1
|
Newton S, Aguilar C, Bowl MR. C57BL/6-derived mice and the Cdh23 ahl allele - Background matters. Hear Res 2025; 462:109278. [PMID: 40305983 DOI: 10.1016/j.heares.2025.109278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/31/2025] [Accepted: 04/09/2025] [Indexed: 05/02/2025]
Abstract
C57BL/6-derived mice are the most utilised mice in biomedical research, and yet actually there is no such thing as a generic C57BL/6 mouse. Instead, there are more than 150 C57BL/6-derived sub-strains recognised by the Mouse Genome Informatics (MGI) database, each of which carry sub-strain-specific fixed genetic differences that can potentially lead to phenotypic differences affecting a single, or multiple biological systems. One of the most widely known strain-specific alleles is the Cdh23ahl allele, a single nucleotide change that predisposes C57BL/6-derived mice to a progressive hearing loss that starts in the high-frequency region. As such, this allele is of particular relevance to auditory researchers. However, a recent study, comparing C57BL/6NTac mice with a co-isogenic strain in which the Cdh23ahl allele has been 'repaired' using genome editing, suggests that the Cdh23ahl allele may have a broader effect on phenotype expressivity of mouse mutants impacting not just the auditory system, but other organ systems as well. Here, using the Cdh23ahl allele as an exemplar, we discuss the importance of knowing, understanding and reporting the genetic background of mouse mutants.
Collapse
Affiliation(s)
- Sherylanne Newton
- UCL Ear Institute, University College London, 332 Gray's Inn Road, London WC1 × 8EE, United Kingdom
| | - Carlos Aguilar
- UCL Ear Institute, University College London, 332 Gray's Inn Road, London WC1 × 8EE, United Kingdom
| | - Michael R Bowl
- UCL Ear Institute, University College London, 332 Gray's Inn Road, London WC1 × 8EE, United Kingdom.
| |
Collapse
|
2
|
Quiñones PM, Pei M, Srivastava H, Cobo-Cuan A, Morán MA, Kim BJ, Walker CB, Serafino MJ, Macias-Escriva F, Wang J, Dewey JB, Applegate BE, McGinley MJ, Oghalai JS. The Medial Olivocochlear Efferent Pathway Potentiates Cochlear Amplification in Response to Hearing Loss. J Neurosci 2025; 45:e2103242025. [PMID: 39984203 PMCID: PMC11984096 DOI: 10.1523/jneurosci.2103-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/04/2025] [Accepted: 02/11/2025] [Indexed: 02/23/2025] Open
Abstract
The mammalian cochlea receives efferent feedback from the brain. Many functions for this feedback have been hypothesized, including on short timescales, such as mediating attentional states, and long timescales, such as buffering acoustic trauma. Testing these hypotheses has been impeded by an inability to make direct measurements of efferent effects in awake animals. Here, we assessed the role of the medial olivocochlear (MOC) efferent nerve fibers on cochlear amplification by measuring organ of Corti vibratory responses to sound in both sexes of awake and anesthetized mice. We studied long-term effects by genetically ablating the efferents and/or afferents. Cochlear amplification increased with deafferentation using VGLUT3-/- mice, but only when the efferents were intact, associated with increased activity within OHCs and supporting cells. Removing both the afferents and the efferents using VGLUT3-/- Alpha9-/- mice did not cause this effect. To test for short-term effects, we recorded sound-evoked vibrations while using pupillometry to measure neuromodulatory brain state. We found no state dependence of cochlear amplification or of the auditory brainstem response. However, state dependence was apparent in the downstream inferior colliculus. Thus, MOC efferents upregulate cochlear amplification chronically with hearing loss, but not acutely with brain state fluctuations. This pathway may partially compensate for hearing loss while mediating associated symptoms, such as tinnitus and hyperacusis.
Collapse
Affiliation(s)
- Patricia M Quiñones
- Caruso Department of Otolaryngology - Head and Neck Surgery, University of Southern California, Los Angeles, California 90033
| | - Michelle Pei
- Caruso Department of Otolaryngology - Head and Neck Surgery, University of Southern California, Los Angeles, California 90033
| | - Hemant Srivastava
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Duncan Neurological Research Institute, Texas Children's Hospital, Texas 77030
| | - Ariadna Cobo-Cuan
- Caruso Department of Otolaryngology - Head and Neck Surgery, University of Southern California, Los Angeles, California 90033
| | - Marcela A Morán
- Caruso Department of Otolaryngology - Head and Neck Surgery, University of Southern California, Los Angeles, California 90033
| | - Bong Jik Kim
- Caruso Department of Otolaryngology - Head and Neck Surgery, University of Southern California, Los Angeles, California 90033
- Department of Otolaryngology - Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon 35015, Republic of Korea
| | - Clayton B Walker
- Caruso Department of Otolaryngology - Head and Neck Surgery, University of Southern California, Los Angeles, California 90033
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843
| | - Michael J Serafino
- Caruso Department of Otolaryngology - Head and Neck Surgery, University of Southern California, Los Angeles, California 90033
| | - Frank Macias-Escriva
- Caruso Department of Otolaryngology - Head and Neck Surgery, University of Southern California, Los Angeles, California 90033
| | - Juemei Wang
- Caruso Department of Otolaryngology - Head and Neck Surgery, University of Southern California, Los Angeles, California 90033
| | - James B Dewey
- Caruso Department of Otolaryngology - Head and Neck Surgery, University of Southern California, Los Angeles, California 90033
| | - Brian E Applegate
- Caruso Department of Otolaryngology - Head and Neck Surgery, University of Southern California, Los Angeles, California 90033
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089
| | - Matthew J McGinley
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Duncan Neurological Research Institute, Texas Children's Hospital, Texas 77030
| | - John S Oghalai
- Caruso Department of Otolaryngology - Head and Neck Surgery, University of Southern California, Los Angeles, California 90033
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089
| |
Collapse
|
3
|
Diuba A, Gratias P, Jeffers PWC, Nouvian R, Puel JL, Kujawa SG, Bourien J. Phenotypic changes of auditory nerve fibers after excitotoxicity. Proc Natl Acad Sci U S A 2025; 122:e2412332122. [PMID: 40168123 PMCID: PMC12002199 DOI: 10.1073/pnas.2412332122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 02/25/2025] [Indexed: 04/03/2025] Open
Abstract
There is a substantial body of evidence elucidating the pathophysiological aspects of excitotoxicity in the mammalian cochlea. However, the question of whether the resultant damage is reversible remains unresolved. To replicate an excitotoxic event, we investigated the long-term effects of kainate application in gerbil cochleae. Surprisingly, despite persistent synapse loss, the compound action potential of the auditory nerve fully recovered. This functional retrieval was associated with a phenotypic change in auditory nerve fibers. Thresholds were improved along the tonotopic axis. High-spontaneous rate (SR) fibers largely populated the apical region, while low-SR fibers from the basal region exhibited sound-driven activity indistinguishable from control high-SR fibers. This functional phenotype change may support the full recovery of neural response thresholds and amplitudes after excitotoxicity. Furthermore, hyperresponsiveness of the auditory nerve fibers could be a crucial factor in the development of hyperactivity in the central auditory pathways, a common occurrence following acoustic overstimulation.
Collapse
Affiliation(s)
- Artem Diuba
- Institute for Neuroscience of Montpellier, University of Montpellier, INSERM, Montpellier34000, France
| | - Paul Gratias
- Institute for Neuroscience of Montpellier, University of Montpellier, INSERM, Montpellier34000, France
| | - Penelope W. C. Jeffers
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA02114
- Division of Medical Sciences, Program in Speech and Hearing Bioscience and Technology, Harvard University, Boston, MA02115
| | - Régis Nouvian
- Institute for Neuroscience of Montpellier, University of Montpellier, INSERM, Montpellier34000, France
| | - Jean-Luc Puel
- Institute for Neuroscience of Montpellier, University of Montpellier, INSERM, Montpellier34000, France
- Audiocampus, University of Montpellier, Montpellier34000, France
| | - Sharon G. Kujawa
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA02114
- Division of Medical Sciences, Program in Speech and Hearing Bioscience and Technology, Harvard University, Boston, MA02115
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA02115
| | - Jérôme Bourien
- Institute for Neuroscience of Montpellier, University of Montpellier, INSERM, Montpellier34000, France
- Audiocampus, University of Montpellier, Montpellier34000, France
| |
Collapse
|
4
|
Schvartz-Leyzac KC, McClaskey CM, Dias JW, Pfingst BE, Harris KC. Contributions of Auditory Nerve Density and Synchrony to Speech Understanding in Older Cochlear Implant Users. J Assoc Res Otolaryngol 2025:10.1007/s10162-025-00984-3. [PMID: 40183851 DOI: 10.1007/s10162-025-00984-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/19/2025] [Indexed: 04/05/2025] Open
Abstract
PURPOSE The majority of adult cochlear implant (CI) recipients are over the age of 65, and previous research in non-implanted older adults shows that auditory nerve (AN) pathophysiology contributes to senescent declines in speech understanding. However, age-related changes to AN structure and function have not yet been explored as a contributory factor to poorer speech understanding outcomes in older CI users. Here, we explore how estimates of AN disengagement (i.e., AN density) and dyssynchrony in CI users contribute to poorer speech recognition performance observed in older CI users. METHODS We examined electrically evoked compound action potentials (ECAPs) in 47 adult (Male = 25) CI recipients. We measured the interphase gap (IPG) effect for the amplitude-growth function (AGF) slope and the N1-P2 interpeak latency as independent metrics of AN density and dyssynchrony, respectively. RESULTS Estimates of AN density and dyssynchrony worsen with increasing age in older CI listeners. These measures were not significantly correlated with one another, but were independently related to speech recognition in noise performance. Lower ECAP IPG effect values (lower density of AN fibers) are observed in older CI users. Longer N1-P2 interpeak latency values (poorer neural synchrony) are also observed in older CI users. When controlling for listener age, poorer AN dyssynchrony contributes to declines in speech-recognition-in-noise performance in CI users. CONCLUSION These results suggest that AN dyssynchrony rather than density is the primary contributing factor to age-related declines in speech understanding in CI users. These results have important implications for better understanding neural contributions to speech understanding in adult CI users.
Collapse
Affiliation(s)
- Kara C Schvartz-Leyzac
- Department of Otolaryngology-Head & Neck Surgery, Medical University of South Carolina, Charleston, SC, 29425, USA.
| | - Carolyn M McClaskey
- Department of Otolaryngology-Head & Neck Surgery, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - James W Dias
- Department of Otolaryngology-Head & Neck Surgery, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Bryan E Pfingst
- Department of Otolaryngology-Head & Neck Surgery, Medical Science II, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kelly C Harris
- Department of Otolaryngology-Head & Neck Surgery, Medical University of South Carolina, Charleston, SC, 29425, USA
| |
Collapse
|
5
|
Erra A, Chen J, Miller CM, Chrysostomou E, Barret S, Kassim YM, Friedman RA, Lauer A, Ceriani F, Marcotti W, Carroll C, Manor U. An Open-Source Deep Learning-Based GUI Toolbox for Automated Auditory Brainstem Response Analyses (ABRA). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.06.20.599815. [PMID: 38948763 PMCID: PMC11213013 DOI: 10.1101/2024.06.20.599815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Hearing loss is a pervasive global health challenge with profound impacts on communication, cognitive function, and quality of life. Recent studies have established age-related hearing loss as a significant risk factor for dementia, highlighting the importance of hearing loss research. Auditory brainstem responses (ABRs), which are electrophysiological recordings of synchronized neural activity from the auditory nerve and brainstem, serve as in vivo readouts for sensory hair cell, synaptic integrity, hearing sensitivity, and other key features of auditory pathway functionality, making them highly valuable for both basic neuroscience research and clinical diagnostics. Despite their utility, traditional ABR analyses rely heavily on subjective manual interpretation, leading to considerable variability and limiting reproducibility across studies. Here, we introduce Auditory Brainstem Response Analyzer (ABRA), a novel open-source graphical user interface powered by deep learning, which automates and standardizes ABR waveform analysis. ABRA employs convolutional neural networks trained on diverse datasets collected from multiple experimental settings, achieving rapid and unbiased extraction of key ABR metrics, including peak amplitude, latency, and auditory threshold estimates. We demonstrate that ABRA's deep learning models provide performance comparable to expert human annotators while dramatically reducing analysis time and enhancing reproducibility across datasets from different laboratories. By bridging hearing research, sensory neuroscience, and advanced computational techniques, ABRA facilitates broader interdisciplinary insights into auditory function. An online version of the tool is available for use at no cost at https://abra.ucsd.edu.
Collapse
Affiliation(s)
- Abhijeeth Erra
- Data Institute, University of San Francisco, San Francisco, CA
| | - Jeffrey Chen
- Data Institute, University of San Francisco, San Francisco, CA
| | - Cayla M. Miller
- Dept. of Cell & Developmental Biology, University of California San Diego, La Jolla, CA
| | - Elena Chrysostomou
- Dept. of Cell & Developmental Biology, University of California San Diego, La Jolla, CA
| | - Shannon Barret
- Dept. of Cell & Developmental Biology, University of California San Diego, La Jolla, CA
| | - Yasmin M. Kassim
- Dept. of Cell & Developmental Biology, University of California San Diego, La Jolla, CA
| | - Rick A. Friedman
- Dept. of Otolaryngology, University of California San Diego, La Jolla, CA
| | - Amanda Lauer
- Depts. of Otolaryngology-Head and Neck Surgery and Neuroscience and Center for Functional Anatomy and Evolution, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Federico Ceriani
- School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Walter Marcotti
- School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Cody Carroll
- Data Institute, University of San Francisco, San Francisco, CA
- Dept. of Mathematics and Statistics, University of San Francisco, San Francisco, CA
| | - Uri Manor
- Dept. of Cell & Developmental Biology, University of California San Diego, La Jolla, CA
- Dept. of Otolaryngology, University of California San Diego, La Jolla, CA
- Halıcıoğlu Data Science Institute, University of California San Diego, La Jolla, CA
| |
Collapse
|
6
|
Agboola OS, Deng M, Hu Z. In vitro generation of spiral ganglion neurons from embryonic stem cells. Hum Cell 2025; 38:68. [PMID: 40069509 DOI: 10.1007/s13577-025-01194-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/26/2025] [Indexed: 04/02/2025]
Abstract
Spiral ganglion neurons (SGNs) are crucial for transmitting auditory signals from the inner ear to the brainstem, playing a pivotal role in the peripheral hearing process. However, SGNs are usually damaged by a variety of insults, which causes permanent hearing loss. Generating SGNs from stem cells represents a promising strategy for advancing cell-replacement therapies to treat sensorineural hearing loss. SGNs comprise two subtypes of neurons (types 1 and 2); however, it remains a challenge to regenerate SGN subtypes. This study aimed to investigate the generation and characterization of SGN subtype neurons induced from embryonic stem cells (ESCs) in vitro. ESCs were cultured and treated with retinoic acid, followed by neuronal induction. The differentiated cells showed protein expressions of multiple neuronal markers, suggesting the generation of neuron-like cells. Protein expressions of vGlut-1 and GATA-3 indicate the generation of glutamatergic otic neuron-like cells. ESC-derived neuron-like cells cultured for 6 days showed co-expressions of calretinin, calbindin, and POU4F1 antibodies, suggesting an early stage of SGN subtype induction. However, 14-day in vitro induction generated cells showing two distinct SGN subtypes: a group of cells expressed calretinin (subtype 1a/2 precursor), and the other group expressed calbindin and POU4F1 (subtype 1b/c). These results suggest that in vitro generation of SGN subtypes from ESCs is culture time dependent.
Collapse
Affiliation(s)
- Oluwafemi S Agboola
- Department of Otolaryngology-HNS, Wayne State University School of Medicine, Detroit, MI, USA
- John D Dingell VA Medical Center, Detroit, MI, USA
| | - Meng Deng
- Department of Otolaryngology-HNS, Wayne State University School of Medicine, Detroit, MI, USA
- John D Dingell VA Medical Center, Detroit, MI, USA
| | - Zhengqing Hu
- Department of Otolaryngology-HNS, Wayne State University School of Medicine, Detroit, MI, USA.
- John D Dingell VA Medical Center, Detroit, MI, USA.
| |
Collapse
|
7
|
Dillard LK, Humes LE, Matthews LJ, Dubno JR. Noise Exposure History and Age-Related Changes to Hearing. JAMA Otolaryngol Head Neck Surg 2025; 151:228-235. [PMID: 39786765 PMCID: PMC11907306 DOI: 10.1001/jamaoto.2024.4768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 11/10/2024] [Indexed: 01/12/2025]
Abstract
Importance Noise exposure is a major modifiable risk factor for hearing loss, yet it is not known whether it affects the rate of hearing decline in aging. Objective To determine the association of noise exposure history with the rate of pure-tone threshold change per year. Design, Setting, and Participants This longitudinal cohort study was conducted in the ongoing community-based Medical University of South Carolina Longitudinal Cohort Study of Age-Related Hearing Loss (1988 to present with the sample based in Charleston, South Carolina, and surrounding area). Following a comprehensive baseline examination, participants attended annual examinations, during which audiometric data were collected. Participants with audiometric data from at least 2 examinations and noise exposure history data were included in the study. Data were analyzed between September 2023 and July 2024. Exposure Noise exposure history, determined by a self-reported questionnaire and history of military service, was categorized as no/little, some, or high exposure. Main Outcomes and Measures Outcome measures were individual audiometric thresholds (0.25 kHz to 8.0 kHz) and pure-tone average (PTA) of thresholds at frequencies 0.5 kHz, 1.0 kHz, 2.0 kHz, and 4.0 kHz, averaged bilaterally. Linear mixed regression models were used to estimate the association of age (per every 1 additional year) with the rate of threshold change at each frequency and PTA, for each noise exposure category. The association of noise exposure with the rate of annual threshold change was determined by an interaction term of age (longitudinal time variable) and noise exposure in regression models. Results Of 1347 participants, the mean (SD) baseline age was 63 (14) years, and 772 (57%) were female. The mean (SD) follow-up time was 5.1 (5.7) years. Compared to the no/little noise exposure group, groups with some and high noise exposure had significantly higher baseline thresholds from 2.0 kHz to 8.0 kHz and PTA, and 1.0 kHz to 8.0 kHz and PTA, respectively. Those with high noise exposure (vs no/little) showed higher rates of threshold change per year at 1.0 kHz and 2.0 kHz. Participants with some and high noise exposure showed lower rates of change per year at 3.0 kHz to 8.0 kHz and 4.0 kHz to 8.0 kHz, respectively, where hearing loss had already occurred. The rate of PTA change per year did not differ across noise exposure groups. Conclusions and Relevance In this cohort study, noise exposure was associated with poorer baseline hearing and higher rates of annual decline at some midfrequencies. Noise exposure can have immediate and potentially long-term negative impacts on hearing.
Collapse
Affiliation(s)
- Lauren K. Dillard
- Department of Otolaryngology–Head and Neck Surgery, Medical University of South Carolina, Charleston
| | - Larry E. Humes
- Department of Speech, Language and Hearing Sciences, Indiana University, Bloomington
| | - Lois J. Matthews
- Department of Otolaryngology–Head and Neck Surgery, Medical University of South Carolina, Charleston
| | - Judy R. Dubno
- Department of Otolaryngology–Head and Neck Surgery, Medical University of South Carolina, Charleston
| |
Collapse
|
8
|
Jiang D, Huang XH, Fang K, Zhao MH, Li Y, Hu HT, Fang LQ, Gao H, Zhou J. Peripheral-central correlation study of acupuncture for chronic tinnitus study protocol for a randomized controlled trial. Front Med (Lausanne) 2025; 12:1543023. [PMID: 40018352 PMCID: PMC11865056 DOI: 10.3389/fmed.2025.1543023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 02/03/2025] [Indexed: 03/01/2025] Open
Abstract
Purpose (1) Exploring the evaluation and correlation of peripheral central auditory function in patients with chronic tinnitus. (2) Evaluation of the cumulative effect of acupuncture on peripheral central auditory function in patients with chronic tinnitus. Method Our research is structured as a regulated and randomized trial with assessor blinding. Seventy-two participants who qualify with chronic tinnitus will be allocated in a 1:1 ratio to either the acupuncture group or the sham acupuncture group. Additionally, we will recruit 15 healthy individuals as subjects for data collection to observe the correlation of peripheral-central auditory function under different physiological states. Result Clinical result metrics encompass the Tinnitus Handicap Inventory (THI), ABR testing, and fNIRS data collection. Evaluations will be carried out at baseline, after 10 treatment sessions. Conclusion This research are anticipated to improve our comprehension of the effectiveness and fundamental processes of acupuncture in addressing persistent tinnitus and deeply explain the mechanism of action of the acupuncture method on chronic tinnitus. Clinical trial registration ClinicalTrials.gov, identifier NCT06401993.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hong Gao
- Department of Acupuncture and Moxibustion, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jie Zhou
- Department of Acupuncture and Moxibustion, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
9
|
Garrett M, Vasilkov V, Mauermann M, Devolder P, Wilson JL, Gonzales L, Henry KS, Verhulst S. Deciphering Compromised Speech-in-Noise Intelligibility in Older Listeners: The Role of Cochlear Synaptopathy. eNeuro 2025; 12:ENEURO.0182-24.2024. [PMID: 39788732 PMCID: PMC11842038 DOI: 10.1523/eneuro.0182-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 11/29/2024] [Accepted: 12/16/2024] [Indexed: 01/12/2025] Open
Abstract
Speech intelligibility declines with age and sensorineural hearing damage (SNHL). However, it remains unclear whether cochlear synaptopathy (CS), a recently discovered form of SNHL, significantly contributes to this issue. CS refers to damaged auditory-nerve synapses that innervate the inner hair cells and there is currently no go-to diagnostic test available. Furthermore, age-related hearing damage can comprise various aspects (e.g., hair cell damage, CS) that each can play a role in impaired sound perception. To explore the link between cochlear damage and speech intelligibility deficits, this study examines the role of CS for word recognition among older listeners. We first validated an envelope-following response (EFR) marker for CS using a Budgerigar model. We then applied this marker in human experiments, while restricting the speech material's frequency content to ensure that both the EFR and the behavioral tasks engaged similar cochlear frequency regions. Following this approach, we identified the relative contribution of hearing sensitivity and CS to speech intelligibility in two age-matched (65-year-old) groups with clinically normal (n = 15, 8 females) or impaired audiograms (n = 13, 8 females). Compared to a young normal-hearing control group (n = 13, 7 females), the older groups demonstrated lower EFR responses and impaired speech reception thresholds. We conclude that age-related CS reduces supra-threshold temporal envelope coding with subsequent speech coding deficits in noise that cannot be explained based on hearing sensitivity alone.
Collapse
Affiliation(s)
- Markus Garrett
- Medizinische Physik and Cluster of Excellence "Hearing4all", Department of Medical Physics and Acoustics, Carl von Ossietzky University of Oldenburg, Oldenburg 26129, Germany
| | - Viacheslav Vasilkov
- Hearing Technology @ WAVES, Department of Information Technology, Ghent University, Zwijnaarde 9052, Belgium
| | - Manfred Mauermann
- Medizinische Physik and Cluster of Excellence "Hearing4all", Department of Medical Physics and Acoustics, Carl von Ossietzky University of Oldenburg, Oldenburg 26129, Germany
| | - Pauline Devolder
- Hearing Technology @ WAVES, Department of Information Technology, Ghent University, Zwijnaarde 9052, Belgium
| | - John L Wilson
- Department of Otolaryngology, University of Rochester, Rochester, New York 14642
- Department of Neuroscience, University of Rochester, Rochester, New York 14642
| | - Leslie Gonzales
- Department of Neuroscience, University of Rochester, Rochester, New York 14642
| | - Kenneth S Henry
- Department of Otolaryngology, University of Rochester, Rochester, New York 14642
- Department of Neuroscience, University of Rochester, Rochester, New York 14642
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627
| | - Sarah Verhulst
- Hearing Technology @ WAVES, Department of Information Technology, Ghent University, Zwijnaarde 9052, Belgium
| |
Collapse
|
10
|
Sitko AA, Frank MM, Romero GE, Hunt M, Goodrich LV. Lateral olivocochlear neurons modulate cochlear responses to noise exposure. Proc Natl Acad Sci U S A 2025; 122:e2404558122. [PMID: 39854232 PMCID: PMC11789013 DOI: 10.1073/pnas.2404558122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 12/04/2024] [Indexed: 01/26/2025] Open
Abstract
The sense of hearing originates in the cochlea, which detects sounds across dynamic sensory environments. Like other peripheral organs, the cochlea is subjected to environmental insults, including loud, damage-inducing sounds. In response to internal and external stimuli, the central nervous system directly modulates cochlear function through olivocochlear neurons (OCNs), which are located in the brainstem and innervate the cochlear sensory epithelium. One population of OCNs, the lateral olivocochlear (LOC) neurons, target spiral ganglion neurons (SGNs), the primary sensory neurons of the ear. LOCs alter their transmitter expression for days to weeks in response to noise exposure (NE), suggesting that they could tune SGN excitability over long time periods in response to auditory experience. To examine how LOCs affect auditory function after NE, we characterized OCN transcriptional profiles and found transient LOC-specific gene expression changes after NE, including upregulation of multiple neuropeptide-encoding genes. Next, by generating intersectional mouse lines that selectively target LOCs, we chemogenetically ablated LOCs and assayed auditory responses at baseline and after NE. Compared to controls, mice with reduced LOC innervation showed greater NE-induced functional deficits 1 d later and had worse auditory function after a 2-wk recovery period. The number of remaining presynaptic puncta at the SGN synapse with inner hair cells did not differ between control and LOC-ablated animals, suggesting that the primary role of LOCs after NE is likely not to protect but instead to compensate, ensuring that SGN function is enhanced during periods of need.
Collapse
Affiliation(s)
- Austen A. Sitko
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | | | | | - Mackenzie Hunt
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | - Lisa V. Goodrich
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| |
Collapse
|
11
|
Oestreicher D, Malpede AM, Reitmeier A, Bräuer CP, Schoch L, Strenzke N, Pangrsic T. Noise-induced ribbon synapse loss in the mouse basal cochlear region does not reduce inner hair cell exocytosis. Front Cell Neurosci 2025; 18:1523978. [PMID: 39839350 PMCID: PMC11747652 DOI: 10.3389/fncel.2024.1523978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 12/20/2024] [Indexed: 01/23/2025] Open
Abstract
Noise-induced hearing loss is one of the most common forms of hearing loss in adults and also one of the most common occupational diseases. Extensive previous work has shown that the highly sensitive synapses of the inner hair cells (IHCs) may be the first target for irreparable damage and permanent loss in the noise-exposed cochlea, more precisely in the cochlear base. However, how such synaptic loss affects the synaptic physiology of the IHCs in this particularly vulnerable part of the cochlea has not yet been investigated. To address this question, we exposed 3-4-week-old C57BL/6J mice to 8-16 kHz noise for 2 h under isoflurane anesthesia. We then employed hearing measurements, immunohistochemistry and patch-clamp to assess IHC synaptic function. Two noise sound pressure levels (SPLs) were used to evoke acute hearing threshold elevations with different levels of recovery 2 weeks post-exposure. Regardless of noise intensity, the exposure resulted in a loss of approximately 25-36% of ribbon synapses in the basal portions of the cochlea that persisted 2 weeks after exposure. Perforated patch-clamp recordings were made in the IHCs of the basal regions of the cochlea where the greatest synaptic losses were observed. Depolarization-evoked calcium currents in IHCs 2 weeks after exposure were slightly but not significantly smaller as compared to controls from age-matched non-exposed animals. Exocytic changes monitored as changes in membrane capacitance did not follow that trend and remained similar to controls despite significant loss of ribbons, likely reflecting increased exocytosis at the remaining synapses. Additionally, we report for the first time that acute application of isoflurane reduces IHC calcium currents, which may have implications for noise-induced IHC synaptic loss.
Collapse
Affiliation(s)
- David Oestreicher
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Alfonso Mauro Malpede
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Annalena Reitmeier
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Carolin Paula Bräuer
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Laura Schoch
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Nicola Strenzke
- Auditory Systems Physiology Group, Institute for Auditory Neuroscience, InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
| | - Tina Pangrsic
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
| |
Collapse
|
12
|
Sun J, Sai N, Zhang T, Tang C, Fan S, Wang Q, Liu D, Zeng X, Li J, Guo W, Yang S, Han W. Repeated low-intensity noise exposure exacerbates age-related hearing loss via RAGE signaling pathway. Neurobiol Dis 2025; 204:106768. [PMID: 39694338 DOI: 10.1016/j.nbd.2024.106768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/11/2024] [Accepted: 12/11/2024] [Indexed: 12/20/2024] Open
Abstract
Repeated low-intensity noise exposure is prevalent in industrialized societies. It has long been considered risk-free until recent evidence suggests that the temporary threshold shift (TTS) induced by such exposure might be a high-risk factor for hearing loss. This study was conducted to further investigate the manner in which repeated low-intensity noise exposure contributed to hearing damage. Two-month-old C57BL/6 J mice were exposed to white noise at 96 dB SPL for 8 h per day over 7 days to induce TTS. Auditory brainstem response (ABR) was monitored to assess changes in hearing thresholds, tracking the effects of noise exposure until the mice reached 12 months of age. Our results indicated that noise-exposed mice exhibited accelerated age-related hearing loss spanning from high to low frequencies. Proteomics analysis revealed an upregulation in the receptor for the advanced glycation end-products (RAGE) signaling pathway, which was associated with an activated inflammatory response, vascular injury, and mitochondrial and synaptic dysfunction. Further analysis confirmed increased levels of inflammatory cytokines in the cochlear lymph fluid and significant macrophages infiltration in the cochlear lateral wall, accompanied by hyperpermeability of the blood-labyrinth barrier. Additionally, degenerated mitochondria in the outer hair cells and decreased synaptic ribbons in the inner hair cells were also observed. These pathological changes indicated that noise exposure damages the cochlear cellular components, increasing the cochlear susceptibility to age-related stress. Our findings suggest that TTS caused by repeated low-intensity noise exposure correlates with a severe sensorineural hearing loss during aging; targeting the RAGE signaling pathway may be a promising strategy to mitigate damage from low-intensity noise and slow down the progression of age-related hearing loss.
Collapse
Affiliation(s)
- Jianbin Sun
- Senior Department of Otorhinolaryngology Head and Neck Surgery, The 6th Medical Center, Chinese PLA General Hospital, Medical School of Chinese PLA, State Key Laboratory of Hearing and Balance Science, National Clinical Research Center for Otorhinolaryngologic Diseases, Beijing 100048, China; Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi Provincial Key Laboratory for Precision Diagnosis and Treatment of Otorhinolaryngology, Xi'an 710004, China
| | - Na Sai
- Senior Department of Otorhinolaryngology Head and Neck Surgery, The 6th Medical Center, Chinese PLA General Hospital, Medical School of Chinese PLA, State Key Laboratory of Hearing and Balance Science, National Clinical Research Center for Otorhinolaryngologic Diseases, Beijing 100048, China
| | - Tong Zhang
- Senior Department of Otorhinolaryngology Head and Neck Surgery, The 6th Medical Center, Chinese PLA General Hospital, Medical School of Chinese PLA, State Key Laboratory of Hearing and Balance Science, National Clinical Research Center for Otorhinolaryngologic Diseases, Beijing 100048, China
| | - Chaoying Tang
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Shuhang Fan
- Senior Department of Otorhinolaryngology Head and Neck Surgery, The 6th Medical Center, Chinese PLA General Hospital, Medical School of Chinese PLA, State Key Laboratory of Hearing and Balance Science, National Clinical Research Center for Otorhinolaryngologic Diseases, Beijing 100048, China
| | - Qin Wang
- Senior Department of Otorhinolaryngology Head and Neck Surgery, The 6th Medical Center, Chinese PLA General Hospital, Medical School of Chinese PLA, State Key Laboratory of Hearing and Balance Science, National Clinical Research Center for Otorhinolaryngologic Diseases, Beijing 100048, China
| | - Da Liu
- Senior Department of Otorhinolaryngology Head and Neck Surgery, The 6th Medical Center, Chinese PLA General Hospital, Medical School of Chinese PLA, State Key Laboratory of Hearing and Balance Science, National Clinical Research Center for Otorhinolaryngologic Diseases, Beijing 100048, China
| | - Xianhai Zeng
- Department of Otorhinolaryngology, Long gang Otorhinolaryngology Hospital, Shenzhen Key Laboratory of Otorhinolaryngology, Shenzhen, Guangdong 518172, China
| | - Juanjuan Li
- Department of Otorhinolaryngology, Long gang Otorhinolaryngology Hospital, Shenzhen Key Laboratory of Otorhinolaryngology, Shenzhen, Guangdong 518172, China
| | - Weiwei Guo
- Senior Department of Otorhinolaryngology Head and Neck Surgery, The 6th Medical Center, Chinese PLA General Hospital, Medical School of Chinese PLA, State Key Laboratory of Hearing and Balance Science, National Clinical Research Center for Otorhinolaryngologic Diseases, Beijing 100048, China.
| | - Shiming Yang
- Senior Department of Otorhinolaryngology Head and Neck Surgery, The 6th Medical Center, Chinese PLA General Hospital, Medical School of Chinese PLA, State Key Laboratory of Hearing and Balance Science, National Clinical Research Center for Otorhinolaryngologic Diseases, Beijing 100048, China.
| | - Weiju Han
- Senior Department of Otorhinolaryngology Head and Neck Surgery, The 6th Medical Center, Chinese PLA General Hospital, Medical School of Chinese PLA, State Key Laboratory of Hearing and Balance Science, National Clinical Research Center for Otorhinolaryngologic Diseases, Beijing 100048, China.
| |
Collapse
|
13
|
Perez P, Tsai TH, Hawks J, Barbone HM, Pinkl J, Thirumala P, Bao J. Hearing Loss in the Unoperated Ear After High-Speed Drilling in Otologic and Skull Base Surgery. Otol Neurotol 2024; 45:993-997. [PMID: 39207294 DOI: 10.1097/mao.0000000000004316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
OBJECTIVE To evaluate if permanent hearing loss occurred in the unoperated ear of patients undergoing otologic and skull base surgery with high-speed otologic drilling. STUDY DESIGN We retrospectively studied 250 patients (mean age 57.8 yr; 120 males, and 130 females) undergoing otologic or skull base surgery with high-speed drilling between 2013 and 2019. SETTING The University of Pittsburgh Medical Center. PATIENTS We evaluated preoperative and postoperative audiograms for patients undergoing surgery for cochlear implantation (95 patients, 38.0%), cholesteatoma or chronic ear disease (88 patients, 35.2%), repair of lateral skull base encephalocele (26 patients, 10.4%), resection of vestibular schwannoma or meningioma of the cerebellopontine angle (23 patients, 9.2%), lateral temporal bone resection (8 patients, 3.2%), microvascular decompression (7 patients, 2.8%), or other operations involving a high-speed otologic drill (3 patients, 1.2%). MAIN OUTCOME MEASURES Hearing threshold shift, measured as the difference between postoperative threshold and preoperative threshold for each frequency. The association of age, gender, tested frequency, and surgery type with hearing threshold shift was investigated with analysis of covariance. RESULTS A total of 102 patients (40.8%) had a 10-dB or greater worsening of their hearing in at least one frequency on their postoperative audiogram in the contralateral, unoperated ear. One hundred six subjects (42.4%) had no change in hearing of 10 dB or greater at any frequency. Among patients with longitudinal postoperative audiograms, accelerated age-related hearing loss was observed in low frequencies. CONCLUSIONS A significant number of patients demonstrated poorer hearing thresholds in the contralateral, unoperated ear after otologic and skull base surgery.
Collapse
Affiliation(s)
- Philip Perez
- Department of Otolaryngology, Head & Neck Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Tsung-Heng Tsai
- Department of Mathematical Sciences, Kent State University, Kent, Ohio
| | - John Hawks
- Gateway Biotechnology Inc., St. Louis, Missouri
| | | | - Joey Pinkl
- Gateway Biotechnology Inc., St. Louis, Missouri
| | - Partha Thirumala
- Departments of Neurological Surgery and Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | |
Collapse
|
14
|
Patro C, Monfiletto A, Singer A, Srinivasan NK, Mishra SK. Midlife Speech Perception Deficits: Impact of Extended High-Frequency Hearing, Peripheral Neural Function, and Cognitive Abilities. Ear Hear 2024; 45:1149-1164. [PMID: 38556645 DOI: 10.1097/aud.0000000000001504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
OBJECTIVES The objectives of the present study were to investigate the effects of age-related changes in extended high-frequency (EHF) hearing, peripheral neural function, working memory, and executive function on speech perception deficits in middle-aged individuals with clinically normal hearing. DESIGN We administered a comprehensive assessment battery to 37 participants spanning the age range of 20 to 56 years. This battery encompassed various evaluations, including standard and EHF pure-tone audiometry, ranging from 0.25 to 16 kHz. In addition, we conducted auditory brainstem response assessments with varying stimulation rates and levels, a spatial release from masking (SRM) task, and cognitive evaluations that involved the Trail Making test (TMT) for assessing executive function and the Abbreviated Reading Span test (ARST) for measuring working memory. RESULTS The results indicated a decline in hearing sensitivities at EHFs and an increase in completion times for the TMT with age. In addition, as age increased, there was a corresponding decrease in the amount of SRM. The declines in SRM were associated with age-related declines in hearing sensitivity at EHFs and TMT performance. While we observed an age-related decline in wave I responses, this decline was primarily driven by age-related reductions in EHF thresholds. In addition, the results obtained using the ARST did not show an age-related decline. Neither the auditory brainstem response results nor ARST scores were correlated with the amount of SRM. CONCLUSIONS These findings suggest that speech perception deficits in middle age are primarily linked to declines in EHF hearing and executive function, rather than cochlear synaptopathy or working memory.
Collapse
Affiliation(s)
- Chhayakanta Patro
- Department of Speech Language Pathology & Audiology, Towson University, Towson, Maryland, USA
| | - Angela Monfiletto
- Department of Speech Language Pathology & Audiology, Towson University, Towson, Maryland, USA
| | - Aviya Singer
- Department of Speech Language Pathology & Audiology, Towson University, Towson, Maryland, USA
| | - Nirmal Kumar Srinivasan
- Department of Speech Language Pathology & Audiology, Towson University, Towson, Maryland, USA
| | - Srikanta Kumar Mishra
- Department of Speech, Language and Hearing Sciences, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
15
|
Fujihira H, Yamagishi S, Furukawa S, Kashino M. Auditory brainstem response to paired clicks as a candidate marker of cochlear synaptopathy in humans. Clin Neurophysiol 2024; 165:44-54. [PMID: 38959535 DOI: 10.1016/j.clinph.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/02/2024] [Accepted: 06/08/2024] [Indexed: 07/05/2024]
Abstract
OBJECTIVE This study aimed to evaluate whether auditory brainstem response (ABR) using a paired-click stimulation paradigm could serve as a tool for detecting cochlear synaptopathy (CS). METHODS The ABRs to single-clicks and paired-clicks with various inter-click intervals (ICIs) and scores for word intelligibility in degraded listening conditions were obtained from 57 adults with normal hearing. The wave I peak amplitude and root mean square values for the post-wave I response within a range delayed from the wave I peak (referred to as the RMSpost-w1) were calculated for the single- and second-click responses. RESULTS The wave I peak amplitudes did not correlate with age except for the second-click responses at an ICI of 7 ms, and the word intelligibility scores. However, we found that the RMSpost-w1 values for the second-click responses significantly decreased with increasing age. Moreover, the RMSpost-w1 values for the second-click responses at an ICI of 5 ms correlated significantly with the scores for word intelligibility in degraded listening conditions. CONCLUSIONS The magnitude of the post-wave I response for the second-click response could serve as a tool for detecting CS in humans. SIGNIFICANCE Our findings shed new light on the analytical methods of ABR for quantifying CS.
Collapse
Affiliation(s)
- Haruna Fujihira
- NTT Communication Science Laboratories, Atsugi, Kanagawa, Japan; Department of Informatics, Faculty of Information Science and Electrical Engineering, Kyushu University, Fukuoka, Japan.
| | | | - Shigeto Furukawa
- NTT Communication Science Laboratories, Atsugi, Kanagawa, Japan; Graduate School of Public Health, Shizuoka Graduate University of Public Health, Shizuoka, Japan; Speech-Language-Hearing Center, Shizuoka General Hospital, Shizuoka, Japan
| | - Makio Kashino
- NTT Communication Science Laboratories, Atsugi, Kanagawa, Japan
| |
Collapse
|
16
|
Schmidt FH, Dörmann A, Ehrt K, Grossmann W, Mlynski R, Zhang L. The curvature quantification of wave I in auditory brainstem responses detects cochlear synaptopathy in human beings. Eur Arch Otorhinolaryngol 2024; 281:4735-4746. [PMID: 38703194 DOI: 10.1007/s00405-024-08699-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/18/2024] [Indexed: 05/06/2024]
Abstract
PURPOSE Patients with age-related hearing loss complain often about reduced speech perception in adverse listening environment. Studies on animals have suggested that cochlear synaptopathy may be one of the primary mechanisms responsible for this phenomenon. A decreased wave I amplitude in supra-threshold auditory brainstem response (ABR) can diagnose this pathology non-invasively. However, the interpretation of the wave I amplitude in humans remains controversial. Recent studies in mice have established a robust and reliable mathematic algorithm, i.e., curve curvature quantification, for detecting cochlear synaptopathy. This study aimed to determine whether the curve curvature has sufficient test-retest reliability to detect cochlear synaptopathy in aging humans. METHODS Healthy participants were recruited into this prospective study. All subjects underwent an audiogram examination with standard and extended high frequencies ranging from 0.125 to 16 kHz and an ABR with a stimulus of 80 dB nHL click. The peak amplitude, peak latency, curvature at the peak, and the area under the curve of wave I were calculated and analyzed. RESULTS A total of 80 individuals with normal hearing, aged 18 to 61 years, participated in this study, with a mean age of 26.4 years. Pearson correlation analysis showed a significant negative correlation between curvature and age, as well as between curvature and extended high frequency (EHF) threshold (10-16 kHz). Additionally, the same correlation was observed between age and area as well as age and EHF threshold. The model comparison demonstrated that the curvature at the peak of wave I is the best metric to correlate with EHF threshold. CONCLUSION The curvature at the peak of wave I is the most sensitive metric for detecting cochlear synaptopathy in humans and may be applied in routine diagnostics to detect early degenerations of the auditory nerve.
Collapse
Affiliation(s)
- Florian Herrmann Schmidt
- Department of Otorhinolaryngology, Head and Neck Surgery, Otto Körner, Rostock University Medical Center, Doberaner Strasse 137-139, 18057, Rostock, Germany
| | - Alexander Dörmann
- Department of Otorhinolaryngology, Head and Neck Surgery, Otto Körner, Rostock University Medical Center, Doberaner Strasse 137-139, 18057, Rostock, Germany
| | - Karsten Ehrt
- Department of Otorhinolaryngology, Head and Neck Surgery, Otto Körner, Rostock University Medical Center, Doberaner Strasse 137-139, 18057, Rostock, Germany
| | - Wilma Grossmann
- Department of Otorhinolaryngology, Head and Neck Surgery, Otto Körner, Rostock University Medical Center, Doberaner Strasse 137-139, 18057, Rostock, Germany
| | - Robert Mlynski
- Department of Otorhinolaryngology, Head and Neck Surgery, Otto Körner, Rostock University Medical Center, Doberaner Strasse 137-139, 18057, Rostock, Germany
| | - Lichun Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Otto Körner, Rostock University Medical Center, Doberaner Strasse 137-139, 18057, Rostock, Germany.
| |
Collapse
|
17
|
Arjmandi MK, Neils-Strunjas J, Nemati S, Fridriksson J, Newman-Norlund S, Newman-Norlund R, Bonilha L. Age-Related Hearing Loss, Cognitive Decline, and Social Interaction: Testing a Framework. JOURNAL OF SPEECH, LANGUAGE, AND HEARING RESEARCH : JSLHR 2024; 67:2743-2760. [PMID: 38995870 DOI: 10.1044/2024_jslhr-23-00810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
PURPOSE Aging increases risk for hearing loss, cognitive decline, and social isolation; however, the nature of their interconnection remains unclear. This study examined the interplay between age-related hearing loss, cognitive decline, and social isolation in adults by testing the ability to understand speech in background noise, a challenge frequently reported by many older adults. METHOD We analyzed data collected from 128 adults (20-79 years of age, Mage = 51 years) recruited as part of the Aging Brain Cohort at the University of South Carolina repository. The participants underwent testing for hearing, cognition, and social interaction, which included pure-tone audiometry, a words-in-noise (WIN) test, a hearing questionnaire (Speech, Spatial and Qualities of Hearing Scale [SSQ12]), a social questionnaire (Patient-Reported Outcomes Measurement Information System-57 Social), and the Montreal Cognitive Assessment. We used a single pure-tone average (PTA) threshold value and a single WIN threshold value for each participant because there were no differences on average between the left and right ears. RESULTS Poorer hearing was significantly associated with cognitive decline, through both PTA and WIN thresholds, with a stronger association observed for WIN threshold. Adults with poorer hearing also exhibited greater social isolation, as evidenced by their WIN threshold and SSQ12 score, although not through PTA. This connection was more pronounced with the WIN threshold than with the SSQ12 score. Cognition was not related to social isolation, suggesting that social isolation is affected more by the ability to understand words in noise than by cognition in a nondemented population. CONCLUSIONS Understanding speech in challenging auditory environments rather than mere threshold detection is strongly linked to social isolation and cognitive decline. Thus, inclusion of a word-recognition-in-noise test and a social isolation survey in clinical settings is warranted. SUPPLEMENTAL MATERIAL https://doi.org/10.23641/asha.26237060.
Collapse
Affiliation(s)
- Meisam K Arjmandi
- Department of Communication Sciences and Disorders, University of South Carolina, Columbia
- Institute for Mind and Brain, University of South Carolina, Columbia
| | - Jean Neils-Strunjas
- Department of Communication Sciences and Disorders, University of South Carolina, Columbia
| | - Samaneh Nemati
- Department of Communication Sciences and Disorders, University of South Carolina, Columbia
| | - Julius Fridriksson
- Department of Communication Sciences and Disorders, University of South Carolina, Columbia
- Institute for Mind and Brain, University of South Carolina, Columbia
| | - Sarah Newman-Norlund
- Department of Communication Sciences and Disorders, University of South Carolina, Columbia
| | - Roger Newman-Norlund
- Department of Communication Sciences and Disorders, University of South Carolina, Columbia
- Institute for Mind and Brain, University of South Carolina, Columbia
| | | |
Collapse
|
18
|
Ono M, Ito T. Hearing loss-related altered neuronal activity in the inferior colliculus. Hear Res 2024; 449:109033. [PMID: 38797036 DOI: 10.1016/j.heares.2024.109033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/01/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024]
Abstract
Hearing loss is well known to cause plastic changes in the central auditory system and pathological changes such as tinnitus and hyperacusis. Impairment of inner ear functions is the main cause of hearing loss. In aged individuals, not only inner ear dysfunction but also senescence of the central nervous system is the cause of malfunction of the auditory system. In most cases of hearing loss, the activity of the auditory nerve is reduced, but that of the successive auditory centers is increased in a compensatory way. It has been reported that activity changes occur in the inferior colliculus (IC), a critical nexus of the auditory pathway. The IC integrates the inputs from the brainstem and drives the higher auditory centers. Since abnormal activity in the IC is likely to affect auditory perception, it is crucial to elucidate the neuronal mechanism to induce the activity changes of IC neurons with hearing loss. This review outlines recent findings on hearing-loss-induced plastic changes in the IC and brainstem auditory neuronal circuits and discusses what neuronal mechanisms underlie hearing-loss-induced changes in the activity of IC neurons. Considering the different causes of hearing loss, we discuss age-related hearing loss separately from other forms of hearing loss (non-age-related hearing loss). In general, the main plastic change of IC neurons caused by both age-related and non-age-related hearing loss is increased central gain. However, plastic changes in the IC caused by age-related hearing loss seem to be more complex than those caused by non-age-related hearing loss.
Collapse
Affiliation(s)
- Munenori Ono
- Department of Physiology, School of Medicine, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan.
| | - Tetsufumi Ito
- Systems Function and Morphology, University of Toyama, Toyama 930-0194, Japan.
| |
Collapse
|
19
|
Zhang B, Hu Y, Du H, Han S, Ren L, Cheng H, Wang Y, Gao X, Zheng S, Cui Q, Tian L, Liu T, Sun J, Chai R. Tissue engineering strategies for spiral ganglion neuron protection and regeneration. J Nanobiotechnology 2024; 22:458. [PMID: 39085923 PMCID: PMC11293049 DOI: 10.1186/s12951-024-02742-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024] Open
Abstract
Cochlear implants can directly activate the auditory system's primary sensory neurons, the spiral ganglion neurons (SGNs), via circumvention of defective cochlear hair cells. This bypass restores auditory input to the brainstem. SGN loss etiologies are complex, with limited mammalian regeneration. Protecting and revitalizing SGN is critical. Tissue engineering offers a novel therapeutic strategy, utilizing seed cells, biomolecules, and scaffold materials to create a cellular environment and regulate molecular cues. This review encapsulates the spectrum of both human and animal research, collating the factors contributing to SGN loss, the latest advancements in the utilization of exogenous stem cells for auditory nerve repair and preservation, the taxonomy and mechanism of action of standard biomolecules, and the architectural components of scaffold materials tailored for the inner ear. Furthermore, we delineate the potential and benefits of the biohybrid neural interface, an incipient technology in the realm of implantable devices. Nonetheless, tissue engineering requires refined cell selection and differentiation protocols for consistent SGN quality. In addition, strategies to improve stem cell survival, scaffold biocompatibility, and molecular cue timing are essential for biohybrid neural interface integration.
Collapse
Affiliation(s)
- Bin Zhang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yangnan Hu
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| | - Haoliang Du
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing University, Nanjing, 210008, China
| | - Shanying Han
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Lei Ren
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Hong Cheng
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Yusong Wang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Xin Gao
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Shasha Zheng
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Qingyue Cui
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Lei Tian
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
| | - Tingting Liu
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
| | - Jiaqiang Sun
- Department of Otolaryngology-Head and Neck Surgery, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230001, China.
| | - Renjie Chai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Department of Neurology, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing, China.
- Southeast University Shenzhen Research Institute, Shenzhen, 518063, China.
| |
Collapse
|
20
|
Sung CYW, Hayase N, Yuen PST, Lee J, Fernandez K, Hu X, Cheng H, Star RA, Warchol ME, Cunningham LL. Macrophage depletion protects against cisplatin-induced ototoxicity and nephrotoxicity. SCIENCE ADVANCES 2024; 10:eadk9878. [PMID: 39047106 PMCID: PMC11268410 DOI: 10.1126/sciadv.adk9878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 06/18/2024] [Indexed: 07/27/2024]
Abstract
Cisplatin is a widely used anticancer drug with notable side effects including ototoxicity and nephrotoxicity. Macrophages, the major resident immune cells in the cochlea and kidney, are important drivers of both inflammatory and tissue repair responses. To investigate the roles of macrophages in cisplatin-induced toxicities, we used PLX3397, a U.S. Food and Drug Administration-approved inhibitor of the colony-stimulating factor 1 receptor, to eliminate tissue-resident macrophages. Mice treated with cisplatin alone had considerable hearing loss (ototoxicity) and kidney injury (nephrotoxicity). Macrophage ablation resulted in significantly reduced hearing loss and had greater outer hair cell survival. Macrophage ablation also protected against cisplatin-induced nephrotoxicity, as evidenced by markedly reduced tubular injury and fibrosis. Mechanistically, our data suggest that the protective effect of macrophage ablation against cisplatin-induced ototoxicity and nephrotoxicity is mediated by reduced platinum accumulation in both the inner ear and the kidney. Together, our data indicate that ablation of tissue-resident macrophages represents an important strategy for mitigating cisplatin-induced ototoxicity and nephrotoxicity.
Collapse
Affiliation(s)
- Cathy Yea Won Sung
- Laboratory of Hearing Biology and Therapeutics, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, MD, USA
| | - Naoki Hayase
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - Peter S. T. Yuen
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - John Lee
- Laboratory of Hearing Biology and Therapeutics, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, MD, USA
| | - Katharine Fernandez
- Laboratory of Hearing Biology and Therapeutics, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, MD, USA
| | - Xuzhen Hu
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - Hui Cheng
- Bioinformatics and Biostatistics Collaboration Core, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, MD, USA
| | - Robert A. Star
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - Mark E. Warchol
- Department of Otolaryngology, School of Medicine, Washington University, Saint Louis, MO, USA
| | - Lisa L. Cunningham
- Laboratory of Hearing Biology and Therapeutics, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, MD, USA
| |
Collapse
|
21
|
Ding D, Manohar S, Kador PF, Salvi R. Multifunctional redox modulator prevents blast-induced loss of cochlear and vestibular hair cells and auditory spiral ganglion neurons. Sci Rep 2024; 14:15296. [PMID: 38961203 PMCID: PMC11222375 DOI: 10.1038/s41598-024-66406-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/01/2024] [Indexed: 07/05/2024] Open
Abstract
Blast wave exposure, a leading cause of hearing loss and balance dysfunction among military personnel, arises primarily from direct mechanical damage to the mechanosensory hair cells and supporting structures or indirectly through excessive oxidative stress. We previously reported that HK-2, an orally active, multifunctional redox modulator (MFRM), was highly effective in reducing both hearing loss and hair cells loss in rats exposed to a moderate intensity workday noise that likely damages the cochlea primarily from oxidative stress versus direct mechanical trauma. To determine if HK-2 could also protect cochlear and vestibular cells from damage caused primarily from direct blast-induced mechanical trauma versus oxidative stress, we exposed rats to six blasts of 186 dB peak SPL. The rats were divided into four groups: (B) blast alone, (BEP) blast plus earplugs, (BHK-2) blast plus HK-2 and (BEPHK-2) blast plus earplugs plus HK-2. HK-2 was orally administered at 50 mg/kg/d from 7-days before to 30-day after the blast exposure. Cochlear and vestibular tissues were harvested 60-d post-exposure and evaluated for loss of outer hair cells (OHC), inner hair cells (IHC), auditory nerve fibers (ANF), spiral ganglion neurons (SGN) and vestibular hair cells in the saccule, utricle and semicircular canals. In the untreated blast-exposed group (B), massive losses occurred to OHC, IHC, ANF, SGN and only the vestibular hair cells in the striola region of the saccule. In contrast, rats treated with HK-2 (BHK-2) sustained significantly less OHC (67%) and IHC (57%) loss compared to the B group. OHC and IHC losses were smallest in the BEPHK-2 group, but not significantly different from the BEP group indicating lack of protective synergy between EP and HK-2. There was no loss of ANF, SGN or saccular hair cells in the BHK-2, BEP and BEPHK-2 groups. Thus, HK-2 not only significantly reduced OHC and IHC damage, but completely prevented loss of ANF, SGN and saccule hair cells. The powerful protective effects of this oral MFRM make HK-2 an extremely promising candidate for human clinical trials.
Collapse
Affiliation(s)
- Dalian Ding
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, 14214, USA
| | | | | | - Richard Salvi
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, 14214, USA.
| |
Collapse
|
22
|
Lutze RD, Ingersoll MA, Thotam A, Joseph A, Fernandes J, Teitz T. ERK1/2 Inhibition via the Oral Administration of Tizaterkib Alleviates Noise-Induced Hearing Loss While Tempering down the Immune Response. Int J Mol Sci 2024; 25:6305. [PMID: 38928015 PMCID: PMC11204379 DOI: 10.3390/ijms25126305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/02/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
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 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.
Collapse
Affiliation(s)
- Richard D. Lutze
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA; (R.D.L.); (M.A.I.); (A.T.); (A.J.); (J.F.)
| | - Matthew A. Ingersoll
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA; (R.D.L.); (M.A.I.); (A.T.); (A.J.); (J.F.)
| | - Alena Thotam
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA; (R.D.L.); (M.A.I.); (A.T.); (A.J.); (J.F.)
| | - Anjali Joseph
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA; (R.D.L.); (M.A.I.); (A.T.); (A.J.); (J.F.)
| | - Joshua Fernandes
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA; (R.D.L.); (M.A.I.); (A.T.); (A.J.); (J.F.)
| | - Tal Teitz
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA; (R.D.L.); (M.A.I.); (A.T.); (A.J.); (J.F.)
- The Scintillon Research Institute, San Diego, CA 92121, USA
| |
Collapse
|
23
|
Zhao HB, Liu LM, Mei L, Quinonez AT, Roberts RA, Lu X. Prevention and treatment of noise-induced hearing loss and cochlear synapse degeneration by potassium channel blockers in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.597382. [PMID: 38895254 PMCID: PMC11185602 DOI: 10.1101/2024.06.04.597382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Noise can induce hearing loss. In particularly, noise can induce cochlear synapse degeneration leading to hidden hearing loss, which is the most common type of hearing disorders in the clinic. Currently, there is no pharmacological treatment, particularly, no post-exposure (i.e., therapeutic) treatment available in the clinic. Here, we report that systematic administration of K + channel blockers before or after noise exposure could significantly attenuate NIHL and synapse degeneration. After systematic administration of a general K-channel blocker tetraethylammonium (TEA), the elevation of auditory brainstem response (ABR) thresholds after noise-exposure significantly reduced, and the active cochlear mechanics significantly improved. The therapeutic effect was further improved as the post-exposure administration time extending to 3 days. BK channel is a predominant K + channel in the inner hair cells. Systematic administration of a BK channel blocker GAL-021 after noise exposure also ameliorated hearing loss and improved hearing behavioral responses tested by acoustic startle response (ASR). Finally, both TEA and GAL-021 significantly attenuated noise-induced ribbon synapse degeneration. These data demonstrate that K + -channel blockers can prevent and treat NIHL and cochlear synapse degeneration. Our finding may aid in developing therapeutic strategies for post-exposure treatment of NIHL and synapse degeneration. Significance Statement Noise is a common deafness factor affecting more 100 million people in the United States. So far, there is no pharmacological treatment available. We show here that administration of K + channel blockers after noise exposure could attenuate noise-induced hearing loss and synapse degeneration, and improved behavioral responses. This is the first time to real the K + channel blockers that could treat noise-induced hearing loss and cochlear synaptopathy after noise exposure.
Collapse
|
24
|
Dias JW, McClaskey CM, Alvey AP, Lawson A, Matthews LJ, Dubno JR, Harris KC. Effects of age and noise exposure history on auditory nerve response amplitudes: A systematic review, study, and meta-analysis. Hear Res 2024; 447:109010. [PMID: 38744019 PMCID: PMC11135078 DOI: 10.1016/j.heares.2024.109010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/08/2024] [Accepted: 04/18/2024] [Indexed: 05/16/2024]
Abstract
Auditory nerve (AN) function has been hypothesized to deteriorate with age and noise exposure. Here, we perform a systematic review of published studies and find that the evidence for age-related deficits in AN function is largely consistent across the literature, but there are inconsistent findings among studies of noise exposure history. Further, evidence from animal studies suggests that the greatest deficits in AN response amplitudes are found in noise-exposed aged mice, but a test of the interaction between effects of age and noise exposure on AN function has not been conducted in humans. We report a study of our own examining differences in the response amplitude of the compound action potential N1 (CAP N1) between younger and older adults with and without a self-reported history of noise exposure in a large sample of human participants (63 younger adults 18-30 years of age, 103 older adults 50-86 years of age). CAP N1 response amplitudes were smaller in older than younger adults. Noise exposure history did not appear to predict CAP N1 response amplitudes, nor did the effect of noise exposure history interact with age. We then incorporated our results into two meta-analyses of published studies of age and noise exposure history effects on AN response amplitudes in neurotypical human samples. The meta-analyses found that age effects across studies are robust (r = -0.407), but noise exposure effects are weak (r = -0.152). We conclude that noise exposure effects may be highly variable depending on sample characteristics, study design, and statistical approach, and researchers should be cautious when interpreting results. The underlying pathology of age-related and noise-induced changes in AN function are difficult to determine in living humans, creating a need for longitudinal studies of changes in AN function across the lifespan and histological examination of the AN from temporal bones collected post-mortem.
Collapse
Affiliation(s)
- James W Dias
- Department of Otolaryngology - Head and Neck Surgery, Medical University of South Carolina, 135 Rutledge Avenue, MSC 550, Charleston, SC 29425-5500, United States.
| | - Carolyn M McClaskey
- Department of Otolaryngology - Head and Neck Surgery, Medical University of South Carolina, 135 Rutledge Avenue, MSC 550, Charleston, SC 29425-5500, United States
| | - April P Alvey
- Department of Otolaryngology - Head and Neck Surgery, Medical University of South Carolina, 135 Rutledge Avenue, MSC 550, Charleston, SC 29425-5500, United States
| | - Abigail Lawson
- Department of Otolaryngology - Head and Neck Surgery, Medical University of South Carolina, 135 Rutledge Avenue, MSC 550, Charleston, SC 29425-5500, United States
| | - Lois J Matthews
- Department of Otolaryngology - Head and Neck Surgery, Medical University of South Carolina, 135 Rutledge Avenue, MSC 550, Charleston, SC 29425-5500, United States
| | - Judy R Dubno
- Department of Otolaryngology - Head and Neck Surgery, Medical University of South Carolina, 135 Rutledge Avenue, MSC 550, Charleston, SC 29425-5500, United States
| | - Kelly C Harris
- Department of Otolaryngology - Head and Neck Surgery, Medical University of South Carolina, 135 Rutledge Avenue, MSC 550, Charleston, SC 29425-5500, United States
| |
Collapse
|
25
|
Gill NB, Dowker-Key PD, Hedrick M, Bettaieb A. Unveiling the Role of Oxidative Stress in Cochlear Hair Cell Death: Prospective Phytochemical Therapeutics against Sensorineural Hearing Loss. Int J Mol Sci 2024; 25:4272. [PMID: 38673858 PMCID: PMC11050722 DOI: 10.3390/ijms25084272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/31/2024] [Accepted: 04/07/2024] [Indexed: 04/28/2024] Open
Abstract
Hearing loss represents a multifaceted and pervasive challenge that deeply impacts various aspects of an individual's life, spanning psychological, emotional, social, and economic realms. Understanding the molecular underpinnings that orchestrate hearing loss remains paramount in the quest for effective therapeutic strategies. This review aims to expound upon the physiological, biochemical, and molecular aspects of hearing loss, with a specific focus on its correlation with diabetes. Within this context, phytochemicals have surfaced as prospective contenders in the pursuit of potential adjuvant therapies. These compounds exhibit noteworthy antioxidant and anti-inflammatory properties, which hold the potential to counteract the detrimental effects induced by oxidative stress and inflammation-prominent contributors to hearing impairment. Furthermore, this review offers an up-to-date exploration of the diverse molecular pathways modulated by these compounds. However, the dynamic landscape of their efficacy warrants recognition as an ongoing investigative topic, inherently contingent upon specific experimental models. Ultimately, to ascertain the genuine potential of phytochemicals as agents in hearing loss treatment, a comprehensive grasp of the molecular mechanisms at play, coupled with rigorous clinical investigations, stands as an imperative quest.
Collapse
Affiliation(s)
- Nicholas B. Gill
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Presley D. Dowker-Key
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Mark Hedrick
- Department of Audiology & Speech Pathology, The University of Tennessee Health Science Center, Knoxville, TN 37996-0240, USA
| | - Ahmed Bettaieb
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
- Graduate School of Genome Science and Technology, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
- Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| |
Collapse
|
26
|
Dias JW, McClaskey CM, Alvey AP, Lawson A, Matthews LJ, Dubno JR, Harris KC. Effects of Age and Noise Exposure History on Auditory Nerve Response Amplitudes: A Systematic Review, Study, and Meta-Analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.20.585882. [PMID: 38585917 PMCID: PMC10996537 DOI: 10.1101/2024.03.20.585882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Auditory nerve (AN) function has been hypothesized to deteriorate with age and noise exposure. Here, we perform a systematic review of published studies and find that the evidence for age-related deficits in AN function is largely consistent across the literature, but there are inconsistent findings among studies of noise exposure history. Further, evidence from animal studies suggests that the greatest deficits in AN response amplitudes are found in noise-exposed aged mice, but a test of the interaction between effects of age and noise exposure on AN function has not been conducted in humans. We report a study of our own examining differences in the response amplitude of the compound action potential N1 (CAP N1) between younger and older adults with and without a self-reported history of noise exposure in a large sample of human participants (63 younger adults 18-30 years of age, 103 older adults 50-86 years of age). CAP N1 response amplitudes were smaller in older than younger adults. Noise exposure history did not appear to predict CAP N1 response amplitudes, nor did the effect of noise exposure history interact with age. We then incorporated our results into two meta-analyses of published studies of age and noise exposure history effects on AN response amplitudes in neurotypical human samples. The meta-analyses found that age effects across studies are robust (r=-0.407), but noise-exposure effects are weak (r=-0.152). We conclude that noise-exposure effects may be highly variable depending on sample characteristics, study design, and statistical approach, and researchers should be cautious when interpreting results. The underlying pathology of age-related and noise-induced changes in AN function are difficult to determine in living humans, creating a need for longitudinal studies of changes in AN function across the lifespan and histological examination of the AN from temporal bones collected post-mortem.
Collapse
Affiliation(s)
- James W Dias
- Medical University of South Carolina Department of Otolaryngology - Head and Neck Surgery
| | - Carolyn M McClaskey
- Medical University of South Carolina Department of Otolaryngology - Head and Neck Surgery
| | - April P Alvey
- Medical University of South Carolina Department of Otolaryngology - Head and Neck Surgery
| | - Abigail Lawson
- Medical University of South Carolina Department of Otolaryngology - Head and Neck Surgery
| | - Lois J Matthews
- Medical University of South Carolina Department of Otolaryngology - Head and Neck Surgery
| | - Judy R Dubno
- Medical University of South Carolina Department of Otolaryngology - Head and Neck Surgery
| | - Kelly C Harris
- Medical University of South Carolina Department of Otolaryngology - Head and Neck Surgery
| |
Collapse
|
27
|
Cordeiro FP, Marchiori LL, Teixeira DC, Andraus RA, Poli-Frederico RC. Plasma Levels of Interleukin 2 (IL-2) Associated with Hearing Loss Evaluation in the Elderly. Noise Health 2024; 26:165-173. [PMID: 38904818 PMCID: PMC11530100 DOI: 10.4103/nah.nah_3_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 12/13/2024] [Accepted: 04/01/2024] [Indexed: 06/22/2024] Open
Abstract
CONTEXT Presbycusis can be mediated by the effects of inflammatory processes on the auditory system, and these aging biological mechanisms remain poorly studied. AIMS The aim of this study was to determine whether plasma biomarkers are associated with hearing disorders caused by aging in the elderly. SETTINGS AND DESIGN Cross-sectional study with 106 participants in the Active Aging Project, 93 (88%) females and 13 (12%) males, with an average age of 70 years. METHODS AND MATERIAL Audiological evaluation was performed with pure tone audiometry and collection of peripheral blood for the measurement of plasma levels of interleukins 2, 4, 6, and 10, tumor necrosis factor-α, and interferon-γ by means of flow cytometry. STATISTICAL ANALYSIS USED The SPSS (v.0, SPSS Inc., Chicago, USA) was used for the analysis of the data obtained. For all data analyzed, the significance level adopted was P < 0.05 and 95% confidence interval. RESULTS There were statistically significant correlations between male and IL-2 (P = 0.031; rs = 0.210), mean II of the right ear (P = 0.004; rs = 0.279), longer in years (P = 0.002; rs = 0.307) and in hours (P = 0.004; rs = 0.281) of noise exposure also in males. CONCLUSIONS In the present study, there was an association between the male gender and higher plasma levels of IL-2, an increase in the average hearing in the right ear, and greater time in years and hours of exposure to noise. There was a predominance of mild sensorineural hearing loss and worsening of hearing related to age, characteristics of presbycusis.
Collapse
Affiliation(s)
- Fernanda P. Cordeiro
- Student of theMaster’s and Doctor’s Degree in Rehabilitation Sciences, UEL-UNOPAR. https://orcid.org/0000-0003-4489-2568
| | - Luciana L.M. Marchiori
- Interdisciplinary Laboratory for Intervention in Health Promotion LIIPS/UNICESUMAR-Maringá, Paraná, Brazil https://orcid.org/0000-0002-9026-0468
| | - Denilson C. Teixeira
- Research Center in Health Science, State University of Londrina, Paraná, Brazil https://orcid.org/0000-0002-6299-0252
| | - Rodrigo A.C. Andraus
- Professor of the Master’s and Doctor’s Degree Associated in Rehabilitation Sciences UEL-UNOPAR https://orcid.org/0000-0002-3849-0872
| | - Regina Célia Poli-Frederico
- Professor of the Master’s and Doctor’s Degree Associated in Rehabilitation Sciences UEL-UNOPAR. https://orcid.org/0000-0003-4631-4606
| |
Collapse
|
28
|
Hong J, Dai P, Liang H, Sun G, Qi W, Bi Y. Extrasynaptic distribution of NMDA receptors in cochlear inner hair cell afferent signaling complex. J Chem Neuroanat 2024; 137:102417. [PMID: 38570170 DOI: 10.1016/j.jchemneu.2024.102417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/30/2024] [Accepted: 03/30/2024] [Indexed: 04/05/2024]
Abstract
OBJECTIVE The distribution and role of NMDA receptors is unclear in the afferent signaling complex of the cochlea. The present study aimed to examine the distribution of NMDA receptors in cochlear afferent signaling complex of the adult mouse, and their relationship with ribbon synapses of inner hair cells (IHCs) and GABAergic efferent terminals of the lateral olivocochlear (LOC). METHODS Immunofluorescence staining in combination with confocal microscopy was used to investigate the distribution of glutamatergic NMDA and AMPA receptors in afferent terminals of SGNs, and their relationship with ribbon synapses of IHCs and GABAergic efferent terminals of LOC. RESULTS Terminals with AMPA receptors along with Ribbons of IHC formed afferent synapses in the basal pole of IHCs, and those with NMDA receptors were mainly distributed longitudinally in the IHCs nuclei region. Significant difference was found in the distribution of NMDA and AMPA receptors in IHC afferent signaling complex (P<0.05). Some GABAergic terminals colocalized with NMDA receptors at the IHC nucleus region (P>0.05). CONCLUSION There is significant difference in the distribution of NMDA and AMPA receptors in cochlear afferent signaling complex. NMDA receptors are present in the extra-synaptic region of ribbon synapses of IHCs, and they are related to GABA efferent terminals of the afferent signaling complex.
Collapse
Affiliation(s)
- Juan Hong
- Department of Otorhinolaryngology, Huashan Hospital of Fudan University, Shanghai 200040, China.
| | - Peidong Dai
- ENT Institute, Eye & ENT Hospital of Fudan University; NHC Hearing Medicine Key Laboratory (Fudan University), Shanghai 200031, China
| | - Huazheng Liang
- Monash Suzhou Research Institute, Suzhou Industrial Park, Jiangsu, China
| | - Guangbin Sun
- Department of Otorhinolaryngology, Huashan Hospital of Fudan University, Shanghai 200040, China
| | - Weidong Qi
- Department of Otorhinolaryngology, Huashan Hospital of Fudan University, Shanghai 200040, China
| | - Yong Bi
- Department of Neurology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China.
| |
Collapse
|
29
|
Liu J, Stohl J, Overath T. Hidden hearing loss: Fifteen years at a glance. Hear Res 2024; 443:108967. [PMID: 38335624 DOI: 10.1016/j.heares.2024.108967] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 01/15/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024]
Abstract
Hearing loss affects approximately 18% of the population worldwide. Hearing difficulties in noisy environments without accompanying audiometric threshold shifts likely affect an even larger percentage of the global population. One of the potential causes of hidden hearing loss is cochlear synaptopathy, the loss of synapses between inner hair cells (IHC) and auditory nerve fibers (ANF). These synapses are the most vulnerable structures in the cochlea to noise exposure or aging. The loss of synapses causes auditory deafferentation, i.e., the loss of auditory afferent information, whose downstream effect is the loss of information that is sent to higher-order auditory processing stages. Understanding the physiological and perceptual effects of this early auditory deafferentation might inform interventions to prevent later, more severe hearing loss. In the past decade, a large body of work has been devoted to better understand hidden hearing loss, including the causes of hidden hearing loss, their corresponding impact on the auditory pathway, and the use of auditory physiological measures for clinical diagnosis of auditory deafferentation. This review synthesizes the findings from studies in humans and animals to answer some of the key questions in the field, and it points to gaps in knowledge that warrant more investigation. Specifically, recent studies suggest that some electrophysiological measures have the potential to function as indicators of hidden hearing loss in humans, but more research is needed for these measures to be included as part of a clinical test battery.
Collapse
Affiliation(s)
- Jiayue Liu
- Department of Psychology and Neuroscience, Duke University, Durham, USA.
| | - Joshua Stohl
- North American Research Laboratory, MED-EL Corporation, Durham, USA
| | - Tobias Overath
- Department of Psychology and Neuroscience, Duke University, Durham, USA
| |
Collapse
|
30
|
Lu Y, Liu J, Li B, Wang H, Wang F, Wang S, Wu H, Han H, Hua Y. Spatial patterns of noise-induced inner hair cell ribbon loss in the mouse mid-cochlea. iScience 2024; 27:108825. [PMID: 38313060 PMCID: PMC10835352 DOI: 10.1016/j.isci.2024.108825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/16/2023] [Accepted: 01/03/2024] [Indexed: 02/06/2024] Open
Abstract
In the mammalian cochlea, moderate acoustic overexposure leads to loss of ribbon-type synapse between the inner hair cell (IHC) and its postsynaptic spiral ganglion neuron (SGN), causing a reduced dynamic range of hearing but not a permanent threshold elevation. A prevailing view is that such ribbon loss (known as synaptopathy) selectively impacts the low-spontaneous-rate and high-threshold SGN fibers contacting predominantly the modiolar IHC face. However, the spatial pattern of synaptopathy remains scarcely characterized in the most sensitive mid-cochlear region, where two morphological subtypes of IHC with distinct ribbon size gradients coexist. Here, we used volume electron microscopy to investigate noise exposure-related changes in the mouse IHCs with and without ribbon loss. Our quantifications reveal that IHC subtypes differ in the worst-hit area of synaptopathy. Moreover, we show relative enrichment of mitochondria in the surviving SGN terminals, providing key experimental evidence for the long-proposed role of SGN-terminal mitochondria in synaptic vulnerability.
Collapse
Affiliation(s)
- Yan Lu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People’s Hospital, Shanghai 200125, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai 200125, China
- Shanghai Institute of Precision Medicine, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Jing Liu
- Laboratory of Brain Atlas and Brain-inspired Intelligence, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Bei Li
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People’s Hospital, Shanghai 200125, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai 200125, China
| | - Haoyu Wang
- Shanghai Institute of Precision Medicine, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Fangfang Wang
- Shanghai Institute of Precision Medicine, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Shengxiong Wang
- Shanghai Institute of Precision Medicine, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Hao Wu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People’s Hospital, Shanghai 200125, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai 200125, China
- Shanghai Institute of Precision Medicine, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Hua Han
- Laboratory of Brain Atlas and Brain-inspired Intelligence, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing 101408, China
- State Key Laboratory of Multimodal Artificial Intelligence Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Yunfeng Hua
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People’s Hospital, Shanghai 200125, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai 200125, China
- Shanghai Institute of Precision Medicine, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| |
Collapse
|
31
|
Chai B, Holland ML, Camposeo EL, King K, Schvartz-Leyzac KC. Patient and Device Factors Contributing to Electrically Evoked Stapedial Reflex Thresholds in Cochlear Implanted Adults. Audiol Neurootol 2024; 29:263-270. [PMID: 38342083 PMCID: PMC11305975 DOI: 10.1159/000535058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 10/22/2023] [Indexed: 02/13/2024] Open
Abstract
INTRODUCTION Optimal cochlear implant (CI) outcomes are due to, at least in part, appropriate device programming. Objective measures, such as electrically evoked stapedial reflex thresholds (ESRTs), can be used to more accurately set programming levels. However, underlying factors that contribute to ESRT levels are not well understood. The objective of the current study was to analyze how demographic variables of patient sex and age, along with CI electrode location, influence ESRTs in adult CI recipients. METHODS A single institution retrospective review was performed. Electronic medical records, CI programming records, and clinic database of postoperative computerized tomography were reviewed to gather information regarding patient demographics, ESRTs, and electrode array metrics including medial-lateral distance and scalar location. Linear mixed models were constructed to determine how demographic variables and electrode position influence ESRTs recorded in 138 adult CI recipients. RESULTS ESRTs were significantly affected by recipient age, with older listeners demonstrating higher ESRT levels. On average, males had higher ESRT levels when compared to females. In a subset of the study sample, ESRT levels increased with increasing medial-lateral distance; however, there was not a statistically significant effect of electrode type (lateral/straight arrays compared to perimodiolar arrays). ESRTs were not affected by scalar location. DISCUSSION/CONCLUSIONS The results suggest that key demographic and electrode position characteristics influence the level of ESRTs in adult CI recipients. While ESRTs are widely used to assist with CI programming, underlying factors are not well understood. The significant factors of aging and sex could be due to middle ear mechanics or neural health differences. However, further data are needed to better understand these associations.
Collapse
Affiliation(s)
- Brianna Chai
- Department of Otolaryngology- Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Mary Laing Holland
- Department of Otolaryngology- Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Elizabeth L Camposeo
- Department of Otolaryngology- Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kaylene King
- Department of Otolaryngology- Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kara C Schvartz-Leyzac
- Department of Otolaryngology- Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
32
|
Charlton PE, Burke K, Kobrina A, Lauer AM, Dent ML. The perception of ultrasonic vocalizations by laboratory mice following intense noise exposures. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2024; 155:867-878. [PMID: 38310604 PMCID: PMC10838193 DOI: 10.1121/10.0024614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 01/02/2024] [Accepted: 01/11/2024] [Indexed: 02/06/2024]
Abstract
Noise-induced hearing loss interacts with age, sex, and listening conditions to affect individuals' perception of ecologically relevant stimuli like speech. The present experiments assessed the impact of age and sex on vocalization detection by noise-exposed mice trained to detect a downsweep or complex ultrasonic vocalization in quiet or in the presence of a noise background. Daily thresholds before and following intense noise exposure were collected longitudinally and compared across several factors. All mice, regardless of age, sex, listening condition, or stimulus type showed their poorest behavioral sensitivity immediately after the noise exposure. There were varying degrees of recovery over time and across factors. Old-aged mice had greater threshold shifts and less recovery compared to middle-aged mice. Mice had larger threshold shifts and less recovery for downsweeps than for complex vocalizations. Female mice were more sensitive, had smaller post-noise shifts, and had better recovery than males. Thresholds in noise were higher and less variable than thresholds in quiet, but there were comparable shifts and recovery. In mice, as in humans, the perception of ecologically relevant stimuli suffers after an intense noise exposure, and results differ from simple tone detection findings.
Collapse
Affiliation(s)
- Payton E Charlton
- Department of Psychology, University at Buffalo, The State University of New York, Buffalo, New York 14260, USA
| | - Kali Burke
- Department of Psychology, University at Buffalo, The State University of New York, Buffalo, New York 14260, USA
| | - Anastasiya Kobrina
- Department of Psychology, University at Buffalo, The State University of New York, Buffalo, New York 14260, USA
| | - Amanda M Lauer
- Department of Otolaryngology-Head and Neck Surgery and Center for Hearing and Balance, Johns Hopkins University, Baltimore, Maryland 21287, USA
| | - Micheal L Dent
- Department of Psychology, University at Buffalo, The State University of New York, Buffalo, New York 14260, USA
| |
Collapse
|
33
|
Sørensen M, Pershagen G, Thacher JD, Lanki T, Wicki B, Röösli M, Vienneau D, Cantuaria ML, Schmidt JH, Aasvang GM, Al-Kindi S, Osborne MT, Wenzel P, Sastre J, Fleming I, Schulz R, Hahad O, Kuntic M, Zielonka J, Sies H, Grune T, Frenis K, Münzel T, Daiber A. Health position paper and redox perspectives - Disease burden by transportation noise. Redox Biol 2024; 69:102995. [PMID: 38142584 PMCID: PMC10788624 DOI: 10.1016/j.redox.2023.102995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/07/2023] [Accepted: 12/10/2023] [Indexed: 12/26/2023] Open
Abstract
Transportation noise is a ubiquitous urban exposure. In 2018, the World Health Organization concluded that chronic exposure to road traffic noise is a risk factor for ischemic heart disease. In contrast, they concluded that the quality of evidence for a link to other diseases was very low to moderate. Since then, several studies on the impact of noise on various diseases have been published. Also, studies investigating the mechanistic pathways underlying noise-induced health effects are emerging. We review the current evidence regarding effects of noise on health and the related disease-mechanisms. Several high-quality cohort studies consistently found road traffic noise to be associated with a higher risk of ischemic heart disease, heart failure, diabetes, and all-cause mortality. Furthermore, recent studies have indicated that road traffic and railway noise may increase the risk of diseases not commonly investigated in an environmental noise context, including breast cancer, dementia, and tinnitus. The harmful effects of noise are related to activation of a physiological stress response and nighttime sleep disturbance. Oxidative stress and inflammation downstream of stress hormone signaling and dysregulated circadian rhythms are identified as major disease-relevant pathomechanistic drivers. We discuss the role of reactive oxygen species and present results from antioxidant interventions. Lastly, we provide an overview of oxidative stress markers and adverse redox processes reported for noise-exposed animals and humans. This position paper summarizes all available epidemiological, clinical, and preclinical evidence of transportation noise as an important environmental risk factor for public health and discusses its implications on the population level.
Collapse
Affiliation(s)
- Mette Sørensen
- Work, Environment and Cancer, Danish Cancer Institute, Copenhagen, Denmark; Department of Natural Science and Environment, Roskilde University, Denmark.
| | - Göran Pershagen
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jesse Daniel Thacher
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Timo Lanki
- Department of Health Security, Finnish Institute for Health and Welfare, Kuopio, Finland; School of Medicine, University of Eastern Finland, Kuopio, Finland; Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Benedikt Wicki
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Martin Röösli
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Danielle Vienneau
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Manuella Lech Cantuaria
- Work, Environment and Cancer, Danish Cancer Institute, Copenhagen, Denmark; Research Unit for ORL - Head & Neck Surgery and Audiology, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Jesper Hvass Schmidt
- Research Unit for ORL - Head & Neck Surgery and Audiology, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Gunn Marit Aasvang
- Department of Air Quality and Noise, Norwegian Institute of Public Health, Oslo, Norway
| | - Sadeer Al-Kindi
- Department of Medicine, University Hospitals, Harrington Heart & Vascular Institute, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH, 44106, USA
| | - Michael T Osborne
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, MA, USA; Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Philip Wenzel
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany; Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Juan Sastre
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Spain
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt Am Main, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - Rainer Schulz
- Institute of Physiology, Faculty of Medicine, Justus-Liebig University, Gießen, 35392, Gießen, Germany
| | - Omar Hahad
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Marin Kuntic
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Jacek Zielonka
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Helmut Sies
- Institute for Biochemistry and Molecular Biology I, Faculty of Medicine, Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Katie Frenis
- Hematology/Oncology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA; Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
| | - Thomas Münzel
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany; Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany; Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany.
| |
Collapse
|
34
|
Xie R, Wang M, Zhang C. Mechanisms of age-related hearing loss at the auditory nerve central synapses and postsynaptic neurons in the cochlear nucleus. Hear Res 2024; 442:108935. [PMID: 38113793 PMCID: PMC10842789 DOI: 10.1016/j.heares.2023.108935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 12/21/2023]
Abstract
Sound information is transduced from mechanical vibration to electrical signals in the cochlea, conveyed to and further processed in the brain to form auditory perception. During the process, spiral ganglion neurons (SGNs) are the key cells that connect the peripheral and central auditory systems by receiving information from hair cells in the cochlea and transmitting it to neurons of the cochlear nucleus (CN). Decades of research in the cochlea greatly improved our understanding of SGN function under normal and pathological conditions, especially about the roles of different subtypes of SGNs and their peripheral synapses. However, it remains less clear how SGN central terminals or auditory nerve (AN) synapses connect to CN neurons, and ultimately how peripheral pathology links to structural alterations and functional deficits in the central auditory nervous system. This review discusses recent progress about the morphological and physiological properties of different subtypes of AN synapses and associated postsynaptic CN neurons, their changes during aging, and the potential mechanisms underlying age-related hearing loss.
Collapse
Affiliation(s)
- Ruili Xie
- Department of Otolaryngology, The Ohio State University, 420 W 12th Ave, Columbus OH 43210, USA; Department of Neuroscience, The Ohio State University, 420W 12th Ave, Columbus, OH 43210, USA.
| | - Meijian Wang
- Department of Otolaryngology, The Ohio State University, 420 W 12th Ave, Columbus OH 43210, USA
| | - Chuangeng Zhang
- Department of Otolaryngology, The Ohio State University, 420 W 12th Ave, Columbus OH 43210, USA
| |
Collapse
|
35
|
Kurioka T, Mizutari K, Satoh Y, Kobayashi Y, Shiotani A. Blast-Induced Central Auditory Neurodegeneration Affects Tinnitus Development Regardless of Peripheral Cochlear Damage. J Neurotrauma 2024; 41:499-513. [PMID: 37795561 DOI: 10.1089/neu.2023.0259] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2023] Open
Abstract
Blast exposure causes serious complications, the most common of which are ear-related symptoms such as hearing loss and tinnitus. The blast shock waves can cause neurodegeneration of the auditory pathway in the brainstem, as well as the cochlea, which is the primary receptor for hearing, leading to blast-induced tinnitus. However, it is still unclear which lesion is more dominant in triggering tinnitus, the peripheral cochlea or the brainstem lesion owing to the complex pathophysiology and the difficulty in objectively measuring tinnitus. Recently, gap detection tests have been developed and are potentially well-suited for determining the presence of tinnitus. In this study, we investigated whether the peripheral cochlea or the central nervous system has a dominant effect on the generation of tinnitus using a blast-exposed mouse model with or without earplugs, which prevent cochlear damage from a blast transmitted via the external auditory canal. The results showed that the earplug (+) group, in which the cochlea was neither physiologically nor histologically damaged, showed a similar extent of tinnitus behavior in a gap prepulse inhibition of acoustic startle reflex test as the earplug (-) group, in which the explosion caused a cochlear synaptic loss in the inner hair cells and demyelination of auditory neurons. In contrast, both excitatory synapses labeled with VGLUT-1 and inhibitory synapses labeled with GAD65 were reduced in the ventral cochlear nucleus, and demyelination in the medial nucleus of the trapezoid body was observed in both groups. These disruptions significantly correlated with the presence of tinnitus behavior regardless of cochlear damage. These results indicate that the lesion in the brainstem could be dominant to the cochlear lesion in the development of tinnitus following blast exposure.
Collapse
Affiliation(s)
- Takaomi Kurioka
- Department of Otolaryngology, Head, and Neck Surgery, National Defense Medical College, Saitama, Japan
| | - Kunio Mizutari
- Department of Otolaryngology, Head, and Neck Surgery, National Defense Medical College, Saitama, Japan
| | - Yasushi Satoh
- Department of Biochemistry, National Defense Medical College, Saitama, Japan
| | - Yasushi Kobayashi
- Department of Anatomy, National Defense Medical College, Saitama, Japan
| | - Akihiro Shiotani
- Department of Otolaryngology, Head, and Neck Surgery, National Defense Medical College, Saitama, Japan
| |
Collapse
|
36
|
Rincon Sabatino S, Rivero A, Sangaletti R, Dietrich WD, Hoffer ME, King CS, Rajguru SM. Targeted therapeutic hypothermia protects against noise induced hearing loss. Front Neurosci 2024; 17:1296458. [PMID: 38292902 PMCID: PMC10826421 DOI: 10.3389/fnins.2023.1296458] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/14/2023] [Indexed: 02/01/2024] Open
Abstract
Introduction Exposure to occupational or recreational loud noise activates multiple biological regulatory circuits and damages the cochlea, causing permanent changes in hearing sensitivity. Currently, no effective clinical therapy is available for the treatment or mitigation of noise-induced hearing loss (NIHL). Here, we describe an application of localized and non-invasive therapeutic hypothermia and targeted temperature management of the inner ear to prevent NIHL. Methods We developed a custom-designed cooling neck collar to reduce the temperature of the inner ear by 3-4°C post-injury to deliver mild therapeutic hypothermia. Results This localized and non-invasive therapeutic hypothermia successfully mitigated NIHL in rats. Our results show that mild hypothermia can be applied quickly and safely to the inner ear following noise exposure. We show that localized hypothermia after NIHL preserves residual hearing and rescues noise-induced synaptopathy over a period of months. Discussion This study establishes a minimally-invasive therapeutic paradigm with a high potential for rapid translation to the clinic for long-term preservation of hearing health.
Collapse
Affiliation(s)
| | - Andrea Rivero
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
| | - Rachele Sangaletti
- The Miami Project to Cure Paralysis, University of Miami, Coral Gables, FL, United States
| | - W. Dalton Dietrich
- Department of Otolaryngology, University of Miami, Coral Gables, FL, United States
| | - Michael E. Hoffer
- The Miami Project to Cure Paralysis, University of Miami, Coral Gables, FL, United States
| | | | - Suhrud M. Rajguru
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
- The Miami Project to Cure Paralysis, University of Miami, Coral Gables, FL, United States
- RestorEar Devices LLC, Bozeman, MT, United States
| |
Collapse
|
37
|
Yang ZJ, Zhao CL, Liang WQ, Chen ZR, Du ZD, Gong SS. ROS-induced oxidative stress and mitochondrial dysfunction: a possible mechanism responsible for noise-induced ribbon synaptic damage. Am J Transl Res 2024; 16:272-284. [PMID: 38322575 PMCID: PMC10839402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/05/2024] [Indexed: 02/08/2024]
Abstract
Evidence suggests that damage to the ribbon synapses (RS) may be the main cause of auditory dysfunction in noise-induced hearing loss (NIHL). Oxidative stress is implicated in the pathophysiology of synaptic damage. However, the relationship between oxidative stress and RS damage in NIHL remains unclear. To investigate the hypothesis that noise-induced oxidative stress is a key factor in synaptic damage within the inner ear, we conducted a study using mice subjected to single or repeated noise exposure (NE). We assessed auditory function using auditory brainstem response (ABR) test and examined cochlear morphology by immunofluorescence staining. The results showed that mice that experienced a single NE exhibited a threshold shift and recovered within two weeks. The ABR wave I latencies were prolonged, and the amplitudes decreased, suggesting RS dysfunction. These changes were also demonstrated by the loss of RS as evidenced by immunofluorescence staining. However, we observed threshold shifts that did not return to baseline levels following secondary NE. Additionally, ABR wave I latencies and amplitudes exhibited notable changes. Immunofluorescence staining indicated not only severe damage to RS but also loss of outer hair cells. We also noted decreased T-AOC, ATP, and mitochondrial membrane potential levels, alongside increased hydrogen peroxide concentrations post-NE. Furthermore, the expression levels of 4-HNE and 8-OHdG in the cochlea were notably elevated. Collectively, our findings suggest that the production of reactive oxygen species leads to oxidative damage in the cochlea. This mitochondrial dysfunction consequently contributes to the loss of RS, precipitating an early onset of NIHL.
Collapse
Affiliation(s)
- Zi-Jing Yang
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical UniversityBeijing 100050, China
- Clinical Center for Hearing Loss, Capital Medical UniversityBeijing 100050, China
| | - Chun-Li Zhao
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical UniversityBeijing 100050, China
- Clinical Center for Hearing Loss, Capital Medical UniversityBeijing 100050, China
| | - Wen-Qi Liang
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical UniversityBeijing 100050, China
- Clinical Center for Hearing Loss, Capital Medical UniversityBeijing 100050, China
| | - Zhong-Rui Chen
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical UniversityBeijing 100050, China
- Clinical Center for Hearing Loss, Capital Medical UniversityBeijing 100050, China
| | - Zheng-De Du
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical UniversityBeijing 100050, China
- Clinical Center for Hearing Loss, Capital Medical UniversityBeijing 100050, China
| | - Shu-Sheng Gong
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical UniversityBeijing 100050, China
- Clinical Center for Hearing Loss, Capital Medical UniversityBeijing 100050, China
| |
Collapse
|
38
|
Austin TT, Thomas CL, Warren B. Auditory robustness and resilience in the aging auditory system of the desert locust. Neurobiol Aging 2024; 133:39-50. [PMID: 37913625 DOI: 10.1016/j.neurobiolaging.2023.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/19/2023] [Accepted: 09/19/2023] [Indexed: 11/03/2023]
Abstract
After overexposure to loud music, we experience a decrease in our ability to hear (robustness), which usually recovers (resilience). Here, we exploited the amenable auditory system of the desert locust, Schistocerca gregaria, to measure how robustness and resilience depend on age. We found that gene expression changes are dominated by age as opposed to noise exposure. We measured sound-evoked nerve activity for young and aged locusts directly, after 24 hours and 48 hours after noise exposure. We found that both young and aged locusts recovered their auditory nerve function over 48 hours. We also measured the sound-evoked transduction current in individual auditory neurons, and although the transduction current magnitude recovered in the young locusts after noise exposure, it failed to recover in the aged locusts. A plastic mechanism compensates for the decreased transduction current in aged locusts. We suggest key genes upregulated in young noise-exposed locusts that mediate robustness to noise exposure and find potential candidates responsible for compensatory mechanisms in the auditory neurons of aged noise-exposed locusts.
Collapse
Affiliation(s)
- Thomas T Austin
- Neurogenetics Group, College of Life Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Christian L Thomas
- Neurogenetics Group, College of Life Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Ben Warren
- Neurogenetics Group, College of Life Sciences, University of Leicester, Leicester LE1 7RH, UK.
| |
Collapse
|
39
|
Warren B, Eberl D. What can insects teach us about hearing loss? J Physiol 2024; 602:297-316. [PMID: 38128023 DOI: 10.1113/jp281281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
Over the last three decades, insects have been utilized to provide a deep and fundamental understanding of many human diseases and disorders. Here, we present arguments for insects as models to understand general principles underlying hearing loss. Despite ∼600 million years since the last common ancestor of vertebrates and invertebrates, we share an overwhelming degree of genetic homology particularly with respect to auditory organ development and maintenance. Despite the anatomical differences between human and insect auditory organs, both share physiological principles of operation. We explain why these observations are expected and highlight areas in hearing loss research in which insects can provide insight. We start by briefly introducing the evolutionary journey of auditory organs, the reasons for using insect auditory organs for hearing loss research, and the tools and approaches available in insects. Then, the first half of the review focuses on auditory development and auditory disorders with a genetic cause. The second half analyses the physiological and genetic consequences of ageing and short- and long-term changes as a result of noise exposure. We finish with complex age and noise interactions in auditory systems. In this review, we present some of the evidence and arguments to support the use of insects to study mechanisms and potential treatments for hearing loss in humans. Obviously, insects cannot fully substitute for all aspects of human auditory function and loss of function, although there are many important questions that can be addressed in an animal model for which there are important ethical, practical and experimental advantages.
Collapse
Affiliation(s)
- Ben Warren
- Neurogenetics Group, College of Life Sciences, University of Leicester, Leicester, UK
| | - Daniel Eberl
- Department of Biology, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
40
|
Aarhus L, Molaug I, Engdahl B. No accelerated 20-year hearing decline after occupational noise exposure has ceased: The HUNT study. Am J Ind Med 2024; 67:10-17. [PMID: 37830428 DOI: 10.1002/ajim.23543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/06/2023] [Accepted: 10/02/2023] [Indexed: 10/14/2023]
Abstract
OBJECTIVES It has been suggested that noise exposure can accelerate hearing decline after the noise exposure has ceased. We aimed to assess long-term hearing decline in persons with and without prior occupational noise exposure. METHODS We conducted a population-based longitudinal study in Norway using the Trøndelag Health Study (HUNT) from 1996 to 1998 (baseline) and from 2017 to 2019 (follow-up). The sample included 1648 participants with baseline age ≥55 years (42% men, mean age 60 years) and <5 years occupational noise exposure after baseline. We analyzed the association between occupational noise exposure before baseline and mean hearing decline between 1998 and 2018 (20-year decline) at each frequency, adjusted for age, sex, education, and impulse noise exposure before baseline. RESULTS Occupational noise exposure before baseline (N = 603) was associated with baseline hearing loss, but not with later accelerated 20-year decline, at any frequency. Noise-exposed persons had less subsequent 20-year decline at 3 kHz than did nonexposed. Restricting the noise-exposed group to persons who also had a baseline Coles notch (hearing thresholds at 3, 4, or 6 kHz of 10 dB or more compared with thresholds at 1 or 2 kHz and 6 or 8 kHz; N = 211), the exposed group showed less 20-year decline at both 3 and 4 kHz, as well as less accelerated 20-year decline at 8 kHz, compared with the nonexposed. CONCLUSION Our large long-term longitudinal study shows no increased risk of continuing hearing decline after occupational noise exposure has ceased. The finding supports a conclusion that ear damage stops when the noise exposure is ended.
Collapse
Affiliation(s)
- Lisa Aarhus
- Department of Occupational Medicine and Epidemiology, National Institute of Occupational Health, Oslo, Norway
- Department of Internal Medicine, Diakonhjemmet Hospital, Oslo, Norway
| | - Ina Molaug
- Department of Occupational Medicine and Epidemiology, National Institute of Occupational Health, Oslo, Norway
| | - Bo Engdahl
- Department of Physical Health and Ageing, Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
41
|
Diao T, Ma X, Fang X, Duan M, Yu L. Compensation in neuro-system related to age-related hearing loss. Acta Otolaryngol 2024; 144:30-34. [PMID: 38265951 DOI: 10.1080/00016489.2023.2295400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 12/10/2023] [Indexed: 01/26/2024]
Abstract
BACKGROUND Age-related hearing loss (ARHL) is a major cause of chronic disability among the elderly. Individuals with ARHL not only have trouble hearing sounds, but also with speech perception. As the perception of auditory information is reliant on integration between widespread brain networks to interpret auditory stimuli, both auditory and extra-auditory systems which mainly include visual, motor and attention systems, play an important role in compensating for ARHL. OBJECTIVES To better understand the compensatory mechanism of ARHL and inspire better interventions that may alleviate ARHL. METHODS We mainly focus on the existing information on ARHL-related central compensation. The compensatory effects of hearing aids (HAs) and cochlear implants (CIs) on ARHL were also discussed. RESULTS Studies have shown that ARHL can induce cochlear hair cell damage or loss and cochlear synaptopathy, which could induce central compensation including compensation of auditory and extra-auditory neural networks. The use of HAs and CIs can improve bottom-up processing by enabling 'better' input to the auditory pathways and then to the cortex by enhancing the diminished auditory signal. CONCLUSIONS The central compensation of ARHL and its possible correlation with HAs and CIs are current hotspots in the field and should be given focus in future research.
Collapse
Affiliation(s)
- Tongxiang Diao
- Department of Otolaryngology, Head and Neck Surgery, People's Hospital, Peking University, Beijing, China
| | - Xin Ma
- Department of Otolaryngology, Head and Neck Surgery, People's Hospital, Peking University, Beijing, China
| | - Xuan Fang
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Maoli Duan
- Department of Clinical Science, Intervention and Technology, Karolinska Institute, Stockholm, Sweden
- Department of Otolaryngology, Head and Neck Surgery & Audiology and Neurotology, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden
| | - Lisheng Yu
- Department of Otolaryngology, Head and Neck Surgery, People's Hospital, Peking University, Beijing, China
| |
Collapse
|
42
|
De Poortere N, Verhulst S, Degeest S, Keshishzadeh S, Dhooge I, Keppler H. Evaluation of Lifetime Noise Exposure History Reporting. JOURNAL OF SPEECH, LANGUAGE, AND HEARING RESEARCH : JSLHR 2023; 66:5129-5151. [PMID: 37988687 DOI: 10.1044/2023_jslhr-23-00266] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
PURPOSE The purpose of this study is to critically evaluate lifetime noise exposure history (LNEH) reporting. First, two different approaches to evaluate the cumulative LNEH were compared. Second, individual LNEH was associated with the subjects' hearing status. Third, loudness estimates of exposure activities, by means of Jokitulppo- and Ferguson-based exposure levels, were compared with dosimeter sound-level measurements. METHOD One hundred one young adults completed the questionnaires, and a subgroup of 30 subjects underwent audiological assessment. Pure-tone audiometry, speech-in-noise intelligibility, distortion product otoacoustic emissions, auditory brainstem responses, and envelope following responses were included. Fifteen out of the 30 subjects took part in a noisy activity while wearing a dosimeter. RESULTS First, results demonstrate that the structured questionnaire yielded a greater amount of information pertaining to the diverse activities, surpassing the insights obtained from an open-ended questionnaire. Second, no significant correlations between audiological assessment and LNEH were found. Lastly, the results indicate that Ferguson-based exposure levels offer a more precise estimation of the actual exposure levels, in contrast to Jokitulppo-based estimates. CONCLUSIONS We propose several recommendations for determining the LNEH. First, it is vital to define accurate loudness categories and corresponding allocated levels, with a preference for the loudness levels proposed by Ferguson et al. (2019), as identified in this study. Second, a structured questionnaire regarding LNEH is recommended, discouraging open-ended questioning. Third, it is essential to include a separate category exclusively addressing work-related activities, encompassing various activities for more accurate surveying.
Collapse
Affiliation(s)
- Nele De Poortere
- Department of Rehabilitation Sciences-Audiology, Ghent University, Belgium
| | - Sarah Verhulst
- Department of Information Technology-Hearing Technology at WAVES, Ghent University, Belgium
| | - Sofie Degeest
- Department of Rehabilitation Sciences-Audiology, Ghent University, Belgium
| | - Sarineh Keshishzadeh
- Department of Information Technology-Hearing Technology at WAVES, Ghent University, Belgium
| | - Ingeborg Dhooge
- Department of Ear, Nose and Throat, Ghent University Hospital, Belgium
- Department of Head and Skin, Ghent University, Belgium
| | - Hannah Keppler
- Department of Rehabilitation Sciences-Audiology, Ghent University, Belgium
- Department of Head and Skin, Ghent University, Belgium
| |
Collapse
|
43
|
Boussaty EC, Tedeschi N, Novotny M, Ninoyu Y, Du E, Draf C, Zhang Y, Manor U, Scheuermann RH, Friedman R. Cochlear transcriptome analysis of an outbred mouse population (CFW). Front Cell Neurosci 2023; 17:1256619. [PMID: 38094513 PMCID: PMC10716316 DOI: 10.3389/fncel.2023.1256619] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/11/2023] [Indexed: 12/20/2023] Open
Abstract
Age-related hearing loss (ARHL) is the most common cause of hearing loss and one of the most prevalent conditions affecting the elderly worldwide. Despite evidence from our lab and others about its polygenic nature, little is known about the specific genes, cell types, and pathways involved in ARHL, impeding the development of therapeutic interventions. In this manuscript, we describe, for the first time, the complete cell-type specific transcriptome of the aging mouse cochlea using snRNA-seq in an outbred mouse model in relation to auditory threshold variation. Cochlear cell types were identified using unsupervised clustering and annotated via a three-tiered approach-first by linking to expression of known marker genes, then using the NSForest algorithm to select minimum cluster-specific marker genes and reduce dimensional feature space for statistical comparison of our clusters with existing publicly-available data sets on the gEAR website, and finally, by validating and refining the annotations using Multiplexed Error Robust Fluorescence In Situ Hybridization (MERFISH) and the cluster-specific marker genes as probes. We report on 60 unique cell-types expanding the number of defined cochlear cell types by more than two times. Importantly, we show significant specific cell type increases and decreases associated with loss of hearing acuity implicating specific subsets of hair cell subtypes, ganglion cell subtypes, and cell subtypes within the stria vascularis in this model of ARHL. These results provide a view into the cellular and molecular mechanisms responsible for age-related hearing loss and pathways for therapeutic targeting.
Collapse
Affiliation(s)
- Ely Cheikh Boussaty
- Department of Otolaryngology, University of California, San Diego, La Jolla, CA, United States
| | - Neil Tedeschi
- J. Craig Venter Institute, La Jolla, CA, United States
| | - Mark Novotny
- J. Craig Venter Institute, La Jolla, CA, United States
| | - Yuzuru Ninoyu
- Department of Otolaryngology, University of California, San Diego, La Jolla, CA, United States
| | - Eric Du
- Department of Otolaryngology, University of California, San Diego, La Jolla, CA, United States
| | - Clara Draf
- Department of Otolaryngology, University of California, San Diego, La Jolla, CA, United States
| | - Yun Zhang
- J. Craig Venter Institute, La Jolla, CA, United States
| | - Uri Manor
- Department of Cell and Developmental Biology, University of California San Diego, Salk Institute for Biological Studies, Waitt Advanced Biophotonics Center, La Jolla, CA, United States
| | - Richard H. Scheuermann
- J. Craig Venter Institute, La Jolla, CA, United States
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States
| | - Rick Friedman
- Department of Otolaryngology, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
44
|
Sung CYW, Hayase N, Yuen PS, Lee J, Fernandez K, Hu X, Cheng H, Star RA, Warchol ME, Cunningham LL. Macrophage Depletion Protects Against Cisplatin-Induced Ototoxicity and Nephrotoxicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.16.567274. [PMID: 38014097 PMCID: PMC10680818 DOI: 10.1101/2023.11.16.567274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Cisplatin is a widely used and highly effective anti-cancer drug with significant side effects including ototoxicity and nephrotoxicity. Macrophages, the major resident immune cells in the cochlea and kidney, are important drivers of both inflammatory and tissue repair responses. To investigate the roles of macrophages in cisplatin-induced ototoxicity and nephrotoxicity, we used PLX3397, an FDA-approved inhibitor of the colony-stimulating factor 1 receptor (CSF1R), to eliminate tissue-resident macrophages during the course of cisplatin administration. Mice treated with cisplatin alone (cisplatin/vehicle) had significant hearing loss (ototoxicity) as well as kidney injury (nephrotoxicity). Macrophage ablation using PLX3397 resulted in significantly reduced hearing loss measured by auditory brainstem responses (ABR) and distortion-product otoacoustic emissions (DPOAE). Sensory hair cells in the cochlea were protected against cisplatin-induced death in mice treated with PLX3397. Macrophage ablation also protected against cisplatin-induced nephrotoxicity, as evidenced by markedly reduced tubular injury and fibrosis as well as reduced plasma blood urea nitrogen (BUN) and neutrophil gelatinase-associated lipocalin (NGAL) levels. Mechanistically, our data suggest that the protective effect of macrophage ablation against cisplatin-induced ototoxicity and nephrotoxicity is mediated by reduced platinum accumulation in both the inner ear and the kidney. Together our data indicate that ablation of tissue-resident macrophages represents a novel strategy for mitigating cisplatin-induced ototoxicity and nephrotoxicity.
Collapse
Affiliation(s)
- Cathy Yea Won Sung
- Laboratory of Hearing Biology and Therapeutics, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, Maryland, USA
| | - Naoki Hayase
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Peter S.T. Yuen
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - John Lee
- Laboratory of Hearing Biology and Therapeutics, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, Maryland, USA
| | - Katharine Fernandez
- Laboratory of Hearing Biology and Therapeutics, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, Maryland, USA
| | - Xuzhen Hu
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Hui Cheng
- Bioinformatics and Biostatistics Collaboration Core, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, Maryland, USA
| | - Robert A. Star
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Mark E. Warchol
- Washington University, Department of Otolaryngology, School of Medicine, Saint Louis, MO
| | - Lisa L. Cunningham
- Laboratory of Hearing Biology and Therapeutics, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, Maryland, USA
| |
Collapse
|
45
|
Lau IH, Vasconcelos RO. Noise-induced damage in the zebrafish inner ear endorgans: evidence for higher acoustic sensitivity of saccular and lagenar hair cells. J Exp Biol 2023; 226:jeb245992. [PMID: 37767687 DOI: 10.1242/jeb.245992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023]
Abstract
The three otolithic endorgans of the inner ear are known to be involved in sound detection in different teleost fishes, yet their relative roles for auditory-vestibular functions within the same species remain uncertain. In zebrafish (Danio rerio), the saccule and utricle are thought to play key functions in encoding auditory and vestibular information, respectively, but the biological function of the lagena is not clear. We hypothesized that the zebrafish saccule serves as a primary auditory endorgan, making it more vulnerable to noise exposure, and that the lagena might have an auditory function given its connectivity to the saccule and the dominant vestibular function of the utricle. We compared the impact of acoustic trauma (continuous white noise at 168 dB for 24 h) between the sensory epithelia of the three otolithic endorgans. Noise treatment caused hair cell loss in both the saccule and lagena but not in the utricle. This effect was identified immediately after acoustic treatment and did not increase 24 h post-trauma. Furthermore, hair cell loss was accompanied by a reduction in presynaptic activity measured based on ribeye b presence, but mainly in the saccule, supporting its main contribution for noise-induced hearing loss. Our findings support the hypothesis that the saccule plays a major role in sound detection and that the lagena is also acoustically affected, extending the species hearing dynamic range.
Collapse
Affiliation(s)
- Ieng Hou Lau
- Institute of Science and Environment, University of Saint Joseph, Macao, S.A.R., China
| | - Raquel O Vasconcelos
- Institute of Science and Environment, University of Saint Joseph, Macao, S.A.R., China
- MARE - Marine and Environmental Sciences Centre/ARNET - Aquatic Research Network, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisbon, Portugal
- EPCV-Department of Life Sciences, Lusófona University, 1749-024 Lisbon, Portugal
| |
Collapse
|
46
|
Brokaw EB, S Brungart D, M Byrne R, A Flamme G, Gupta R, Jokel CR, Kujawa SG, Lalis L, L McKinley R, Murphy WJ, W Spencer R, J Smalt C, F Zagadou B. Recommendations for a Military Health System Auditory Blast Injury Prevention Standard. Mil Med 2023; 188:176-184. [PMID: 37948248 DOI: 10.1093/milmed/usad078] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/06/2023] [Accepted: 03/22/2023] [Indexed: 11/12/2023] Open
Abstract
INTRODUCTION Although existing auditory injury prevention standards benefit warfighters, the Department of Defense could do more to understand and address auditory injuries (e.g., hearing loss, tinnitus, and central processing deficits) among service members. The Blast Injury Prevention Standards Recommendation (BIPSR) Process is designed to address the needs of all the Military Services for biomedically valid Military Health System (MHS) Blast Injury Prevention Standards. MATERIALS AND METHODS Through the BIPSR Process, stakeholders provided their intended uses and requested functionalities for an MHS Blast Injury Prevention Standard. The BIPSR Process established a broad-based, non-advocacy panel of auditory injury Subject Matter Expert (SME) Panel with members drawn from industry, academia, and government. The SME Panel selected evaluation factors, weighted priorities, and then evaluated the resulting candidate MHS Auditory Blast Injury Prevention Standards against the evaluation criteria. The SME Panel members provided rationales for their decisions, documented discussions, and used iterative rounds of feedback to promote consensus building among members. The BIPSR Process used multi-attribute utility theory to combine members' evaluations and compare the candidate standards. RESULTS The SME Panel identified and collated information about existing auditory injury datasets to identify gaps and promote data sharing and comprehensive evaluations of standards for preventing auditory blast injury. The panel evaluated the candidate standards and developed recommendations for an MHS Blast Injury Prevention Standard. CONCLUSIONS The BIPSR Process illuminated important characteristics, capabilities, and limitations of candidate standards and existing datasets (e.g., limited human exposure data to evaluate the validity of injury prediction) for auditory blast injury prevention. The evaluation resulted in the recommendation to use the 8-hour Equivalent Level (LAeq8hr) as the interim MHS Auditory Blast Injury Prevention Standard while the community performs additional research to fill critical knowledge gaps.
Collapse
Affiliation(s)
| | | | | | - Greg A Flamme
- Stephenson and Stephenson Research and Consulting, LLC, Forest Grove, OR 97116, USA
| | - Raj Gupta
- U.S. Army Medical Research and Development Command, Frederick, MD 21702-501, USA
| | - Charles R Jokel
- Defense Centers for Public Health-Aberdeen, Aberdeen Proving Ground, MD 21010-5403, USA
| | | | - Lisa Lalis
- The MITRE Corporation, McLean, VA 22102, USA
| | | | - William J Murphy
- Stephenson and Stephenson Research and Consulting, LLC, Forest Grove, OR 97116, USA
| | | | - Christopher J Smalt
- Massachusetts Institute of Technology Lincoln Laboratory, 244 Wood St, Lexington, Massachusetts 02421, USA
| | | |
Collapse
|
47
|
Lutze RD, Ingersoll MA, Thotam A, Joseph A, Fernandes J, Teitz T. ERK1/2 Inhibition Alleviates Noise-Induced Hearing Loss While Tempering Down the Immune Response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.18.563007. [PMID: 37905140 PMCID: PMC10614960 DOI: 10.1101/2023.10.18.563007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Noise-induced hearing loss (NIHL) is a major cause of hearing impairment, yet no FDA-approved drugs exist to prevent it. Targeting the mitogen activated protein kinase (MAPK) cellular pathway has emerged as a promising approach to attenuate NIHL. Tizaterkib is an orally bioavailable, highly specific ERK1/2 inhibitor, currently in Phase-1 anticancer clinical trials. Here, we tested tizaterkib's efficacy against permanent NIHL in mice at doses equivalent to what humans are currently prescribed in clinical trials. The drug given orally 24 hours after noise exposure, protected an average of 20-25 dB SPL in three frequencies, in female and male mice, had a therapeutic window >50, and did not confer additional protection to KSR1 genetic knockout mice, showing the drug works through the MAPK pathway. Tizaterkib shielded from noise-induced cochlear synaptopathy, and a 3-day, twice daily, treatment with the drug was the optimal determined regimen. Importantly, tizaterkib was shown to decrease the number of CD45 and CD68 positive immune cells in the cochlea following noise exposure, which could be part of the protective mechanism of MAPK inhibition.
Collapse
Affiliation(s)
- Richard D. Lutze
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Matthew A. Ingersoll
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Alena Thotam
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Anjali Joseph
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Joshua Fernandes
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Tal Teitz
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE 68178, USA
| |
Collapse
|
48
|
Snapp HA, Vanlooy L, Kuzbyt B, Kolberg C, Laffitte-Lopez D, Rajguru S. Peripheral vestibular loss in noise-exposed firefighters. Front Integr Neurosci 2023; 17:1236661. [PMID: 37849955 PMCID: PMC10577377 DOI: 10.3389/fnint.2023.1236661] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/25/2023] [Indexed: 10/19/2023] Open
Abstract
Introduction Occupational workers are increasingly aware of the risk of noise overexposure to the auditory system but lack awareness about potential risks to the vestibular system. The purpose of this study was to investigate changes in vestibular end organ function in a known at-risk noise-exposed population, firefighters compared to age- and sex-matched controls using electrophysiologic measures of cervical vestibular evoked myogenic potentials (cVEMP). Methods A cross-sectional observational study compared cVEMP response characteristics in 38 noise-exposed firefighters. Firefighters were grouped by years of exposure in the fire service. The cVEMP responses were compared within firefighter groups and between firefighters and age- and sex-matched controls. Dependent variables included the response characteristics of amplitude, latency and threshold. Results cVEMP response amplitudes were significantly decreased in firefighters compared to their age- and sex-matched controls. Threshold of the cVEMP responses were significantly higher in firefighters compared to controls and firefighters had a higher incidence of absent cVEMP responses compared to controls. Response amplitudes decreased with increasing years in the fire-service at an increased rate compared to their age- and sex-matched controls. Latency of the cVEMP response was not significantly different in firefighters compared to controls. These findings are consistent with both animal and human studies suggesting noise-induced changes in the sacculocollic pathway. Discussion In the absence of any reported vestibular symptoms or auditory indicators of noise-induced hearing loss, these early effects on the vestibular system point to a potential hidden vestibular loss.
Collapse
Affiliation(s)
- Hillary Anne Snapp
- Department of Otolaryngology, University of Miami, Miami, FL, United States
| | - Lindsey Vanlooy
- Department of Otolaryngology, University of Miami, Miami, FL, United States
| | - Brianna Kuzbyt
- Department of Otolaryngology, University of Miami, Miami, FL, United States
| | - Courtney Kolberg
- Department of Otolaryngology, University of Miami, Miami, FL, United States
| | | | - Suhrud Rajguru
- Department of Otolaryngology, University of Miami, Miami, FL, United States
- Department of Biomedical Engineering, University of Miami, Miami, FL, United States
- Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL, United States
| |
Collapse
|
49
|
Wu PZ, O'Malley JT, Liberman MC. Neural Degeneration in Normal-Aging Human Cochleas: Machine-Learning Counts and 3D Mapping in Archival Sections. J Assoc Res Otolaryngol 2023; 24:499-511. [PMID: 37957485 PMCID: PMC10695900 DOI: 10.1007/s10162-023-00909-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 09/03/2023] [Indexed: 11/15/2023] Open
Abstract
Quantifying the survival patterns of spiral ganglion cells (SGCs), the cell bodies of auditory-nerve fibers, is critical to studies of sensorineural hearing loss, especially in human temporal bones. The classic method of manual counting is tedious, and, although stereology approaches can be faster, they can only be used to estimate total cell numbers per cochlea. Here, a machine-learning algorithm that automatically identifies, counts, and maps the SGCs in digitized images of semi-serial human temporal-bone sections not only speeds the analysis, with no loss of accuracy, but also allows 3D visualization of the SGCs and fine-grained mapping to cochlear frequency. Applying the algorithm to 62 normal-aging human ears shows significantly faster degeneration of SGCs in the basal than the apical half of the cochlea. Comparison to fiber counts in the same ears shows that the fraction of surviving SGCs lacking a peripheral axon steadily increases with age, reaching more than 50% in the apical cochlea and almost 66% in basal regions.
Collapse
Affiliation(s)
- Pei-Zhe Wu
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear Infirmary, 243 Charles St., Boston, MA, 02114-3096, USA.
- Department of Otolaryngology, Harvard Medical School, Boston, MA, 02115, USA.
| | - Jennifer T O'Malley
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear Infirmary, 243 Charles St., Boston, MA, 02114-3096, USA
- Department of Otolaryngology, Harvard Medical School, Boston, MA, 02115, USA
| | - M Charles Liberman
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear Infirmary, 243 Charles St., Boston, MA, 02114-3096, USA
- Department of Otolaryngology, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
50
|
Hu S, Sun Q, Xu F, Jiang N, Gao J. Age-related hearing loss and its potential drug candidates: a systematic review. Chin Med 2023; 18:121. [PMID: 37730634 PMCID: PMC10512576 DOI: 10.1186/s13020-023-00825-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 08/25/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND Age-related hearing loss (ARHL) is one of the main illnesses afflicting the aged population and has a significant negative impact on society, economy, and health. However, there is presently no appropriate therapeutic treatment of ARHL due to the absence of comprehensive trials. OBJECTIVES The goal of this review is to systematically evaluate and analyze recent statistics on the pathologic classifications, risk factors, treatment strategies, and drug candidates of ARHL, including that from traditional Chinese medicine (TCM), to provide potential new approaches for preventing and treating ARHL. METHODS Literature related to ARHL was conducted in databases such as PubMed, WOS, China National Knowledge Infrastructure (CNKI), and Wanfang from the establishment of the database to Jan, 2023. The pathology, causal factor, pathophysiological mechanism, treatment strategy, and the drug candidate of ARHL were extracted and pooled for synthesis. RESULTS Many hypotheses about the etiology of ARHL are based on genetic and environmental elements. Most of the current research on the pathology of ARHL focuses on oxidative damage, mitochondrial dysfunction, inflammation, cochlear blood flow, ion homeostasis, etc. In TCM, herbs belonging to the kidney, lung, and liver meridians exhibit good hearing protection. Seven herbs belonging to the kidney meridian, 9 belonging to the lung meridian, and 4 belonging to the liver meridian were ultimately retrieved in this review, such as Polygonum multiflorum Thunb., Panax ginseng C.A. Mey, and Pueraria lobata (Willd.) Ohwi. Their active compounds, 2,3,4',5-Tetrahydroxystilbene-2-O-D-glucoside, ginsenoside Rb1, and puerarin, may act as the molecular substance for their anti-ARHL efficacy, and show anti-oxidative, neuroprotective, anti-inflammatory, anti-apoptotic, or mitochondrial protective effects. CONCLUSION Anti-oxidants, modulators of mitochondrial function, anti-inflammation agents, vasodilators, K+ channel openers, Ca2+ channel blockers, JNK inhibitors, and nerve growth factors/neurotrophic factors all contribute to hearing protection, and herbs are an important source of potential anti-ARHL drugs.
Collapse
Affiliation(s)
- Shiyu Hu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China
| | - Qingru Sun
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China
| | - Fei Xu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China
| | - Ninghua Jiang
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, 314000, Zhejiang, People's Republic of China
| | - Jianli Gao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China.
| |
Collapse
|