Nothing Special   »   [go: up one dir, main page]

You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (148)

Search Parameters:
Keywords = noise-induced hearing loss

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
27 pages, 6803 KiB  
Article
Longitudinal Effect of Music Exposure on Hearing Among Vocal Performance Students
by Rachel L. Lowrance, Charles J. Nudelman, Yvonne Gonzales Redman and Pasquale Bottalico
Acoustics 2024, 6(4), 1047-1073; https://doi.org/10.3390/acoustics6040057 - 25 Nov 2024
Viewed by 695
Abstract
The effects of music exposure on vocal performers remain relatively unknown. This study aimed to assess the immediate and long-term effects of music and singing practice on the peripheral auditory system of vocal performers using otoscopy, pure-tone audiometry, and noise dosimetry. The hearing [...] Read more.
The effects of music exposure on vocal performers remain relatively unknown. This study aimed to assess the immediate and long-term effects of music and singing practice on the peripheral auditory system of vocal performers using otoscopy, pure-tone audiometry, and noise dosimetry. The hearing status, sound pressure levels (SPLs), and sound doses of 12 vocal performers with normal hearing at the study’s onset were evaluated. Pre- and post-study questionnaires regarding the participants’ otologic health and music-making activities, as well as repeated hearing evaluations, were implemented. Additionally, noise dosimetry was conducted on each participant’s most vocally active day of the week. Audiometric assessments generally revealed normal hearing thresholds, with some exceptions. Half of the participants exhibited elevated low-frequency thresholds and over half of the participants displayed emerging audiometric “notches” at 6000 Hz. Noise dosimetry measurements indicated that most of the participants were consistently exposed to SPLs during music-making activities that exceeded recommended limits. Questionnaire responses highlighted that the participants often engaged in extra-curricular music-making activities, frequently with piano accompaniment, and with little to no use of hearing protection devices. A few of the participants reported histories of otologic issues and potential hearing problems. Full article
(This article belongs to the Special Issue Vibration and Noise (2nd Edition))
Show Figures

Figure 1

Figure 1
<p>Histograms of the time history of instantaneous SPLs per participant for Spring 2023. The three curves represent the three main sound sources/activities identified by the Gaussian mixture model.</p>
Full article ">Figure 2
<p>Mean background noise, speech/choir, and singing per participant for Fall 2021. The orange-white striped bins represent choir activity, while the solid orange bins represent speech levels.</p>
Full article ">Figure 3
<p>Mean background noise, speech/choir, and singing per participant for Spring 2022. The orange-white striped bins represent choir activity, while the solid orange bins represent speech levels.</p>
Full article ">Figure 4
<p>Mean background noise, speech/choir, and singing per participant for Fall 2022. The orange-white striped bins represent choir activity, while the solid orange bins represent speech levels.</p>
Full article ">Figure 5
<p>Mean background noise, speech/choir, and singing per participant for Spring 2023.</p>
Full article ">Figure 6
<p>Equivalent levels (<span class="html-italic">Leq</span>) and maximum duration of exposure per participant for Fall 2021.</p>
Full article ">Figure 7
<p>Equivalent levels (<span class="html-italic">Leq</span>) and maximum duration of exposure per participant for Spring 2022. The dashed lines indicate the NIOSH recommended exposure limit of 85 dBA and 8 working hours.</p>
Full article ">Figure 8
<p>Equivalent levels (<span class="html-italic">Leq</span>) and maximum duration of exposure per participant for Fall 2022. The dashed lines indicate the NIOSH recommended exposure limit of 85 dBA and 8 working hours.</p>
Full article ">Figure 9
<p>Equivalent levels (<span class="html-italic">Leq</span>) and maximum duration of exposure per participant for Spring 2023. The dashed lines indicate the NIOSH recommended exposure limit of 85 dBA and 8 working hours.</p>
Full article ">Figure A1
<p>Yearly audiograms for Participant 1.</p>
Full article ">Figure A2
<p>Yearly audiograms for Participant 2.</p>
Full article ">Figure A3
<p>Yearly audiograms for Participant 3.</p>
Full article ">Figure A4
<p>Yearly audiograms for Participant 4.</p>
Full article ">Figure A5
<p>Yearly audiograms for Participant 5.</p>
Full article ">Figure A6
<p>Yearly audiograms for Participant 6.</p>
Full article ">Figure A7
<p>Yearly audiograms for Participant 7.</p>
Full article ">Figure A8
<p>Yearly audiograms for Participant 8.</p>
Full article ">Figure A9
<p>Yearly audiograms for Participant 9.</p>
Full article ">Figure A10
<p>Yearly audiograms for Participant 10.</p>
Full article ">Figure A11
<p>Yearly audiograms for Participant 11.</p>
Full article ">Figure A12
<p>Yearly audiograms for Participant 12.</p>
Full article ">
12 pages, 1440 KiB  
Article
Health Risks Due to Co-Exposure to Noise and Respirable Crystalline Silica Among Workers in the Open-Pit Mining Industry—Results of a Preliminary Study
by Iryna Myshchenko, Małgorzata Pawlaczyk-Luszczynska, Adam Dudarewicz and Alicja Bortkiewicz
Toxics 2024, 12(11), 781; https://doi.org/10.3390/toxics12110781 - 27 Oct 2024
Viewed by 715
Abstract
Occupational exposure to carcinogenic respirable crystalline silica and noise requires a deeper understanding and an assessment of the possible health risks caused by their combined action. Data on individual exposure to respirable crystalline silica (RCS) and occupational noise (ON) was collected among 44 [...] Read more.
Occupational exposure to carcinogenic respirable crystalline silica and noise requires a deeper understanding and an assessment of the possible health risks caused by their combined action. Data on individual exposure to respirable crystalline silica (RCS) and occupational noise (ON) was collected among 44 open-pit miners. The study group was divided into two groups according to the job tasks performed. The individual exposure, exceeding of maximum admissible concentration/intensity, and predicted hearing thresholds (HTs) (according to ISO 1999:2013) were compared between the groups directly participating in the technological process (group 1; N = 23) and performing auxiliary, supervising, or laboratory activities (group 2; N = 21). All the analysed indices were significantly higher for group 1; therefore, the job category may predict ON and RCS exposure among open-pit miners. A statistically significant relationship (rs = 0.66, p < 0.05) was found between the time-weighted average (TWA) 8 h RCS and individual daily noise exposure levels. Exposure to noise in the course of employment causes the risk of hearing impairment (mean HTs for 2, 3, and 4 kHz > 25 dB) up to 74% and 4.4% (in the case of groups 1 and 2, respectively). Further studies are needed before conclusions concerning the effects of co-exposure to ON and RCS on open-pit miners can be made. Full article
(This article belongs to the Topic New Research in Work-Related Diseases, Safety and Health)
Show Figures

Figure 1

Figure 1
<p>The number and percentage of subjects within group 1 (<b>a</b>) and group 2 (<b>b</b>) at the studied workplaces.</p>
Full article ">Figure 2
<p>The concentrations of respirable crystalline silica and the noise exposure levels among the open-pit mining workers (N = 44).</p>
Full article ">Figure 3
<p>Results of the risk assessment of hearing impairment expressed as mean hearing threshold level at the frequencies of 1, 2, and 3 kHz &gt; 25 dB (<b>a</b>) and &gt;45 dB (<b>b</b>). The calculations are based on the energy mean and maximum values of daily noise exposure levels (i.e., L<sub>EX,8h,mean</sub> and L<sub>EX,8h,max</sub>, respectively).</p>
Full article ">Figure 4
<p>Results of the risk assessment of hearing impairment expressed as mean hearing threshold level at the frequencies of 2, 3, and 4 kHz &gt; 25 dB (<b>a</b>) and &gt; 45 dB (<b>b</b>). The calculations are based on the energy mean and maximum values of daily noise exposure levels (i.e., L<sub>EX,8h,mean</sub> and L<sub>EX,8h,max</sub>, respectively).</p>
Full article ">
30 pages, 1339 KiB  
Systematic Review
Pathogenesis and New Pharmacological Approaches to Noise-Induced Hearing Loss: A Systematic Review
by Francisco Javier Santaolalla Sanchez, Juan David Gutierrez Posso, Francisco Santaolalla Montoya, Javier Aitor Zabala, Ane Arrizabalaga-Iriondo, Miren Revuelta and Ana Sánchez del Rey
Antioxidants 2024, 13(9), 1105; https://doi.org/10.3390/antiox13091105 - 12 Sep 2024
Viewed by 1307
Abstract
Noise-induced hearing loss (NIHL) is responsible for significant adverse effects on cognition, quality of life and work, social relationships, motor skills, and other psychological aspects. The severity of NIHL depends on individual patient characteristics, sound intensity, and mainly the duration of sound exposure. [...] Read more.
Noise-induced hearing loss (NIHL) is responsible for significant adverse effects on cognition, quality of life and work, social relationships, motor skills, and other psychological aspects. The severity of NIHL depends on individual patient characteristics, sound intensity, and mainly the duration of sound exposure. NIHL leads to the production of a reactive oxygen (ROS) inflammatory response and the activation of apoptotic pathways, DNA fragmentation, and cell death. In this situation, antioxidants can interact with free radicals as well as anti-apoptotics or anti-inflammatory substances and stop the reaction before vital molecules are damaged. Therefore, the aim of this study was to analyze the effects of different pharmacological treatments, focusing on exogenous antioxidants, anti-inflammatories, and anti-apoptotics to reduce the cellular damage caused by acoustic trauma in the inner ear. Experimental animal studies using these molecules have shown that they protect hair cells and reduce hearing loss due to acoustic trauma. However, there is a need for more conclusive evidence demonstrating the protective effects of antioxidant/anti-inflammatory or anti-apoptotic drugs’ administration, the timeline in which they exert their pharmacological action, and the dose in which they should be used in order to consider them as therapeutic drugs. Further studies are needed to fully understand the potential of these drugs as they may be a promising option to prevent and treat noise-induced hearing loss. Full article
(This article belongs to the Special Issue Oxidative Stress in Hearing Loss)
Show Figures

Figure 1

Figure 1
<p>Systematic review strategy flowchart.</p>
Full article ">Figure 2
<p>Systematic review strategy flowchart.</p>
Full article ">
10 pages, 806 KiB  
Article
P1 and N1 Characteristics in Individuals with Normal Hearing and Hearing Loss, and Cochlear Implant Users: A Pilot Study
by Hye Yoon Seol, Soojin Kang, Sungkean Kim, Jihoo Kim, Euijin Kim, Sung Hwa Hong and Il Joon Moon
J. Clin. Med. 2024, 13(16), 4941; https://doi.org/10.3390/jcm13164941 - 22 Aug 2024
Viewed by 807
Abstract
Background: It has been reported in many previous studies that the lack of auditory input due to hearing loss (HL) can induce changes in the brain. However, most of these studies have focused on individuals with pre-lingual HL and have predominantly compared the [...] Read more.
Background: It has been reported in many previous studies that the lack of auditory input due to hearing loss (HL) can induce changes in the brain. However, most of these studies have focused on individuals with pre-lingual HL and have predominantly compared the characteristics of those with normal hearing (NH) to cochlear implant (CI) users in children. This study examined the visual and auditory evoked potential characteristics in NH listeners, individuals with bilateral HL, and CI users, including those with single-sided deafness. Methods: A total of sixteen participants (seven NH listeners, four individuals with bilateral sensorineural HL, and five CI users) completed speech testing in quiet and noise and evoked potential testing. For speech testing, the Korean version of the Hearing in Noise Test was used to assess individuals’ speech understanding ability in quiet and in noise (noise from the front, +90 degrees, and −90 degrees). For evoked potential testing, visual and auditory (1000 Hz, /ba/, and /da/) evoked potentials were measured. Results: The results showed that CI users understood speech better than those with HL in all conditions except for the noise from +90 and −90 degrees. In the CI group, a decrease in P1 amplitudes was noted across all channels after implantation. The NH group exhibited the highest amplitudes, followed by the HL group, with the CI group (post-CI) showing the lowest amplitudes. In terms of auditory evoked potentials, the smallest amplitude was observed in the pre-CI condition regardless of the type of stimulus. Conclusions: To the best of our knowledge, this is the first study that examined visual and auditory evoked potentials based on various hearing profiles. The characteristics of evoked potentials varied across participant groups, and further studies with CI users are necessary, as there are significant challenges in collecting and analyzing evoked potentials due to artifact issues on the CI side. Full article
Show Figures

Figure 1

Figure 1
<p>Grand average waveforms for VEP.</p>
Full article ">Figure 2
<p>Grand average waveforms for AEP.</p>
Full article ">
14 pages, 3650 KiB  
Article
Effects of Castanopsis echinocarpa on Sensorineural Hearing Loss via Neuronal Gene Regulation
by Isabel Rodriguez, Youn Hee Nam, Sung Woo Shin, Gyeong Jin Seo, Na Woo Kim, Wanlapa Nuankaew, Do Hoon Kim, Yu Hwa Park, Hwa Yeon Lee, Xi Hui Peng, Bin Na Hong and Tong Ho Kang
Nutrients 2024, 16(16), 2716; https://doi.org/10.3390/nu16162716 - 15 Aug 2024
Viewed by 934
Abstract
Sensorineural hearing loss (SNHL), characterized by damage to the inner ear or auditory nerve, is a prevalent auditory disorder. This study explores the potential of Castanopsis echinocarpa (CAE) as a therapeutic agent for SNHL. In vivo experiments were conducted using zebrafish and mouse [...] Read more.
Sensorineural hearing loss (SNHL), characterized by damage to the inner ear or auditory nerve, is a prevalent auditory disorder. This study explores the potential of Castanopsis echinocarpa (CAE) as a therapeutic agent for SNHL. In vivo experiments were conducted using zebrafish and mouse models. Zebrafish with neomycin-induced ototoxicity were treated with CAE, resulting in otic hair cell protection with an EC50 of 0.49 µg/mL and a therapeutic index of 1020. CAE treatment improved auditory function and protected cochlear sensory cells in a mouse model after noise-induced hearing loss (NIHL). RNA sequencing of NIHL mouse cochleae revealed that CAE up-regulates genes involved in neurotransmitter synthesis, secretion, transport, and neuronal survival. Real-time qPCR validation showed that NIHL decreased the mRNA expression of genes related to neuronal function, such as Gabra1, Gad1, Slc32a1, CaMK2b, CaMKIV, and Slc17a7, while the CAE treatment significantly elevated these levels. In conclusion, our findings provide strong evidence that CAE protects against hearing loss by promoting sensory cell protection and enhancing the expression of genes critical for neuronal function and survival. Full article
(This article belongs to the Special Issue Effects of Plant Extracts on Human Health)
Show Figures

Figure 1

Figure 1
<p>CAE’s efficacy on otic hair cell protection after neomycin-induced ototoxicity in zebrafish model. (<b>A</b>) Number of otic hair cells in the untreated group (NM) and the treated groups (0.5, 1, 5, and 10 µg/mL of CAE). (<b>B</b>) Fluorescence images of otic hair cells in the normal (NOR), control (NM), and treated groups. Hair cells were stained with YO-PRO-1 at 0.1%. Data are presented as means ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 (control vs. treated groups). <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 (normal vs. control group). <span class="html-italic">n</span> = 10 per group.</p>
Full article ">Figure 2
<p>Dose–response curves and therapeutic index of CAE. (<b>A</b>) The EC<sub>50</sub> value of CAE in neomycin (NM)-induced ototoxicity was defined as 0.497 µg/mL. (<b>B</b>) The LC<sub>50</sub> value of zebrafish embryos exposed to CAE for 48 h was defined as 500 µg/mL. (<b>C</b>) The therapeutic index (TI) of CAE was calculated to be 1020, indicating a high level of drug safety. Data are presented as means ± SEM. Conc. = concentration; EC<sub>50</sub> = 50% effective concentration; LC<sub>50</sub> = 50% lethal concentration. <span class="html-italic">n</span> = 10 per group for EC<sub>50</sub>; <span class="html-italic">n</span> = 20 per group for LC<sub>50</sub>.</p>
Full article ">Figure 3
<p>Toxicity evaluation of CAE based on zebrafish embryo testing. Data represent 48 h of exposure. (<b>A</b>) Hatching rate of zebrafish embryos exposed to CAE at varying concentrations: 10–1000 µg/mL. (<b>B</b>) Heartbeat rate (beats per minute) of zebrafish treated with CAE at varying concentrations: 10–400 µg/mL. (<b>C</b>) Body length of zebrafish treated with CAE at varying concentrations: 10–400 µg/mL. Data are presented as means ± SEM. <span class="html-italic">n</span> = 20 per group.</p>
Full article ">Figure 4
<p>Effects of CAE on auditory function in NIHL mouse model. No-treatment (NIHL) and CAE-treated groups were compared. Auditory brainstem response (ABR) threshold shifts with click stimulus (<b>A</b>), 8 kHz tone burst (<b>B</b>), and 16 kHz tone burst (<b>C</b>) in mouse model at 10 days (10 D) and 20 days (20 D) after noise insult. Data are presented as means ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 (NIHL group vs. CAE-treated groups). CAE 100, 100 mg/kg; CAE 300, 300 mg/kg; CAE 500, 500 mg/kg. <span class="html-italic">n</span> = 10 per group.</p>
Full article ">Figure 5
<p>CAE alleviated cochlear hair cell damage in NIHL mice. (<b>A</b>) Outer hair cell (OHC) survival in 1 mm segments from the apex, middle, and base of the cochlea (<span class="html-italic">n</span> = 6 per group). (<b>B</b>) Fluorescence images of the outer (OHC) and inner (IHC) hair cells at the apex, middle, and base of the cochlea by Rhodamine phalloidin staining. Scale bar = 50 µm. <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 (normal group vs. NIHL group). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 (NIHL group vs. CAE-treated group). NOR = normal. CAE 100, 100 mg/kg. White triangles indicate the locations where the loss of outer hair cells occurred.</p>
Full article ">Figure 6
<p>Differential gene expression induced by CAE treatment in the cochlea of NIHL mice (<span class="html-italic">n</span> = 3 per group) using Reactome Pathway analysis. Heat map based on RNA-seq analysis of gene expression in the mouse cochlea and Venn diagram showing the overlap of RNA-seq results for the regulated gene set of CAE vs. NIHL group. Genes were categorized into CAE-induced and CAE-repressed groups. Of the total genes, 211 were significantly altered by CAE treatment (false discovery rate (FDR) adjusted Q &lt; 0.01, |fold change| ≥ 2.0).</p>
Full article ">Figure 7
<p>Functional categorization of genes up-regulated by CAE 100 mg/kg using Reactome Pathway analysis (<span class="html-italic">n</span> = 3 per group). Heat map generated from RNA-seq data showing gene sets involved in transmission across chemical synapses (<span class="html-italic">p</span> value 8.74 × 10<sup>−21</sup>; Q value 1.21 × 10<sup>−18</sup>; Enrichment score 101.5087) and the neuronal system (<span class="html-italic">p</span> value 2.92 × 10<sup>−22</sup>; Q value 8.09 × 10<sup>−20</sup>; Enrichment score 110.3959).</p>
Full article ">Figure 8
<p>Changes in the expression of neuronal function-related genes in NIHL mice treated with CAE 100 mg/kg. Gene expression changes were evaluated by qPCR 20 days after noise insult. The effects of CAE treatment on genes related to inhibitory synaptic transmission (<b>A</b>–<b>C</b>), neuronal survival (<b>D</b>,<b>E</b>), and synaptic function (<b>E</b>,<b>F</b>) are shown. Data are presented as means ± SEM. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 (NOR group vs. NIHL group); ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 (NIHL group vs. CAE-treated group). NOR = normal.</p>
Full article ">
26 pages, 1810 KiB  
Review
Roles of Sirtuins in Hearing Protection
by Chail Koo, Claus-Peter Richter and Xiaodong Tan
Pharmaceuticals 2024, 17(8), 998; https://doi.org/10.3390/ph17080998 - 28 Jul 2024
Viewed by 1420
Abstract
Hearing loss is a health crisis that affects more than 60 million Americans. Currently, sodium thiosulfate is the only drug approved by the Food and Drug Administration (FDA) to counter hearing loss. Sirtuins were proposed as therapeutic targets in the search for new [...] Read more.
Hearing loss is a health crisis that affects more than 60 million Americans. Currently, sodium thiosulfate is the only drug approved by the Food and Drug Administration (FDA) to counter hearing loss. Sirtuins were proposed as therapeutic targets in the search for new compounds or drugs to prevent or cure age-, noise-, or drug-induced hearing loss. Sirtuins are proteins involved in metabolic regulation with the potential to ameliorate sensorineural hearing loss. The mammalian sirtuin family includes seven members, SIRT1-7. This paper is a literature review on the sirtuins and their protective roles in sensorineural hearing loss. Literature search on the NCBI PubMed database and NUsearch included the keywords ‘sirtuin’ and ‘hearing’. Studies on sirtuins without relevance to hearing and studies on hearing without relevance to sirtuins were excluded. Only primary research articles with data on sirtuin expression and physiologic auditory tests were considered. The literature review identified 183 records on sirtuins and hearing. After removing duplicates, eighty-one records remained. After screening for eligibility criteria, there were forty-eight primary research articles with statistically significant data relevant to sirtuins and hearing. Overall, SIRT1 (n = 29) was the most studied sirtuin paralog. Over the last two decades, research on sirtuins and hearing has largely focused on age-, noise-, and drug-induced hearing loss. Past and current studies highlight the role of sirtuins as a mediator of redox homeostasis. However, more studies need to be conducted on the involvement of SIRT2 and SIRT4-7 in hearing protection. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Flow diagram representing the screening process for articles investigating sirtuins in hearing loss. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 2
<p>Sirtuin and hearing loss research over time by the number of publications in each sirtuin paralog * Up to May 2024.</p>
Full article ">Figure 3
<p>Potential roles of sirtuins in hearing loss. SIRT1 and SIRT3 can modulate ROS in the pathologies leading to hearing loss, evidenced by studies in cell cultures and animal models. The major mechanism is the inhibition of intrinsic apoptosis pathway in IHCs, OHCs, and SGNs. Autophagy and mitophagy, which are triggered by pathways involving SIRT1 and SIRT3, can keep the cells under the threshold of caspase-3 activity sufficient for triggering apoptosis, providing protective roles. Inhibition of SIRT2 may be beneficial in NIHL. It is unclear if SIRT4, SIRT5, and SIRT6 are involved in hearing loss or hearing protection. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">
18 pages, 1437 KiB  
Review
Hearing Loss and Oxidative Stress: A Comprehensive Review
by A. Maniaci, L. La Via, J. R. Lechien, G. Sangiorgio, G. Iannella, G. Magliulo, A. Pace, Q. Mat, S. Lavalle and M. Lentini
Antioxidants 2024, 13(7), 842; https://doi.org/10.3390/antiox13070842 - 14 Jul 2024
Cited by 2 | Viewed by 1997
Abstract
Hearing loss is a prevalent condition affecting millions of people worldwide. Hearing loss has been linked to oxidative stress as a major factor in its onset and progression. The goal of this thorough analysis is to investigate the connection between oxidative stress and [...] Read more.
Hearing loss is a prevalent condition affecting millions of people worldwide. Hearing loss has been linked to oxidative stress as a major factor in its onset and progression. The goal of this thorough analysis is to investigate the connection between oxidative stress and hearing loss, with an emphasis on the underlying mechanisms and possible treatments. The review addressed the many forms of hearing loss, the role of reactive oxygen species (ROS) in causing damage to the cochlea, and the auditory system’s antioxidant defensive mechanisms. The review also goes over the available data that support the use of antioxidants and other methods to lessen hearing loss brought on by oxidative stress. We found that oxidative stress is implicated in multiple types of hearing loss, including age-related, noise-induced, and ototoxic hearing impairment. The cochlea’s unique anatomical and physiological characteristics, such as high metabolic activity and limited blood supply, make it particularly susceptible to oxidative damage. Antioxidant therapies have shown promising results in both animal models and clinical studies for preventing and mitigating hearing loss. Emerging therapeutic approaches, including targeted drug delivery systems and gene therapy, offer new possibilities for addressing oxidative stress in the auditory system. The significance of this review lies in its comprehensive analysis of the intricate relationship between oxidative stress and hearing loss. By synthesizing current knowledge and identifying gaps in understanding, this review provides valuable insights for both researchers and clinicians. It highlights the potential of antioxidant-based interventions and emphasizes the need for further research into personalized treatment strategies. Our findings on oxidative stress mechanisms may also affect clinical practice and future research directions. This review serves as a foundation for developing novel therapeutic approaches and may inform evidence-based strategies for the prevention and treatment of hearing loss, ultimately contributing to improved quality of life for millions affected by this condition worldwide. Full article
(This article belongs to the Special Issue Oxidative Stress in Hearing Loss)
Show Figures

Figure 1

Figure 1
<p>Flow diagram describing the literature research protocol.</p>
Full article ">Figure 2
<p>Flow diagram for molecular mechanisms of Oxidative stress related to Hearing loss development.</p>
Full article ">Figure 3
<p>Molecular mechanism of oxidative stress in hearing loss.</p>
Full article ">
20 pages, 1872 KiB  
Article
Efficacy and Mechanisms of Antioxidant Compounds and Combinations Thereof against Cisplatin-Induced Hearing Loss in a Rat Model
by Liliana Carles, Alejandro Gibaja, Verena Scheper, Juan C. Alvarado, Carlos Almodovar, Thomas Lenarz and José M. Juiz
Antioxidants 2024, 13(7), 761; https://doi.org/10.3390/antiox13070761 - 24 Jun 2024
Viewed by 962
Abstract
Cisplatin is an election chemotherapeutic agent used for many cancer treatments. Its cytotoxicity against neoplastic cells is mirrored by that taking place in healthy cells and tissues, resulting in serious adverse events. A very frequent one is ototoxicity, causing hearing loss which may [...] Read more.
Cisplatin is an election chemotherapeutic agent used for many cancer treatments. Its cytotoxicity against neoplastic cells is mirrored by that taking place in healthy cells and tissues, resulting in serious adverse events. A very frequent one is ototoxicity, causing hearing loss which may permanently affect quality of life after successful oncologic treatments. Exacerbated oxidative stress is a main cytotoxic mechanism of cisplatin, including ototoxicity. Previous reports have shown antioxidant protection against cisplatin ototoxicity, but there is a lack of comparative studies on the otoprotectant activity and mechanism of antioxidant formulations. Here, we show evidence that a cocktail of vitamins A, C, and E along with Mg++ (ACEMg), previously shown to protect against noise-induced hearing loss, reverses auditory threshold shifts, promotes outer hair cell survival, and attenuates oxidative stress in the cochlea after cisplatin treatment, thus protecting against extreme cisplatin ototoxicity in rats. The addition of 500 mg N-acetylcysteine (NAC), which, administered individually, also shows significant attenuation of cisplatin ototoxicity, to the ACEMg formulation results in functional degradation of ACEMg otoprotection. Mg++ administered alone, as MgSO4, also prevents cisplatin ototoxicity, but in combination with 500 mg NAC, otoprotection is also greatly degraded. Increasing the dose of NAC to 1000 mg also results in dramatic loss of otoprotection activity compared with 500 mg NAC. These findings support that single antioxidants or antioxidant combinations, particularly ACEMg in this experimental series, have significant otoprotection efficacy against cisplatin ototoxicity. However, an excess of combined antioxidants and/or elevated doses, above a yet-to-be-defined “antioxidation threshold”, results in unrecoverable redox imbalance with loss of otoprotectant activity. Full article
(This article belongs to the Section Health Outcomes of Antioxidants and Oxidative Stress)
Show Figures

Figure 1

Figure 1
<p>Average threshold shifts in the different antioxidant treatment groups. ABR recordings were performed 7 days after starting the corresponding treatments and 2 days after cisplatin injection. Group II-Cis has been illustrated outside the scale of the graph, representing the undetectability of auditory evoked potentials in this group at any of the frequencies or intensities studied. Overall, Group IX-ACEMg showed the smallest threshold shifts of all tested treatments. At the other end were Group VI-NAC500 + ACEMg and Group IV-NAC1000, with significant threshold shifts spanning throughout most or all tested frequencies. This suggests that excess antioxidant concentrations and/or bioavailability may override redox balance, leading to diminished antioxidant treatment efficacy. N.S.: statistically not significant <span class="html-italic">p</span>-values relative to normal control baseline in Group I-Control. Significant <span class="html-italic">p</span>-values relative to normal control baseline in Group I-Control are shown as: # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.005, ### <span class="html-italic">p</span> &lt; 0.001. Significant <span class="html-italic">p</span>-values relative to Group IX-ACEMg in Group VII-ACE are shown as blue and yellow asterisks (*), respectively (<span class="html-italic">p</span> &lt; 0.05). The broken line in Group VI indicates data obtained from a single animal and therefore not subject to statistical analysis (see text).</p>
Full article ">Figure 2
<p>Line graphs (cytocochleograms) showing outer hair cell loss and preservation in rat cochleae from the different experimental groups. Each color line represents one cochlea, and “n” is the total number of cochleae from individual animals used for cell counts in each treatment group after eliminating defective cochlear turn samples. The black line is the average percentage of outer hair cells as a function of distance from the apex. Notice individual cases in which there is virtually no OHC loss. It is interesting that they are mostly in treatment groups providing better antioxidant otoprotection. They may represent cases of exceptional sensitivity to antioxidant otoprotection in the context of natural biological variability or, alternatively, limited sensitivity to cisplatin ototoxicity. It is worth noting that the Cis group, NAC1000, and NAC500 + ACE + Mg do not show such individual outliers.</p>
Full article ">Figure 3
<p>Bar graph showing the average percentage of the apical to basal length of the organ of Corti with complete preservation of OHCs (green bars), partial loss (yellow bar), or complete loss (red bar). In Group IX-ACEMg, Group VIII-Mg, and Group VII-ACE, the relative length of the organ of Corti with 100% OHC loss is significantly reduced, whereas the apical segment with maximum preservation of OHCs is longer. Notice that Group IV-NAC1000 and Group VI-NAC500 + ACE + Mg did not show significant differences with Group II-Cis in OHC survival patterns. (*) Statistical significance of <span class="html-italic">p</span> values relative to cisplatin, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Fluorescent immunolocalization of the oxidative stress marker 3-NT in the cochlea after cisplatin ototoxicity in comparison with the different antioxidant treatments in this study. (<b>A</b>) Immunolabeling for 3-NT (red) with DAPI counterstaining (blue) in representative sections of the apical turn of the organ of Corti from rats of the different experimental groups. Notice very low or low levels of 3-NT immunolabeling in Group IX-ACEMg, Group VIII-Mg, Group VII-ACE, Group III-NAC500, and also Group V-NAC500 + Mg, comparable to Group I-control. Group IX-NAC1000 and Group VI-NAC500 + ACE + Mg show 3-NT immunostaining visually similar to Group II-Cis. (<b>B</b>) Relative intensity levels of 3-NT immunolabeling in OHC regions from cochlear sections of animals from the different experimental groups. (*) Statistical significance of <span class="html-italic">p</span>-values, relative to the Group II-Cis, * <span class="html-italic">p</span> &lt; 0.05. (#) Statistical significance of <span class="html-italic">p</span>-values relative to the Group I-Control. # <span class="html-italic">p</span> &lt; 0.05, ### <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
22 pages, 3720 KiB  
Article
ERK1/2 Inhibition via the Oral Administration of Tizaterkib Alleviates Noise-Induced Hearing Loss While Tempering down the Immune Response
by Richard D. Lutze, Matthew A. Ingersoll, Alena Thotam, Anjali Joseph, Joshua Fernandes and Tal Teitz
Int. J. Mol. Sci. 2024, 25(12), 6305; https://doi.org/10.3390/ijms25126305 - 7 Jun 2024
Cited by 3 | Viewed by 1410
Abstract
Noise-induced hearing loss (NIHL) is a major cause of hearing impairment and is linked to dementia and mental health conditions, yet no FDA-approved drugs exist to prevent it. Downregulating the mitogen-activated protein kinase (MAPK) cellular pathway has emerged as a promising approach to [...] Read more.
Noise-induced hearing loss (NIHL) is a major cause of hearing impairment and is linked to dementia and mental health conditions, yet no FDA-approved drugs exist to prevent it. Downregulating the mitogen-activated protein kinase (MAPK) cellular pathway has emerged as a promising approach to attenuate NIHL, but the molecular targets and the mechanism of protection are not fully understood. Here, we tested specifically the role of the kinases ERK1/2 in noise otoprotection using a newly developed, highly specific ERK1/2 inhibitor, tizaterkib, in preclinical animal models. Tizaterkib is currently being tested in phase 1 clinical trials for cancer treatment and has high oral bioavailability and low predicted systemic toxicity in mice and humans. In this study, we performed dose–response measurements of tizaterkib’s efficacy against permanent NIHL in adult FVB/NJ mice, and its minimum effective dose (0.5 mg/kg/bw), therapeutic index (>50), and window of opportunity (<48 h) were determined. The drug, administered orally twice daily for 3 days, 24 h after 2 h of 100 dB or 106 dB SPL noise exposure, at a dose equivalent to what is prescribed currently for humans in clinical trials, conferred an average protection of 20–25 dB SPL in both female and male mice. The drug shielded mice from the noise-induced synaptic damage which occurs following loud noise exposure. Equally interesting, tizaterkib was shown to decrease the number of CD45- and CD68-positive immune cells in the mouse cochlea following noise exposure. This study suggests that repurposing tizaterkib and the ERK1/2 kinases’ inhibition could be a promising strategy for the treatment of NIHL. Full article
(This article belongs to the Special Issue Hearing Loss: Molecular Biological Insights)
Show Figures

Figure 1

Figure 1
<p>AZD0364 protects mice from noise-induced hearing loss when administered 45 min before noise exposure. (<b>A</b>) Molecular structure of tizaterkib. (<b>B</b>) Schedule of administration of noise exposure and tizaterkib treatments in FVB mice. Mice were given their first treatment of tizaterkib via an oral gavage 45 min before noise exposure. Mice were treated with the drug for a total of 3 days, twice a day, and exposed to noise once. (<b>C</b>) ABR threshold shifts following procedure in (<b>B</b>). Shaded region is the frequency range of the noise exposure. (<b>D</b>) Representative post-noise-exposure ABRs of noise-alone- and noise + tizaterkib-treated mice. (<b>E</b>) Percent weight change of different experimental cohorts throughout the 14-day protocol shown in (<b>B</b>). Noise + Carrier (red), noise + tizaterkib (green), tizaterkib alone (blue), and carrier (black). Data shown as means ± SEM; ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 compared to noise alone by two-way ANOVA with a Bonferroni post hoc test. The color of the asterisks indicates the statistical significance of the treatment group with that same color compared to noise + carrier treated mice. <span class="html-italic">n</span> = 9–10 mice.</p>
Full article ">Figure 2
<p>Tizaterkib protects mice from noise-induced hearing loss when administered 24 h after noise exposure. (<b>A</b>) Schedule of noise exposure and tizaterkib, which began 24 h after noise exposure. Mice were treated with varying concentrations of tizaterkib twice a day for 3 whole days. (<b>B</b>) ABR threshold shifts following procedure in (<b>A</b>), with 25 and 5 mg/kg tizaterkib given to separate groups. Shaded region is the frequency range of the noise exposure. (<b>C</b>) ABR threshold shifts following the procedure in (<b>A</b>) with 0.5 mg/kg administered to mice. (<b>D</b>) ABR threshold shifts following the procedure in (<b>A</b>) with the 0.1 mg/kg tizaterkib treatment. (<b>E</b>) Dose–response curve of tizaterkib protection from noise-induced hearing loss at 16 kHz, with 100% protection as a 0 dB SPL threshold shift. (<b>F</b>) ABR threshold shifts of males and females, graphed separately, that were treated with tizaterkib or carrier following the procedure in (<b>A</b>). (<b>G</b>) Representative ABR traces of noise-alone- and noise + tizaterkib-treated mice. Noise + carrier (red), noise + tizaterkib (green), noise + 5 mg/kg tizaterkib (purple), tizaterkib alone (blue), and carrier (black). Data shown as means ± SEM; * <span class="html-italic">p</span> &lt; 0.05 and *** <span class="html-italic">p</span> &lt; 0.001 compared to noise alone by two-way ANOVA with a Bonferroni post hoc test. The color of the asterisks indicates the statistical significance of the treatment group with that same color compared to noise + carrier treated mice. <span class="html-italic">n</span> = 9–10 mice.</p>
Full article ">Figure 3
<p>Tizaterkib protects mice from noise-induced synaptopathy in the 8 and 16 kHz regions. (<b>A</b>) Representative images of whole-mount cochlear sections stained with myosin VI (green) and Ctbp2 (red) in the 8 kHz region. (<b>B</b>) Number of Ctbp2 puncta per IHC in the 8 kHz region. (<b>C</b>) Representative images of whole-mount cochlear sections in the 16 kHz region. (<b>D</b>) Number of Ctbp2 puncta per IHC in the 16 kHz region. (<b>E</b>) ABR wave 1 amplitude for 16 kHz from the post experimental ABR recordings shown in <a href="#ijms-25-06305-f002" class="html-fig">Figure 2</a>C. The wave 1 amplitude was measured from 60–90 dB. Data shown as means ± SEM; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 compared to noise alone by one-way ANOVA with a Bonferroni post hoc test. Tizaterkib alone (blue), carrier (black), noise alone (red), noise + tizaterkib (green). <span class="html-italic">n</span> = 9 mice.</p>
Full article ">Figure 4
<p>Tizaterkib protects from noise-induced hearing loss when mice are exposed to 106 dB. (<b>A</b>) Schedule of noise exposure and tizaterkib treatment. Mice were exposed to 106 dB SPL for 2 h and tizaterkib treatment started 24 h after noise exposure. Mice were treated for 3 whole days, twice a day. (<b>B</b>) ABR threshold shifts following the protocol in (<b>A</b>). Shaded region is the frequency range of the noise exposure. (<b>C</b>) Representative ABR traces of noise-alone- and noise + tizaterkib-treated mice following the 106 dB SPL noise exposure. (<b>D</b>) DPOAE threshold shifts following the protocol in (<b>A</b>). Noise + carrier (red), noise + tizaterkib (green), tizaterkib alone (blue), and carrier (black). Data shown as means ± SEM; * <span class="html-italic">p</span> &lt; 0.05 and *** <span class="html-italic">p</span> &lt; 0.001 compared to noise alone by two-way ANOVA with a Bonferroni post hoc test. The color of the asterisks indicates the statistical significance of the treatment group with that same color compared to noise + carrier treated mice. <span class="html-italic">n</span> = 13 mice.</p>
Full article ">Figure 5
<p>Tizaterkib treatment phenocopies the resistance to noise-induced hearing loss measured in the KSR1 KO mouse model. (<b>A</b>) KSR1 is a scaffolding protein for RAF, MEK, and ERK which enables the efficient transmission of MAPK signals. (<b>B</b>) Schedule of noise exposure and 5 mg/kg tizaterkib treatment in KSR1 WT and KO mice. Mice were exposed to 100 dB SPL for 2 h and tizaterkib treatment began 24 h after noise exposure. Mice were treated with tizaterkib or carrier twice a day for 3 whole days. (<b>C</b>) ABR threshold shifts following the protocol in (<b>B</b>). Shaded region is the frequency range of the noise exposure. WT + noise (red), KO + noise + tizaterkib (blue), WT + noise + tizaterkib (green), KO + noise (purple), and WT + carrier alone (black). Data shown as means ± SEM; * <span class="html-italic">p</span> &lt; 0.05 and *** <span class="html-italic">p</span> &lt; 0.001 compared to noise alone by two-way ANOVA with a Bonferroni post hoc test. The color of the asterisks indicates the statistical significance of the treatment group with that same color compared to noise + carrier treated mice. <span class="html-italic">n</span> = 5–6 mice.</p>
Full article ">Figure 6
<p>Three days of treatment beginning 24 h after noise exposure produces the optimal protection in terms of tizaterkib administration. (<b>A</b>) Schedule of administration for noise exposure and tizaterkib treatment. Treatment with tizaterkib began 48 h after noise exposure and mice were treated for 3 days, twice a day. (<b>B</b>) ABR threshold shifts following the protocol in (<b>A</b>). Shaded region is the frequency range of the noise exposure. Noise alone (red), noise + tizaterkib (green), carrier (black), and tizaterkib alone (blue). (<b>C</b>) Schedule of administration of noise exposure and tizaterkib treatment. Treatment began 24 h after noise exposure and one cohort was treated for 1 day, one cohort was treated for 2 days, and another cohort was treated for 3 days. (<b>D</b>) ABR threshold shifts following the protocol in (<b>C</b>). Noise alone (red), 1-day treatment + noise (purple), 2-day treatment + noise (blue), 3-day treatment + noise (green), carrier alone (black). Data shown as means ± SEM; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 compared to noise alone by two-way ANOVA with a Bonferroni post hoc test. The color of the asterisks indicates statistical significance of the treatment group with respect to that same color compared to noise + carrier-treated mice. <span class="html-italic">n</span> = 6–10 mice.</p>
Full article ">Figure 7
<p>Tizaterkib treatment lowers the number of CD45-positive cells in the cochlea 4 days post noise exposure. (<b>A</b>) Representative low-magnification images of cochlear cryosections stained with CD45 (red) and DAPI (blue). The treatment protocol shown in <a href="#ijms-25-06305-f002" class="html-fig">Figure 2</a>A was utilized and mice were sacrificed 4 days after noise exposure, 1 h after the final tizaterkib treatment. (<b>B</b>) Quantification of the CD45-positive cells in the cochlear sections in (<b>A</b>). (<b>C</b>) Higher magnification of the images shown in (<b>A</b>) of the scala tympani. (<b>D</b>) Quantification of CD45-positive cells in the walls of the scala tympani as presented in (<b>C</b>). (<b>E</b>) Representative images of cochlear cryosections of the stria vascularis following noise and tizaterkib treatment. (<b>F</b>) Quantification of CD45-positive cells per experimental group from the images in (<b>E</b>). Carrier (black), tizaterkib alone (blue), noise alone (red), noise + tizaterkib (green). Data shown as means ± SEM, * <span class="html-italic">p</span> &lt; 0.05 and *** <span class="html-italic">p</span> &lt; 0.001 compared to noise alone by one-way ANOVA with a Bonferroni post hoc test. <span class="html-italic">n</span> = 3–6 mice, with 3 sections each per mouse.</p>
Full article ">Figure 8
<p>Tizaterkib treatment lowers the amount of CD45 and CD68 in the cochlea 6 days post noise exposure. (<b>A</b>) Western blots showing the amount of CD45 and CD68 in the cochlea following noise exposure and tizaterkib treatment. The same treatment protocol shown in <a href="#ijms-25-06305-f002" class="html-fig">Figure 2</a>A was utilized and mice were sacrificed 6 days after noise exposure. (<b>B</b>) CD45/GAPDH ratio, normalized to the carrier-alone lane. Band intensities were measured using ImageJ software (version 1.54g). (<b>C</b>) CD68/GAPDH ratio, normalized to the carrier-alone lane. Data shown as means ± SEM, * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 compared to noise alone by one-way ANOVA with a Bonferroni post hoc test. The experimental groups, from left to right, are as follows: carrier alone, tizaterkib alone, noise alone, and noise + tizaterkib. Each group had the cochleae from 5 mice (10 cochleae) pooled together to make the tissue lysate. <span class="html-italic">n</span> = 5.</p>
Full article ">
17 pages, 4901 KiB  
Article
Trametinib, a MEK1/2 Inhibitor, Protects Mice from Cisplatin- and Noise-Induced Hearing Loss
by Richard D. Lutze, Matthew A. Ingersoll, Regina G. Kelmann and Tal Teitz
Pharmaceuticals 2024, 17(6), 735; https://doi.org/10.3390/ph17060735 - 5 Jun 2024
Viewed by 1094
Abstract
Hearing loss is one of the most common types of disability; however, there is only one FDA-approved drug to prevent any type of hearing loss. Treatment with the highly effective chemotherapy agent, cisplatin, and exposure to high-decibel noises are two of the most [...] Read more.
Hearing loss is one of the most common types of disability; however, there is only one FDA-approved drug to prevent any type of hearing loss. Treatment with the highly effective chemotherapy agent, cisplatin, and exposure to high-decibel noises are two of the most common causes of hearing loss. The mitogen-activated protein kinase (MAPK) pathway, a phosphorylation cascade consisting of RAF, MEK1/2, and ERK1/2, has been implicated in both types of hearing loss. Pharmacologically inhibiting BRAF or ERK1/2 is protective against noise- and cisplatin-induced hearing loss in multiple mouse models. Trametinib, a MEK1/2 inhibitor, protects from cisplatin-induced outer hair cell death in mouse cochlear explants; however, to the best of our knowledge, inhibiting MEK1/2 has not yet been shown to be protective against hearing loss in vivo. In this study, we demonstrate that trametinib protects against cisplatin-induced hearing loss in a translationally relevant mouse model and does not interfere with cisplatin’s tumor-killing efficacy in cancer cell lines. Higher doses of trametinib were toxic to mice when combined with cisplatin, but lower doses of the drug were protective against hearing loss without any known toxicity. Trametinib also protected mice from noise-induced hearing loss and synaptic damage. This study shows that MEK1/2 inhibition protects against both insults of hearing loss, as well as that targeting all three kinases in the MAPK pathway protects mice from cisplatin- and noise-induced hearing loss. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Figure 1

Figure 1
<p>Trametinib inhibits MAPK activity in the HEI-OC1 cell line and does not interfere with cisplatin’s tumor-killing ability in cancer cell lines. (<b>A</b>) Schematic of the MAPK phosphorylation cascade in which dabrafenib inhibits BRAF, trametinib inhibits MEK1/2, and tizaterkib inhibits ERK1/2. (<b>B</b>) Representative Western blots of HEI-OC1 cell lysates treated with medium, cisplatin, and different concentrations of trametinib. Treatment groups from left to right are as follows: medium alone, 1 μM trametinib alone, 50 μM cisplatin alone, 50 μM cisplatin + 0.01 μM trametinib, 50 μM cisplatin + 0.1 μM trametinib, and 50 μM cisplatin + 1 μM trametinib. (<b>C</b>) Quantification of Western blots represented in (<b>B</b>). A total of 4 separate experiments were performed. The ratio of pERK to GAPDH was measured for each individual lane and all groups were then normalized to the medium-alone treatment group. Data shown as means ± SEM. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001 compared to cisplatin-alone treatment. All groups were compared to one another by one-way ANOVA with Bonferroni post hoc test. (<b>D</b>) Percentage of cell viability for SK-N-AS cell line treated with cisplatin and various concentrations of trametinib starting at 4.57 nM and going up to 30 μM in increments of 3-fold increases. (<b>E</b>) Percentage of cell viability for SH-SY5Y cell line treated with cisplatin and various concentrations of trametinib as mentioned in (<b>D</b>). (<b>F</b>) Percentage of cell viability for A549 cell line treated with cisplatin and various concentrations of trametinib as mentioned in (<b>D</b>). Medium alone (black), cisplatin alone (red), trametinib alone (Blue), and trametinib + cisplatin (blue and red checkered pattern). All wells treated with cisplatin had the same concentration of cisplatin, and increasing concentrations of trametinib were used starting at 4.57 nM going up to 30 μM going from left to right. All treatments were normalized to medium-alone-treated cells and compared to the cisplatin-alone treatment. Data shown as mean ± SEM; ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 compared to cisplatin alone by one-way ANOVA with Bonferroni post hoc test.</p>
Full article ">Figure 2
<p>Trametinib protects adult mice from cisplatin-induced hearing loss in a clinically relevant mouse model. (<b>A</b>) Schedule of administration of trametinib and cisplatin in a translational, multi-cycle cisplatin treatment protocol using CBA/CaJ mice. Each cycle consisted of four days of treatment with 3 mg/kg cisplatin in the morning and five days of treatment with 1.0, 0.2, or 0.1 mg/kg trametinib in the morning and evening. A 9-day recovery period followed the 5 days of treatment. This cycle was repeated a total of 3 times. Auditory testing occurred before treatment began and immediately after cycle 3 (day 42). (<b>B</b>) ABR threshold shifts recorded immediately after the completion of cycle 3 (day 42) in protocol shown in (<b>A</b>). (<b>C</b>) Amplitudes of ABR wave 1 at 16 kHz from (<b>B</b>). (<b>D</b>) DPOAE threshold shifts recorded after the completion of cycle 3 (day 42) in protocol shown in (<b>A</b>). Carrier alone (black), 1 mg/kg trametinib alone (orange), 0.2 mg/kg trametinib alone (yellow), cisplatin alone (red), 1 mg/kg trametinib + cisplatin (blue), 0.2 mg/kg trametinib + cisplatin (green), and 0.1 mg/kg trametinib + cisplatin (purple). Data shown as means ± SEM; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 compared to cisplatin alone by two-way ANOVA with Bonferroni post hoc test.</p>
Full article ">Figure 2 Cont.
<p>Trametinib protects adult mice from cisplatin-induced hearing loss in a clinically relevant mouse model. (<b>A</b>) Schedule of administration of trametinib and cisplatin in a translational, multi-cycle cisplatin treatment protocol using CBA/CaJ mice. Each cycle consisted of four days of treatment with 3 mg/kg cisplatin in the morning and five days of treatment with 1.0, 0.2, or 0.1 mg/kg trametinib in the morning and evening. A 9-day recovery period followed the 5 days of treatment. This cycle was repeated a total of 3 times. Auditory testing occurred before treatment began and immediately after cycle 3 (day 42). (<b>B</b>) ABR threshold shifts recorded immediately after the completion of cycle 3 (day 42) in protocol shown in (<b>A</b>). (<b>C</b>) Amplitudes of ABR wave 1 at 16 kHz from (<b>B</b>). (<b>D</b>) DPOAE threshold shifts recorded after the completion of cycle 3 (day 42) in protocol shown in (<b>A</b>). Carrier alone (black), 1 mg/kg trametinib alone (orange), 0.2 mg/kg trametinib alone (yellow), cisplatin alone (red), 1 mg/kg trametinib + cisplatin (blue), 0.2 mg/kg trametinib + cisplatin (green), and 0.1 mg/kg trametinib + cisplatin (purple). Data shown as means ± SEM; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 compared to cisplatin alone by two-way ANOVA with Bonferroni post hoc test.</p>
Full article ">Figure 3
<p>Trametinib protects against cisplatin-induced OHC loss in the multi-cycle cisplatin treatment protocol. (<b>A</b>) Representative whole-mount cochlear sections stained with myosin VI to visualize hair cells. Treatment groups from top to bottom are as follows: carrier alone, 1 mg/kg trametinib alone, cisplatin alone, 1 mg/kg trametinib + cisplatin, and 0.2 mg/kg trametinib + cisplatin. Apical turn is shown on left, middle turn in the middle, and basal turn on the right. (<b>B</b>) Quantification of the number of outer hair cells per 160 μm per section for apical turn, middle turn, and basal turn of cochlea. Carrier alone (black), 1 mg/kg trametinib alone (orange), cisplatin alone (red), 1 mg/kg trametinib + cisplatin (blue), and 0.2 mg/kg trametinib + cisplatin (green). Data shown as means ± SEM; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001 compared to cisplatin alone by two-way ANOVA with Bonferroni post hoc test. <span class="html-italic">n</span> = 4–5 mice.</p>
Full article ">Figure 4
<p>Trametinib confers slight protection from cisplatin-induced weight loss but co-treatment of higher doses of trametinib with cisplatin caused mouse death. (<b>A</b>) Weight loss over the 42-day treatment protocol shown in <a href="#pharmaceuticals-17-00735-f002" class="html-fig">Figure 2</a>A. Data shown as means ± SEM; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt;0.01, and *** <span class="html-italic">p</span> &lt; 0.001 compared to cisplatin alone by two-way ANOVA with Bonferroni post hoc test. (<b>B</b>) Kaplan–Meier survival curves of mouse cohorts going to day 42 following protocol in <a href="#pharmaceuticals-17-00735-f002" class="html-fig">Figure 2</a>A. Carrier alone (black), 1 mg/kg trametinib alone (orange), 0.2 mg/kg trametinib alone (yellow), cisplatin alone (red), 1 mg/kg trametinib + cisplatin (blue), and 0.2 mg/kg trametinib + cisplatin (green).</p>
Full article ">Figure 5
<p>Trametinib protects against noise-induced hearing loss and ribbon synapse loss following noise exposure. (<b>A</b>) Noise exposure and treatment protocol. ABR pre-hearing tests were performed and then mice were exposed to 100 dB SPL noise for 2 h. Starting 24 h after noise exposure, mice were treated with 3.15 mg/kg trametinib twice a day for 3 total days, once in the morning and once at night. Fourteen days after noise exposure, ABR hearing tests were performed again to determine the amount of hearing loss for each mouse. (<b>B</b>) ABR threshold shifts from the treatment protocol shown in (<b>A</b>). Carrier alone (black), trametinib alone (green), noise alone (red), and trametinib + noise (blue). Data shown as means ± SEM; *** <span class="html-italic">p</span> &lt; 0.001 compared to noise alone by two-way ANOVA with Bonferroni post hoc test. (<b>C</b>) Representative confocal images of whole-mount cochlear sections stained with myosin VI (green) and Ctbp2 (red). (<b>D</b>) Quantification of the average number of Ctbp2 puncta per IHC for each treatment group. Trametinib alone (black), noise alone (red), and noise + trametinib (blue). Data shown as means ± SEM; * <span class="html-italic">p</span> &lt; 0.05 and *** <span class="html-italic">p</span> &lt; 0.001 with all groups compared to one another by one-way ANOVA with Bonferroni post hoc test.</p>
Full article ">
17 pages, 575 KiB  
Review
The Current State of Proteomics and Metabolomics for Inner Ear Health and Disease
by Motahare Khorrami, Christopher Pastras, Paul A. Haynes, Mehdi Mirzaei and Mohsen Asadnia
Proteomes 2024, 12(2), 17; https://doi.org/10.3390/proteomes12020017 - 4 Jun 2024
Viewed by 1677
Abstract
Characterising inner ear disorders represents a significant challenge due to a lack of reliable experimental procedures and identified biomarkers. It is also difficult to access the complex microenvironments of the inner ear and investigate specific pathological indicators through conventional techniques. Omics technologies have [...] Read more.
Characterising inner ear disorders represents a significant challenge due to a lack of reliable experimental procedures and identified biomarkers. It is also difficult to access the complex microenvironments of the inner ear and investigate specific pathological indicators through conventional techniques. Omics technologies have the potential to play a vital role in revolutionising the diagnosis of ear disorders by providing a comprehensive understanding of biological systems at various molecular levels. These approaches reveal valuable information about biomolecular signatures within the cochlear tissue or fluids such as the perilymphatic and endolymphatic fluid. Proteomics identifies changes in protein abundance, while metabolomics explores metabolic products and pathways, aiding the characterisation and early diagnosis of diseases. Although there are different methods for identifying and quantifying biomolecules, mass spectrometry, as part of proteomics and metabolomics analysis, could be utilised as an effective instrument for understanding different inner ear disorders. This study aims to review the literature on the application of proteomic and metabolomic approaches by specifically focusing on Meniere’s disease, ototoxicity, noise-induced hearing loss, and vestibular schwannoma. Determining potential protein and metabolite biomarkers may be helpful for the diagnosis and treatment of inner ear problems. Full article
(This article belongs to the Special Issue Quantitative Proteomics: Techniques and Applications)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The anatomy of the human ear includes the external, middle, and inner ear (comprising the cochlea and vestibular system). Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">
32 pages, 7815 KiB  
Article
Neural Adaptation at Stimulus Onset and Speed of Neural Processing as Critical Contributors to Speech Comprehension Independent of Hearing Threshold or Age
by Jakob Schirmer, Stephan Wolpert, Konrad Dapper, Moritz Rühle, Jakob Wertz, Marjoleen Wouters, Therese Eldh, Katharina Bader, Wibke Singer, Etienne Gaudrain, Deniz Başkent, Sarah Verhulst, Christoph Braun, Lukas Rüttiger, Matthias H. J. Munk, Ernst Dalhoff and Marlies Knipper
J. Clin. Med. 2024, 13(9), 2725; https://doi.org/10.3390/jcm13092725 - 6 May 2024
Cited by 1 | Viewed by 1241
Abstract
Background: It is assumed that speech comprehension deficits in background noise are caused by age-related or acquired hearing loss. Methods: We examined young, middle-aged, and older individuals with and without hearing threshold loss using pure-tone (PT) audiometry, short-pulsed distortion-product otoacoustic emissions [...] Read more.
Background: It is assumed that speech comprehension deficits in background noise are caused by age-related or acquired hearing loss. Methods: We examined young, middle-aged, and older individuals with and without hearing threshold loss using pure-tone (PT) audiometry, short-pulsed distortion-product otoacoustic emissions (pDPOAEs), auditory brainstem responses (ABRs), auditory steady-state responses (ASSRs), speech comprehension (OLSA), and syllable discrimination in quiet and noise. Results: A noticeable decline of hearing sensitivity in extended high-frequency regions and its influence on low-frequency-induced ABRs was striking. When testing for differences in OLSA thresholds normalized for PT thresholds (PTTs), marked differences in speech comprehension ability exist not only in noise, but also in quiet, and they exist throughout the whole age range investigated. Listeners with poor speech comprehension in quiet exhibited a relatively lower pDPOAE and, thus, cochlear amplifier performance independent of PTT, smaller and delayed ABRs, and lower performance in vowel-phoneme discrimination below phase-locking limits (/o/-/u/). When OLSA was tested in noise, listeners with poor speech comprehension independent of PTT had larger pDPOAEs and, thus, cochlear amplifier performance, larger ASSR amplitudes, and higher uncomfortable loudness levels, all linked with lower performance of vowel-phoneme discrimination above the phase-locking limit (/i/-/y/). Conslusions: This study indicates that listening in noise in humans has a sizable disadvantage in envelope coding when basilar-membrane compression is compromised. Clearly, and in contrast to previous assumptions, both good and poor speech comprehension can exist independently of differences in PTTs and age, a phenomenon that urgently requires improved techniques to diagnose sound processing at stimulus onset in the clinical routine. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Elevated hearing thresholds correlated with age, in particular at high frequencies: (<b>A</b>) Individual (grey lines) and mean (black line) PTTs for the three age groups young (left), middle-aged (center), and older (right) used for PTAs of four different frequency ranges: low frequencies “PTA-LF” (0.125–1 kHz), main-language region “PTA4” (0.5–4 kHz), high frequencies “PTA-HF” (6–10 kHz), and extended high frequencies “PTA-EHF” (11.2–16 kHz), illustrated in red on the abscissa of the left-most audiogram. The group mean thresholds are plotted in black (young: circles; middle-aged: triangles; older: squares). (<b>B</b>) Scatterplots for individual hearing thresholds as a function of age, split into the four PTA frequency ranges. The shaded area delineates the age range of the middle-aged group. <span class="html-italic">p</span>-Values (Pearson’s correlation): p(PTA-LF) = 0.000016; p(PTA4) &lt; 0.00001; p(PTA-HF) &lt; 0.00001, p(PTA-EHF) &lt; 0.00001.</p>
Full article ">Figure 2
<p>(<b>A</b>) Power spectrum of the OLSA speech material (broadband, black curve), of high-pass filtered speech (OLSA-HP, red shaded area), and low-pass filtered speech (OLSA-LP, blue shaded area), shown peak-normalized to 0 dB and 1/f-corrected. The four different PTA frequency ranges are depicted: low frequencies “PTA-LF” (0.125–1 kHz), “PTA4” (0.5–4 kHz), high frequencies “PTA-HF” [6–10 kHz], and extended high frequencies “PTA-EHF” (11.2–16 kHz). (<b>B</b>,<b>C</b>) The influence of noise on OLSA SRT<sub>50</sub> was examined using differently filtered speech material. Noise conditions in (<b>B</b>) quiet and (<b>C</b>) ipsilateral noise. Columns provide results for broadband and filtered OLSA stimuli. OLSA SRTs are provided as a function of age.</p>
Full article ">Figure 3
<p>ABR as a function of age, pure-tone averages, and speech comprehension: (<b>A</b>) ABR wave amplitude and latencies grouped by age. Circles represent young, triangles middle-aged, and squares older participants. (<b>B</b>–<b>D</b>) ABR wave amplitudes and latencies grouped for participants with low (green) and high (pink) thresholds of PTA4 (<b>B</b>), PTA-HF (<b>C</b>), and PTA-EHF (<b>D</b>).</p>
Full article ">Figure 4
<p>OLSA speech reception threshold SRT<sub>50</sub> (dB SPL; <span class="html-italic">y</span>-axes) for differently filtered OLSA stimuli ((<b>A</b>,<b>E</b>) broadband, (<b>B</b>,<b>F</b>) low pass, and (<b>C</b>,<b>D</b>,<b>G</b>,<b>H</b>) high-pass) as a function of PTA4 (<b>A</b>,<b>E</b>), PTA-LF (<b>B</b>,<b>F</b>), PTA-HF(<b>C</b>,<b>G</b>), and PTA-EHF (<b>D</b>,<b>H</b>) (<span class="html-italic">x</span>-axes). (<b>A</b>–<b>D</b>) provide results obtained in quiet (n = 89), (<b>E</b>–<b>H</b>) under ipsilateral (n = 63) noise condition. Regression lines are plotted in black and include y-intersections and R<sup>2</sup> values. The different colors assign each subject to one of the three speech comprehension groups: good (blue), standard (grey), and poor (orange).</p>
Full article ">Figure 5
<p>Subjective hearing evaluation by age and speech comprehension: (<b>A</b>) shows age groups and (<b>B</b>) groups according to objective speech comprehension performance based on OLSA thresholds corrected by PNOTs. <span class="html-italic">y</span>-axis: subjective evaluation, <span class="html-italic">x</span>-axis: percentage of all responses given by all participants in age groups (<b>A</b>) and in PNOT groups (<b>B</b>). Participants were asked to rate their hearing as excellent, very good, good, moderate, or bad (<span class="html-italic">y</span>-axis labels).</p>
Full article ">Figure 6
<p>(<b>A</b>) ASSR response amplitudes in µV averaged for 4 and 6 kHz carriers as a function of participant age in years. The blue, grey, and orange-colored symbols refer to the good, standard, and poor speech comprehension groups, respectively. (<b>B</b>) Median (horizontal bar) and individual participants (symbols) ASSR amplitude averaged for 4 and 6 kHz carriers (Mean, left), 4 kHz carrier (middle), and 6 kHz carrier (right) for the quiet listening condition (upper row), or in ipsilateral noise (lower row). Numbers in brackets indicate the number of participants included in the analyses. (<b>C</b>) Regression line (black) of the dependence of OLSA SRT<sub>50</sub> in ipsilateral noise on ASSR amplitudes (averaged for 4 and 6 kHz carriers) normalized for PTT. The y-intersection, R<sup>2</sup> value, and <span class="html-italic">p</span>-value of regression are given close to the trend line.</p>
Full article ">Figure 7
<p>(<b>A</b>,<b>E</b>) L<sub>EDPT</sub> acceptance rates, (<b>B</b>,<b>F</b>) PTT, (<b>C</b>,<b>G</b>) L<sub>EDPT</sub>, (<b>D</b>,<b>H</b>) L<sub>EDPT</sub>-to-PTT difference for left and right ears are compared between good (blue) and poor (orange) speech-in-quiet comprehension performers. Participants with good speech-in-quiet performance (blue) showed higher acceptance rates (<b>A</b>), equal PTT (<b>B</b>), inconclusive L<sub>EDPT</sub> (<b>C</b>), but a consistent 3 dB better threshold for L<sub>EDPT</sub>-to-PTT although on the right ear with only <span class="html-italic">p</span> = 0.084 (<b>D</b>). Estimated distortion-product thresholds (L<sub>EDPT</sub>) in relation to PTT, when participants are grouped with respect to their speech-in-ipsilateral-noise performance (<b>E</b>) L<sub>EDPT</sub> acceptance rates, (<b>F</b>) PTT, (<b>G</b>) L<sub>EDPT</sub>, (<b>H</b>) L<sub>EDPT</sub>-to-PTT difference for left and right ears are compared between good (blue) and poor (orange) speech-in-noise comprehension performers. Participants with good speech-in-noise performance (blue) show reduced acceptance rates, reduced PTT and L<sub>EDPT</sub>, but no difference for L<sub>EDPT</sub>-to-PTT.</p>
Full article ">Figure 8
<p>ABR wave amplitude as a function of ABR wave latency in participants matched for PTA thresholds and grouped for good (blue), standard (grey), or poor (orange) speech comprehension in quiet. Significant shifts in latency in poor comprehension, in comparison to the group with good speech comprehension were observed (ABR wave I latency: n = 29, 27, 24, <span class="html-italic">p</span> = 0.218242; wave II latency: n = 24, 22, 16, <span class="html-italic">p</span> = 0.007707, wave III latency: n = 30, 28, 26, <span class="html-italic">p</span> = 0.182784; wave V latency: n = 30, 28, 28, <span class="html-italic">p</span> = 0.026617 and wave VI latency: n = 27, 27, 24, <span class="html-italic">p</span> = 0.001055).</p>
Full article ">Figure 9
<p>Syllable-discrimination scores in relation to speech comprehension. The scores for four pairs of phonemes (/o/-/u/, /i/-/y/, /du/-/bu/, /di/-/bi/) are segregated for participants with poor (orange), good (blue), and standard (grey) speech comprehension selected by PNOT in quiet (<b>A</b>), and ipsilateral noise (<b>B</b>). Each plot consists of a boxplot with perceptual performance [% correct] as a function of PNOT (x-axis). Finally, there is a graphical representation of the significance assessed by Mann–Whitney U tests (<a href="#app1-jcm-13-02725" class="html-app">Supplementary Table S4</a>), significant differences are shown as asterisks with a color code reflecting the three groups.</p>
Full article ">Figure 10
<p>Good and poor speech comprehension in quiet differs from good and poor speech comprehension in ipsilateral noise in the discrimination ability of formant contrasts below PLL (requiring TFS coding), and above PLL (requiring TENV coding). In quiet, poor speech comprehension is associated with poor discrimination below the PLL (e.g., for /o/-/u/), while good speech comprehension is associated with good discrimination above the PLL (e.g., for /i/-/y/). In ipsilateral noise, poor speech comprehension is associated with lower performance for discriminating phoneme pairs with formant contrasts above PLL (/i/-/y/, above 1500 Hz), while good speech comprehension is associated with good discrimination of formants below the PLL (/o/-/u/, below 1500 Hz).</p>
Full article ">
17 pages, 682 KiB  
Review
Role of Oxidative Stress in Sensorineural Hearing Loss
by Masato Teraoka, Naohito Hato, Haruhiko Inufusa and Fukka You
Int. J. Mol. Sci. 2024, 25(8), 4146; https://doi.org/10.3390/ijms25084146 - 9 Apr 2024
Cited by 6 | Viewed by 3063
Abstract
Hearing is essential for communication, and its loss can cause a serious disruption to one’s social life. Hearing loss is also recognized as a major risk factor for dementia; therefore, addressing hearing loss is a pressing global issue. Sensorineural hearing loss, the predominant [...] Read more.
Hearing is essential for communication, and its loss can cause a serious disruption to one’s social life. Hearing loss is also recognized as a major risk factor for dementia; therefore, addressing hearing loss is a pressing global issue. Sensorineural hearing loss, the predominant type of hearing loss, is mainly due to damage to the inner ear along with a variety of pathologies including ischemia, noise, trauma, aging, and ototoxic drugs. In addition to genetic factors, oxidative stress has been identified as a common mechanism underlying several cochlear pathologies. The cochlea, which plays a major role in auditory function, requires high-energy metabolism and is, therefore, highly susceptible to oxidative stress, particularly in the mitochondria. Based on these pathological findings, the potential of antioxidants for the treatment of hearing loss has been demonstrated in several animal studies. However, results from human studies are insufficient, and future clinical trials are required. This review discusses the relationship between sensorineural hearing loss and reactive oxidative species (ROS), with particular emphasis on age-related hearing loss, noise-induced hearing loss, and ischemia–reperfusion injury. Based on these mechanisms, the current status and future perspectives of ROS-targeted therapy for sensorineural hearing loss are described. Full article
(This article belongs to the Special Issue Oxidative Stress and Antioxidants in Human Diseases)
Show Figures

Figure 1

Figure 1
<p>Summary figure of mitochondrial oxidative stress in relation to hearing loss. When the balance between the production and elimination of ROS is disrupted, cellular physiology is affected. Mitochondria play an important role in ROS production. Excessive levels of ROS, caused by external factors such as noise or ototoxic drugs, compromise the antioxidant defenses of hair cells, induce apoptosis, and cause inflammation, resulting in permanent cochlear degeneration.</p>
Full article ">
14 pages, 3993 KiB  
Article
Lercanidipine’s Antioxidative Effect Prevents Noise-Induced Hearing Loss
by Zhaoqi Guo, E Tian, Sen Chen, Jun Wang, Jingyu Chen, Weijia Kong, Debbie C. Crans, Yisheng Lu and Sulin Zhang
Antioxidants 2024, 13(3), 327; https://doi.org/10.3390/antiox13030327 - 7 Mar 2024
Cited by 1 | Viewed by 1698
Abstract
Noise-induced hearing loss (NIHL) is a prevalent form of adult hearing impairment, characterized by oxidative damage to auditory sensory hair cells. Although certain dihydropyridines, the L-type calcium channel blockers, exhibit protective properties against such damage, the ability of third-generation dihydropryidines like lercanidipine to [...] Read more.
Noise-induced hearing loss (NIHL) is a prevalent form of adult hearing impairment, characterized by oxidative damage to auditory sensory hair cells. Although certain dihydropyridines, the L-type calcium channel blockers, exhibit protective properties against such damage, the ability of third-generation dihydropryidines like lercanidipine to mitigate NIHL remains unclear.We utilized glucose oxidase (GO)-treated OC1 cell lines and cochlear explants to evaluate the protective influence of lercanidipine on hair cells. To further investigate its effectiveness, we exposed noise-stimulated mice in vivo and analyzed their hearing thresholds. Additionally, we assessed the antioxidative capabilities of lercanidipine by examining oxidation-related enzyme expression and levels of oxidative stress markers, including 3-nitrotyrosine (3NT) and 4-hydroxynonenal (4HNE). Our findings demonstrate that lercanidipine significantly reduces the adverse impacts of GO on both OC-1 cell viability (0.3 to 2.5 µM) and outer hair cell (OHC) survival in basal turn cochlear explants (7 µM). These results are associated with increased mRNA expression of antioxidant enzyme genes (HO-1, SOD1/2, and Txnrd1), along with decreased expression of oxidase genes (COX-2, iNOS). Crucially, lercanidipine administration prior to, and following, noise exposure effectively ameliorates NIHL, as evidenced by lowered hearing thresholds and preserved OHC populations in the basal turn, 14 days post-noise stimulation at 110 dB SPL. Moreover, our observations indicate that lercanidipine’s antioxidative action persists even three days after simultaneous drug and noise treatments, based on 3-nitrotyrosine and 4-hydroxynonenal immunostaining in the basal turn. Based on these findings, we propose that lercanidipine has the capacity to alleviate NIHL and safeguard OHC survival in the basal turn, potentially via its antioxidative mechanism. These results suggest that lercanidipine holds promise as a clinically viable option for preventing NIHL in affected individuals. Full article
(This article belongs to the Special Issue Oxidative Stress in Hearing Loss)
Show Figures

Figure 1

Figure 1
<p>Lercanidipine alleviates the detrimental effects of GO on OC-1 cell viability. (<b>A</b>,<b>B</b>) The dose-dependence of OC-1 viability on GO and lercanidipine, where the half-maximal inhibitory concentration (IC50) was 7.5 U/L and 4.52 μM, respectively. (<b>C</b>) 0.3 to 2.5 μM lercanidipine alleviated the cytotoxity of GO treatment (24 h, 10 U/L) on OC-1 cell, while 5 to 10 μM lercanidipine did not provide any protection. (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001. <span class="html-italic">n</span> = 3).</p>
Full article ">Figure 2
<p>Lercanidipine reduces GO-induced OHC loss in the cochlear explant. (<b>A</b>) After 24 h culturing, cochlear explants were treated without GO, with GO or GO + lercanidipine for 8 h. Representative images of OHC and IHC from the three cochlea turns labeled with phalloidin (red). White triangles indicate cell loss. No impairment was observed in the IHC (<b>B</b>), while lercanidipine reversed the GO-induced OHC loss (<b>C</b>). (** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001. <span class="html-italic">n</span> = 4 mice).</p>
Full article ">Figure 3
<p>Lercanidipine inhibits oxidative and promotes antioxidative enzyme gene mRNA expression after GO treatment of cochlea explant. The mRNA levels of oxidative-related genes, Cox-2 and iNOS increased in the GO group, which could be reversed by lercanidipine (<b>A</b>,<b>B</b>). The mRNA levels of antioxidative-related genes, HO-1, SOD1, SOD2, and Txnrd1, increased in the GO + lercanidipine group, compared to the GO group (<b>C</b>–<b>F</b>). (* <span class="html-italic">p</span> &lt; 0.05, ns: no significance, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001. <span class="html-italic">n</span> = 3).</p>
Full article ">Figure 4
<p>Lercanidipine attenuates noise-induced hearing loss. Fourteen days after one-hour noise exposure, the hearing threshold in the noise group was elevated, which was partially reversed by lercanidipine treatment (i.p. injection, 6 mg/kg, once before noise and once a day in post-noise 1, 2, 3 days). (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001. <span class="html-italic">n</span> = 3).</p>
Full article ">Figure 5
<p>Lercanidipine attenuates noise-induced hair cell loss in OHC. (<b>A</b>) Representative images showing OHC from the three turns of the cochlea labeled with phalloidin (red) and myosin 7a (green) 14 days after 1 h noise exposure with (noise + lercanidipine group) or without (noise group) lercanidipine i.p. injection (6 mg/kg) 1 h in advance. Arrowheads indicate the missing hair cells in three turns. (<b>B</b>) No difference was observed in the IHC survival rate 14 days after noise exposure. (<b>C</b>) Lercanidipine reversed noise-induced OHC loss in the basal turn. (**** <span class="html-italic">p</span> &lt; 0.0001. <span class="html-italic">n</span> = 3).</p>
Full article ">Figure 6
<p>Lercanidipine reduces oxidative stress in the OHC induced by noise. 3-NT (<b>A</b>) or 4-HNE (<b>C</b>) were coimmunostained with phalloidin in the basal turn OHCs 3 d after noise exposure with (treatment group, once before noise and once a day in post-noise 1, 2, 3 days) or without (noise group) i.p. lercanidipine injection (6 mg/kg). Quantification of 3-NT (<b>B</b>) and 4-HNE (<b>D</b>) fluorescent intensity in OHCs showed a significant decrease in the noise + lercanidipine groups. (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001. <span class="html-italic">n</span> = 3 mice).</p>
Full article ">Figure 7
<p>Chemical structures of the lercanidipine.</p>
Full article ">
13 pages, 923 KiB  
Article
Association of Head Injury, Neck Injury or Acoustic Trauma on Phenotype of Ménière’s Disease
by Ilmari Pyykkö, Vinay, Artur Vetkas, Jing Zou and Vinaya Manchaiah
Audiol. Res. 2024, 14(1), 204-216; https://doi.org/10.3390/audiolres14010019 - 17 Feb 2024
Cited by 1 | Viewed by 1624
Abstract
The aim of the present study was to investigate adverse effects of head injury, neck trauma, and chronic noise exposure on the complaint profile in people with Ménière’s disease (MD). The study used a retrospective design. Register data of 912 patients with MD [...] Read more.
The aim of the present study was to investigate adverse effects of head injury, neck trauma, and chronic noise exposure on the complaint profile in people with Ménière’s disease (MD). The study used a retrospective design. Register data of 912 patients with MD from the Finnish Ménière Federation database were studied. The data comprised case histories of traumatic brain injury (TBI), neck trauma and occupational noise exposure, MD specific complaints, impact related questions, and the E-Qol health-related quality of life instrument. TBI was classified based on mild, moderate, and severe categories of transient loss of consciousness (TLoC). The mean age of the participants was 60.2 years, the mean duration of the disease was 12.6 years, and 78.7% were females. Logistic regression analysis, linear correlation, and pairwise comparisons were used in evaluating the associations. 19.2% of the participants with MD had a history of TBI. The phenotype of participants with TBI was associated with frequent vestibular drop attacks (VDA), presyncope, headache-associated vertigo, and a reduction in the E-QoL. Logistic regression analysis explained the variability of mild TBI in 6.8%. A history of neck trauma was present in 10.8% of the participants. Neck trauma associated with vertigo (NTwV) was seen in 47 and not associated with vertigo in 52 participants. The phenotype of NTwV was associated with balance problems, VDA, physical strain-induced vertigo, and hyperacusia. Logistic regression analysis explained 8.7% of the variability of the complaint profile. Occupational noise exposure was recorded in 25.4% of the participants and correlated with the greater impact of tinnitus, hyperacusis, and hearing loss. Neither the frequency, duration, or severity of vertigo or nausea were significantly different between the baseline group and the TBI, NTwV, or noise-exposure groups. The results indicate that TBI and NTwV are common among MD patients and may cause a confounder effect. Full article
Show Figures

Figure 1

Figure 1
<p>Frequency of vestibular drop attacks (VDA) in subjects with TBI and in the control group (<b>upper</b>) and of presyncope in subjects with TBI and the control group (<b>lower</b>).</p>
Full article ">Figure 2
<p>Health-related quality of life (E-QoL-5D-3L) in MD and severity of TBI.</p>
Full article ">Figure 3
<p>Health-related quality of life (E-QoL-5D-3L) among study participants with and without NTwV.</p>
Full article ">Figure 4
<p>Health-related quality of life (E-QoL-5D-3L) among study participants with and without noise exposure.</p>
Full article ">
Back to TopTop