7,919
Views
46
CrossRef citations to date
0
Altmetric
Review Articles

Effects of noise exposure on auditory brainstem response and speech-in-noise tasks: a review of the literature

Pages S3-S32 | Received 11 Jun 2018, Accepted 04 Oct 2018, Published online: 18 Dec 2018

Abstract

Objective: Short-term noise exposure that induces transient changes in thresholds has induced permanent cochlear synaptopathy in multiple species. Here, the literature was reviewed to gain translational insight into the relationships between noise exposure, ABR metrics, speech-in-noise performance and TTS in humans.

Design: PubMed-based literature search, retrieval and review of full-text articles. Study Sample: Peer-reviewed literature identified using PubMed search.

Results: Permanent occupational noise-induced hearing loss (NIHL) is frequently accompanied by abnormal ABR amplitude and latency. In the absence of NIHL, there are mixed results for relationships between noise exposure and ABR metrics. Interpretation of speech-in-noise deficits is difficult as both cochlear synaptopathy and outer hair cell (OHC) loss can drive deficits. Reductions in Wave I amplitude during TTS may reflect temporary OHC pathology rather than cochlear synaptopathy. Use of diverse protocols across studies reduces the ability to compare outcomes across studies.

Conclusions: Longitudinal ABR and speech-in-noise data collected using consistent protocols are needed. Although speech-in-noise testing may not reflect cochlear synaptopathy, speech-in-noise testing should be completed as part of a comprehensive test battery to provide the objective measurement of patient difficulty.

Introduction

Exposure to loud sound has the potential to damage cells in the inner ear. It has long been known that the outer hair cells (OHCs) are particularly vulnerable to noise injury (Wang, Hirose, and Liberman Citation2002; for review, see Hu Citation2012), as well as other diverse insults such as occupational chemical exposure (for review, see Morata and Johnson Citation2012) and ototoxic drugs including aminoglycoside antibiotics and chemotherapeutics (for review, see Campbell and Le Prell Citation2018). More recently, it has become clear that the synapses connecting the inner hair cells (IHCs) to the auditory nerve dendrites are also vulnerable to loss as a consequence of noise (for review, see Kujawa and Liberman Citation2015), aminoglycoside antibiotics (Hinojosa and Lerner, Citation1987) and aging (Sergeyenko et al., Citation2013).

Hearing loss that occurs after exposure to loud sound may recover, in which case it is a temporary threshold shift (TTS) or the hearing loss may not resolve, in which case it is a permanent threshold shift (PTS) (for review, see Ryan et al. Citation2016). The classic literature on human TTS includes the measurement of TTS 2 minutes after the end of the noise exposure (TTS2) (Yates, Ramsey, and Holland Citation1976; Stephenson and Wall Citation1984). Threshold shifts at later times are a smaller, recovering, TTS (for review, see Ward Citation1970; Melnick Citation1991). It is more difficult to capture TTS2 in animal studies given the need to anaesthetise the animal prior to testing. In this review, TTS2 is used to denote classic 2-minute post-noise testing, and TTS without a specific time notation attached refers to a recovering TTS assessed at a longer post-noise time. A typical paradigm when using an animal model is to expose an awake animal to intense sound; post exposure, sound-evoked auditory brainstem response (ABR) thresholds are measured over the days and weeks after the noise exposure. Distortion product otoacoustic emission (DPOAE) thresholds may be monitored in place of or in combination with ABR thresholds, or DPOAE amplitude may be monitored. At the conclusion of the experiment, cochlear tissues are harvested and the OHC and IHC populations are counted.

One of the paradigms that has become increasingly common in recent years is the use of a short-term (two to four hours) sound exposure, with changes in both DPOAE and ABR thresholds and amplitude measured approximately 24 hours after the exposure and at one- to two-week intervals thereafter to monitor any permanent changes (Kujawa and Liberman Citation2006, Citation2009). Using such paradigms, there is now an elegant and convincing documentation of permanent auditory nerve synaptic pathology “hidden” behind normal hair cell counts, with a parallel decrease in ABR Wave I amplitude “hidden” behind normal threshold sensitivity. In this context, hidden synapse loss refers to a loss of synapses that occurs despite intact OHC and IHC populations – i.e. synapse loss cannot be inferred from the conventional OHC and IHC counts (as these cell counts are normal). The Wave I amplitude decreases have been termed “hidden hearing loss” as threshold measurements, the most common measurement of function, return to baseline, even though the amplitude of Wave I is decreased. The use of the term hidden hearing loss to specifically refer to decreased Wave I amplitude was proposed by Schaette and McAlpine (Citation2011). Since then, this term has also sometimes been used within the literature to refer to auditory dysfunction that is speculated to accompany the cochlear synaptopathy and accompanying decreased Wave I amplitude (Kujawa & Liberman, Citation2015).

Taken together, after noise exposure resulting in TTS, but no OHC loss and no PTS, permanent decreases in the number of synaptic connections between the IHCs and auditory nerve fibres resulting in decreased Wave I amplitude can be substantive, but will go undetected and unreported based on conventional reporting that is limited to threshold measures and counts of IHCs and OHCs. Fundamental to the interpretation of hidden hearing loss, i.e. a cochlear synaptopathy-driven functional deficit, the test battery must document that the middle ear conduction system is intact, typically accomplished using tympanometry, and there must be documentation that the OHC population has not been compromised, in order to attribute any observed functional deficits to the inferred “hidden” synapse loss.

Effects of noise on cochlear hair cells

Noise exposure can result in stereocilia damage or, in more severe cases, mechanical trauma to the OHCs or the organ of Corti itself (Henderson, Hamernik, and Sitler Citation1974; Henderson and Hamernik Citation1986; Wang, Hirose, and Liberman Citation2002). Noise-induced damage to the OHCs compromises threshold sensitivity; the electromotile action of the OHC population provides up to 40 dB of gain and the OHCs are therefore labelled the “cochlear amplifier” (Dallos and Evans Citation1995; Dallos, Zheng, and Cheatham Citation2006; Ashmore et al. Citation2010). If the OHCs are damaged, the loss of the cochlear amplifier will result in a reduced input to the IHCs. The IHCs have typically been documented to be less vulnerable to noise injury than the OHCs (Wang, Hirose, and Liberman Citation2002; Chen and Fechter Citation2003); however, Mulders, Chin and Robertson (Citation2018) recently argued that noise-induced injury to the IHCs plays a significant role in the Wave I amplitude reductions that are labelled hidden hearing loss. OHC loss shows only a moderate correlation with PTS, at least in part because OHCs may be present (living) but impaired (Chen and Fechter Citation2003).

With respect to monitoring noise-induced damage to the OHCs, a common strategy for monitoring OHC function is the use of DPOAE tests. Not only are DPOAEs well known for use identifying OHC damage due to cisplatin or aminoglycoside treatments (for recent review, see Campbell and Le Prell Citation2018), DPOAEs have shown high sensitivity to noise injury with deficits in DPOAE amplitude reportedly associated with occupational noise (Seixas et al. Citation2004; Korres et al. Citation2009; Seixas et al. Citation2012; Boger, Sampaio, and Oliveira Citation2017), recreational music player use (Santaolalla Montoya et al. Citation2008; Lee et al. Citation2014) and military service (de Souza Chelminski Barreto et al. Citation2011). Observations that noise-induced deficits in DPOAE and click-evoked otoacoustic emission (CEOAE) thresholds significantly exceeded noise-induced TTS have been used to suggest that OAEs are a more sensitive measure of noise injury than the audiogram itself (Attias and Bresloff Citation1996). DPOAE amplitude decreases may provide an early warning of pre-clinical damage and increased vulnerability for hearing loss (Lapsley Miller et al. Citation2006; Lapsley Miller and Marshall Citation2007). Despite their strengths and utility, it must be remembered that normal DPOAEs can be recorded even in the presence of OHC damage, and thus, normal DPOAEs do not necessarily imply the OHC population is not damaged (Subramaniam et al. Citation1994a, Citation1994b; Chen and Fechter Citation2003). Taken together, normal OAEs do not confirm the absence of OHC pathology, but they provide reasonable assurance that the cochlear amplifier is relatively intact.

Documentation of DPOAE amplitude as a measure of OHC integrity may be less important for inferences regarding cochlear synaptopathy if neural function is measured using higher level sound stimuli. At higher sound pressure levels, the OHCs do not provide any significant cochlear gain. Studies by Ruggero and Rich (Citation1991), Fridberger et al. (Citation2002), and Earl and Chertoff (Citation2012) provide evidence that compromised OHC function does not significantly decrease the magnitude of auditory nerve output in response to high-intensity stimuli (for additional discussion, see Adelman, Weinberger, and Sohmer Citation2010). Thus, studies including high-level stimuli sufficient to “bypass” the influence of OHCs are less reliant on use of DPOAE data to confirm normal OHC function when attempting to make inferences about the integrity of the auditory nerve pathway. In essence, if the stimulus level used for ABR recordings is sufficiently intense to stimulate IHCs and trigger neural firing directly, the interpretation of changes in ABR amplitude may be less complicated. However, if higher-level sound is used as a stimulus, the medial olivochlear (MOC) efferent reflex (Müller et al. Citation2005; Sun Citation2008; Verschooten et al. Citation2017) and the middle ear reflex (Sun Citation2008) may be stimulated, either of which can have the net effect of reducing sound-driven auditory nerve discharge.

Pathological neural consequences of noise that induces TTS

Detailed reviews of the literature on noise exposure, cochlear synaptopathy and permanent auditory nerve pathology clearly describe the potential for permanent pathology in the ascending neural pathway in the absence of noise-induced PTS (for reviews, see Plack, Barker and Prendergast Citation2014; Kujawa and Liberman Citation2015; Hickox et al. Citation2017; Kobel et al. Citation2017; Liberman Citation2017; Liberman and Kujawa Citation2017; Barbee et al. Citation2018; Le Prell Citation2018). Whereas loss of cochlear OHCs results in poorer sound detection thresholds (hearing loss) (for review see Saunders et al. Citation1991), a significant proportion of the IHC population can be lost without comprising thresholds (Lobarinas, Salvi and Ding Citation2013). As noted above, pathology that is limited to the IHCs, their synaptic connections to the auditory nerve dendrites and the auditory nerve itself have therefore been described as “hidden” as these pathologies can occur in the absence of PTS (see Schaette and McAlpine Citation2011). In rodent models, noise exposure that induces a TTS of 40 dB or greater (measured the day after the noise exposure) has resulted in permanent auditory nerve injury in multiple studies, even though complete post-noise threshold recovery was observed.

Noise-induced cochlear synaptopathy has been observed across diverse animal models including mice (Kujawa and Liberman Citation2009; Wang and Ren Citation2012; Fernandez et al. Citation2015), guinea pigs (Lin et al. Citation2011; Furman, Kujawa, and Liberman Citation2013), chinchillas (Hickman et al. Citation2018), rats (Altschuler et al. Citation2018) and nonhuman primates (Valero et al. Citation2017). These exposures typically employ a short-term noise exposure, with rodent subjects typically exposed to octave band noise for two-hours. Across rodents, the two-hour duration sound exposure level needed to induce cochlear synaptopathy has varied, with higher sound levels needed in the guinea pig (106 dB SPL, see Lin et al. Citation2011) and rat (109 dB SPL, see Lobarinas, Spankovich, and Le Prell Citation2017) than in the mouse (100 dB SPL, see Kujawa and Liberman, Citation2009; Fernandez et al. Citation2015). In the absence of noise exposure, age-related cochlear synaptopathy is also reliably observed. This age-related pathology has been reported in mice (Sergeyenko et al. Citation2013), rats (Möhrle et al. Citation2016) and gerbils (Viana et al. Citation2015; Gleich, Semmler, and Strutz Citation2016). One significant issue that has been identified long after recovery from an earlier TTS is the long-term slowly progressive expansion of synapse loss from the immediately damaged higher-frequency regions (at 22.6 and 32 kHz in the mouse cochlea) to the previously intact lower-frequency cochlear regions (at 5.6 and 11.3 kHz in the mouse cochlea) (Fernandez et al. Citation2015). A second significant issue is the slowly progressive loss of spiral ganglion neurons, with the total observed loss exceeding the otherwise expected age-related decline in spiral ganglion numbers (Kujawa and Liberman, Citation2006; Lin et al. Citation2011). Multiple recent reviews of these pathological consequences are available (Kujawa and Liberman Citation2015; Liberman and Kujawa, Citation2017), including a recent review of the extent to which permanent auditory nerve pathology can occur in parallel with cochlear hair cell pathology (Hickox et al. Citation2017).

Based on the above results, it has been suggested that the risks of TTS in humans exposed to intense sound may be greater than previously assumed (Kujawa and Liberman, Citation2006; Kujawa and Liberman, Citation2009). In addition, there has been fairly broad speculation that hidden neuropathic damage (immediate cochlear synaptopathy followed by slow spiral ganglion cell loss) could explain the disproportionate difficulties experienced by some individuals processing speech in noisy environments (Kujawa and Liberman, Citation2009; Lin et al. Citation2011; Makary et al. Citation2011). The relationships between TTS2, TTS recovery, auditory pathology and supra-threshold functional deficits are a topic of significant interest to scientists and clinicians, and there are concerns about the implications for occupational injury to workers who may be at risk for TTS (for additional discussion, see Kujawa and Liberman Citation2009; Kujawa Citation2014; Kujawa and Liberman Citation2015; Liberman and Kujawa Citation2017).

Audiometric monitoring for occupational noise-induced hearing loss

The federal noise regulations in the United States [i.e. 29 CFR 1910.95 enforced by the US Occupational Safety and Health Administration (OSHA)] and national regulations in other countries include annual audiometric testing requirements so that the development of NIHL can be detected and additional hearing loss prevented. Even if a worker wears a hearing protection device (HPD; typically earplugs or earmuffs), it is possible that annual audiometric testing will reflect temporary changes in hearing. Thus, if a new hearing loss is detected (relative to the baseline audiogram on file for that worker), there will be typically be some effort to complete a retest either immediately or after a noise-free interval to confirm the repeatability and/or permanence of the change.

Per 29 CFR 1904.10, hearing loss is required to be reported as an occupational noise injury when 1) a standard threshold shift (STS) is documented during annual audiometric testing (with STS defined as a permanent threshold change averaging 10 dB or more at the frequencies of 2, 3 and 4 kHz), 2) the average threshold at the 2, 3 and 4 kHz frequencies is 25 dB HL or poorer, and 3) exposure in the workplace may have caused or contributed to the hearing loss (OSHA Citation1983). Other agencies have provided different guidance in an effort to support earlier detection of NIHL. For example, the US National Institute on Occupational Safety and Health (NIOSH) advocates that a “significant threshold shift” be defined as a 15-dB threshold shift at 0.5, 1, 2, 3, 4 or 6 kHz, with an immediate retest used to confirm that the observed shift is not a function of test–retest reliability (NIOSH Citation1998). With earlier detection, earlier intervention is possible, with the goal of minimising additional hearing loss.

Although the OSHA regulations do not require testing at 8 kHz or above, poorer high-frequency hearing has been reported in workers exposed to occupational noise. Deficits have been reported within the extended high-frequency (EHF) range of 10 kHz and above in workers exposed to occupational noise (Hallmo, Borchgrevink, and Mair Citation1995; Korres et al. Citation2008; Riga et al. Citation2010; Mehrparvar et al. Citation2014), personal audio system device users (Le Prell et al. Citation2013; Sulaiman, Husain, and Seluakumaran Citation2015; Kumar et al. Citation2017), those with a history of musical training (Schmidt et al. Citation1994; Gonçalves et al. Citation2013; Liberman et al. Citation2016), frequent concert goers (Grose, Buss, and Hall Citation2017), those with acoustic trauma during military service (Balatsouras, Homsioglou, and Danielidis Citation2005; Buchler, Kompis, and Hotz Citation2012) and individuals with higher lifetime noise exposure (Prendergast et al. Citation2017a; Yeend et al. Citation2017). NIOSH recommends testing through 8 kHz. Inclusion of EHF frequencies above 8 kHz in monitoring programmes would require careful consideration of the effects of age at EHF frequencies, as these are significant (Stelmachowicz et al. Citation1989).

Although occupational noise reporting requirements are largely intended to capture the rate of new PTS injuries at the 2, 3 and 4 kHz frequencies (i.e. hearing loss meeting STS and other criteria laid out in 29 CFR 1904.10), there have also been efforts to understand and perhaps even regulate, noise injury based on TTS. For example, the Committee on Hearing, Bioacoustics, and Biomechanics (CHABA) proposed a limit for exposure to impulse noise (gunfire) based on generation of a specific amount of TTS, with the maximum allowable TTS defined as 10 dB at 1 kHz and below, 15 dB at 2 kHz, or 20 dB at 3 kHz and above (CHABA Citation1968, Citation1992). It has been suggested that these strategies have the potential for application to occupational noise as well. Specifically, Kryter et al. (Citation1966) proposed that TTS2 might serve as a measure “that will correlate with the ability of a single-day's exposure to produce a noise-induced, permanent threshold shift (NIPTS), if it is repeated on a near-daily basis, over a course of about ten years”.

There are continued efforts to understand the extent to which TTS is predictive for a later PTS (Fritze Citation1981; Moshammer et al. Citation2015; Chan, Ho, and Ryan Citation2016). One potential application of this work is the screening of workers for TTS vulnerability to identify those at increased risk of PTS; this was suggested by Moshammer et al. (Citation2015) based on their observation that workers with a larger TTS, as measured at least 10 min after a standardised noise exposure, also developed greater PTS during long-term follow-up (average follow-up time was 13 years, with testing at 3–5-year intervals). Taken together, although the cumulative effect of repeated daily occupational noise exposure on the auditory nerve currently remains unknown, it is clear that noise-induced cochlear synaptopathy has been observed in the absence of PTS in multiple animal models. If cochlear synaptopathy was to develop as a consequence of exposure to occupational noise, neither the threshold-based STS strategies mandated by OSHA nor the “red flag” early detection strategies recommended by NIOSH would be sensitive to IHC injury, cochlear synaptopathy or other hidden auditory nerve injuries (for additional discussion, see Kujawa and Liberman, Citation2009; Kujawa Citation2014; Kujawa and Liberman Citation2015; Liberman and Kujawa Citation2017). The translational relevance of cochlear synaptopathy to noise-exposed workers is a topic that remains actively under discussion (see Dobie and Humes Citation2017; Murphy and Le Prell Citation2017). While much of the literature on occupational NIHL is limited to threshold deficits, a number of studies include descriptions of ABR waveforms, perhaps providing an opportunity for potential insights into underlying noise-induced pathology.

Non-pathological TTS exposures

In studies in which rodent subjects have had a smaller TTS, i.e., a maximum of 20–30-dB TTS 24-hour post-noise, measurable synaptic pathology generally has not been documented (Hickox and Liberman, Citation2014; Fernandez et al. Citation2015; Jensen et al. Citation2015). Although it is appealing to infer that shorter or less intense exposures that result in these smaller TTS deficits are “safe” based on the lack of pathology in rodent models, the rodent studies have not been designed to allow broad conclusions regarding safety as they typically include only a single exposure. Because human exposures are likely to be repeated, from a practical perspective, an exposure is only “safe” if it can be repeated without injury emerging after subsequent exposures.

A small number of studies have combined repeated exposure paradigms with synapse counts in animal models (Wang and Ren Citation2012; Gannouni et al. Citation2015; Mannstrom, Kirkegaard, and Ulfendahl Citation2015). However, none of these repeated exposure paradigms were designed to provide insight into the effects of daily exposure to occupational noise over extended periods of time. Indeed, humans are likely to be exposed to intense sound repeatedly across their lifespan regardless of whether sound exposure is occupational, non-occupational or related to military service. Because the experimental data do not address whether repeated exposures, each resulting in a small, transient TTS, will ultimately result in permanent auditory nerve pathology akin to that associated with a single-larger TTS, the potential risk for human populations is a topic of interest.

Human cochlear synaptopathy: direct measurements and inferred pathology

The identification of noise-induced cochlear synaptopathy requires cochlear histology; thus, there are very few studies in which cochlear synaptopathy has been confirmed in humans. There is one report in which progressive age-related cochlear synaptopathy was suggested based on quantification of synapses in five human temporal bones from donors ranging in age from 54 to 89 years old (Viana et al. Citation2015). The observation of fewer synaptic connections in human temporal bones from older donors complements earlier reports describing age-related decreases in myelinated nerve fibres. Felder and Schrott-Fischer (Citation1995), for example, reported an age-related loss of nerve fibres in nine temporal bones from eight temporal bone donors; this loss was in addition to the expected hair cell loss that was also observed. These early observations were extended by Makary et al. (Citation2011), who documented selective age-related decreases in human cochlear spiral ganglion cell survival, in the absence of hair cell loss, in 100 temporal bones. The age-related neural and synaptic pathologies observed in human temporal bones parallel systematic observations of age-related cochlear synaptopathy in mice have not been exposed to noise and do not have other risk factors for hearing loss (Sergeyenko et al. Citation2013). The amplitude of Wave I of the ABR is highly correlated with synapse loss in mice (Sergeyenko et al. Citation2013). Thus, it is intriguing that age-related declines in ABR waveform amplitude in humans are well known from other human studies without histology components (Konrad-Martin et al. Citation2012; Skoe et al. Citation2015).

Speech-in-noise tests: a role in diagnosis of cochlear synaptopathy?

The role of IHC loss

The most commonly hypothesised functional effect of noise-induced neuropathic damage is difficulty understanding speech in noisy environments (Kujawa and Liberman Citation2009; Lin et al. Citation2011; Makary et al. Citation2011; see also the detailed discussions by Plack, Barker, and Prendergast Citation2014; Plack et al. Citation2016; Pienkowski Citation2017). Compelling evidence that selective loss of IHCs compromises hearing in noise was provided using carboplatin-induced lesions of the chinchilla IHC population in combination with a psychophysical tone in noise detection task (Lobarinas, Salvi, and Ding Citation2016). Consistent with this, Vinay and Moore (Citation2007) interpreted elevated thresholds on the threshold equalising noise (specified as dB hearing level) [TEN(HL)] test as perhaps reflecting the loss of neural synchrony, as their participants were previously diagnosed with auditory neuropathy. During the TEN(HL) test, pure-tone targets are presented in background noise, with the background noise spectrally shaped to interfere with off-frequency listening (Moore et al. Citation2000; Moore, Glasberg, and Stone Citation2004). In contrast to these observations, modelling of the effects of synapse loss on basic perceptual tasks suggests that the effects of 50% loss of synapses would be barely measurable – for example, a just-noticeable difference of 1 dB is modelled to increase to 1.4 dB with 50% loss of synapses (Oxenham Citation2016). The recent detailed review by Carney (Citation2018) argues against a direct role for cochlear synaptopathy in the coding of moderate to high-level speech sounds, pointing instead to interactions between cochlear gain (by OHCs) and IHC saturation as well as central processing within the brainstem and midbrain.

The role of OHC loss

Because damage to the OHC population is often observed in parallel with neural or synaptic pathologies (for detailed review, see Hickox et al. Citation2017), the extent to which speech-in-noise test outcomes (or more generically, signal-in-noise test outcomes) will be influenced by OHC loss must be considered in parallel to considerations of the effects of cochlear synaptopathy. Measurements of auditory nerve discharge in response to vowel sounds (in animal models) clearly indicates that impairment of both the OHCs and the IHCs can contribute to degraded representation of speech sounds by the auditory nerve (Bruce, Sachs and Young Citation2003). There is also a fairly extensive literature that suggests OHC damage may directly contribute to speech-in-noise deficits (see, for example, Leger, Moore and Lorenzi Citation2012; Summers et al. Citation2013; Hoben et al. Citation2017). Indirectly supporting a role of the OHCs in perception of speech in noise, the MOC system, which modulates the OHC response to sound, may aid speech perception in noise (Kumar and Vanaja Citation2004).

Specifically probing the role of the OHCs, Lutman and Saunders (Citation1992) reported no reliable differences in click-evoked OAEs when patients with complaints of difficulty understanding speech in noise (diagnosed as obscure auditory dysfunction) were compared to matched controls. As noted above, however, it is possible to obtain normal OAEs even in the presence of OHC damage, reducing confidence in assumptions that the OHC population had not been subtly damaged in the patients with obscure auditory dysfunction. Another possible approach is to use statistical techniques to account for OAE amplitude. For example, Ridley et al. (Citation2018) assessed participants with diverse noise exposure and diverse thresholds using the TEN(HL) test and reported that deficits detecting tones in noise were greater than expected after statistically adjusting for the expected effects of threshold shift and OAE amplitude. Bramhall et al. (Citation2015) also used statistical techniques to account for multi-variate interactions. They measured speech-in-noise using the Quick Sentences in Noise (QuickSin) test in addition to measuring the amplitude of Wave I of the ABR and found reduced ABR Wave I amplitudes as a function of increasing age. ABR Wave I amplitude was also associated with decreased performance on the QuickSin, but only in combination with poorer pure-tone thresholds. DPOAE data were not collected and the potential contributions of OHC loss are thus unknown.

The role of test difficulty

A major factor that must be taken into account in evaluating the literature for evidence of performance deficits is the specific perceptual task used in each investigation. A wide range of tests are available (for review and discussion, see Le Prell and Brungart Citation2016; Le Prell and Lobarinas Citation2016). Across tests, as the difficulty increases, performance of the participants decreases, and it is increasingly likely that subtle deficits will emerge even in normal hearing listeners when difficult listening tests are used (see Wilson, McArdle, and Smith Citation2007b).

One of the tests specifically targeted to children is the Bamford-Kowal-Bench Speech-in-Noise (BKB-SIN) test, in which the BKB sentence lists (Bench, Kowal, and Bambford Citation1979) are presented against four-talker babble with SNRs ranging from 21 dB SNR (easiest) to –6 dB SNR (hardest) (for review, see Etymotic Research Citation2005; Schafer Citation2010). Sentence levels systematically decrease from 96 dB SPL to 90 dB SPL, and the background babble level increases by 3 dB per sentence, making the test progressively harder with each additional sentence. A second commonly used sentence-based test is the Hearing in Noise Test (HINT), which also uses the BKB sentence-based speech materials, but with sentences presented at various sound levels in a background of spectrally matched masking noise presented at a fixed level of 72 dBA (Nilsson, Soli, and Sullivan Citation1994; Vermiglio Citation2008). The QuickSin is also a sentence-based test, with sentences presented at 70-dB HL, while the level of the four talker babble background is adjusted from 15-dB SNR (easiest) to 0-dB SNR (hardest) (Killion et al. Citation2004; McArdle and Wilson Citation2006). Finally, there are word-based tests, such as the Word in Noise (WIN) test. During testing with the WIN, NU-6 words are presented in multiple talker babble, with a female speaker against a background babble that includes six competing female voices. The babble is set at 80 dB SPL, and the target word levels decrease from 104 dB SPL (24 dB SNR) to 80 dB SPL (0 dB SNR) in 4-dB decrements, with five words per condition (Wilson, Carnell, and Cleghorn Citation2007a; Wilson and McArdle Citation2007). There are significant correlations with respect to performance across tests (Wilson, McArdle, and Smith Citation2007b). Although highly correlated, listeners typically do better on the BKB-SIN and the HINT (with sentences providing context) than the QuickSin or the WIN (which do not provide context from sentences) (Wilson, McArdle, and Smith Citation2007b). A suggested benefit of the WIN over the QuickSin is the reduced reliance on memory, cognition or other linguistic skills, as the participant repeats only the target word, rather than working to recall multiple words per sentence (Wilson, McArdle, and Smith Citation2007b).

Summary

The data generally suggest that damage to the OHCs and damage to the IHCs both have the potential to compromise speech-in-noise performance. To attribute deficits to cochlear synaptopathy, it is therefore necessary to exclude potential OHC pathology that could contribute to deficits on the speech-in-noise test, or otherwise correct for OHC dysfunction using statistical techniques. Caution is warranted as experimenters should assure deficits are related to poorer perceptual ability and not memory or cognition deficits. Taken together, the utility of speech-in-noise deficits as part of the differential diagnosis of the site of lesion within the organ of Corti remains questionable. As stated by Liberman and Kujawa (Citation2017), this area of research is in its infancy, but is crucial to the translation of findings to humans.

Purpose of the current review

As outlined above, there is significant interest in the potential strategies for diagnosis of cochlear synaptopathy in human patients, and a need to define the risk factors for cochlear synaptopathy in humans. The issue of noise-induced cochlear synaptopathy has been discussed in the context of a public health issue, based on suggestions that much of the population is at risk given the prevalence of many common non-occupational leisure noise exposures (Jensen et al. Citation2015; Liberman Citation2015). The issue of noise-induced cochlear synaptopathy has also been discussed as a potential occupational health issue, in that the current noise regulations define a threshold-based monitoring approach that will not be sensitive for diagnosing cochlear synaptopathy (Kujawa and Liberman Citation2009; Kujawa and Liberman Citation2015; Liberman and Kujawa Citation2017). Multiple research laboratories around the world have therefore initiated efforts to identify evidence that is consistent with cochlear synaptopathy and any accompanying supra-threshold deficits, with different electrophysiological tests and different functional tasks being used across laboratories and scientific teams (see data tables, below). The purpose of the summary tables provided in this review is to provide detailed information on study protocols and results within four specific topic areas: 1) the relationship between occupational noise exposure and ABR metrics, 2) the relationship between non-occupational noise exposure and ABR metrics, 3) the relationship between noise exposure history and speech-in-noise performance and 4) the relationship between noise exposure, TTS and ABR metrics.

Methods

Multiple PubMed literature searches were completed using diverse combinations of search terms including for example ABR and occupational noise, evoked potential and occupational noise, ABR and noise-induced hearing loss, evoked potential and noise-induced hearing loss, hidden hearing loss, cochlea and synaptopathy, noise-induced hearing loss and speech in noise, occupational noise and speech in noise, temporary threshold shift and hearing, temporary threshold shift and ABR, temporary threshold shift and evoked potential, temporary threshold shift and speech in noise, etc. PubMed is a free resource, developed and maintained by the National Center for Biotechnology Information at the National Library of Medicine. With more than 28 million citations available across thousands of journals, it provides a comprehensive search tool for articles dated back to at least 1966, with less consistent inclusion of references prior to that time. Although chapters, proceedings and government reports will not be identified via PubMed, peer-reviewed literature can be readily identified.

The author reviewed the resulting article title lists for relevance and carefully read abstracts of potentially relevant articles to determine whether the study included human participants with a history of noise exposure and then to confirm that the design included either sound-evoked auditory nerve response measurement (ABR or CAP) or a speech-in-noise test. All articles that appeared to be relevant were downloaded from the University of Texas at Dallas (UTD) library or requested through the UTD Inter-Library Loan service if the full-text was not available through the UTD library. Articles written in any language other than English were excluded from the full-text review. In addition to the PubMed search strategy, all full-text articles were reviewed for references to other published data relevant to the current review. Initial PubMed searches were completed in May of 2018; an additional search for new publications was completed in September 2018.

Noise exposure history was only grossly defined in the majority of the studies identified; in those cases where sound exposure levels or duration were provided by the authors, this information has been included in the data table. Although it would be helpful if noise exposures were characterised as short term/long term, continuous/intermittent/impulsive, single/repeated, with sound levels and sound spectra provided, very little information was provided in any of the retrieved studies discussed below. Variability in the accuracy of the noise exposure categorizations, including the accuracy of recall during the collection of self-reported noise exposure data, may contribute to differences in results across studies. Variable use of HPDs over the course of an occupational career, or across non-occupational events and activities, has the potential to influence deficits; reporting of HPD use is sporadic across reports, and even in those cases where HPD use was reported, it is not known if HPDs were used correctly or consistently.

Results

Relationship between occupational noise exposure and ABR metrics

A summary of the studies reporting ABR data for diverse noise-exposed worker populations is provided in . The most common test paradigm included the placement of electrodes on the forehead and mastoid and use of alternating polarity click stimuli. Stimulus presentation rates were variable, ranging from 10.3/sec to 71/sec. For the subset of studies reporting the total number of presentations averaged into the final response, 1024 and 2048 were most commonly reported. Only a small number of studies reported replication of the waveforms to assure repeatability. There was no consistent reporting convention for sound levels used during testing with five sound level measurement protocols (dB HL, dB nHL, dB SL, dB peSPL and dB SPL) used in the 13 reports listed in . As per Hall (Citation1992, see Chapter 4: Effect of stimulus factors), the most common convention within a clinical setting is to report stimulus levels in dB nHL (dB relative to normal hearing population), but all five stimulus measurement strategies are appropriate. For typical click stimulus conditions (0.1 msec click presented at rate of 10–20/sec), a 0 dB nHL click would correspond to click levels of 36.4-dB peak SPL and 29.9 dB SPL (Hall Citation1992). If those conversion factors are applied to the sound levels listed in , then sound levels across studies were on the order of 70 dB nHL (Almadori et al. Citation1988; Chen, Chiang, and Chen Citation1992; Donaldson and Ruth, Citation1996; Thakur, Anand, and Banerjee Citation2004), 75 dB nHL (Attias and Pratt, Citation1984; Konrad-Martin et al. Citation2012), 80 dB nHL (Samelli et al. Citation2012; Karawani et al. Citation2015; Pushpalatha and Konadath, Citation2016) or 90 dB nHL (Attias et al. Citation1996; Noorhassim, Kaga, and Nishimura Citation1996; Xu et al. Citation1998).

Table 1. Relationship between occupational noise exposure and auditory brainstem response (ABR) metrics.

Across studies, there were multiple reports that the latency for Waves I, III, and/or V were delayed in workers exposed to noise, with a smaller number of studies measuring or reporting the amplitude for Waves I, III or V. One exception to this pattern of results was observed for the professional musicians assessed by Samelli et al. (Citation2012), with no significant deficits in ABR Waves I, III or V detected in professional musicians with or without hearing loss. A small number of additional studies assessed ABR metrics in military populations with a significant noise exposure history, but the effects of noise could not be readily extracted. First, a study by Attias et al. (Citation1996), assessing the relationships between ABR amplitude and tinnitus, did not include a noise-free control group. Second, a study by Konrad-Martin et al. (Citation2012), assessing the relationships between ABR amplitude and aging, reported that noise exposure was included in the statistical model, but the effects of noise were not specifically reported or discussed.

Although prolonged ABR waveform latencies were commonly reported across studies, most of these studies also reported that the noise-exposed worker cohorts had significant hearing loss. The presence of a permanent NIHL suggests OHC pathology accompanied any neuropathic change that might potentially be inferred from the ABR latency increases and other atypical ABR results listed in . Decreased DPOAE amplitude is well documented in noise-exposed populations (Seixas et al., Citation2004; Korres et al. Citation2009; Seixas et al. Citation2012; Boger, Sampaio, and Oliveira Citation2017). As seen in , many of the studies employing ABR metrics to assess potential effects of occupational noise exposures did not include the parallel collection of DPOAE data, complicating the interpretation of the observed changes in ABR amplitude and latency.

The clinical utility of DPOAEs was not generally recognised prior to the 1990s (Lonsbury-Martin and Martin, Citation1990), and thus, it is not surprising that DPOAEs were not conducted at least in the earlier studies listed within . Nonetheless, with stimulus levels of 70–100 dB nHL (i.e., approx 100–130 dB SPL), the studies listed in presumably used stimuli that were sufficiently intense to stimulate IHCs and trigger neural firing directly via the passive mechanics of the basilar membrane. At these sound levels, the passive mechanics of the cochlea dominate after the active process has saturated (see models and discussion in Johnstone, Patuzzi, and Yates Citation1986), and thus one might speculate that neural pathology contributed to delayed ABR latencies and reduced ABR amplitudes, even if permanent threshold deficits were caused by undocumented OHC loss. Although speculative, this interpretation is consistent with the conclusion by Hickox et al. (Citation2017) that OHC loss and cochlear synaptopathy are likely to occur in parallel. Given this interpretation of potential neural pathology in workers exposed to occupational noise, pre-clinical research in animal models should directly assess the potential for synapse loss with lower level repeated exposures modelled after occupational noise. Longitudinal studies of workers would also be helpful in determining effects of noise on the auditory nerve in workers exposed to occupational noise. Variation in vulnerability to NIHL is considerable in humans (see for example the 10th versus the 90th percentiles in International Standard Organization Citation2013) and variability is also well documented in animal models (Maison and Liberman Citation2000).

Relationship between non-occupational noise exposure and ABR metrics

summarises methods and results from studies assessing potential cochlear synaptopathy in human participants using ABR Wave I amplitude as a metric. Whereas studies of cochlear synaptopathy in rodent models have typically relied on short-term noise exposure paradigms that induce a large TTS lasting at least 24 hours, many of the sounds that human populations are commonly exposed to are likely to induce a smaller and shorter duration TTS. Changes in hearing in those attending concerts or clubs are commonly on the order of 10 dB immediately after the event (Opperman et al. Citation2006; Derebery et al. Citation2012; Ramakers et al. Citation2016). Moreover, threshold recovery is likely to be complete within 24 hours of the event for most attendees (Grinn et al. Citation2017). Concerns about the hazards of the repetition of such exposure, in combination with other intense sound encountered on a regular basis, led Liberman (Citation2015) to suggest that the cochlear synaptopathy observed in animals “raises questions about the risks of routine exposure to loud music at concerts and clubs and via personal listening devices”. Similarly, Jensen et al. (Citation2015) pointed to the increasing sales of portable listening devices and similarly suggested a population of at-risk adolescents based on the use of such devices. As per , this has resulted in a number of relatively more recent investigations focussing on participants with less noise exposure, better hearing thresholds and intact DPOAE function.

Table 2. Relationship between non-occupational noise exposure and auditory brainstem response (ABR) metrics.

Consistent with the results summarised in , the most common test paradigm involves placement of electrodes on the forehead and mastoid and use of alternating polarity click stimuli, although the use of foil-wrapped “TipTrode” ear canal electrodes is increasingly common and a number of studies now include tone bursts in addition to click stimuli. The total number of presentations averaged into the final response was highly variable across studies, with stimulus averaging ranging from 500 (Grinn et al. Citation2017) to 12,500 (Valderrama et al. Citation2018). As discussed by Valderrama et al. (Citation2018), averaging across larger numbers of stimulus presentations improves data quality, with improved signal to noise ratios (SNR) for waveforms. According to Hall (Citation1992), the most robust improvement in SNR is obtained over the initial 1000 sweeps (from the start of averaging to the completion of 1000 sweeps), with lessor improvement from 1000 to 2000 sweeps. However, near threshold, as many as 4000 sweeps may be necessary to accurately distinguish small neural responses from the noise floor (for discussion see Chapter 5, Effect of acquisition factors, in Hall Citation1992).

Stimulus rates were also highly variable. Although most studies used either 11/sec or 27/sec rates, Skoe and Tufts (Citation2018) assessed responses with stimulus presentation rates ranging from 3.4/sec to 91.24/sec. For click rates of 3.1 to 21.1/sec, AP amplitude and latency should be relatively constant, but amplitude progressively decreases and latency progressively increases as click rates increase from 31.1 to 91.1/sec (Hall Citation1992; for illustration of these changes, see his Figure 4–21). Effects of click rate on ABR Wave I amplitude and latency are similar; responses are generally equivalent when click rates are below 20/sec but as click rates increase, amplitude and latency progressively change for Waves I, III and V (Hall, Citation1992; for illustration of these changes, see his Figure 4–22).

As shown in , there have been mixed results across studies assessing potential relationships between non-occupational noise exposure and ABR Wave I amplitude. Several studies assessing this phenomena obtained results that were consistent with synaptic and/or auditory nerve pathology based on observations of decreased ABR Wave I amplitude as a function of noise exposure (Stamper and Johnson, Citation2015a; Bramhall et al. Citation2017; Valderrama et al. Citation2018). The decreased Wave I amplitudes observed by Stamper and Johnson (Citation2015a) were generally attributed to recreational noise, which was estimated based on self-reported exposure in the past 12-months. The decreased Wave I amplitudes observed by Valderrama et al. (Citation2018) were observed in association with increasing lifetime noise exposure, which accrues both as a function of leisure and work-related activities, although the relationship was no longer statistically significant if a single outlier was removed from the analysis. In contrast, the decreased Wave I amplitudes reported by Bramhall et al. (Citation2017) were generally attributed to recreational firearm use (civilians) or service-related firearm use (military personnel), with participants distributed into four discrete groups based on use of firearms and exposure to other intense sound.

In addition to reports of associations between Wave I amplitude and historic noise exposure, data expressed as normalised evoked potential metrics (such as the SP/AP ratio, and ABR Wave V/I ratio) have been interpreted as reflecting potential noise-induced cochlear synaptopathy and/or auditory nerve pathology (Liberman et al. Citation2016; Grose, Buss, and Hall Citation2017). The decreased SP/AP ratios observed by Liberman et al. (Citation2016) were generally attributed to musical rehearsals and performance associated with enrolment in a music conservatory, whereas the decreased Wave V/I ratios reported by Grose, Buss, and Hall (Citation2017) were specifically attributed to frequent concert attendance (which was one component of the enrolment criteria). In another recent study with normal hearing participants, the latency of the ABR waveforms was delayed even though amplitude did not appear to have been affected by previous noise exposure (Skoe and Tufts Citation2018); participants in this investigation were distributed within low-noise and high-noise groups based on the results of one-week of body worn dosimetry data collection.

In contrast to these positive results, a number of other studies have not found evidence of decreased ABR Wave I amplitude or changes in latency as a function of either lifetime noise exposure or the previous 12-month exposure period. In studies completed by Spankovich et al. (Citation2017), Fulbright et al. (Citation2017), Grinn et al. (Citation2017) and, most recently, Ridley et al. (Citation2018), noise exposure was estimated following Stamper and Johnson (Citation2015a), with noise exposure over the previous 12 months surveyed using the Noise Exposure Questionnaire (NEQ) and changes from previous years queried. No relationships between noise exposure and Wave I amplitude were detected in any of these studies. A comprehensive series of studies completed at the University of Manchester used detailed interview data to calculate a single comprehensive lifetime noise score (Guest et al. Citation2017; Prendergast et al. Citation2017; Prendergast, Millman et al.,2017b; Guest et al. Citation2018; Prendergast et al. Citation2018). No relationships between noise exposure and Wave I amplitude were detected in any of these studies. As part of the comprehensive analysis by Prendergast et al. (Citation2017a), it was noted that initial observations suggested that ABR wave V latency increased with increasing noise exposure (for the 80 dB peSPL click) and that frequency following response (FFR) signal-to-noise ratios decreased as a function of noise exposure (in males but not females), but neither of these correlations remained significant after controlling for the effects of age. Given observations of age-related synaptopathy in the absence of noise exposure, it is critical to adjust for age when samples with a wide age range are recruited. As per , most studies have recruited participants in their 20’s–30’s, with a smaller number of studies enrolling participants in their 40’s or 50’s.

Relationship between noise exposure and speech-in-noise performance

The most commonly hypothesised functional effect of noise-induced neuropathic damage is difficulty understanding speech in noisy environments (Kujawa and Liberman Citation2009; Lin et al. Citation2011; Makary et al. Citation2011; see also the detailed discussions by Plack, Barker, and Prendergast Citation2014; Plack et al. Citation2016; Le Prell and Clavier Citation2017; Pienkowski Citation2017; Le Prell Citation2018). therefore includes a summary of studies that have assessed the relationships between speech-in-noise and noise exposure history. Although some of these studies included ABR assessments (and those studies are therefore repeated from ), the collection of ABR data was not required for inclusion in . The literature search strategy for was specifically based on speech-in-noise deficits analysed as a function of noise exposure history. If other signal-in-noise tests were completed in parallel with speech-in-noise tests, those results are also listed in . However, a systematic search and review of outcomes across all articles reporting signal-in-noise outcomes were outside the scope of the current review and studies that included signal-in-noise tests, but not speech-in-noise tests, are not included here.

Table 3. Relationship between noise exposure history and speech-in-noise test metrics.

To be included in , normal (or near normal) hearing for participants was required. Certainly, there are a number of studies in which workers with permanent NIHL have poorer word-in-noise test outcomes (Smoorenburg Citation1992; Leensen, de Laat, and Dreschler Citation2011; Leensen et al. Citation2011; Jansen et al. Citation2014). Some of these deficits are related to audibility of the test signals, although even after correcting for audibility, deficits are often detected (for discussion, see Le Prell and Brungart Citation2016). specifically illustrates the various patterns of deficits that have been reported in worker populations with small (i.e., 5–10 dB) deficits relative to controls (Alvord Citation1983; Kujala et al. Citation2004; Kumar, Ameenudin, and Sangamanatha 2012; Hope, Luxon, and Bamiou Citation2013), and other populations with normal hearing.

The studies by Fulbright et al. (Citation2017), Grinn et al. (Citation2017), Le Prell et al. (Citation2018) and Bramhall, Konrad-Martin, and McMillan (Citation2018) used the relatively difficult WIN test (described above). There were no relationships between WIN threshold and noise exposure history (Fulbright et al. Citation2017; Grinn et al. Citation2017; Le Prell et al. Citation2018) or WIN threshold and Wave I amplitude (Bramhall, Konrad-Martin, and McMillan Citation2018). In the studies by Yeend et al. (Citation2017) and Valderrama et al. (Citation2018), the Listening in Spatialized Noise – Sentences High Cue Condition (LiSN-S) was used. During the LiSN-S test, two-talker masker noise is presented at 61 dB SPL 90 degrees relative to the listener, and the target speech is delivered at 68 dB SPL, 0 degrees relative to the listener. Neither study reported relationships between LiSN-S performance and noise history, although Valderrama et al. (Citation2018) did report that longer ABR interpeak latencies and reduced central gain (less growth of Wave-V amplitude relative to Wave-I amplitude) was associated with poorer performance on this test. As part of a comprehensive test battery, Yeend et al. (Citation2017) also completed the National Acoustic Laboratories Dynamic Conversations Test (NAL-DCT), a temporal fine structure task, the TEN test, and an amplitude modulation test. There was no evidence that noise exposure history was associated with auditory processing ability on any of these tests.

One of the most carefully controlled investigations to date is that of Guest et al. (Citation2018). They recruited individuals self-reporting deficits perceiving speech in noise, verified deficits using the Coordinate Response Measure (CRM) test and assessed relationships between both self-reported and verified speech-in-noise deficits and ABR Wave-I amplitude, as well as lifetime noise exposure history, with no statistically significant relationships detected. During the CRM test, participants must listen for a colour and number in the presence of 80 dB SPL background noise. Masker noise is delivered via two speakers each at 60 degrees relative to the listener, and the target speech is delivered at 0 degrees relative to the listener, with the level varied in 2-dB increments to determine threshold. The CRM test was also used by Prendergast, Millman et al. (Citation2017). No differences were detected on the CRM test, or a variety of other tests including the digit triplet test, during which three spoken digits with varied sound levels are presented sequentially in background noise at 40 or 80 dB SPL. Frequency difference limens, intensity difference limens, interaural phase difference discrimination, amplitude modulation detection thresholds, localisation and musical consonance task were also assessed, with no statistically significant findings although some weak trends were noted on some measures.

Grose et al. (Citation2017) used the BKB sentences with the target words filtered from 1–2 kHz or 3000–6000 kHz and corresponding speech-shaped noise with an equivalent spectral envelope as a masking stimulus. The filtered speech was presented at 60 and 80 dB SPL, and the level of the background babble varied in 2 dB steps to determine threshold. No differences in performance on the BKB sentences were detected when those with a history of frequent concert attendance (defined as at least 25 concerts within the past year, and at least 40 within the past two years) were compared to those with minimal concert attendance. Other psychoacoustic tests completed as part of the study by Grose et al. (Citation2017) included temporal modulation detection, spectral modulation detection and sensitivity to interaural phase, with no relationships with concert attendance detected.

In contrast to these largely negative findings, Liberman et al. (Citation2016) reported poorer speech-in-noise performance in participants with more noise exposure using a difficult custom task, including the identification of NU-6 words at 35 dB HL at an SNR of 0 dB or 5 dB, with or without digital time compression (45% or 65% of original duration), and with 0.3-sec reverberation added. Participants in that study had poorer hearing at frequencies above 8 kHz and a larger SP/AP ratio, which was driven by decreased SP amplitude in the higher-noise participants (Liberman et al. Citation2016). Given these results, it seems possible that studies using the most difficult listening tasks may show greater sensitivity for detection of the relationships of interest. For example, the study of deficits on an envelope-processing task in a group exposed to concert noise revealed poorer performance in concert-goers than controls, but only when the tests were conducted at levels below 20 dB SL; there were no deficits detected when the listening test was performed at higher sound sensation levels (Stone, Moore, and Greenish Citation2008). Consistent with the hypothesis that only the most difficult tests will reveal deficits, there was limited evidence for changes in function on a signal-in-noise test when rats were tested in difficult listening conditions before TTS was induced and after thresholds had recovered to baseline (Lobarinas, Spankovich, and Le Prell Citation2017). Functional changes were only induced in those cases in which TTS was robust (40–50 dB, 24-hour post-noise), and listening in noise deficits were only observed at the subset of frequencies at which a permanent noise-induced decrease in ABR Wave I amplitude was measured. Moreover, deficits were measured only in the most difficult SNR conditions.

The studies included in include quantitative speech-in-noise tests; efforts to assess self-reported difficulty hearing in noise have also been used, both in an effort to understand the prevalence of this clinical complaint (Gilliver et al. Citation2017) and in efforts to recruit participant populations with deficits (Guest et al. Citation2018). Statistically significant associations between exposure to noise and self-reported hearing difficulty were detected in a large epidemiological analysis, suggesting potential utility for qualitative metrics (Spankovich et al. Citation2018). One survey that has been used to explore potential relationships between ABR Wave I amplitude and perceived difficulties in noise is the Speech, Spatial, and Qualities of Hearing Scale (SSQ), developed by Gatehouse and Noble (Citation2004) and used by Yeend et al. (Citation2017) and Prendergast, Millman et al. (Citation2017). The SSQ qualitatively assesses perceived hearing difficulty in settings ranging from ideal listening conditions (one-on-one conversation in quiet listening conditions) to difficult listening conditions (group conversations in noisy environments). Neither Yeend et al. (Citation2017) nor Prendergast, Millman et al. (Citation2017) found statistically significant relationships between lifetime noise exposure and overall SSQ scores. Efforts to validate self-reported difficulty using the SSQ against quantitative speech-in-noise test measures have had limited success (Banh, Singh, and Pichora-Fuller Citation2012; Fredriksson et al. Citation2016). Taken together, at this time, there is no “gold standard” for either qualitative speech-in-noise surveys or quantitative speech-in-noise testing (for discussion, see Le Prell and Lobarinas Citation2015; Le Prell and Brungart Citation2016; Le Prell and Clavier Citation2017; Le Prell Citation2018).

Relationship between short-term noise exposure resulting in TTS and ABR amplitude change

In contrast to the studies described in , in which noise exposure was assessed retrospectively, creating the opportunity for errors in subject recall to confound results, there are a small number of studies in which temporary changes in ABR Wave-I amplitude have been documented in humans after controlled noise exposures. These studies are described in . Unfortunately, there have been virtually no efforts to document changes in OHC function using OAEs in these TTS studies (with the exception of Grinn et al. Citation2017, who found no reliable deficits in either OAE or ABR-Wave-I amplitude at the one-day post-noise test), preventing any insight into whether compromised OHC function contributed to the observed changes in sound-evoked ABR. Similarly, there has been virtually no effort to identify potential supra-threshold deficits in any of these TTS studies; supra-threshold deficits could include tinnitus, hyperacusis, listening in noise deficits, temporal processing deficits, localisation errors or other auditory processing deficits. Because cochlear synaptopathy, when observed in animals, is both immediate and permanent, it seems unlikely that any of the studies listed in provide evidence of a temporary cochlear synaptopathy, as all changes in evoked potential metrics fully recovered. Temporary injury to the OHCs during the period of post-noise deficits is the more parsimonious interpretation based on well-documented recovery of the OHC population after mild or moderate noise exposures.

Table 4. Relationship between short-term noise exposure resulting in temporary threshold shift (TTS) and auditory brainstem response (ABR) amplitude changes.

The data from Lichtenhan and Chertoff (Citation2008) are particularly notable here given the unique design of their study (i.e. experimentally controlled/standardised noise bandwidth to induce TTS in humans) and their detailed description of noise-induced changes in the sound-evoked compound action potential (CAP). Here, the CAP provided a measurement of the sound-evoked auditory nerve response using tympanic electrodes rather than the more distal electrode placements used during ABR tests. Lichtenhan and Chertoff (Citation2008) reported that CAP amplitude in response to high-level sound stimuli was less impacted by TTS than when induced by lower-level sound stimuli. They inferred that at the lower stimulus levels, at which healthy OHCs amplified the cochlear response prior to noise exposure, post-noise CAP deficits were potentially a consequence of OHC trauma. In contrast, for CAP responses to the higher stimulus levels outside the range at which cochlear amplification occurs, noise-induced CAP deficits were not observed; post-noise CAP amplitude was equivalent to pre-noise baseline measures at the higher sound levels. Level-dependent analyses such as this are potentially helpful in distinguishing potential OHC pathology from potential IHC, synapse or auditory nerve pathology.

Discussion

Relationships between occupational noise and ABR metrics ()

The studies listed in clearly demonstrate that deficits in the amplitude and latency of ABR Wave I have been detected in a variety of studies enrolling workers exposed to occupational noise as participants. The lack of DPOAE data precludes the distinguishing of effects of OHC loss and cochlear synaptopathy or progressive loss of spiral ganglion cells. However, detection of deficits at higher stimulus levels, at which the passive mechanics of the cochlea dominate after the active process has saturated, allows speculation that neural pathology contributed to delayed ABR latencies and reduced ABR amplitudes, even if permanent threshold deficits were caused by undocumented OHC loss. There is an urgent need for research in animal models using exposures that model occupational noise injury and assess the potential for synapse loss with lower-level repeated exposures. Within subjects, ABR waveforms are highly reliable (Grinn et al. Citation2017; Prendergast et al. Citation2018), and longitudinal studies of workers are needed.

Relationships between non-occupational noise and ABR metrics ()

documents a small number of studies that have revealed associations between noise exposure and deficits in the amplitude and latency of ABR Wave I (Stamper and Johnson, Citation2015b; Bramhall et al. Citation2017; Valderrama et al. Citation2018) or other related metrics (Liberman Citation2015; Grose, Buss and Hall Citation2017). However, a number of studies treating noise exposure as a continuous variable, rather than dichotomising subjects into groups, failed to find any relationships between noise history and Wave I amplitude (Fulbright et al. Citation2017; Grinn et al. Citation2017; Prendergast et al. Citation2017a; Spankovich et al. Citation2017). Taken together, it seems likely that outcomes in studies such as these are highly dependent on both the specific protocols and the noise exposure of the enrolled participants. A major unknown is the role of high frequency loss, at frequencies above 8 kHz, in the above results, as hearing loss at the higher frequencies was consistently observed across studies.

Relationships between noise exposure and speech-in-noise metrics ()

The studies listed in were highly variable with respect to outcomes. Several studies enrolling workers exposed to occupational noise, who had not yet developed significant hearing loss, revealed differences between the noise-exposed workers and controls (Alvord Citation1983; Kujala et al. Citation2004; Kumar, Ameenudin, and Sangamanatha Citation2012; Hope, Luxon, and Bamiou Citation2013). A study employing an extremely difficult speech in noise test revealed deficits associated with musical training (Liberman Citation2015), whereas a second study comparing frequent concert goers to those rarely attending concerts found no differences in speech in noise function (Grose, Buss, and Hall Citation2017). As in , a number of studies treating noise exposure as a continuous variable failed to find any relationships between noise history and outcomes on speech in noise tests (Fulbright et al. Citation2017; Grinn et al. Citation2017; Prendergast, Millman et al. Citation2017; Yeend et al. Citation2017; Valderrama et al. Citation2018). It is likely that outcomes in studies such as these are highly dependent on both the specific protocols and the noise exposure of the enrolled participants.

Short-term noise, TTS and ABR amplitude change in humans ()

The studies listed in are important in that the noise insults were clearly defined and constant across participants, eliminating the significant unknowns associated with subject recall. If the short-term noise exposure paradigms used in these studies revealed permanent changes in ABR Wave I or associate metrics, in the absence of changes in OAEs, cochlear synaptopathy would be suggested. Most studies did not include OAEs; however, none of these studies revealed permanent changes in ABR Wave I or associated metrics, suggesting cochlear synaptopathy was unlikely. Those results are not surprising, as none of the studies listed in resulted in the larger and longer lasting TTS deficits that have been associated with cochlear synaptopathy in animal models. In animal studies in which cochlear synaptopathy has been documented, the pathology has been both immediate and permanent.

It seems reasonable to speculate that the temporary changes in ABR Wave-I amplitude reported in could reflect temporary decreases in OHC gain, while OHCs recover from reversible injuries. TTS studies completed using a music player model revealed decreases in DPOAE amplitude that paralleled changes in threshold sensitivity, with recovery of DPOAE amplitude and audiometric thresholds proceeding in parallel across the post-music test times (Le Prell et al. Citation2012; Le Prell et al. Citation2016). Alternatively, the temporary changes reported in may reflect a reversible auditory nerve dendritic swelling in the absence of synapse loss. Swelling of the auditory nerve dendrites under the IHCs has been shown to be reversible after infusion of excitotoxic substances such as AMPA and kainite (Pujol et al. Citation1990; Puel et al. Citation1994, Citation1995). Recovery of both auditory nerve dendrite swelling and AP (or ABR Wave-I) amplitude is also observed after noise exposure (Puel et al. Citation1998; Yamasoba et al. Citation2005). Recovery of the swelling and regained growth of ABR amplitude, with complete return to baseline, is observed even when swelling is induced for extended periods during chronic infusion of excitotoxic substances such as AMPA (Le Prell et al. Citation2004). In other words, excitotoxicity induced by noise or chemicals (AMPA, kainite) presumably has not resulted in permanent cochlear synaptopathy as the amplitude of the auditory nerve evoked responses returns to baseline. Taken together, additional research combining ABR and OAE protocols will be necessary to understand the likely pathology associated with TTS in humans participating in studies such as those in .

Additional comments on the differential diagnosis of cochlear synaptopathy

Given that a confirmed diagnosis of cochlear synaptopathy requires post-mortem histological analysis, there has been and will continue to be significant interest in the development of an in vivo test battery that would allow cochlear synaptopathy to be correctly inferred based on patterns of results across tests. To identify selective cochlear synaptopathy (a loss of synapses in the absence of middle ear and OHC pathology), the test battery must document that the middle ear conduction system is intact, typically accomplished using tympanometry, and there must be documentation that the OHC population has not been compromised. If cochlear synaptopathy occurs in parallel with OHC loss, diagnosis will be more difficult. These issues are of significant interest not only scientifically, to improve research design, but also to audiologists and otolaryngologists (Lin Citation2016; Hall Citation2017) who commonly report interest in cochlear synaptopathy as it relates to patient complaints and patient care. Given the variety of test batteries and test protocols being used across research studies, what can be inferred at this time, regarding clinical and scientific best practices?

As a starting point for patient care, the Joint Audiology Committee on Clinical Practice Algorithms and Statements is helpful (American Speech Language Hearing Association Citation1999); this committee included representatives from the American Speech-Language-Hearing Association (ASHA), the American Academy of Audiology (AAA) and the Department of Veterans Affairs (VA). With the caveat that the components of the assessment are dictated by patient need and may vary across patients, the following examination components have been identified: history, appropriate physical examination (eg otoscopy), cerumen management, air conduction pure-tone thresholds with appropriate masking, bone conduction pure-tone thresholds with appropriate masking, speech thresholds with appropriate masking, speech recognition measures with appropriate masking, acoustic immittance (tympanometry/acoustic reflex thresholds), acoustic reflex decay, rehabilitative needs assessment, communication inventory, otoacoustic emissions, high-frequency audiometry and either speech or pure-tone Stenger.

For differential diagnosis of cochlear synaptopathy, a careful patient history, otoscopy, and tympanometry, will be the basic starting point, to gain insight into patient risk factors, and the health and function of the outer and middle ear conductive system. Pure-tone air and bone conduction are also warranted, to determine the extent to which patient complaints are related to audibility issues, which may be appropriate to resolve with amplification. Completion of both air and bone testing will provide insight into conductive and sensorineural components of any observed hearing loss and guide recommendations. Pure-tone testing should include EHF audiometry given the reliable observation of high-frequency hearing deficits in association with Wave I amplitude deficits.

Speech detection thresholds can be measured as part of a conventional clinical test battery for the purpose of validating puretone threshold measurements (“cross-check principle”) and word recognition in quiet can be considered, but these tests likely are not critical for differential diagnosis of cochlear synaptopathy. Word recognition scores are a supra-threshold test, commonly completed at 40 dB above the SRT, to determine the patient's best possible performance in easy listening conditions. Speech recognition thresholds (SRT) can also be measured; this test uses spondee words, which have two syllables pronounced with equal emphasis (“toothbrush”). The SRT is the minimum signal level at which the listener can correctly identify 50% of the speech material presented (Plomp and Mimpen Citation1979).

OAE tests

Over the course of this review, there has been significant discussion of the use of OAEs to assess the health of the OHCs. Given that the amplitude of these emissions varies across a 40-dB range even within normal hearing listeners, screening is not adequate; a diagnostic protocol should be used. As new equipment allowing higher frequencies to be routinely tested becomes available, testing through 12 kHz has begun to be incorporated into research protocols and should be considered as part of both research and clinical test batteries in order to gain insight into the health of the basal cochlea. With a baseline established, changes in cochlear health over time can be monitored (assuming patients have intact hearing and/or intact DPOAEs at these higher frequencies; if responses are absent at baseline, tests do not need to be repeated). In research studies where data are considered at the group level, rather than on an individual basis, protocols using multivariate statistical analyses in an effort to control for differences in DPOAE strength should be considered (following Bramhall et al. Citation2017; Bramhall, Konrad-Martin, and McMillan Citation2018). Such protocols are of significant interest as DPOAE amplitude can vary across a wide range even within a highly homogeneous normal-hearing population. For example, Le Prell et al. (Citation2018) recently documented DPOAE amplitudes over a 30-dB range within a primarily female, Caucasian sample of young adults. Limiting the already homogeneous sample to just those participants with thresholds of 10 dB HL or better, DP amplitude still ranges from 0 dB SNR (responses indistinguishable from the noise floor) to as much as 40 dB above the noise floor, with the majority of the data ranging from 10 dB SNR to 30 dB SNR (see Figure 2 in the report by Le Prell Citation2018, for illustration).

Middle ear reflex tests

Although there is not yet sufficient evidence to advocate middle ear reflex testing for the purpose of diagnosing cochlear synaptopathy, testing should be considered as part of the comprehensive battery. The comparison of ipsilateral and contralateral evoked responses from the two ears has long provided not only a cross-check of the audiogram, but also insight into potential conductive, cochlear and retro-cochlear pathologies (Jerger, Jerger, and Hall Citation1979; Prasher and Cohen Citation1993; Neary et al. Citation1996). New wideband acoustic immittance devices increase the speed and power of such tests (Schairer et al. Citation2013). With respect to the differential diagnosis of cochlear synaptopathy, the amplitude of the middle ear muscle reflex has been successfully associated with cochlear synaptopathy in mouse models (Valero, Hancock and Liberman Citation2016; Valero et al. Citation2018). Middle ear muscle reflexes are weak, or absent, in a subset of the population (Flamme et al. Citation2017; McGregor et al. Citation2018), leading several investigators to suggest that noise exposure resulting in cochlear synaptopathic injury could provide one explanation for individual variability in human participants (Wojtczak, Beim, and Oxenham Citation2017; McGregor et al. Citation2018). There is also the thought that the amount of energy reaching the cochlea is a function of the wide band reflectance or admittance. If more energy is reaching the cochlea, then there is a greater risk of exposure. Consistent with these suggestions, reflex decay has been associated with speech recognition impairment and hearing loss in noise exposed workers (Duarte et al. Citation2015). As additional data begin to emerge in humans, it will be important to carefully consider the interpretation of any detected correlations. It is not only clear that noise exposure can compromise middle ear reflex amplitude (Valero, Hancock, and Liberman Citation2016; Valero et al. Citation2018), but also that a more robust middle ear reflex more effectively reduces the transmission of noise to the inner ear (Karlovich et al. Citation1977; Borg, Nilsson, and Engstrom Citation1983). Based on those data, the acoustic reflex has been suggested to directly mediate vulnerability to TTS2 (Karlovich et al. Citation1977) as well as permanent NIHL (Borg, Nilsson, and Engstrom Citation1983) in noise-exposed patients and populations.

ABR and other evoked potentials

A major unknown at this time is the specific sound-evoked potentials that might be used during differential diagnosis of cochlear synaptopathy. There is a strong correlation between cochlear synaptopathy and the amplitude of Wave-I of the ABR in rodent ears in which OHC function is intact (Sergeyenko et al. Citation2013), but human patients and participants have diverse risk factors for OHC loss and the OHC population may be damaged. Although the amplitude of Wave-I of the ABR serves as the gold standard within animal models, this has not been accepted as best practice for either human research studies or clinical test batteries used by those attempting differential diagnosis of patients. In addition to ABR protocol differences, differences in the specific populations and methods used to estimate previous noise exposure almost certainly contribute to the mixed outcomes across humans studies.

A number of alternative evoked potential metrics continue to be evaluated for potential use in human test batteries. Wave-V of the ABR, for example, has been suggested as a metric for cochlear synaptopathy in humans, with the specific measures utilised to date including measurement of ABR Wave-V latency (Verhulst et al. Citation2016; Skoe and Tufts Citation2018), ABR Wave-V latency during masking noise (Mehraei et al. Citation2016), and ABR Wave-V latency changes during forward masking (Mehraei et al. Citation2017). Although there have been reports in which the amplitude of ABR Wave-I relative to the amplitude of ABR Wave-V shows deficits (Verhulst et al. Citation2016; Grose, Buss, and Hall Citation2017), any Wave-I reliability issues presumably also impact this measure. However, it is worth note that Prendergast et al. (Citation2018) carefully measured test–retest reliability for Waves I and V, and the SP, and found that the amplitudes of Waves I and V were highly reliable within subjects, whereas SP amplitude was less reliable. Several investigators have normalised the amplitude of the AP relative to the amplitude of the SP (i.e. SP/AP ratio) (Nam and Won, Citation2004; Liberman et al. Citation2016; Ridley et al. Citation2018); if the SP is not reliable, this metric will be less useful. Other evoked potential assessments considered to date include the FFR (Prendergast et al. Citation2017a), and the envelope following response (EFR) (Bharadwaj et al. Citation2015; Shaheen et al. Citation2015; Grose, Buss, and Hall, Citation2017; Guest et al. Citation2017; Paul, Bruce, and Roberts Citation2017; Guest et al. Citation2018), although statistically significant differences have not been detected either as a function of lifetime noise exposure (Prendergast et al. Citation2017a) or frequent concert/musical event attendance (Grose, Buss, and Hall Citation2017).

Speech in noise tests

A final consideration is the use and interpretation of speech in noise tests both in research studies and in patient care. Recommendations for speech in noise testing were long ago provided by Carhart and Tillman (Citation1970), and these tests continue to be widely advocated as a metric that better captures real-world patient complaints regarding difficulties understanding speech in noisy backgrounds (Soli Citation2008; Wilson Citation2011; Vermiglio et al. Citation2012; Brungart, Sheffield, and Kubli Citation2014). As argued above, speech in noise deficits can emerge with either OHC loss or IHC loss/auditory nerve dys-sychrony. Thus, speech in noise deficits are not confirmatory for cochlear synaptopathy. However, regardless of whether speech in noise difficulties are ultimately determined to be caused by OHC pathology, cochlear synaptopathy or some combination of cumulative pathological changes in the inner ear, the patient’s complaints should be assessed, and the only way to quantify their difficulty is to complete speech in noise testing. With a baseline in hand, increasing deficits can be detected, and, equally important, benefits of amplification or rehabilitation can be documented. It is important that efforts continue with respect to better understanding the relationships between noise exposure and auditory dysfunction in difficult listening environments. The identification of specific functional impairments as a consequence of noise exposure, emerging prior to permanent threshold shift, has the potential to guide new evidence-based screening and monitoring strategies regardless of whether the specific functional deficits are a consequence of OHC loss or cochlear synaptopathy.

Conclusions and challenges to the field

New data from animal subjects exposed to chronic noise through daily exposure paradigms are urgently needed as occupational exposure is composed of lower daily doses repeated on a daily basis over weeks, months and years. The extent to which synaptic pathology, OHC loss or mixed pathologies, will be induced by these chronic exposure histories is not known, as this condition has not been tested in animal studies (for discussion, see Dobie and Humes Citation2017; Murphy and Le Prell Citation2017). Non-occupational noise history generally has not been associated with decreasing ABR Wave I amplitude (Fulbright et al. Citation2017; Grinn et al. Citation2017; Prendergast et al. Citation2017a; Spankovich et al. Citation2017), although repeated exposure to loud music (Liberman et al. Citation2016; Grose, Buss, and Hall Citation2017) and exposure to firearm discharge (Bramhall et al. Citation2017) appear to be associated with changes in auditory nerve discharge in human participants. Although ABR amplitude in humans is more variable than that observed in rodents, ABR Wave-I amplitude appears to have high test–retest reliability (Prendergast et al. Citation2018) and it can be reliably monitored longitudinally within subjects for potential changes over time (Grinn et al. Citation2017). Longitudinal data are urgently needed to understand the potential for reduction of human ABR Wave-I amplitude or other derived measures as a function of aging or noise exposure. Diverse subject populations are needed, to fully characterise where risk begins, and how risk grows, with different real-world exposures and diverse real-world risk factors. Although the time, cost, equipment and training necessary for the collection of ABR data make it unlikely that such monitoring could readily be adopted as a monitoring tool across hearing loss prevention programmes (for discussion, see Skoe and Tufts Citation2018), such data have the potential to significantly contribute to our understanding of the mechanisms of noise injury. Speech in noise data is also urgently needed. Scientific documentation of deficits as a consequence of noise exposure is a necessary precursor for evidence-based suggestions for updated testing requirements. In addition to continued efforts to understand the extent to which noise exposure history affects speech in noise understanding, the relationships between noise exposure and high-frequency hearing loss should also continue to be explored.

A final caveat and call for research involves the need to assess the potential for spontaneous recovery of synapses. Although this generally has not been documented in pre-clinical noise-induced cochlear synaptopathy models using rodents as subjects, there are a small number of studies reporting recovery of the synaptic ribbons over a one-month period following noise exposure (Liu et al. Citation2012; Shi, Liu, He, et al. Citation2013; Shi, Liu, Wang et al. Citation2015). As discussed by Liberman and Kujawa (Citation2017), additional research will be necessary to reconcile the differences in results across studies. If cochlear synaptopathy is ultimately determined to be reversible, the potential for temporary cochlear synaptopathic damage will be difficult, if not impossible, to distinguish from temporary OHC damage and temporary excitotoxic swelling. Finally, regardless of whether cochlear synaptopathy is induced by noise, or a function of aging, new research is necessary to fully identify mechanisms associated with drug-induced regeneration of synapses (Wan et al. Citation2014; Wan and Corfas Citation2015; Suzuki, Corfas, and Liberman Citation2016). These observations of synaptogenesis raise hope that a “cure” could be available, if human cochlear synaptopathy becomes possible to diagnose using test batteries including elements such as those described here.

Declaration of interest

Funding for previously completed studies on temporary threshold shift in humans was provided by U01 DC 008423 from the National Institutes of Health – National Institute on Deafness and Other Communication Disorders (NIH-NIDCD), as well as contracts to the University of Florida awarded by Sound Pharmaceuticals, Inc. and Edison Pharmaceuticals, Inc. Support for the preparation of this manuscript was provided by the Emilie and Phil Schepps Distinguished Professorship in Hearing Science at the University of Texas at Dallas.

Abbreviations
ABR=

auditory brainstem response

AP=

action potential

BKB-SIN=

Bamford-Kowal-Bench Speech-in-Noise

CAP=

compound action potential

CEOAE=

click-evoked otoacoustic emission

CHABA=

Committee on Hearing, Bioacoustics, and Biomechanics

CRM=

Coordinate Response Measure test

dB=

decibel

dBA=

A-weighted decibel

dB HL=

decibel hearing level

dB nHL=

decibel hearing level relative to a normal hearing population

dB S/B=

decibel signal to babble ratio

dB SL=

decibel sensation level (dB above threshold)

dB SPL=

decibel sound pressure level

dB peSPL=

decibel peak-equivalent sound pressure level

DPOAE=

distortion product otoacoustic emission

ECochG=

Electrocochleography

EHF=

extended high frequency

FFR=

frequency following response

HINT=

Hearing in Noise Test

HPD=

hearing protection device

Hz=

hertz

IHC=

inner hair cell

kHz=

kilohertz

LiSN-S=

Listening in Spatialized Noise-Sentences High Cue Condition

NEQ=

noise exposure questionnaire

NIHL=

noise-induced hearing loss

NIOSH=

National Institute on Occupational Safety and Health

NU-6=

Northwestern University Auditory Test Number 6

OHC=

outer hair cell

OSHA=

Occupational Safety and Health Administration

PTS=

permanent threshold shift

QuickSin=

Quick Sentences in Noise

SNR=

signal-to-noise ratio

SRT=

speech recognition threshold

SSQ=

Speech, Spatial, and Qualities of Hearing Scale

STS=

standard threshold shift

TEN(HL)=

threshold equalising noise (specified as dB hearing level)

TTS=

temporary threshold shift

UTD=

University of Texas at Dallas

WIN=

Word-in-Noise test

References

  • Adelman, C., J. M. Weinberger, and H. Sohmer. 2010. “How Are the Inner Hair Cells and Auditory Nerve Fibers Activated without the Mediation of the Outer Hair Cells and the Cochlear Amplifier?” Journal of Basic and Clinical Physiology and Pharmacology 21 (3): 231–40.
  • Almadori, G., F. Ottaviani, G. Paludetti, M. Rosignoli, L. Gallucci, L. D'Alatri, and G. Vergoni. 1988. “Auditory Brainstem Responses in Noise-Induced Permanent Hearing Loss.” Audiology: Official Organ of the International Society of Audiology 27 (1): 36–41.
  • Altschuler, R. A., K. Halsey, A. Kanicki, C. Martin, D. Prieskorn., S. DeRemer, D. F. Dolan. 2018. “Small Arms Fire-like Noise: Effects on Hearing Loss, Gap Detection and the Influence of Preventive Treatment.” Neuroscience. Doi: 10.1016/j.neuroscience.2018.07.027.
  • Alvord, L. S. 1983. “Cochlear Dysfunction in "Normal-Hearing" Patients with History of Noise Exposure.” Ear and Hearing 4 (5):247–50.
  • American Speech Language Hearing Association. 1999. Joint audiology committee clinical practice statements and guidelines [Guidelines]; available from www.asha.org/policy; see https://www.asha.org/policy/gl1999-00013/. Accessed on October 4, 2018
  • Ashmore, J., P. Avan, W. E. Brownell, P. Dallos, K. Dierkes, R. Fettiplace, K. Grosh., et al. 2010. “The Remarkable Cochlear Amplifier.” Hearing Research 266 (1–2):1–17.
  • Attias, J., and I. Bresloff. 1996. “Noise Induced Temporary Otoacoustic Emission Shifts.” Journal of Basic and Clinical Physiology and Pharmacology 7 (3):221–33.
  • Attias, J., and H. Pratt. 1984. “Auditory Evoked Potentials and Audiological Follow-up of Subjects Developing Noise-Induced Permanent Threshold Shift.” Audiology: Official Organ of the International Society of Audiology 23 (5):498–508.
  • Attias, J., and H. Pratt. 1986. “Follow-up of Auditory-Evoked Potentials and Temporary Threshold Shift in Subjects Developing Noise-Induced Permanent Hearing Loss.” Audiology: Official Organ of the International Society of Audiology 25 (2):116–23.
  • Attias, J., H. Pratt, I. Reshef, I. Bresloff, G. Horowitz, A. Polyakov, Z. Shemesh., et al. 1996. “Detailed Analysis of Auditory Brainstem Responses in Patients with Noise-Induced Tinnitus.” Audiology: Official Organ of the International Society of Audiology 35 (5):259–70.
  • Balatsouras, D. G., E. Homsioglou, and V. Danielidis. 2005. “Extended High-Frequency Audiometry in Patients with Acoustic Trauma.” Clinical Otolaryngology: Official Journal of Ent-UK; Official Journal of Netherlands Society for Oto-Rhino-Laryngology & Cervico-Facial Surgery 30 (3):249–54.
  • Banh, J., G. Singh, and M. K. Pichora-Fuller. 2012. “Age Affects Responses on the Speech, Spatial, and Qualities of Hearing Scale (SSQ) by Adults with Minimal Audiometric Loss.” Journal of the American Academy of Audiology 23 (2): 81–91. quiz 139-140.
  • Barbee, C. M., J. A. James, J. H. Park, E. M. Smith, C. E. Johnson, S. Clifton, J. L. Danhauer., et al. 2018. “Effectiveness of Auditory Measures for Detecting Hidden Hearing Loss and/or Cochlear Synaptopathy: A Systematic Review.” Seminars in Hearing 39 (2):172–209.
  • Beach, E. F., M. Gilliver, and W. Williams. 2013. Sound Check Australia: a citizen science approach to noise & hearing conservation research. Paper Presented at the National Hearing Conservation Association Conference.
  • Bench, J., A. Kowal, and J. Bamford. 1979. “The BKB (Bamford-Kowal-Bench) Sentence Lists for Partially-Hearing Children.” British Journal of Audiology 13 (3): 108–12.
  • Bharadwaj, H. M., S. Masud, G. Mehraei, S. Verhulst, and B. G. Shinn-Cunningham. 2015. “Individual Differences Reveal Correlates of Hidden Hearing Deficits.” The Journal of Neuroscience: The Official Journal of the Society for Neuroscience 35 (5): 2161–72.
  • Boger, M. E., A. L. L. Sampaio, and C. Oliveira. 2017. “Analysis of Hearing and Tinnitus in Workers Exposed to Occupational Noise.” The International Tinnitus Journal 20 (2): 88–92.
  • Borg, E., R. Nilsson, and B. Engstrom. 1983. “Effect of the Acoustic Reflex on Inner Ear Damage Induced by Industrial Noise.” Acta Oto-Laryngologica 96 (5–6): 361–9.
  • Botte, M. C., H. A. Beagley, and R. Chocholle. 1976. “Temporary Threshold Shift and the Auditory Evoked Response. A Preliminary Report.” Clinical Otolaryngology and Allied Sciences 1 (2): 143–6.
  • Botte, M. C., H. A. Beagley, and R. Chocholle. 1979. “Temporary Threshold Shift, Loudness, and Auditory Evoked Potentials.” Clinical Otolaryngology and Allied Sciences 4 (1): 49–56.
  • Bramhall, N., B. Ong, J. Ko, and M. Parker. 2015. “Speech Perception Ability in Noise Is Correlated with Auditory Brainstem Response Wave I Amplitude.” Journal of the American Academy of Audiology 26 (5): 509–17.
  • Bramhall, N. F., D. Konrad-Martin, and G. P. McMillan. 2018. “Tinnitus and Auditory Perception after a History of Noise Exposure: Relationship to Auditory Brainstem Response Measures.” Ear and Hearing 39 (5): 881–94.
  • Bramhall, N. F., D. Konrad-Martin, G. P. McMillan, and S. E. Griest. 2017. “Auditory Brainstem Response Altered in Humans with Noise Exposure despite Normal Outer Hair Cell Function.” Ear and Hearing 38 (1): e1–e12.
  • Bruce, I. C., M. B. Sachs, and E. D. Young. 2003. “An Auditory-Periphery Model of the Effects of Acoustic Trauma on Auditory Nerve Responses.” Journal of the Acoustical Society of America 113 (1): 369–88.
  • Brungart, D. S., B. M. Sheffield, and L. R. Kubli. 2014. “Development of a Test Battery for Evaluating Speech Perception in Complex Listening Environments.” Journal of the Acoustical Society of America 136 (2): 777–90.
  • Buchler, M., M. Kompis, and M. A. Hotz. 2012. “Extended Frequency Range Hearing Thresholds and Otoacoustic Emissions in Acute Acoustic Trauma.” Otology & Neurotology: Official Publication of the American Otological Society, American Neurotology Society [and] European Academy of Otology and Neurotology 33 (8): 1315–22.
  • Campbell, K. C. M., and C. G. Le Prell. 2018. “Drug-Induced Ototoxicity: Diagnosis and Monitoring.” Drug Safety 41 (5): 451–64.
  • Carhart, R., and T. W. Tillman. 1970. “Interaction of Competing Speech Signals with Hearing Losses.” Archives of Otolaryngology (Chicago, Ill.: 1960) 91 (3): 273–9.
  • Carney, L. H. 2018. “Supra-Threshold Hearing and Fluctuation Profiles: Implications for Sensorineural and Hidden Hearing Loss.” Journal of the Association for Research in Otolaryngology 19 (4): 331–352. Doi: 10.1007/s10162-018-0669-5
  • CHABA 1968. Committee on Hearing, Bioacoustics, and Biomechanics: Commission on Behavioral and Social Sciences and Education; National Research Council. Proposed damage-risk criterion for impulse noise (gunfire). Washington D.C.: National Academy of Sciences.
  • CHABA 1992. Committee on Hearing, Bioacoustics, and Biomechanics: Commission on Behavioral and Social Sciences and Education; National Research Council. Hazardous exposure to noise: Working group on hazardous exposure to impulse noise. Washington D.C.: National Academy Press.
  • Chan, P., K. Ho, and A. F. Ryan. 2016. “Impulse Noise Injury Model.” Military Medicine 181 (5 Suppl): 59–69.
  • Chen, G. D., and L. D. Fechter. 2003. “The Relationship between Noise-Induced Hearing Loss and Hair Cell Loss in Rats.” Hearing Research 177 (1–2): 81–90.
  • Chen, T. J., H. C. Chiang, and S. S. Chen. 1992. “Effects of Aircraft Noise on Hearing and Auditory Pathway Function of Airport Employees.” Journal of Occupational Medicine: Official Publication of the Industrial Medical Association 34 (6): 613–9.
  • Dallos, P., and B. N. Evans. 1995. “High-Frequency Motility of Outer Hair Cells and the Cochlear Amplifier.” Science (New York, N.Y.) 267 (5206): 2006–9.
  • Dallos, P., J. Zheng, and M. A. Cheatham. 2006. “Prestin and the Cochlear Amplifier.” The Journal of Physiology 576 (Pt 1): 37–42.
  • de Souza Chelminski Barreto, M. A., C. S. Costa, L. K. de Souza Guarita, C. A. Oliveira, and J. F. Bahmad. 2011. “Auditory Monitoring by Means of Evaluation of the Cochlea in Soldiers of the Brazilian Army Exposed to Impulse Noise.” International Tinnitus Journal 16 (2): 123–9.
  • Derebery, M. J., A. Vermiglio, K. I. Berliner, M. Potthoff, and K. Holguin. 2012. “Facing the Music: Pre- and Postconcert Assessment of Hearing in Teenagers.” Otology & Neurotology: Official Publication of the American Otological Society, American Neurotology Society [and] European Academy of Otology and Neurotology 33 (7): 1136–41.
  • Dobie, R. A., and L. E. Humes. 2017. “Commentary on the Regulatory Implications of Noise-Induced Cochlear Neuropathy.” International Journal of Audiology 56 (sup1): 74–8.
  • Donaldson, G. S., and R. A. Ruth. 1993. “Derived Band Auditory Brain-Stem Response Estimates of Traveling Wave Velocity in Humans. I: Normal-Hearing Subjects.” Journal of the Acoustical Society of America 93 (2): 940–51.
  • Donaldson, G. S., and R. A. Ruth. 1996. “ Derived-Band Auditory Brain-Stem Response Estimates of Traveling Wave Velocity in Humans: II. Subjects with Noise-Induced Hearing Loss and Meniére's Disease.” Journal of Speech and Hearing Research 39 (3):534–45.
  • Duarte, A. S., R. T. Ng, G. M. Carvalho, A. C. Guimarães, L. A. Pinheiro, E. A. Costa, R. J. Gusmão. 2015. “High Levels of Sound Pressure: Acoustic Reflex Thresholds and Auditory Complaints of Workers with Noise Exposure.” Brazilian Journal of Otorhinolaryngology 81 (4): 374–83.
  • Earl, B. R., and M. E. Chertoff. 2012. “Mapping Auditory Nerve Firing Density Using High-Level Compound Action Potentials and High-Pass Noise Masking.” Journal of the Acoustical Society of America 131 (1): 337–52.
  • Etymotic Research 2005. BKB-SIN: Bamford-Kowal-Bench Speech in Noise Test. Elk Grove, IL: Etymotic Research.
  • Fabiani, M., A. Mattioni, M. Saponara, and A. Cordier. 1998. “Auditory Evoked Potentials for the Assessment of Noise Induced Hearing Loss.” Scandinavian Audiology. Supplementum 48: 147–53.
  • Felder, E., and A. Schrott-Fischer. 1995. “Quantitative Evaluation of Myelinated Nerve Fibres and Hair Cells in Cochleae of Humans with Age-Related High-Tone Hearing Loss.” Hearing Research 91 (1–2): 19–32.
  • Fernandez, K. A., P. W. Jeffers, K. Lall, M. C. Liberman, and S. G. Kujawa. 2015. “Aging after Noise Exposure: Acceleration of Cochlear Synaptopathy in "Recovered" Ears.” Journal of Neuroscience: The Official Journal of the Society for Neuroscience 35 (19): 7509–20.
  • Fiałkowska, M. D., G. Janczewski, K. Kochanek, and J. Dawidowicz. 1983. “Studies on the Temporary Effect of Noise on the Auditory Function in Man.” Scandinavian Audiology 12 (4): 295–8.
  • Flamme, G. A., K. K. Deiters, S. M. Tasko, and W. A. Ahroon. 2017. “Acoustic Reflexes Are Common but Not Pervasive: evidence from the National Health and Nutrition Examination Survey, 1999-2012.” International Journal of Audiology 56 (sup1): 52–62.
  • Fredriksson, S., O. Hammar, L. Magnusson, K. Kahari, and K. Persson Waye. 2016. “Validating Self-Reporting of Hearing-Related Symptoms against Pure-Tone Audiometry, Otoacoustic Emission, and Speech Audiometry.” International Journal of Audiology 55 (8):454–62.
  • Fridberger, A., J. Zheng, A. Parthasarathi, T. Ren, and A. Nuttall. 2002. “Loud Sound-Induced Changes in Cochlear Mechanics.” Journal of Neurophysiology 88 (5): 2341–8.
  • Fritze, W. 1981. “[a Method for Prediction of Permanent Threshold Shift (PTS) (author's Transl)].” Laryngologie, Rhinologie, Otologie 60 (10): 512–6.
  • Fulbright, A. N. C. 2016. “Normal Hearing? What Functional Hearing Tests, Noise Exposure History and ABR Wave-I Amplitude Reveal.” Dissertation, Department of Speech, Language, and Hearing Sciences: University of Florida.
  • Fulbright, A. N. C., C. G. Le Prell, S. K. Griffiths, and E. Lobarinas. 2017. “Effects of Recreational Noise on Threshold and Supra-Threshold Measures of Auditory Function.” Seminars in Hearing 38 (04): 298–318.
  • Furman, A. C., S. G. Kujawa, and M. C. Liberman. 2013. “Noise-Induced Cochlear Neuropathy Is Selective for Fibers with Low Spontaneous Rates.” Journal of Neurophysiology 110 (3): 577–86.
  • Gannouni, N., A. Mhamdi, M. Lenoir, J. L. Puel, O. Tebourbi, M. El May, R. Gharbi., et al. 2015. “Assessment of Cochlear Effects and Extra-Auditory Disorders in Male Rats Exposed Repetitively to Low Noise.” La Tunisie Medicale 93 (12):789–94.
  • Gatehouse, S., and W. Noble. 2004. “The Speech, Spatial and Qualities of Hearing Scale (SSQ).” International Journal of Audiology 43 (2): 85–99.
  • Gilliver, M., J. Nguyen, E. F. Beach, and C. Barr. 2017. “Personal Listening Devices in Australia: Patterns of Use and Levels of Risk.” Seminars in Hearing 38 (4): 282–97.
  • Gleich, O., P. Semmler, and J. Strutz. 2016. “Behavioral Auditory Thresholds and Loss of Ribbon Synapses at Inner Hair Cells in Aged Gerbils.” Experimental Gerontology 84: 61–70.
  • Gonçalves, C. G., A. B. Lacerda, B. S. Zeigelboim, J. M. Marques, and D. Luders. 2013. “Auditory Thresholds among Military Musicians: conventional and High Frequency.” CoDAS 25 (2): 181–7.
  • Grinn, S. K., K. B. Wiseman, J. A. Baker, and C. G. Le Prell. 2017. “Hidden Hearing Loss? No Effect of Common Recreational Noise Exposure on Cochlear Nerve Response Amplitude in Humans.” Front Neuroscience 11: 1–24. Doi: 10.3389/fnins.2017.00465/full
  • Grose, J. H., E. Buss, and J. W. Hall. 3rd 2017. “Loud Music Exposure and Cochlear Synaptopathy in Young Adults: Isolated Auditory Brainstem Response Effects but No Perceptual Consequences.” Trends in Hearing 21: 1–8.
  • Guest, H., K. J. Munro, G. Prendergast, S. Howe, and C. J. Plack. 2017. “Tinnitus with a Normal Audiogram: Relation to Noise Exposure but No Evidence for Cochlear Synaptopathy.” Hearing Research 344: 265–74.
  • Guest, H., K. J. Munro, G. Prendergast, R. E. Millman, and C. Plack. 2018. “Impaired Speech Perception in Noise with a Normal Audiogram: No Evidence for Cochlear Synaptopathy and No Relation to Lifetime Noise Exposure.” Hearing Research 364: 142–51.
  • Hall, J. W. III. 1992. Handbook of Auditory Evoked Responses. Boston: Allyn and Bacon.
  • Hall, J. W. III. 2017. “Hidden Hearing Loss: An Audiologist's Perspective.” Hearing Journal 70 (1): 6.
  • Hallmo, P., H. M. Borchgrevink, and I. W. Mair. 1995. “Extended High-Frequency Thresholds in Noise-Induced Hearing Loss.” Scandinavian Audiology 24 (1): 47–52.
  • Henderson, D., and R. P. Hamernik. 1986. “Impulse Noise: Critical Review.” Journal of the Acoustical Society of America 80 (2): 569–84.
  • Henderson, D., R. P. Hamernik, and R. Sitler. 1974. “Audiometric and Anatomical Correlates of Impulse Noise Exposure.” Archives of Otolaryngology (Chicago, Ill. : 1960) 99 (1):62–6.
  • Hickman, T. T., C. Smalt, J. Bobrow, T. Quatieri, and M. C. Liberman. 2018. “Blast-Induced Cochlear Synaptopathy in Chinchillas.” Scientific Reports 8 (1): 10740.
  • Hickox, A. E., E. Larsen, M. G. Heinz, L. Shinobu, and J. P. Whitton. 2017. “Translational Issues in Cochlear synaptopathy.” Hearing Research 349: 164–71.
  • Hickox, A. E., and M. C. Liberman. 2014. “Is Noise-Induced Cochlear Neuropathy Key to the Generation of Hyperacusis or Tinnitus?.” Journal of Neurophysiology 111 (3): 552–64.
  • Hinojosa, R., and S. A. Lerner. 1987. “Cochlear Neural Degeneration without Hair Cell Loss in Two Patients with Aminoglycoside Ototoxicity.” Journal of Infectious Diseases 156 (3): 449–55.
  • Hoben, R., G. Easow, S. Pevzner, and M. A. Parker. 2017. “Outer Hair Cell and Auditory Nerve Function in Speech Recognition in Quiet and in Background Noise.” Frontiers in Neuroscience 11: 157.
  • Hope, A. J., L. M. Luxon, and D. E. Bamiou. 2013. “Effects of Chronic Noise Exposure on Speech-in-Noise Perception in the Presence of Normal Audiometry.” Journal of Laryngology and Otology 127 (3): 233–8.
  • Hu, B. 2012. Noise-induced structural damage in the cochlea. In: C.G. Le Prell, D. Henderson, A.N. Popper & R.R. Fay (eds.) Noise-Induced Hearing Loss: Scientific Advances; Springer Handbook of Auditory Research. New York: Springer; pp. 57–86.
  • International Standard Organization. 2013. Acoustics: Estimation of noise-induced hearing loss (ISO-1999). Geneva, Switzerland: International Standard Organization.
  • Jansen, S., H. Luts, P. Dejonckere, A. van Wieringen, and J. Wouters. 2014. “Exploring the Sensitivity of Speech-in-Noise Tests for Noise-Induced Hearing Loss.” International Journal of Audiology 53 (3): 199–205.
  • Jensen, J. B., A. C. Lysaght, M. C. Liberman, K. Qvortrup, and K. M. Stankovic. 2015. “Immediate and Delayed Cochlear Neuropathy after Noise Exposure in Pubescent Mice.” PLoS One 10 (5):e0125160.
  • Jerger, S., J. Jerger, and J. Hall. 1979. “A New Acoustic Reflex pattern.” Archives of Otolaryngology (Chicago, Ill. : 1960) 105 (1): 24–8.
  • Johnstone, B. M., R. Patuzzi, and G. K. Yates. 1986. “Basilar Membrane Measurements and the Travelling Wave.” Hearing Research 22: 147–53.
  • Jokitulppo, J.,. M. Toivonen, and E. Bjork. 2006. “Estimated Leisure-Time Noise Exposure, Hearing Thresholds, and Hearing Symptoms of Finnish Conscripts.” Military Medicine 171 (2): 112–6.
  • Jokitulppo, J. S., E. A. Bjork, and E. Akaan-Penttila. 1997. “Estimated Leisure Noise Exposure and Hearing Symptoms in Finnish Teenagers.” Scandinavian Audiology 26 (4): 257–62.
  • Karawani, H., J. Attias, R. Shemesh, and B. Nageris. 2015. “Evaluation of Noise-Induced Hearing Loss by Auditory Steady-State and Auditory Brainstem-Evoked Responses.” Clinical Otolaryngology: Official Journal of Ent-UK; Official Journal of Netherlands Society for Oto-Rhino-Laryngology & Cervico-Facial Surgery 40 (6): 672–81.
  • Karlovich, R. S., H. A. Osier, H. N. Gutnick, R. G. Ivey, K. Wolf, S. Schwimmer, M. L. Strennen., et al. 1977. “The Acoustic Reflex and Temporary Threshold Shift: Temporal Characteristics.” Journal of Speech and Hearing Research 20 (3): 565–73.
  • Killion, M. C., P. A. Niquette, G. I. Gudmundsen, L. J. Revit, and S. Banerjee. 2004. “Development of a Quick Speech-in-Noise Test for Measuring Signal-to-Noise Ratio Loss in Normal-Hearing and Hearing-Impaired Listeners.” Journal of the Acoustical Society of America 116 (4 Pt 1): 2395–405.
  • Klein, A. J., and J. H. Mills. 1981. “Physiological and Psychophysical Measures from Humans with Temporary Threshold Shift.” Journal of the Acoustical Society of America 70 (4): 1045–53.
  • Kobel, M., C. G. Le Prell, J. Liu, J. W. Hawks, and J. Bao. 2017. “Noise-Induced Cochlear Synaptopathy: Past Findings and Future Studies.” Hearing Research 349: 148–54.
  • Konrad-Martin, D., M. F. Dille, G. McMillan, S. Griest, D. McDermott, S. A. Fausti, D. F. Austin., et al. 2012. “Age-Related Changes in the Auditory Brainstem Response.” Journal of the American Academy of Audiology 23 (1): 18–35. quiz 74-15.
  • Korres, G. S., D. G. Balatsouras, A. Tzagaroulakis, D. Kandiloros, and E. Ferekidis. 2008. “Extended High-Frequency Audiometry in Subjects Exposed to Occupational Noise.” B-ENT 4 (3): 147–55.
  • Korres, G. S., Balatsouras, D. G., A. Tzagaroulakis, D. Kandiloros, and E. Ferekidou. 2009. “Distortion Product Otoacoustic Emissions in an Industrial Setting.” Noise and Health 11 (43): 103–10.
  • Kryter, K. D., W. D. Ward, J. D. Miller, and D. H. Eldredge. 1966. “Hazardous Exposure to Intermittent and Steady-State Noise.” Journal of the Acoustical Society of America 39 (3): 451–64.
  • Kujala, T., Y. Shtyrov, I. Winkler, M. Saher, M. Tervaniemi, M. Sallinen, W. Teder-Sälejärvi., et al. 2004. “Long-Term Exposure to Noise Impairs Cortical Sound Processing and Attention Control.” Psychophysiology 41 (6): 875–81.
  • Kujawa, S. G. 2014. “Putting the ‘Neural’ Back in Sensorineural: Primary Cochlear Neurodegeneration in Noise and Aging.” Hearing Journal 67 (11): 8.
  • Kujawa, S. G., and M. C. Liberman. 2006. “Acceleration of Age-Related Hearing Loss by Early Noise Exposure: evidence of a Misspent Youth.” Journal of Neuroscience: The Official Journal of the Society for Neuroscience 26 (7): 2115–23.
  • Kujawa, S. G., and M. C. Liberman. 2009. “Adding Insult to Injury: Cochlear Nerve Degeneration after "Temporary" Noise-Induced Hearing Loss.” Journal of Neuroscience: The Official Journal of the Society for Neuroscience 29 (45): 14077–85.
  • Kujawa, S. G., and M. C. Liberman. 2015. “Synaptopathy in the Noise-Exposed and Aging Cochlea: Primary Neural Degeneration in Acquired Sensorineural Hearing Loss.” Hearing Research 330 (Pt B): 191–9.
  • Kumar, P., P. Upadhyay, A. Kumar, S. Kumar, and G. B. Singh. 2017. “Extended High Frequency Audiometry in Users of Personal Listening Devices.” American Journal of Otolaryngology 38 (2): 163–7.
  • Kumar, U. A., S. Ameenudin, and A. V. Sangamanatha. 2012. “Temporal and Speech Processing Skills in Normal Hearing Individuals Exposed to Occupational Noise.” Noise and Health 14 (58): 100–5.
  • Kumar, U. A., and C. S. Vanaja. 2004. “Functioning of Olivocochlear Bundle and Speech Perception in Noise.” Ear and Hearing 25 (2): 142–6.
  • Lapsley Miller, J. A., and L. Marshall. 2007. Otoacoustic Emissions as Preclinical Measures of NIHL and Susceptibility to NIHL. In: M.S. Robinette & T.J. Glattke (eds.) Otoacoustic Emissions, Clinical Application. (Third edn.). New York: Thieme; pp. 321–341.
  • Lapsley Miller, J. A., L. Marshall, L. M. Heller, and L. M. Hughes. 2006. “Low-Level Otoacoustic Emissions May Predict Susceptibility to Noise-Induced Hearing Loss.” Journal of the Acoustical Society of America 120 (1): 280–96.
  • Le Prell, C. G. 2018. Effect of Noise Exposure on Human Auditory Function: Hidden versus Not-so-Hidden Hearing Loss. In: S. Hatzopoulos, A. Ciorba & M. Krumm (eds.) Advances in Audiology, Speech Pathology, and Hearing Science, Volume 1: Clinical Protocols and Hearing Devices. New Jersey: Apple Academic Press, in press.
  • Le Prell, C. G., and D. S. Brungart. 2016. “Speech-in-Noise Tests and Supra-Threshold Auditory Evoked Potentials as Metrics for Noise Damage and Clinical Trial Outcome Measures.” Otology & Neurotology: Official Publication of the American Otological Society, American Neurotology Society [and] European Academy of Otology and Neurotology 37 (8): e295–302.
  • Le Prell, C. G., and O. H. Clavier. 2017. “Effects of Noise on Speech Recognition: Challenges for Communication by Service Members.” Hearing Research 349: 76–89.
  • Le Prell, C. G., S. Dell, B. N. Hensley, J. W. III. Hall, K. C. Campbell. P. J. Antonelli, P. J. Green., et al. 2012. “Digital Music Exposure Reliably Induces Temporary Threshold Shift (TTS) in Normal Hearing Human Subjects.” Ear and Hearing 33: e44–58.
  • Le Prell, C. G., A. Fulbright, C. Spankovich, S. K. Griffiths, E. Lobarinas, K. C. M. Campbell, P. J. Antonelli., et al. 2016. “Dietary Supplement Comprised of β-Carotene, Vitamin C, Vitamin E, and Magnesium: Failure to Prevent Music-Induced Temporary Threshold Shift.” Audiology and Neurotology. Extra 6 (2): 20–39.
  • Le Prell, C. G., and E. Lobarinas. 2015. Strategies for Assessing Antioxidant Efficacy in Clinical Trials. In: J.M. Miller, C.G. Le Prell & L.P. Rybak (eds.) Oxidative Stress in Applied Basic Research and Clinical Practice: Free Radicals in ENT Pathology. New York: Humana Press; pp. 163–192.
  • Le Prell, C. G., and E. Lobarinas. 2016. Clinical and Translational Research: Challenges to the Field. In: C.G. Le Prell, E. Lobarinas, R.R. Fay & A.N. Popper (eds.) Translational Research in Audiology and the Hearing Sciences, Springer Handbook of Auditory Research. New York: Springer; pp. 241–265.
  • Le Prell, C. G., H. W. Siburt, E. Lobarinas, S. K. Griffiths, and C. Spankovich. 2018. “No Reliable Association between Recreational Noise Exposure and Threshold Sensitivity, Distortion Product Otoacoustic Emission Amplitude, or Word-in-Noise Performance in a College Student Population.” Ear and Hearing :1. [Epub ahead of print DOI: 10.1097/aud.0000000000000575].
  • Le Prell, C. G., C. Spankovich, E. Lobarinas, and S. K. Griffiths. 2013. “Extended High-frequency Thresholds in College Students: Effects of Music Player Use and Other Recreational Noise.” Journal of the American Academy of Audiology 24 (8): 725–39.
  • Le Prell, C. G., M. Yagi, K. Kawamoto, L. A. Beyer, G. Atkin, Y. Raphael, D. F. Dolan., et al. 2004. “Chronic Excitotoxicity in the Guinea Pig Cochlea Induces Temporary Functional Deficits without Disrupting Otoacoustic Emissions.” Journal of the Acoustical Society of America 116 (2): 1044–56.
  • Lee, G. J. C., M. Y. Lim, A. Y. W. Kuan, J. H. W. Teo, H. G. Tan, and W. K. Low. 2014. “Relationship between Leisure Noise Exposure and Otoacoustic Emissions in a Young Asian Population.” International Journal of Audiology 53 (7): 462–8.
  • Leensen, M. C., J. A. de Laat, and W. A. Dreschler. 2011. “Speech-in-Noise Screening Tests by Internet, Part 1: test Evaluation for Noise-Induced Hearing Loss Identification.” International Journal of Audiology 50 (11): 823–34.
  • Leensen, M. C., J. A. de Laat, A. F. Snik, and W. A. Dreschler. 2011. “Speech-in-Noise Screening Tests by Internet, Part 2: improving Test Sensitivity for Noise-Induced Hearing Loss.” International Journal of Audiology 50 (11): 835–48.
  • Leger, A. C., B. C. Moore, and C. Lorenzi. 2012. “Abnormal Speech Processing in Frequency Regions Where Absolute Thresholds Are Normal for Listeners with High-Frequency Hearing Loss.” Hearing Research 294 (1–2): 95–103.
  • Liberman, M. C. 2015. “Hidden Hearing Loss.” Scientific American 313 (2): 48–53.
  • Liberman, M. C. 2017. “Noise-Induced and Age-Related Hearing Loss: New Perspectives and Potential Therapies.” F1000Research 6: 927.
  • Liberman, M. C., M. J. Epstein, S. S. Cleveland, H. Wang, and S. F. Maison. 2016. “Toward a Differential Diagnosis of Hidden Hearing Loss in Humans.” PLoS One 11 (9): e0162726.
  • Liberman, M. C., and S. G. Kujawa. 2017. “Cochlear Synaptopathy in Acquired Sensorineural Hearing Loss: Manifestations and Mechanisms.” Hearing Research 349: 138–47.
  • Lichtenhan, J. T., and M. E. Chertoff. 2008. “Temporary Hearing Loss Influences Post-Stimulus Time Histogram and Single Neuron Action Potential Estimates from Human Compound Action Potentials.” Journal of the Acoustical Society of America 123 (4): 2200–12.
  • Lin, C. Y. 2016. “Hidden Hearing Loss: A Clinician's Perspective.” Hearing Journal 69 (11): 6.
  • Lin, H. W., A. C. Furman, S. G. Kujawa, and M. C. Liberman. 2011. “Primary Neural Degeneration in the Guinea Pig Cochlea after Reversible Noise-Induced Threshold Shift.” Journal of the Association for Research in Otolaryngology 12 (5): 605–16.
  • Liu, L.,. H. Wang, L. Shi, A. Almuklass, T. He, S. Aiken, M. Bance., et al. 2012. “Silent Damage of Noise on Cochlear Afferent Innervation in Guinea Pigs and the Impact on Temporal Processing.” PLoS One 7 (11): e49550.
  • Lobarinas, E., R. Salvi, and D. Ding. 2013. “Insensitivity of the Audiogram to Carboplatin Induced Inner Hair Cell Loss in Chinchillas.” Hearing Research 302: 113–20.
  • Lobarinas, E., R. Salvi, and D. Ding. 2016. “Selective Inner Hair Cell Dysfunction in Chinchillas Impairs Hearing-in-Noise in the Absence of Outer Hair Cell Loss.” Journal of the Association for Research in Otolaryngology 17 (2): 89–101.
  • Lobarinas, E., C. Spankovich, and C. G. Le Prell. 2017. “Evidence of Hidden Hearing Loss Following Noise Exposures that Produce Robust TTS and ABR Wave-I Amplitude Reductions.” Hearing Research 349: 155–63.
  • Lonsbury-Martin, B. L., and G. K. Martin. 1990. “The Clinical Utility of Distortion-Product Otoacoustic Emissions.” Ear and Hearing 11 (2): 144–54.
  • Lutman, M. E., A. C. Davis, and M. A. Ferguson. 2008. Epidemiological Evidence for the Effectiveness of the Noise at Work Regulations. Health Safety Executive Research Report RR669, available at http://wwwhsegovuk/research/rrpdf/rr669pdf.
  • Lutman, M. E., and G. H. Saunders. 1992. “Lack of Association between Otoacoustic Emissions and Hearing Difficulty in Subjects with Normal Hearing Thresholds.” Journal of the Acoustical Society of America 92 (2 Pt 1): 1184–5.
  • Maison, S. F., and M. C. Liberman. 2000. “Predicting Vulnerability to Acoustic Injury with a Noninvasive Assay of Olivocochlear Reflex Strength.” Journal of Neuroscience: The Official Journal of the Society for Neuroscience 20 (12): 4701–7.
  • Makary, C. A., J. Shin, S. G. Kujawa, M. C. Liberman, and S. N. Merchant. 2011. “Age-Related Primary Cochlear Neuronal Degeneration in Human Temporal Bones.” Journal of the Association for Research in Otolaryngology 12 (6): 711–7.
  • Mannstrom, P., M. Kirkegaard, and M. Ulfendahl. 2015. “Repeated Moderate Noise Exposure in the Rat–an Early Adulthood Noise Exposure Model.” Journal of the Association for Research in Otolaryngology 16 (6): 763–72.
  • McArdle, R. A., and R. H. Wilson. 2006. “Homogeneity of the 18 QuickSIN Lists.” Journal of the American Academy of Audiology 17 (3): 157–67.
  • McGregor, K. D., G. A. Flamme, S. M. Tasko, K. K. Deiters, W. A. Ahroon, C. L. Themann, W. J. Murphy., et al. 2018. “Acoustic Reflexes Are Common but Not Pervasive: Evidence Using a Diagnostic Middle Ear Analyser.” International Journal of Audiology 57 (sup1): S42–s50.
  • Mehraei, G., A. P. Gallardo, B. G. Shinn-Cunningham, and T. Dau. 2017. “Auditory Brainstem Response Latency in Forward Masking, a Marker of Sensory Deficits in Listeners with Normal Hearing Thresholds.” Hearing Research 346: 34–44.
  • Mehraei, G., A. E. Hickox, H. M. Bharadwaj, H. Goldberg, S. Verhulst, M. C. Liberman, B. G. Shinn-Cunningham., et al. 2016. “Auditory Brainstem Response Latency in Noise as a Marker of Cochlear Synaptopathy.” Journal of Neuroscience: The Official Journal of the Society for Neuroscience 36 (13): 3755–64.
  • Mehrparvar, A. H., S. J. Mirmohammadi, M. H. Davari, M. Mostaghaci, A. Mollasadeghi, M. Bahaloo, S. H. Hashemi., et al. 2014. “Conventional Audiometry, Extended High-Frequency Audiometry, and DPOAE for Early Diagnosis of NIHL.” Iranian Red Crescent Medical Journal 16 (1): e9628.
  • Melnick, W. 1991. “Human Temporary Threshold Shift (TTS) and Damage risk.” Journal of the Acoustical Society of America 90 (1): 147–54.
  • Mills, J. H., R. W. Gengel, C. S. Watson, and J. D. Miller. 1970. “Temporary Changes of the Auditory System Due to Exposure to Noise for One or Two Days.” Journal of the Acoustical Society of America 48 (2): 524–30.
  • Möhrle, D., K. Ni, K. Varakina, D. Bing, S. C. Lee, U. Zimmermann, M. Knipper., et al. 2016. “Loss of Auditory Sensitivity from Inner Hair Cell Synaptopathy Can Be Centrally Compensated in the Young but Not Old Brain.” Neurobiology of Aging 44: 173–84.
  • Moore, B. C., B. R. Glasberg, and M. A. Stone. 2004. “New Version of the TEN Test with Calibrations in dB HL.” Ear and Hearing 25 (5): 478–87.
  • Moore, B. C., M. Huss, D. A. Vickers, B. R. Glasberg, and J. I. Alcantara. 2000. “A Test for the Diagnosis of Dead Regions in the Cochlea.” British Journal of Audiology 34 (4): 205–24.
  • Morata, T. C., and A.-C. Johnson. 2012. Effects of Exposure to Chemicals on Noise-Induced Hearing Loss. In: C. G. Le Prell, D. Henderson, R. R. Fay & A. N. Popper (eds.) Noise-Induced Hearing Loss: Scientific Advances, Springer Handbook of Auditory Research. New York: Springer Science + Business Media, LLC, pp. 223–254.
  • Moshammer, H., M. Kundi, P. Wallner, A. Herbst, A. Feuerstein, and H.-P. Hutter. 2015. “Early Prognosis of Noise-Induced Hearing Loss.” Occupational and Environmental Medicine 72 (2):85–9.
  • Mulders, W., I. L. Chin, and D. Robertson. 2018. “Persistent Hair Cell Malfunction Contributes to Hidden Hearing Loss.” Hearing Research 361:45–51.
  • Müller, J., T. Janssen, G. Heppelmann, and W. Wagner. 2005. “Evidence for a Bipolar Change in Distortion Product Otoacoustic Emissions during Contralateral Acoustic Stimulation in Humans.” Journal of the Acoustical Society of America 118 (6): 3747–56.
  • Murphy, W. J., and C. G. Le Prell. 2017. “Making Sound Waves: selected Papers from the 2016 Annual Conference of the National Hearing Conservation Association.” International Journal of Audiology 56 (sup1): 1–3.
  • Nam, E. C., and J. Y. Won. 2004. “Extratympanic Electrocochleographic Changes on Noise-induced Temporary Threshold Shift .” Otolaryngology-Head and Neck Surgery: Official Journal of American Academy of Otolaryngology-Head and Neck Surgery 130 (4): 437–42.
  • Neary, W. J., V. E. Newton, S. N. Laoide-Kemp, R. T. Ramsden, V. F. Hillier, and S. W. Kan. 1996. “A Clinical, Genetic and Audiological Study of Patients and Families with Unilateral Vestibular Schwannomas. II. Audiological Findings in 93 Patients with Unilateral Vestibular Schwannomas.” Journal of Laryngology and Otology 110 (12): 1120–8.
  • Neitzel, R., N. Seixas, B. Goldman, and W. Daniell. 2004. “Contributions of Non-Occupational Activities to Total Noise Exposure of Construction Workers.” Annals of Occupational Hygiene 48 (5): 463–73.
  • Neitzel, R., N. Seixas, J. Olson, W. Daniell, and B. Goldman. 2004. “Nonoccupational Noise: Exposures Associated with Routine Activities.” Journal of the Acoustical Society of America 115 (1): 237–45.
  • Nilsson, M., S. D. Soli, and J. A. Sullivan. 1994. “Development of the Hearing in Noise Test for the Measurement of Speech Reception Thresholds in Quiet and in Noise.” Journal of the Acoustical Society of America 95 (2):1085–99.
  • NIOSH 1998. Criteria for a Recommended Standard, Occupational Noise Exposure, DHHS (NIOSH) Publication No.98-126. DHHS/CDC/NIOSH, Cincinnati, OH.
  • Noorhassim, I., K. Kaga, and K. Nishimura. 1996. “Pure-Tone Audiometry and Auditory Brainstem Responses in Noise-Induced Deafness.” American Journal of Otolaryngology 17 (1): 31–5.
  • Olsen, S. E. 2004. “Psychological Aspects of Adolescents’ Perceptions and Babits in Noisy Environments.” Doctoral diss., Department of Psychology, Göteborgs Universitet.
  • Opperman, D. A., W. Reifman, R. Schlauch, and S. Levine. 2006. “Incidence of Spontaneous Hearing Threshold Shifts during Modern Concert Performances.” Official journal of Canadian Society of Otolaryngology – Head & Neck Surgery 134: 667–73. Doi: 10.1016/j.otohns.2005.11.039
  • OSHA 1983. 29 CFR 1910.95. Occupational Noise Exposure; Hearing Conservation Amendment; Final Rule, effective 8 March 1983.
  • Oxenham, A. J. 2016. “Predicting the Perceptual Consequences of Hidden Hearing Loss.” Trends in Hearing 20:2331216516686768.
  • Paul, B. T., I. C. Bruce, and L. E. Roberts. 2017. “Evidence That Hidden Hearing Loss Underlies Amplitude Modulation Encoding Deficits in Individuals with and without Tinnitus.” Hearing Research 344: 170–82.
  • Pienkowski, M. 2017. “On the Etiology of Listening Difficulties in Noise despite Clinically Normal Audiograms.” Ear and Hearing 38 (2): 135–48.
  • Plack, C. J., D. Barker, and G. Prendergast. 2014. “Perceptual Consequences of "Hidden" Hearing Loss.” Trends in Hearing 18: 1–11. Doi: 10.1177/2331216514550621
  • Plack, C. J., A. Leger, G. Prendergast, K. Kluk, H. Guest., K. J. Munro., 2016. “Toward a Diagnostic Test for Hidden Hearing Loss.” Trends in Hearing 20: 1–9. Doi: 10.1177/2331216516657466
  • Plomp, R., and A. Mimpen. 1979. “ Speech-reception threshold for sentences as a function of age and noise level.” Journal of the Acoustical Society of America 66 (5): 1333–42.
  • Prasher, D., and M. Cohen. 1993. “Effectiveness of Acoustic Reflex Threshold Criteria in the Diagnosis of Retrocochlear Pathology.” Scandinavian Audiology 22 (1): 11–8.
  • Prendergast, G., H. Guest, K. J. Munro, K. Kluk, A. Léger, D. A. Hall, M. G. Heinz., et al. 2017. “Effects of Noise Exposure on Young Adults with Normal Audiograms I: Electrophysiology.” Hearing Research 344: 68–81. Doi: 10.1016/j.heares.2016.10.028
  • Prendergast, G., R. E. Millman, H. Guest, K. J. Munro, K. Kluk, R. S. Dewey, D. A. Hall., et al. 2017. “Effects of Noise Exposure on Young Adults with Normal Audiograms II: Behavioral Measures.” Hearing Research 356: 74–86. Doi: 10.1016/j.heares.2017.10.007
  • Prendergast, G., W. Tu, H. Guest, R. E. Millman, K. Kluk, S. Couth, K. J. Munro., et al. 2018. “Supra-Threshold Auditory Brainstem Response Amplitudes in Humans: Test-Retest Reliability, Electrode Montage and Noise Exposure.” Hearing Research 364: 38–47.
  • Puel, J. L., R. Pujol, F. Tribillac, S. Ladrech, and M. Eybalin. 1994. “Excitatory Amino Acid Antagonists Protect Cochlear Auditory Neurons from Excitotoxicity.” The Journal of Comparative Neurology 341 (2): 241–56.
  • Puel, J. L., J. Ruel, C. Gervais d'Aldin, and R. Pujol. 1998. “Excitotoxicity and Repair of Cochlear Synapses after Noise-Trauma Induced Hearing Loss.” Neuroreport 9 (9):2109–14.
  • Puel, J.L., Saffiedine, S., Gervais d'Aldin, C. Eybalin, M. and Pujol R. 1995. “Synaptic Regeneration and Functional Recovery after Excitotoxic Injury in the guinea Pig Cochlea.” Comptes Rendus de L'academie Des Sciences. Serie Iii, Sciences de la Vie (1)318: 67–75.
  • Pujol, R., G. Rebillard, J. L. Puel, M. Lenoir, M. Eybalin, and M. Recasens. 1990. “Glutamate Neurotoxicity in the Cochlea: A Possible Consequence of Ischaemic or Anoxic Conditions Occurring in Ageing.” Acta Oto-Laryngologica. Supplementum 476: 32–6.
  • Pushpalatha, Z. V., and Konadath S. 2016. “Auditory Brainstem Responses for Click and CE-Chirp Stimuli in Individuals with and without Occupational Noise Exposure.” Noise & Health 18 (84): 260–5.
  • Ramakers, G. G. J., Kraaijenga, V. J. C. Cattani, G. van Zanten, G. A., and Grolman W. 2016. “Effectiveness of Earplugs in Preventing Recreational Noise-Induced Hearing Loss.” JAMA Otolaryngology–Head & Neck Surgery 142 (6): 551–8.
  • Ridley, C. L., Kopun, J. G. Neely, S. T. Gorga, M. P. and Rasetshwane D. M. 2018. “Using Thresholds in Noise to Identify Hidden Hearing Loss in Humans.” Ear and Hearing 39 (5): 829–44.
  • Riga, M., Korres, G. Balatsouras, D., and Korres S. 2010. “Screening Protocols for the Prevention of Occupational Noise-Induced Hearing Loss: The Role of Conventional and Extended High Frequency Audiometry May Vary according to the Years of Employment.” Medical Science Monitor: International Medical Journal of Experimental and Clinical Research 16 (7): CR352–6.
  • Ruggero, M. A., and Rich N. C.. 1991. “Furosemide Alters Organ of Corti Mechanics: evidence for Feedback of Outer Hair Cells upon the Basilar Membrane.” The Journal of Neuroscience: The Official Journal of the Society for Neuroscience 11 (4):1057–67.
  • Ryan, A. F., Kujawa, S. G. Hammill, T. Le Prell, C., and Kil J.. 2016. “Temporary and Permanent Noise-Induced Threshold Shifts: A Review of Basic and Clinical Observations.” Otology & Neurotology: Official Publication of the American Otological Society, American Neurotology Society [and] European Academy of Otology and Neurotology 37 (8):e271–5.
  • Samelli, A. G., C. G. Matas, R. M. M. Carvallo, R. F. Gomes, C. S. de Beija, F. C. L. Magliaro, C. M. Rabelo., et al. 2012. “Audiological and Electrophysiological Assessment of Professional Pop/Rock Musicians.” Noise & Health 14 (56): 6–12.
  • Santaolalla Montoya, F., Ibarguen, A. M., Vences, A. R., del Rey A. S., and Fernandez J. M. 2008. “Evaluation of Cochlear Function in Normal-Hearing Young Adults Exposed to MP3 Player Noise by Analyzing Transient Evoked Otoacoustic Emissions and Distortion Products.” Journal of Otolaryngology - Head & Neck Surgery [Le Journal D'oto-Rhino-Laryngologie et de Chirurgie Cervico-Faciale] 37 (5): 718–24.
  • Saunders, J. C., Cohen, Y. E., and Szymko Y. M. 1991. “The Structural and Functional Consequences of Acoustic Injury in the Cochlea and Peripheral Auditory System: A Five Year Update.” Journal of the Acoustical Society of America 90 (1): 136–46.
  • Schaette, R., and McAlpine D. 2011. “Tinnitus with a Normal Audiogram: physiological Evidence for Hidden Hearing Loss and Computational Model.” Journal of Neuroscience 31(38): 13452–7.
  • Schafer, E. C. 2010. “Speech Perception in Noise Measures for Children: A Critical Review and Case Studies.” Journal of Educational Audiology 16:4–15. http://www.edaud.org/journal/2010/4-article-10.pdf.
  • Schairer, K. S., Feeney, M. P., and Sanford C. A. 2013. “Acoustic Reflex Measurement.” Ear and Hearing 34 (Suppl 1):43S–7s.
  • Schmidt, J. M., Verschuure, J. and Brocaar M. P.. 1994. “Hearing Loss in Students at a conservatory.” Audiology: Official Organ of the International Society of Audiology 33 (4): 185–94.
  • Seixas, N. S., S. G. Kujawa, S. Norton, L. Sheppard, R. Neitzel, and A. Slee. 2004. “Predictors of Hearing Threshold Levels and Distortion Product Otoacoustic Emissions among Noise Exposed Young Adults.” Occupational and Environmental Medicine 61 (11): 899–907.
  • Seixas, N. S., R. Neitzel, B. Stover, L. Sheppard, P. Feeney, D. Mills, S. Kujawa., et al. 2012. “10-Year Prospective Study of Noise Exposure and Hearing Damage among Construction Workers.” Occupational and Environmental Medicine 69 (9):643–50.
  • Sergeyenko, Y., Lall, K., Liberman, M. C., and Kujawa S. 2013. “Age-Related Cochlear Synaptopathy: an Early-Onset Contributor to Auditory Functional Decline.” Journal of Neuroscience 33 (34): 13686–94.
  • Shaheen, L. A., Valero, M. D., and Liberman M. C. 2015. “Towards a Diagnosis of Cochlear Neuropathy with Envelope following Responses.” Journal of the Association for Research in Otolaryngology 16 (6): 727–45.
  • Shi, L., K. Liu, H. Wang, Y. Zhang, Z. Hong, M. Wang, X. Wang., et al. 2015. “Noise Induced Reversible Changes of Cochlear Ribbon Synapses Contribute to Temporary Hearing Loss in Mice.” Acta Oto-Laryngologica 135 (11): 1093–102.
  • Shi, L., L. Liu, T. He, X. Guo, Z. Yu, S. Yin, J. Wang., et al. 2013. “Ribbon Synapse Plasticity in the Cochleae of Guinea Pigs after Noise-Induced Silent Damage.” PLoS One 8 (12): e81566.
  • Skoe, E., Krizman, J., Anderson, S., and Kraus N. 2015. “Stability and Plasticity of Auditory Brainstem Function across the Lifespan.” Cerebral Cortex (New York, N.Y. : 1991) 25 (6): 1415–26.
  • Skoe, E., and Tufts J. 2018. “Evidence of Noise-Induced Subclinical Hearing Loss Using Auditory Brainstem Responses and Objective Measures of Noise Exposure in Humans.” Hearing Research 361: 80–91.
  • Smoorenburg, G. F. 1992. “Speech Reception in Quiet and in Noisy Conditions by Individuals with Noise-Induced Hearing Loss in Relation to Their Tone Audiogram.” Journal of the Acoustical Society of America 91 (1): 421–37.
  • Sohmer, H., and Pratt H. 1975. “Electrocochleography during Noise-Induced Temporary Threshold Shifts.” Audiology: Official Organ of the International Society of Audiology 14 (2):130–4.
  • Soli, S. D. 2008. “Some Thoughts on Communication Handicap and Hearing Impairment.” International Journal of Audiology (6)47:285–6.
  • Spankovich, C. 2010. Early Indices of Auditory Pathology in Young Adults with Type-I Diabetes Hearing and Speech Sciences. Nashville: Vanderbilt University, pp. 1–132.
  • Spankovich, C., Gonzalez, V. B., Su, D., and Bishop C. E. 2018. “Self Reported Hearing Difficulty, Tinnitus, and Normal Audiometric Thresholds, the National Health and Nutrition Examination Survey 1999-2002.” Hearing Research 358: 30–6.
  • Spankovich, C., C. G. Le Prell, L. J. Hood, and E. Lobarinas. 2017. “Noise History and Auditory Function in Young Adults with and without Type-1 Diabetes.” Ear and Hearing 38 (6): 724–735.
  • Stamper, G. C., and T. A. Johnson. 2015a. “Auditory Function in Normal-Hearing, Noise-Exposed Human Ears.” Ear and Hearing 36 (2):172–84.
  • Stamper, G. C., and T. A. Johnson. 2015b. “Letter to the Editor: Examination of Potential Sex Influences in Auditory Function in Normal-Hearing, Noise-Exposed Human Ears.” Ear and Hearing 36: 172–84.
  • Stelmachowicz, P. G., K. A. Beauchaine, A. Kalberer, and W. Jesteadt. 1989. “Normative Thresholds in the 8- to 20-kHz Range as a Function of Age.” Journal of the Acoustical Society of America 86 (4): 1384–91.
  • Stephenson, M. R., and L. G. Wall. 1984. “Post-Noise-Exposure Auditory Sensitivity and Temporal Integration Recovery Functions.” The Journal of Auditory Research 24 (4): 239–50.
  • Stone, M. A., B. C. Moore, and H. Greenish. 2008. “Discrimination of Envelope Statistics Reveals Evidence of Sub-Clinical Hearing Damage in a Noise-Exposed Population with 'Normal' Hearing Thresholds.” International Journal of Audiology 47 (12): 737–50.
  • Subramanian, M., D. Henderson, and V. Spongr. 1994. “The Relationship among Distortion-Product Otoacoustic Emissions, Evoked Potential Thresholds, and Outer Hair Cells following Interrupted Noise Exposures.” Ear and Hearing 15 (4): 299–309.
  • Subramaniam, M., R. J. Salvi, V. P. Spongr, D. Henderson, and N. L. Powers. 1994. “Changes in Distortion Product Otoacoustic Emissions and Outer Hair Cells following Interrupted Noise Exposures.” Hearing Research 74 (1–2): 204–16.
  • Sulaiman, A. H., R. Husain, and K. Seluakumaran. 2015. “Hearing Risk among Young Personal Listening Device Users: Effects at High-Frequency and Extended High-Frequency Audiogram Thresholds.” The Journal of International Advanced Otology 11 (2): 104–9.
  • Summers, V., M. J. Makashay, S. M. Theodoroff, and M. R. Leek. 2013. “Suprathreshold Auditory Processing and Speech Perception in Noise: hearing-Impaired and Normal-Hearing Listeners.” Journal of the American Academy of Audiology 24 (4): 274–92.
  • Sun, X. M. 2008. “Contralateral Suppression of Distortion Product Otoacoustic Emissions and the Middle-Ear Muscle Reflex in Human Ears.” Hearing Research 237 (1–2): 66–75.
  • Suzuki, J., G. Corfas, and M. C. Liberman. 2016. “Round-Window Delivery of Neurotrophin 3 Regenerates Cochlear Synapses after Acoustic Overexposure.” Scientific Reports 6: 24907.
  • Thakur, L., J. P. Anand, and P. K. Banerjee. 2004. “Auditory Evoked Functions in Ground Crew Working in High Noise Environment of Mumbai Airport.” Indian Journal of Physiology and Pharmacology 48 (4):453–60.
  • Valderrama, J. T., E. F. Beach, I. Yeend, M. Sharma, B. Van Dun, and H. Dillon. 2018. “Effects of Lifetime Noise Exposure on the Middle-Age Human Auditory Brainstem Response, Tinnitus and Speech-in-Noise Intelligibility.” Hearing Research 365: 36–48.
  • Valero, M. D., J. A. Burton, S. N. Hauser, T. A. Hackett, R. Ramachandran, and M. C. Liberman. 2017. “Noise-Induced Cochlear Synaptopathy in Rhesus Monkeys (Macaca mulatta).” Hearing Research 353: 213–23.
  • Valero, M. D., K. E. Hancock, and M. C. Liberman. 2016. “The Middle Ear Muscle Reflex in the Diagnosis of Cochlear Neuropathy.” Hearing Research 332: 29–38.
  • Valero, M. D., K. E. Hancock, S. F. Maison, and M. C. Liberman. 2018. “Effects of Cochlear Synaptopathy on Middle-Ear Muscle Reflexes in Unanesthetized Mice.” Hearing Research 363: 109–18.
  • Verhulst, S., A. Jagadeesh, M. Mauermann, and F. Ernst. 2016. “Individual Differences in Auditory Brainstem Response Wave Characteristics: Relations to Different Aspects of Peripheral Hearing Loss.” Trends in Hearing 20: 1–20.
  • Vermiglio, A. J. 2008. “The American English Hearing in Noise Test.” International Journal of Audiology 47 (6): 386–7.
  • Vermiglio, A. J., S. D. Soli, D. J. Freed, and L. M. Fisher. 2012. “The Relationship between High-Frequency Pure-Tone Hearing Loss, Hearing in Noise Test (HINT) Thresholds, and the Articulation Index.” Journal of the American Academy of Audiology 23 (10): 779–88.
  • Verschooten, E., E. A. Strickland, N. Verhaert, and P. X. Joris. 2017. “Assessment of Ipsilateral Efferent Effects in Human via ECochG.” Frontiers in Neuroscience 11: 331.
  • Viana, L. M., J. T. O'Malley, B. J. Burgess, D. D. Jones, C. A. C. P. Oliveira, F. Santos, S. N. Merchant., et al. 2015. “Cochlear Neuropathy in Human Presbycusis: Confocal Analysis of Hidden Hearing Loss in Post-Mortem Tissue.” Hearing Research 327: 78–88.
  • Vinay and Moore, B. C. J. 2007. “Ten(HL)-Test Results and Psychophysical Tuning Curves for Subjects with Auditory Neuropathy.” International Journal of Audiology 46: 39–46. Doi: 10.1080/14992020601077992
  • Wan, G., and G. Corfas. 2015. “No Longer Falling on Deaf Ears: Mechanisms of Degeneration and Regeneration of Cochlear Ribbon Synapses.” Hearing Research 329: 1–10.
  • Wan, G., M. E. Gomez-Casati, A. R. Gigliello, M. C. Liberman, and G. Corfas. 2014. “Neurotrophin-3 Regulates Ribbon Synapse Density in the Cochlea and Induces Synapse Regeneration after Acoustic Trauma.” eLife 3: e03564. Doi: 10.7554/eLife.03564
  • Wang, Y., K. Hirose, and M. C. Liberman. 2002. “Dynamics of Noise-Induced Cellular Injury and Repair in the Mouse Cochlea.” Journal of the Association for Research in Otolaryngology 3 (3): 248–68.
  • Wang, Y., and C. Ren. 2012. “Effects of Repeated "Benign" Noise Exposures in Young CBA Mice: Shedding Light on Age-Related Hearing Loss.” Journal of the Association for Research in Otolaryngology 13 (4): 505–15.
  • Ward, W. D. 1970. “Temporary Threshold Shift and Damage-Risk Criteria for Intermittent Noise Exposures.” Journal of the Acoustical Society of America 48 (2):561–74.
  • Williams, W., L. Carter, and M. Seeto. 2015. “Pure Tone Hearing Thresholds and Leisure Noise: Is There a Relationship?” Noise & Health 17 (78): 358–63.
  • Wilson, R. H. 2011. “Clinical Experience with the Words-in-Noise Test on 3430 Veterans: Comparisons with Pure-Tone Thresholds and Word Recognition in Quiet.” Journal of the American Academy of Audiology 22 (7): 405–23.
  • Wilson, R. H., C. S. Carnell, and A. L. Cleghorn. 2007a. “The Words-in-Noise (WIN) Test with Multitalker Babble and Speech-Spectrum Noise Maskers.” Journal of the American Academy of Audiology 18 (6): 522–9.
  • Wilson, R. H., and R. McArdle. 2007. “Intra- and Inter-Session Test, Retest Reliability of the Words-in-Noise (WIN) Test.” Journal of the American Academy of Audiology 18 (10):813–25.
  • Wilson, R. H., R. A. McArdle, and S. L. Smith. 2007b. “An Evaluation of the BKB-SIN, HINT, QuickSIN, and WIN Materials on Listeners with Normal Hearing and Listeners with Hearing Loss.” Journal of Speech, Language, and Hearing Research 50 (4): 844–56.
  • Wojtczak, M., J. A. Beim, and A. J. Oxenham. 2017. “Weak Middle-Ear-Muscle Reflex in Humans with Noise-Induced Tinnitus and Normal Hearing May Reflect Cochlear Synaptopathy.” eNeuro 4: 1–8.
  • Xu, Z. M., B. Vinck, E. De Vel, and P. van Cauwenberge. 1998. “Mechanisms in Noise-Induced Permanent Hearing Loss: an Evoked Otoacoustic Emission and Auditory Brainstem Response Study.” The Journal of Laryngology and Otology 112 (12): 1154–61.
  • Yamasoba, T., A. Pourbakht, T. Sakamoto, and M. Suzuki. 2005. “Ebselen Prevents Noise-Induced Excitotoxicity and Temporary Threshold Shift.” Neuroscience Letters 380 (3): 234–8.
  • Yates, J. T., J. D. Ramsey, and J. W. Holland. 1976. “Damage Risk: an Evaluation of the Effects of Exposure to 85 versus 90 dBA of Noise.” Journal of Speech and Hearing Research 19 (2): 216–24.
  • Yeend, I., E. F. Beach, M. Sharma, and H. Dillon. 2017. “The Effects of Noise Exposure and Musical Training on Suprathreshold Auditory Processing and Speech Perception in Noise.” Hearing Research 353: 224–36.