, N.; Mani, K.V.; Shyam, R.; Sharma, R.K.; Singh, A.P.; Selvamurthy, W. Effect of vitamin E supplementation on carbogen-induced amelioration of noise induced hearing loss in man. Noise Health 2011, 13, 452–458. [Google Scholar] [CrossRef]
  296. Yeh, C.W.; Tseng, L.H.; Yang, C.H.; Hwang, C.F. Effects of oral zinc supplementation on patients with noise-induced hearing loss associated tinnitus: A clinical trial. Biomed. J. 2019, 42, 46–52. [Google Scholar] [CrossRef] [PubMed]
  297. Kopke, R.; Allen, K.A.; Henderson, D.; Hoffer, M.; Frenz, D.; Van de Water, T. A radical demise. Toxins and trauma share common pathways in hair cell death. Ann. NY Acad. Sci. 1999, 884, 171–191. [Google Scholar] [CrossRef]
  298. Coleman, J.K.; Kopke, R.D.; Liu, J.; Ge, X.; Harper, E.A.; Jones, G.E.; Cater, T.L.; Jackson, R.L. Pharmacological rescue of noise induced hearing loss using N-acetylcysteine and acetyl-L-carnitine. Hear. Res. 2007, 226, 104–113. [Google Scholar] [CrossRef]
  299. Aruoma, O.I.; Halliwell, B.; Hoey, B.M.; Butler, J. The antioxidant action of N-acetylcysteine: Its reaction with hydrogen peroxide, hydroxyl radical, superoxide, and hypochlorous acid. Free. Radic. Biol. Med. 1989, 6, 593–597. [Google Scholar] [CrossRef]
  300. Lin, C.Y.; Wu, J.L.; Shih, T.S.; Tsai, P.J.; Sun, Y.M.; Ma, M.C.; Guo, Y.L. N-Acetyl-cysteine against noise-induced temporary threshold shift in male workers. Hear. Res. 2010, 269, 42–47. [Google Scholar] [CrossRef] [PubMed]
  301. Kopke, R.; Slade, M.D.; Jackson, R.; Hammill, T.; Fausti, S.; Lonsbury-Martin, B.; Sanderson, A.; Dreisbach, L.; Rabinowitz, P.; Torre, P.; et al. Efficacy and safety of N-acetylcysteine in prevention of noise induced hearing loss: A randomized clinical trial. Hear. Res. 2015, 323, 40–50. [Google Scholar] [CrossRef] [PubMed]
  302. Niu, X.; Canlon, B. Protective mechanisms of sound conditioning. Adv. Otorhinolaryngol. 2002, 59, 96–105. [Google Scholar]
  303. Henderson, D.; McFadden, S.L.; Liu, C.C.; Hight, N.; Zheng, X.Y. The role of antioxidants in protection from impulse noise. Ann. NY Acad. Sci. 1999, 884, 368–380. [Google Scholar] [CrossRef]
  304. Jacono, A.A.; Hu, B.; Kopke, R.D.; Henderson, D.; Van De Water, T.R.; Steinman, H.M. Changes in cochlear antioxidant enzyme activity after sound conditioning and noise exposure in the chinchilla. Hear. Res. 1998, 117, 31–38. [Google Scholar] [CrossRef]
  305. Harris, K.C.; Bielefeld, E.; Hu, B.H.; Henderson, D. Increased resistance to free radical damage induced by low-level sound conditioning. Hear. Res. 2006, 213, 118–129. [Google Scholar] [CrossRef] [PubMed]
  306. Kil, J.; Harruff, E.E.; Longenecker, R.J. Development of ebselen for the treatment of sensorineural hearing loss and tinnitus. Hear. Res. 2022, 413, 108209. [Google Scholar] [CrossRef]
  307. Kil, J.; Pierce, C.; Tran, H.; Gu, R.; Lynch, E.D. Ebselen treatment reduces noise induced hearing loss via the mimicry and induction of glutathione peroxidase. Hear. Res. 2007, 226, 44–51. [Google Scholar] [CrossRef]
  308. Lynch, E.D.; Gu, R.; Pierce, C.; Kil, J. Ebselen-mediated protection from single and repeated noise exposure in rat. Laryngoscope 2004, 114, 333–337. [Google Scholar] [CrossRef]
  309. Kil, J.; Lobarinas, E.; Spankovich, C.; Griffiths, S.K.; Antonelli, P.J.; Lynch, E.D.; Le Prell, C.G. Safety and efficacy of ebselen for the prevention of noise-induced hearing loss: A randomised, double-blind, placebo-controlled, phase 2 trial. Lancet 2017, 390, 969–979. [Google Scholar] [CrossRef]
  310. Psillas, G.; Pavlidis, P.; Karvelis, I.; Kekes, G.; Vital, V.; Constantinidis, J. Potential efficacy of early treatment of acute acoustic trauma with steroids and piracetam after gunshot noise. Eur. Arch. Otorhinolaryngol. 2008, 265, 1465–1469. [Google Scholar] [CrossRef] [PubMed]
  311. Zloczower, E.; Tsur, N.; Hershkovich, S.; Fink, N.; Marom, T. Efficacy of oral steroids for acute acoustic trauma. Audiol. Neurotol. 2022, 27, 312–320. [Google Scholar] [CrossRef]
  312. Rhee, T.M.; Hwang, D.; Lee, J.S.; Park, J.; Lee, J.M. Addition of hyperbaric oxygen therapy vs medical therapy alone for idiopathic sudden sensorineural hearing loss: A systematic review and meta-analysis. JAMA Otolaryngol. Head. Neck Surg. 2018, 144, 1153–1161. [Google Scholar] [CrossRef] [PubMed]
  313. Suzuki, H.; Hashida, K.; Nguyen, K.-H.; Hohchi, N.; Katoh, A.; Koizumi, H.; Ohbuchi, T. Efficacy of intratympanic steroid administration on idiopathic sudden sensorineural hearing loss in comparison with hyperbaric oxygen therapy. Laryngoscope 2012, 122, 1154–1157. [Google Scholar] [CrossRef]
  314. Oya, M.; Tadano, Y.; Takihata, Y.; Ikomi, F.; Tokunaga, T. Utility of hyperbaric oxygen therapy for acute acoustic trauma: 20 years’ experience at the japan maritime self-defense force undersea medical center. Int. Arch. Otorhinolaryngol. 2019, 23, e408–e414. [Google Scholar] [CrossRef] [PubMed]
  315. Ylikoski, J.; Mrena, R.; Makitie, A.; Kuokkanen, J.; Pirvola, U.; Savolainen, S. Hyperbaric oxygen therapy seems to enhance recovery from acute acoustic trauma. Acta Otolaryngol. 2008, 128, 1110–1115. [Google Scholar] [CrossRef]
  316. Salihoğlu, M.; Ay, H.; Cincik, H.; Cekin, E.; Cesmeci, E.; Memis, A.; Uzun, G.; Altundag, A.; Simsek, K. Efficiency of hyperbaric oxygen and steroid therapy in treatment of hearing loss following acoustic trauma. Undersea Hyperb. Med. 2015, 42, 539–546. [Google Scholar] [PubMed]
  317. Fakhry, N.; Rostain, J.C.; Cazals, Y. Hyperbaric oxygenation with corticoid in experimental acoustic trauma. Hear. Res. 2007, 230, 88–92. [Google Scholar] [CrossRef]
  318. Lafère, P.; Vanhoutte, D.; Germonprè, P. Hyperbaric oxygen therapy for acute noise-induced hearing loss: Evaluation of different treatment regimens. Diving Hyperb. Med. 2010, 40, 63–67. [Google Scholar]
  319. Bayoumy, A.B.; van der Veen, E.L.; van Ooij, P.A.M.; Besseling-Hansen, F.S.; Koch, D.A.A.; Stegeman, I.; de Ru, J.A. Effect of hyperbaric oxygen therapy and corticosteroid therapy in military personnel with acute acoustic trauma. BMJ Mil. Health 2020, 166, 243–248. [Google Scholar] [CrossRef] [PubMed]
  320. Garcia, S.L.; Smith, K.J.; Palmer, C. Cost-effectiveness analysis of a military hearing conservation program. Mil. Med. 2018, 183, e547–e553. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Examples of noise exposure levels in occupational and non-occupational settings. Data from the United States National Institute for Occupational Safety and Health [30] and the Hearing Health Foundation [31].
Figure 1. Examples of noise exposure levels in occupational and non-occupational settings. Data from the United States National Institute for Occupational Safety and Health [30] and the Hearing Health Foundation [31].
Jcm 12 02347 g001
Figure 2. Anatomical structures of the inner ear impacted by NIHL. Three-panel diagram illustrating (a) cross section of gross outer, middle, and inner ear anatomy; (b) cross-sectional anatomy of the cochlea; (c) cellular-level anatomy of the sensory epithelium of the cochlea (organ of Corti). Original illustrations by Christine Gralapp and used with permission.
Figure 2. Anatomical structures of the inner ear impacted by NIHL. Three-panel diagram illustrating (a) cross section of gross outer, middle, and inner ear anatomy; (b) cross-sectional anatomy of the cochlea; (c) cellular-level anatomy of the sensory epithelium of the cochlea (organ of Corti). Original illustrations by Christine Gralapp and used with permission.
Jcm 12 02347 g002
Figure 3. Mechanisms of damage in NIHL. Abbreviations: ROS, reactive oxygen species; SGN, spiral ganglion neurite. Created with BioRender (www.biorender.com; accessed on 7 February 2023).
Figure 3. Mechanisms of damage in NIHL. Abbreviations: ROS, reactive oxygen species; SGN, spiral ganglion neurite. Created with BioRender (www.biorender.com; accessed on 7 February 2023).
Jcm 12 02347 g003
Figure 4. Characteristic audiogram of an individual with NIHL. The signature pattern of hearing loss is focused on 3–4 kHz range. Made with Interactive Audiogram Plotter (https://audprof.com/aud_tools/audiogram/; accessed on 1 February 2023).
Figure 4. Characteristic audiogram of an individual with NIHL. The signature pattern of hearing loss is focused on 3–4 kHz range. Made with Interactive Audiogram Plotter (https://audprof.com/aud_tools/audiogram/; accessed on 1 February 2023).
Jcm 12 02347 g004
Table 1. Occupational noise exposure limits recommended by NIOSH and OSHA.
Table 1. Occupational noise exposure limits recommended by NIOSH and OSHA.
Sound Pressure Level (dB)Permissible Exposure Time
NIOSHOSHA
1209 s7 min 30 s
115 28 s15 min
11256 s22 min 48 s
1101 min 29 s30 min
1091 min 53 s34 min 12 s
1063 min 45 s52 min 12 s
1054 min 43 s1 h
1037 min 30 s1 h 18 min
10015 min2 h
9730 min3 h
9547 min 37 s4 h
941 h4 h 36 min
912 h7 h
902 h 31 min8 h
884 h10 h 36 min
858 h16 h
8216 h24 h 18 min
8120 h 10 min27 h 54 min
8025 h 24 min32 h
The NIOSH limits represent clinically safe levels, and the OSHA limits are more liberal to account for practical issues in industry [204,205]. Units of time are standardized for ease of comparison. Abbreviations: dB, decibel; OSHA, Occupational Safety and Health Administration; NIOSH, National Institute for Occupational Safety and Health.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Natarajan, N.; Batts, S.; Stankovic, K.M. Noise-Induced Hearing Loss. J. Clin. Med. 2023, 12, 2347. https://doi.org/10.3390/jcm12062347

AMA Style

Natarajan N, Batts S, Stankovic KM. Noise-Induced Hearing Loss. Journal of Clinical Medicine. 2023; 12(6):2347. https://doi.org/10.3390/jcm12062347

Chicago/Turabian Style

Natarajan, Nirvikalpa, Shelley Batts, and Konstantina M. Stankovic. 2023. "Noise-Induced Hearing Loss" Journal of Clinical Medicine 12, no. 6: 2347. https://doi.org/10.3390/jcm12062347

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop