1
|
Bottom RT, Xu Y, Siebald C, Jung J, Müller U. Defects in hair cells disrupt the development of auditory peripheral circuitry. Nat Commun 2024; 15:10899. [PMID: 39738090 DOI: 10.1038/s41467-024-55275-x] [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: 02/22/2024] [Accepted: 12/05/2024] [Indexed: 01/01/2025] Open
Abstract
Deafness is the most common form of sensory impairment in humans and frequently caused by defects in hair cells of the inner ear. Here we demonstrate that in male mice which model recessive non-syndromic deafness (DFNB6), inactivation of Tmie in hair cells disrupts gene expression in the neurons that innervate them. This includes genes regulating axonal pathfinding and synaptogenesis, two processes that are disrupted in the inner ear of the mutant mice. Similar defects are observed in mouse models for deafness caused by mutations in other genes with primary functions in hair cells. Gene therapy targeting hair cells restores hearing and inner ear circuitry in DFNB6 model mice. We conclude that hair cell function is crucial for the establishment of peripheral auditory circuitry. Treatment modalities for deafness thus need to consider restoration of the function of both hair cells and neurons, even when the primary defect occurs in hair cells.
Collapse
Affiliation(s)
- Riley T Bottom
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Yijun Xu
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Caroline Siebald
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Jinsei Jung
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Ulrich Müller
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
2
|
Jahn KN, Kashiwagura ST, Yousuf MS. Clinical phenotype and management of sound-induced pain: Insights from adults with pain hyperacusis. THE JOURNAL OF PAIN 2024; 27:104741. [PMID: 39586560 DOI: 10.1016/j.jpain.2024.104741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/07/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Pain hyperacusis, also known as noxacusis, causes physical pain in response to sounds that do not bother most people. How sound causes excruciating pain that can last for weeks or months is not well understood, resulting in a lack of effective treatments. To gain insight into the underlying mechanisms of the condition, 32 adults attended a virtual focus group to describe their sound-induced pain. Focus group data were used to develop three follow-up surveys that aimed to identify the most common symptoms of pain hyperacusis as well as the participants' use of therapies for pain relief. All participants endorsed negative effects of pain hyperacusis on psychosocial and physical function. Most reported sound-induced burning (80.77%), stabbing (76.92%), throbbing (73.08%), and pinching (53.85%) sensations that occur either in the ear or elsewhere in the body. Participants have used numerous pharmaceutical and non-pharmaceutical interventions to alleviate their pain with varying degrees of pain relief. Benzodiazepines and nerve blockers emerged as the most effective analgesic options while non-pharmaceutical therapies were largely ineffective. Symptoms and therapeutic approaches were generally consistent with peripheral mechanistic theories of pain hyperacusis (e.g., trigeminal nerve involvement). An interdisciplinary approach to clinical studies and the development of animal models are needed to identify and treat the pathological mechanisms of pain hyperacusis. PERSPECTIVE: This article presents the physical and psychosocial consequences of debilitating sound-induced pain (i.e., pain hyperacusis) and the interventions that sufferers have sought for pain relief. The results are largely consistent with peripheral mechanistic theories (e.g., trigeminal nerve involvement) and will guide future work to investigate neural mechanisms and effective therapies.
Collapse
Affiliation(s)
- Kelly N Jahn
- Department of Speech, Language, and Hearing, The University of Texas at Dallas, 800 W Campbell Rd., Richardson, TX 75080, USA; Callier Center for Communication Disorders, The University of Texas at Dallas, 1966 Inwood Rd., Dallas, TX 75235, USA.
| | - Sean Takamoto Kashiwagura
- Department of Speech, Language, and Hearing, The University of Texas at Dallas, 800 W Campbell Rd., Richardson, TX 75080, USA; Callier Center for Communication Disorders, The University of Texas at Dallas, 1966 Inwood Rd., Dallas, TX 75235, USA.
| | - Muhammad Saad Yousuf
- Department of Neuroscience and Center for Advanced Pain Studies, The University of Texas at Dallas, 860 N. Loop Rd., Richardson, TX 75080, USA.
| |
Collapse
|
3
|
Cederroth CR, Dyhrfjeld-Johnsen J, Canlon B. Pharmacological Approaches to Hearing Loss. Pharmacol Rev 2024; 76:1063-1088. [PMID: 39164117 PMCID: PMC11549935 DOI: 10.1124/pharmrev.124.001195] [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] [Received: 02/14/2024] [Revised: 07/08/2024] [Accepted: 07/16/2024] [Indexed: 08/22/2024] Open
Abstract
Hearing disorders pose significant challenges to individuals experiencing them and their overall quality of life, emphasizing the critical need for advanced pharmacological approaches to address these conditions. Current treatment options often focus on amplification devices, cochlear implants, or other rehabilitative therapies, leaving a substantial gap regarding effective pharmacological interventions. Advancements in our understanding of the molecular and cellular mechanisms involved in hearing disorders induced by noise, aging, and ototoxicity have opened new avenues for drug development, some of which have led to numerous clinical trials, with promising results. The development of optimal drug delivery solutions in animals and humans can also enhance the targeted delivery of medications to the ear. Moreover, large genome studies contributing to a genetic understanding of hearing loss in humans combined with advanced molecular technologies in animal studies have shown a great potential to increase our understanding of the etiologies of hearing loss. The auditory system exhibits circadian rhythms and temporal variations in its physiology, its vulnerability to auditory insults, and its responsiveness to drug treatments. The cochlear clock rhythms are under the control of the glucocorticoid system, and preclinical evidence suggests that the risk/benefit profile of hearing disorder treatments using chronopharmacological approaches would be beneficial. If translatable to the bedside, such approaches may improve the outcome of clinical trials. Ongoing research into the molecular and genetic basis of auditory disorders, coupled with advancements in drug formulation and delivery as well as optimized timing of drug administration, holds great promise of more effective treatments. SIGNIFICANCE STATEMENT: Hearing disorders pose significant challenges to individuals and their overall quality of life, emphasizing the critical need for advanced pharmacological approaches to address these conditions. Ongoing research into the molecular and genetic basis of auditory disorders, coupled with advancements in drug delivery procedures and optimized timing of drug administration, holds the promise of more effective treatments.
Collapse
Affiliation(s)
- Christopher R Cederroth
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| | - Jonas Dyhrfjeld-Johnsen
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| | - Barbara Canlon
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| |
Collapse
|
4
|
Liang C, Zhai TY, Fang S, Chen J, Liu LM, Yu N, Zhao HB. ATP-gated P2x7 receptor is a major channel type at type II auditory nerves and required for hearing sensitivity efferent controlling and noise protection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.618333. [PMID: 39464017 PMCID: PMC11507775 DOI: 10.1101/2024.10.14.618333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Hearing sensitivity and noise protection are mediated and determined by negative feedback of the cochlear efferent system. Type II auditory nerves (ANs) innervate outer hair cells (OHCs) in the cochlea and provide an input to this efferent control. However, little is known about underlying channel information. Here, we report that ATP-gated P2x7 receptor had a predominant expression at type II ANs and the synaptic areas under inner hair cells and OHCs with lateral and medial olivocochlear efferent nerves. Knockout (KO) of P2x7 increased hearing sensitivity with enhanced acoustic startle response (ASR), auditory brainstem response (ABR), and cochlear microphonics (CM) by increasing OHC electromotility, an active cochlear amplifier in mammals. P2x7 KO also increased susceptibility to noise. Middle level noise exposure could impair active cochlear mechanics resulting in permanent hearing loss in P2x7 KO mice. These data demonstrate that P2x7 receptors have a critical role in type II AN function and the cochlear efferent system to control hearing sensitivity; deficiency of P2x7 receptors can impair the cochlear efferent suppression leading to hearing oversensitivity and susceptibility to noise.
Collapse
|
5
|
Jahn KN, Kashiwagura ST, Yousuf MS. Clinical phenotype and management of sound-induced pain: Insights from adults with pain hyperacusis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.19.24309185. [PMID: 38946957 PMCID: PMC11213080 DOI: 10.1101/2024.06.19.24309185] [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
Pain hyperacusis, also known as noxacusis, causes physical pain in response to everyday sounds that do not bother most people. How sound causes excruciating pain that can last for weeks or months in otherwise healthy individuals is not well understood, resulting in a lack of effective treatments. To address this gap, we identified the most salient physical and psychosocial consequences of debilitating sound-induced pain and reviewed the interventions that sufferers have sought for pain relief to gain insights into the underlying mechanisms of the condition. Adults (n = 32) with pain hyperacusis attended a virtual focus group to describe their sound-induced pain. They completed three surveys to identify common symptoms and themes that defined their condition and to describe their use of pharmaceutical and non-pharmaceutical therapies for pain relief. All participants endorsed negative effects of pain hyperacusis on psychosocial and physical function. Most reported sound-induced burning (80.77%), stabbing (76.92%), throbbing (73.08%), and pinching (53.85%) that occurs either in the ear or elsewhere in the body (i.e., referred pain). Participants reported using numerous pharmaceutical and non-pharmaceutical interventions to alleviate their pain with varying degrees of pain relief. Benzodiazepines and nerve blockers emerged as the most effective analgesic options while non-pharmaceutical therapies were largely ineffective. Symptoms of pain hyperacusis and therapeutic approaches are largely consistent with peripheral mechanistic theories of pain hyperacusis (e.g., trigeminal nerve involvement). An interdisciplinary approach to clinical studies and the development of animal models is needed to identify, validate, and treat the pathological mechanisms of pain hyperacusis.
Collapse
Affiliation(s)
- Kelly N Jahn
- Department of Speech, Language, and Hearing, The University of Texas at Dallas, Richardson, TX, USA
- Callier Center for Communication Disorders, The University of Texas at Dallas, Dallas, TX, USA
| | - Sean Takamoto Kashiwagura
- Department of Speech, Language, and Hearing, The University of Texas at Dallas, Richardson, TX, USA
- Callier Center for Communication Disorders, The University of Texas at Dallas, Dallas, TX, USA
| | - Muhammad Saad Yousuf
- Department of Neuroscience and Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
6
|
Cherri D, Formby C, Secor CA, Eddins DA. Counseling Protocol for a Transitional Intervention for Debilitating Hyperacusis. JOURNAL OF SPEECH, LANGUAGE, AND HEARING RESEARCH : JSLHR 2024; 67:1886-1902. [PMID: 38718266 PMCID: PMC11192559 DOI: 10.1044/2023_jslhr-23-00353] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 10/31/2023] [Accepted: 12/18/2023] [Indexed: 06/07/2024]
Abstract
INTRODUCTION This clinical focus article describes a structured counseling protocol for use with protected sound management and therapeutic sound in a transitional intervention for debilitating hyperacusis. The counseling protocol and its associated visual aids are crafted as a teaching tool to educate affected individuals about hyperacusis and encourage their acceptance of a transitional intervention. DESCRIPTION OF COUNSELING COMPONENTS The counseling protocol includes five components. First, the patient's audiometric results are reviewed with the patient, and the transitional intervention is introduced. An overview of peripheral auditory structures and central neural pathways and the concept of central gain are covered in the second and third components. Maladaptive hyper-gain processes within the auditory neural pathways, which underlie the hyperacusis condition, and associated connections with nonauditory processes responsible for negative reactions to hyperacusis are covered in the fourth component. Detrimental effects from misused hearing protection devices (HPDs) and the necessity to wean the patient from overuse of HPDs are also discussed. In the fifth component, the importance of therapeutic sound is introduced as a tool to downregulate hyper-gain activity within the auditory pathways; its implementation in uncontrolled and controlled sound environments is described. It is explained that, over the course of the transitional intervention, recalibration of the hyper-gain processes will be ongoing, leading to restoration of normal homeostasis within the auditory pathways. In turn, associated activation of reactive nonauditory processes, which contribute to hyperacusis-related distress, will be reduced or eliminated. As recalibration progresses, there will be less need for protected sound management and sound therapy. Sound tolerance will improve, hyperacusis will subside, and daily activities in typical healthy sound environments will again become routine. RESULTS AND CONCLUSION The combination of counseling with protected sound management and therapeutic sound is highlighted in companion reports, including a summary of the outcomes of a successful trial of the transitional intervention.
Collapse
Affiliation(s)
- Dana Cherri
- Auditory & Speech Sciences Laboratory, University of South Florida, Tampa
| | - Craig Formby
- Auditory & Speech Sciences Laboratory, University of South Florida, Tampa
- The University of Alabama, Tuscaloosa
| | - Carrie A. Secor
- Auditory & Speech Sciences Laboratory, University of South Florida, Tampa
| | - David A. Eddins
- Auditory & Speech Sciences Laboratory, University of South Florida, Tampa
- University of Central Florida, Orlando
| |
Collapse
|
7
|
Xie X, Li Y, Yan B, Peng Q, Yao R, Deng Q, Li J, Wu Y, Chen S, Yang X, Ma P. Mediation of the JNC/ILC2 pathway in DBP-exacerbated allergic asthma: A molecular toxicological study on neuroimmune positive feedback mechanism. JOURNAL OF HAZARDOUS MATERIALS 2024; 465:133360. [PMID: 38157815 DOI: 10.1016/j.jhazmat.2023.133360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Dibutyl phthalate (DBP), a commonly used plasticizer, has been found to be strongly linked to a consistently high prevalence of allergic diseases, particularly allergic asthma. Previous animal experiments have demonstrated that exposure to DBP can worsen asthma by triggering the production of calcitonin gene-related peptide (CGRP), a neuropeptide in the lung tissue. However, the precise neuroimmune mechanism and pathophysiology of DBP-exacerbated allergic asthma with the assistance of CGRP remain unclear. OBJECTIVE The present study was to investigate the potential pathophysiological mechanism in DBP-exacerbated asthma from the perspective of neural-immune interactions. METHODS AND RESULTS C57BL/6 mice were orally exposed to different concentrations (0.4, 4, 40 mg/kg) of DBP for 28 days. They were then sensitized with OVA and nebulized with OVA for 7 consecutive excitations. To investigate whether DBP exacerbates allergic asthma in OVA induced mice, we analyzed airway hyperresponsiveness and lung histopathology. To investigate the activation of JNC and TRPV1 neurons and the release of CGRP by JNC cells, we measured the levels of TRPV1 channels, calcium inward flow, and downstream neuropeptide CGRP. Results showed that TRPV1 expression, inward calcium flux, and CGRP levels were significantly elevated in the lung tissues of the 40DBP + OVA group, suggesting the release of CGRP by JNC cells. To counteract the detrimental effects of DBP mediated by CGRP, we employed olcegepant (also known as BIBN-4096), a CGRP receptor specific antagonist. Results revealed that 40DBP + OVA + olcegepant led to notable decreases in TRPV1, calcium inward flow, and CGRP expression in lung tissues compare with 40DBP + OVA, further supporting the efficacy of olcegepant. Additionally, we also conducted ILC2 flow sorting and observed that neuropeptide CGRP-activated ILC2 cells have a crucial role as key effector cells in DBP-induced neuroimmune positive feedback regulation. Finally, we examined the protein expression of CGRP, GATA3 and P-GATA3, and found that significant upregulations of CGRP and P-GATA3 in the 40DBP + OVA group, suggest that GATA3 acted as a key regulator of CGRP-activated ILC2. CONCLUSION The aforementioned studies indicate that exposure to DBP can exacerbate allergic asthma, leading to airway inflammation. This exacerbation occurs through the activation of TRPV1 in JNC, resulting in the release of CGRP. The excessive release of CGRP further promotes the release of Th2 cytokines by inducing the activation of ILC2 through GATA phosphorylation. Consequently, this process contributes to the development of airway inflammation and allergic asthma. The increased production of Th2 cytokines also triggers the production of IgE, which interacts with FcεRI on JNC neurons, thereby mediating neuro-immune positive feedback regulation.
Collapse
Affiliation(s)
- Xiaomin Xie
- Key Laboratory of Environmental Related Diseases and One Health, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Yan Li
- Key Laboratory of Environmental Related Diseases and One Health, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China; Department of Pharmacy, Ezhou Central Hospital, Ezhou 436000, China
| | - Biao Yan
- Key Laboratory of Environmental Related Diseases and One Health, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Qi Peng
- Key Laboratory of Environmental Related Diseases and One Health, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Runming Yao
- Joint International Research Laboratory of Green Buildings and Built Environments (Ministry of Education), Chongqing University, Chongqing 400045, China
| | - Qihong Deng
- School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Jinquan Li
- Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yang Wu
- Key Laboratory of Environmental Related Diseases and One Health, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Shaohui Chen
- Key Laboratory of Environmental Related Diseases and One Health, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Xu Yang
- Key Laboratory of Environmental Related Diseases and One Health, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China
| | - Ping Ma
- Key Laboratory of Environmental Related Diseases and One Health, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China.
| |
Collapse
|
8
|
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
|
9
|
Seicol BJ, Guo Z, Garrity K, Xie R. Potential uses of auditory nerve stimulation to modulate immune responses in the inner ear and auditory brainstem. Front Integr Neurosci 2023; 17:1294525. [PMID: 38162822 PMCID: PMC10755874 DOI: 10.3389/fnint.2023.1294525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024] Open
Abstract
Bioelectronic medicine uses electrical stimulation of the nervous system to improve health outcomes throughout the body primarily by regulating immune responses. This concept, however, has yet to be applied systematically to the auditory system. There is growing interest in how cochlear damage and associated neuroinflammation may contribute to hearing loss. In conjunction with recent findings, we propose here a new perspective, which could be applied alongside advancing technologies, to use auditory nerve (AN) stimulation to modulate immune responses in hearing health disorders and following surgeries for auditory implants. In this article we will: (1) review the mechanisms of inflammation in the auditory system in relation to various forms of hearing loss, (2) explore nerve stimulation to reduce inflammation throughout the body and how similar neural-immune circuits likely exist in the auditory system (3) summarize current methods for stimulating the auditory system, particularly the AN, and (4) propose future directions to use bioelectronic medicine to ameliorate harmful immune responses in the inner ear and auditory brainstem to treat refractory conditions. We will illustrate how current knowledge from bioelectronic medicine can be applied to AN stimulation to resolve inflammation associated with implantation and disease. Further, we suggest the necessary steps to get discoveries in this emerging field from bench to bedside. Our vision is a future for AN stimulation that includes additional protocols as well as advances in devices to target and engage neural-immune circuitry for therapeutic benefits.
Collapse
Affiliation(s)
- Benjamin J. Seicol
- Department of Otolaryngology, The Ohio State University, Columbus, OH, United States
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| | - Zixu Guo
- Department of Otolaryngology, The Ohio State University, Columbus, OH, United States
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| | - Katy Garrity
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| | - Ruili Xie
- Department of Otolaryngology, The Ohio State University, Columbus, OH, United States
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
10
|
Wallace MN, Palmer AR. Neural Plasticity in Tinnitus Mechanisms. Brain Sci 2023; 13:1615. [PMID: 38137063 PMCID: PMC10741551 DOI: 10.3390/brainsci13121615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/09/2023] [Accepted: 10/13/2023] [Indexed: 12/24/2023] Open
Abstract
Basic work into neuroplasticity mechanisms in both invertebrate and vertebrate brains, followed by the development of the first animal model of tinnitus, and coupled with clinical studies of tinnitus, meant that, by 1990, Jastreboff [...].
Collapse
Affiliation(s)
- Mark N. Wallace
- Hearing Sciences, Mental Health and Clinical Neurosciences, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK;
| | | |
Collapse
|
11
|
Wang M, Lin S, Xie R. Apical-basal distribution of different subtypes of spiral ganglion neurons in the cochlea and the changes during aging. PLoS One 2023; 18:e0292676. [PMID: 37883357 PMCID: PMC10602254 DOI: 10.1371/journal.pone.0292676] [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] [Received: 08/10/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023] Open
Abstract
Sound information is transmitted from the cochlea to the brain mainly by type I spiral ganglion neurons (SGNs), which consist of different subtypes with distinct physiological properties and selective expression of molecular markers. It remains unclear how these SGN subtypes distribute along the tonotopic axis, and whether the distribution pattern changes during aging that might underlie age-related hearing loss (ARHL). We investigated these questions using immunohistochemistry in three age groups of CBA/CaJ mice of either sex, including 2-5 months (young), 17-19 months (middle-age), and 28-32 months (old). Mouse cochleae were cryo-sectioned and triple-stained using antibodies against Tuj1, calretinin (CR) and calbindin (CB), which are reportedly expressed in all type I, subtype Ia, and subtype Ib SGNs, respectively. Labeled SGNs were classified into four groups based on the expression pattern of stained markers, including CR+ (subtype Ia), CB+ (subtype Ib), CR+CB+ (dual-labeled Ia/Ib), and CR-CB- (subtype Ic) neurons. The distribution of these SGN groups was analyzed in the apex, middle, and base regions of the cochleae. It showed that the prevalence of subtype Ia, Ib and dual-labeled Ia/Ib SGNs are high in the apex and low in the base. In contrast, the distribution pattern is reversed in Ic SGNs. Such frequency-dependent distribution is largely maintained during aging except for a preferential reduction of Ic SGNs, especially in the base. These findings corroborate the prior study based on RNAscope that SGN subtypes show differential vulnerability during aging. It suggests that sound processing of different frequencies involves distinct combinations of SGN subtypes, and the age-dependent loss of Ic SGNs in the base may especially impact high-frequency hearing during ARHL.
Collapse
Affiliation(s)
- Meijian Wang
- Department of Otolaryngology, The Ohio State University, Columbus, OH, United States of Ameirca
| | - Shengyin Lin
- Department of Otolaryngology, The Ohio State University, Columbus, OH, United States of Ameirca
| | - Ruili Xie
- Department of Otolaryngology, The Ohio State University, Columbus, OH, United States of Ameirca
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States of Ameirca
| |
Collapse
|
12
|
Yang Y, Murtha K, Climer LK, Ceriani F, Thompson P, Hornak AJ, Marcotti W, Simmons DD. Oncomodulin regulates spontaneous calcium signalling and maturation of afferent innervation in cochlear outer hair cells. J Physiol 2023; 601:4291-4308. [PMID: 37642186 PMCID: PMC10621907 DOI: 10.1113/jp284690] [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: 03/14/2023] [Accepted: 08/08/2023] [Indexed: 08/31/2023] Open
Abstract
Cochlear outer hair cells (OHCs) are responsible for the exquisite frequency selectivity and sensitivity of mammalian hearing. During development, the maturation of OHC afferent connectivity is refined by coordinated spontaneous Ca2+ activity in both sensory and non-sensory cells. Calcium signalling in neonatal OHCs can be modulated by oncomodulin (OCM, β-parvalbumin), an EF-hand calcium-binding protein. Here, we investigated whether OCM regulates OHC spontaneous Ca2+ activity and afferent connectivity during development. Using a genetically encoded Ca2+ sensor (GCaMP6s) expressed in OHCs in wild-type (Ocm+/+ ) and Ocm knockout (Ocm-/- ) littermates, we found increased spontaneous Ca2+ activity and upregulation of purinergic receptors in OHCs from Ocm-/- cochlea immediately following birth. The afferent synaptic maturation of OHCs was delayed in the absence of OCM, leading to an increased number of ribbon synapses and afferent fibres on Ocm-/- OHCs before hearing onset. We propose that OCM regulates the spontaneous Ca2+ signalling in the developing cochlea and the maturation of OHC afferent innervation. KEY POINTS: Cochlear outer hair cells (OHCs) exhibit spontaneous Ca2+ activity during a narrow period of neonatal development. OHC afferent maturation and connectivity requires spontaneous Ca2+ activity. Oncomodulin (OCM, β-parvalbumin), an EF-hand calcium-binding protein, modulates Ca2+ signals in immature OHCs. Using transgenic mice that endogenously expressed a Ca2+ sensor, GCaMP6s, we found increased spontaneous Ca2+ activity and upregulated purinergic receptors in Ocm-/- OHCs. The maturation of afferent synapses in Ocm-/- OHCs was also delayed, leading to an upregulation of ribbon synapses and afferent fibres in Ocm-/- OHCs before hearing onset. We propose that OCM plays an important role in modulating Ca2+ activity, expression of Ca2+ channels and afferent innervation in developing OHCs.
Collapse
Affiliation(s)
- Yang Yang
- Department of Biology, Baylor University, 101 Bagby Ave, Waco, TX
| | - Kaitlin Murtha
- Department of Biology, Baylor University, 101 Bagby Ave, Waco, TX
| | - Leslie K. Climer
- Department of Biology, Baylor University, 101 Bagby Ave, Waco, TX
| | - Federico Ceriani
- School of Biosciences, University of Sheffield, S10 2TN Sheffield, United Kingdom
| | - Pierce Thompson
- Department of Biology, Baylor University, 101 Bagby Ave, Waco, TX
| | - Aubrey J. Hornak
- Department of Biology, Baylor University, 101 Bagby Ave, Waco, TX
| | - Walter Marcotti
- School of Biosciences, University of Sheffield, S10 2TN Sheffield, United Kingdom
- Sheffield Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Dwayne D. Simmons
- Department of Biology, Baylor University, 101 Bagby Ave, Waco, TX
- School of Biosciences, University of Sheffield, S10 2TN Sheffield, United Kingdom
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
- Department of Psychology and Neuroscience, Baylor University, Waco, TX
| |
Collapse
|
13
|
Tureček R, Melichar A, Králíková M, Hrušková B. The role of GABA B receptors in the subcortical pathways of the mammalian auditory system. Front Endocrinol (Lausanne) 2023; 14:1195038. [PMID: 37635966 PMCID: PMC10456889 DOI: 10.3389/fendo.2023.1195038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
GABAB receptors are G-protein coupled receptors for the inhibitory neurotransmitter GABA. Functional GABAB receptors are formed as heteromers of GABAB1 and GABAB2 subunits, which further associate with various regulatory and signaling proteins to provide receptor complexes with distinct pharmacological and physiological properties. GABAB receptors are widely distributed in nervous tissue, where they are involved in a number of processes and in turn are subject to a number of regulatory mechanisms. In this review, we summarize current knowledge of the cellular distribution and function of the receptors in the inner ear and auditory pathway of the mammalian brainstem and midbrain. The findings suggest that in these regions, GABAB receptors are involved in processes essential for proper auditory function, such as cochlear amplifier modulation, regulation of spontaneous activity, binaural and temporal information processing, and predictive coding. Since impaired GABAergic inhibition has been found to be associated with various forms of hearing loss, GABAB dysfunction could also play a role in some pathologies of the auditory system.
Collapse
Affiliation(s)
- Rostislav Tureček
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czechia
| | - Adolf Melichar
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czechia
- Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Michaela Králíková
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czechia
| | - Bohdana Hrušková
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czechia
| |
Collapse
|
14
|
Kempfle JS, Jung DH. Experimental drugs for the prevention or treatment of sensorineural hearing loss. Expert Opin Investig Drugs 2023; 32:643-654. [PMID: 37598357 DOI: 10.1080/13543784.2023.2242253] [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: 02/15/2023] [Accepted: 07/26/2023] [Indexed: 08/22/2023]
Abstract
INTRODUCTION Sensorineural hearing loss results in irreversible loss of inner ear hair cells and spiral ganglion neurons. Reduced sound detection and speech discrimination can span all ages, and sensorineural hearing rehabilitation is limited to amplification with hearing aids or cochlear implants. Recent insights into experimental drug treatments for inner ear regeneration and otoprotection have paved the way for clinical trials in order to restore a more physiological hearing experience. Paired with the development of innovative minimally invasive approaches for drug delivery to the inner ear, new, emerging treatments for hearing protection and restoration are within reach. AREAS COVERED This expert opinion provides an overview of the latest experimental drug therapies to protect from and to restore sensorineural hearing loss. EXPERT OPINION The degree and type of cellular damage to the cochlea, the responsiveness of remaining, endogenous cells to regenerative treatments, and the duration of drug availability within cochlear fluids will determine the success of hearing protection or restoration.
Collapse
Affiliation(s)
- Judith S Kempfle
- Department of Otolaryngology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Otolaryngology, Head & Neck Surgery, Harvard Medical School, Boston, MA, USA
- Department of Otolaryngology, UMass Memorial Medical Center, Worcester, MA, USA
- Department of Otolaryngology, Head & Neck Surgery, University of Massachusetts Medical School, Worcester, MA, USA
| | - David H Jung
- Department of Otolaryngology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Otolaryngology, Head & Neck Surgery, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
15
|
Yang Y, Murtha K, Climer LK, Ceriani F, Thompson P, Hornak AJ, Marcotti W, Simmons DD. Oncomodulin Regulates Spontaneous Calcium Signaling and Maturation of Afferent Innervation in Cochlear Outer Hair Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.01.529895. [PMID: 36909575 PMCID: PMC10002690 DOI: 10.1101/2023.03.01.529895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Cochlear outer hair cells (OHCs) are responsible for the exquisite frequency selectivity and sensitivity of mammalian hearing. During development, the maturation of OHC afferent connectivity is refined by coordinated spontaneous Ca 2+ activity in both sensory and non-sensory cells. Calcium signaling in neonatal OHCs can be modulated by Oncomodulin (OCM, β-parvalbumin), an EF-hand calcium-binding protein. Here, we investigated whether OCM regulates OHC spontaneous Ca 2+ activity and afferent connectivity during development. Using a genetically encoded Ca 2+ sensor (GCaMP6s) expressed in OHCs in wild-type (Ocm +/+ ) and Ocm knockout (Ocm -/- ) littermates, we found increased spontaneous Ca 2+ activity and upregulation of purinergic receptors in OHCs from GCaMP6s Ocm -/- cochlea immediately following birth. The afferent synaptic maturation of OHCs was delayed in the absence of OCM, leading to an increased number of ribbon synapses and afferent fibers on GCaMP6s Ocm -/- OHCs before hearing onset. We propose that OCM regulates the spontaneous Ca 2+ signaling in the developing cochlea and the maturation of OHC afferent innervation.
Collapse
|
16
|
Sun G, Tang M, Wang X, Li D, Liu W, Qi J, Wang H, Hu B. Generation of human otic neuronal organoids using pluripotent stem cells. Cell Prolif 2023; 56:e13434. [PMID: 36825797 DOI: 10.1111/cpr.13434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
Otic neurons, also known as spiral ganglion neurons (SGNs) in mammalian cochlea, transmit electrical signals from sensory hair cells to cochlear nuclei of the auditory system. SGNs are sensitive to toxic insults, vulnerable to get irreversible damaged and hardly regenerate after damage, causing persistent sensorineural hearing loss. Yet, to get authentic SGNs for research or therapeutic purpose remains challenging. Here we developed a protocol to generate human otic neuronal organoids (hONOs) from human pluripotent stem cells (hESCs), in which hESCs were step-wisely induced to SGNs of the corresponding stages according to their developmental trajectory. The hONOs were enriched for SGN-like cells at early stage, and for both neurons and astrocytes, Schwann cells or supporting cells thereafter. In these hONOs, we also determined the existence of typical Type I and Type II SGNs. Mature hONOs (at differentiation Day 60) formed neural network, featured by giant depolarizing potential (GDP)-like events and rosette-organized regions-elicited calcium traces. Electrophysiological analysis confirmed the existence of glutamate-responsive neurons in these hONOs. The otic neuronal organoids generated in this study provide an ideal model to study SGNs and related disorders, facilitating therapeutic development for sensorineural hearing loss.
Collapse
Affiliation(s)
- Gaoying Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Mingming Tang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Xinyue Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Da Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Wenwen Liu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jianhuan Qi
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Haibo Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Baoyang Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
17
|
Nomdedeu-Sancho G, Alsina B. Wiring the senses: Factors that regulate peripheral axon pathfinding in sensory systems. Dev Dyn 2023; 252:81-103. [PMID: 35972036 PMCID: PMC10087148 DOI: 10.1002/dvdy.523] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 01/04/2023] Open
Abstract
Sensory neurons of the head are the ones that transmit the information about the external world to our brain for its processing. Axons from cranial sensory neurons sense different chemoattractant and chemorepulsive molecules during the journey and in the target tissue to establish the precise innervation with brain neurons and/or receptor cells. Here, we aim to unify and summarize the available information regarding molecular mechanisms guiding the different afferent sensory axons of the head. By putting the information together, we find the use of similar guidance cues in different sensory systems but in distinct combinations. In vertebrates, the number of genes in each family of guidance cues has suffered a great expansion in the genome, providing redundancy, and robustness. We also discuss recently published data involving the role of glia and mechanical forces in shaping the axon paths. Finally, we highlight the remaining questions to be addressed in the field.
Collapse
Affiliation(s)
- Gemma Nomdedeu-Sancho
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona, Barcelona, Spain
| | - Berta Alsina
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona, Barcelona, Spain
| |
Collapse
|
18
|
The purinergic receptors 2X3 on spiral ganglion neurons enhance the medial olivocochlear reflex in mice after long-term moderate noise exposure. Neuroreport 2022; 33:786-790. [PMID: 36367795 DOI: 10.1097/wnr.0000000000001846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Our purpose was to study the expression of purinergic receptors 2X2 (P2X2) and purinergic receptors 2X3 (P2X3) in spiral ganglion neurons (SGNs), the afferent nerves of medial olivocochlear (MOC) reflex, after long-term moderate noise exposure, and its relationship with the enhancement of MOC reflex. Mice were exposed a moderate broadband noise for 4 weeks consecutively. Then mouse hearing functions, including threshold auditory brainstem responses, distortion-product otoacoustic emissions, and MOC reflex, were evaluated and the expression of P2X2 and P2X3 on SGNs were assessed by cochlear immunofluorescence. AF-353 was injected before each noise exposure. Four weeks later, mice were also tested for hearing functions and expression of P2X2 and P2X3 on SGNs. The long-term moderate noise strengthened MOC reflex, and AF-353 reduced it in mice and P2X3 expression on SGNs increased after long-term moderate noise exposure, and AF-353 can downregulate it. The P2X3 on SGNs of mice increased after long-term moderate noise exposure, and the upregulation of it mediated the enhancement of MOC reflex.
Collapse
|
19
|
Sacchetto L, Apa E, Ciorba A, Palma S, Caragli V, Gherpelli C, Monzani D, Genovese E, Nocini R. Psychological Profile and Social Behaviors of Patients with Hyperacusis. J Clin Med 2022; 11:jcm11247317. [PMID: 36555934 PMCID: PMC9784993 DOI: 10.3390/jcm11247317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/02/2022] [Accepted: 12/08/2022] [Indexed: 12/13/2022] Open
Abstract
Increased noise sensitivity refers to the abnormal subjective response to external sounds, with a prevalence of between 8% and 15.2% in the adult population as suggested by epidemiological studies. The basic neural mechanism of hyperacusis still remains obscure, so therapies for this often-devastating symptom remain elusive. The aim of this study was to assess psychological profiles in patients with presbycusis without tinnitus in a perspective case-control design. All subjects were initially submitted to audiological evaluation (tympanometry, recordings of the acoustic reflex thresholds, pure tone audiometry) and subsequently were administered the following questionnaires: the hyperacusis questionnaire (HQ), the brief symptom inventory (BSI), and the modified somatic perception questionnaire (MSPQ). Patients with hyperacusis reported a total score and subscales (attentional, social, and emotional) of the HQ significantly higher than controls. They also reported higher scores of the MSPQ and significantly higher mean values with concern to the somatization, obsessive-compulsive, interpersonal sensitivity, depression, and anxiety subscales of the BSI. These results show that psychological distress, as expressed by higher level of somatic attention, somatization, anxiety, and depression, is a significant factor to consider for a complete diagnosis and effective treatment of hyperacusis. For a correct diagnosis of patients seeking help for hyperacusis, their psychological distress should also be assessed, regardless of their hearing abilities. Further studies are required to investigate the pathological mechanisms that are involved in the onset of hyperacusis in patients with normal hearing and those with sensorineural hearing loss.
Collapse
Affiliation(s)
- Luca Sacchetto
- Ear Nose and Throat (ENT), Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, 37134 Verona, Italy
| | - Enrico Apa
- Audiology, ENT Department, University of Modena and Reggio Emilia, 41100 Modena, Italy
| | - Andrea Ciorba
- ENT & Audiology Unit, Department of Neurosciences, University Hospital of Ferrara, Via Aldo Moro 8, 44124 Cona, Italy
| | - Silvia Palma
- Audiology, Primary Care Department, Ausl Modena, 41121 Modena, Italy
- Correspondence:
| | - Valeria Caragli
- Audiology, ENT Department, University of Modena and Reggio Emilia, 41100 Modena, Italy
| | - Chiara Gherpelli
- Audiology, ENT Department, University of Modena and Reggio Emilia, 41100 Modena, Italy
| | - Daniele Monzani
- Ear Nose and Throat (ENT), Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, 37134 Verona, Italy
| | - Elisabetta Genovese
- Audiology, ENT Department, University of Modena and Reggio Emilia, 41100 Modena, Italy
| | - Riccardo Nocini
- Ear Nose and Throat (ENT), Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, 37134 Verona, Italy
| |
Collapse
|
20
|
Sanders TR, Kelley MW. Specification of neuronal subtypes in the spiral ganglion begins prior to birth in the mouse. Proc Natl Acad Sci U S A 2022; 119:e2203935119. [PMID: 36409884 PMCID: PMC9860252 DOI: 10.1073/pnas.2203935119] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 10/17/2022] [Indexed: 11/22/2022] Open
Abstract
The afferent innervation of the cochlea is comprised of spiral ganglion neurons (SGNs), which are characterized into four subtypes (Type 1A, B, and C and Type 2). However, little is known about the factors and/or processes that determine each subtype. Here, we present a transcriptional analysis of approximately 5,500 single murine SGNs collected across four developmental time points. All four subtypes are transcriptionally identifiable prior to the onset of coordinated spontaneous activity, indicating that the initial specification process is under genetic control. Trajectory analysis indicates that SGNs initially split into two precursor types (Type 1A/2 and Type 1B/C), followed by subsequent splits to give rise to four transcriptionally distinct subtypes. Differential gene expression, pseudotime, and regulon analyses were used to identify candidate transcription factors which may regulate the subtypes specification process. These results provide insights into SGN development and comprise a transcriptional atlas of SGN maturation across the prenatal period.
Collapse
Affiliation(s)
- Tessa R. Sanders
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD20892
| | - Matthew W. Kelley
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD20892
| |
Collapse
|
21
|
Hyperacusis: Loudness Intolerance, Fear, Annoyance and Pain. Hear Res 2022; 426:108648. [DOI: 10.1016/j.heares.2022.108648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/02/2022] [Accepted: 11/07/2022] [Indexed: 11/10/2022]
|
22
|
Mazurek B, Rose M, Schulze H, Dobel C. Systems Medicine Approach for Tinnitus with Comorbid Disorders. Nutrients 2022; 14:nu14204320. [PMID: 36297004 PMCID: PMC9611054 DOI: 10.3390/nu14204320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/26/2022] [Accepted: 10/06/2022] [Indexed: 11/07/2022] Open
Abstract
Despite the fact that chronic diseases usually occur together with a spectrum of possible comorbidities that may differ strongly between patients, they are classically still viewed as distinct disease entities and, consequently, are often treated with uniform therapies. Unfortunately, such an approach does not take into account that different combinations of symptoms and comorbidities may result from different pathological (e.g., environmental, genetic, dietary, etc.) factors, which require specific and individualised therapeutic strategies. In this opinion paper, we aim to put forward a more differentiated, systems medicine approach to disease and patient treatment. To elaborate on this concept, we focus on the interplay of tinnitus, depression, and chronic pain. In our view, these conditions can be characterised by a variety of phenotypes composed of variable sets of symptoms and biomarkers, rather than distinct disease entities. The knowledge of the interplay of such symptoms and biomarkers will provide the key to a deeper, mechanistic understanding of disease pathologies. This paves the way for prediction and prevention of disease pathways, including more personalised and effective treatment strategies.
Collapse
Affiliation(s)
- Birgit Mazurek
- Tinnitus Center, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- Correspondence:
| | - Matthias Rose
- Medical Department, Division of Psychosomatic Medicine, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Holger Schulze
- Department of Otorhinolaryngology–Head and Neck Surgery, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Christian Dobel
- Department of Otorhinolaryngology, Jena University Hospital, 07743 Jena, Germany
| |
Collapse
|
23
|
Cederholm JME, Parley KE, Perera CJ, von Jonquieres G, Pinyon JL, Julien JP, Ryugo DK, Ryan AF, Housley GD. Noise-induced hearing loss vulnerability in type III intermediate filament peripherin gene knockout mice. Front Neurol 2022; 13:962227. [PMID: 36226085 PMCID: PMC9549866 DOI: 10.3389/fneur.2022.962227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
In the post-natal mouse cochlea, type II spiral ganglion neurons (SGNs) innervating the electromotile outer hair cells (OHCs) of the ‘cochlear amplifier' selectively express the type III intermediate filament peripherin gene (Prph). Immunolabeling showed that Prph knockout (KO) mice exhibited disruption of this (outer spiral bundle) afferent innervation, while the radial fiber (type I SGN) innervation of the inner hair cells (~95% of the SGN population) was retained. Functionality of the medial olivocochlear (MOC) efferent innervation of the OHCs was confirmed in the PrphKO, based on suppression of distortion product otoacoustic emissions (DPOAEs) via direct electrical stimulation. However, “contralateral suppression” of the MOC reflex neural circuit, evident as a rapid reduction in cubic DPOAE when noise is presented to the opposite ear in wildtype mice, was substantially disrupted in the PrphKO. Auditory brainstem response (ABR) measurements demonstrated that hearing sensitivity (thresholds and growth-functions) were indistinguishable between wildtype and PrphKO mice. Despite this comparability in sound transduction and strength of the afferent signal to the central auditory pathways, high-intensity, broadband noise exposure (108 dB SPL, 1 h) produced permanent high frequency hearing loss (24–32 kHz) in PrphKO mice but not the wildtype mice, consistent with the attenuated contralateral suppression of the PrphKO. These data support the postulate that auditory neurons expressing Prph contribute to the sensory arm of the otoprotective MOC feedback circuit.
Collapse
Affiliation(s)
- Jennie M. E. Cederholm
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Kristina E. Parley
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Chamini J. Perera
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Georg von Jonquieres
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Jeremy L. Pinyon
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Jean-Pierre Julien
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec, QC, Canada
| | - David K. Ryugo
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- School of Biomedical Sciences, UNSW Sydney, Sydney, NSW, Australia
- Department of Otolaryngology, Head, Neck & Skull Base Surgery, St Vincent's Hospital, Sydney, NSW, Australia
| | - Allen F. Ryan
- Departments of Surgery and Neurosciences, University of California, San Diego, La Jolla, CA, United States
- Veterans Administration Medical Center, La Jolla, CA, United States
| | - Gary D. Housley
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, UNSW Sydney, Sydney, NSW, Australia
- *Correspondence: Gary D. Housley
| |
Collapse
|
24
|
Heuermann ML, Matos S, Hamilton D, Cox BC. Regenerated hair cells in the neonatal cochlea are innervated and the majority co-express markers of both inner and outer hair cells. Front Cell Neurosci 2022; 16:841864. [PMID: 36187289 PMCID: PMC9524252 DOI: 10.3389/fncel.2022.841864] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 08/29/2022] [Indexed: 11/30/2022] Open
Abstract
After a damaging insult, hair cells can spontaneously regenerate from cochlear supporting cells within the first week of life. While the regenerated cells express several markers of immature hair cells and have stereocilia bundles, their capacity to differentiate into inner or outer hair cells, and ability to form new synaptic connections has not been well-described. In addition, while multiple supporting cell subtypes have been implicated as the source of the regenerated hair cells, it is unclear if certain subtypes have a greater propensity to form one hair cell type over another. To investigate this, we used two CreER mouse models to fate-map either the supporting cells located near the inner hair cells (inner phalangeal and border cells) or outer hair cells (Deiters’, inner pillar, and outer pillar cells) along with immunostaining for markers that specify the two hair cell types. We found that supporting cells fate-mapped by both CreER lines responded early to hair cell damage by expressing Atoh1, and are capable of producing regenerated hair cells that express terminal differentiation markers of both inner and outer hair cells. The majority of regenerated hair cells were innervated by neuronal fibers and contained synapses. Unexpectedly, we also found that the majority of the laterally positioned regenerated hair cells aberrantly expressed both the outer hair cell gene, oncomodulin, and the inner hair cell gene, vesicular glutamate transporter 3 (VGlut3). While this work demonstrates that regenerated cells can express markers of both inner and outer hair cells after damage, VGlut3 expression appears to lack the tight control present during embryogenesis, which leads to its inappropriate expression in regenerated cells.
Collapse
Affiliation(s)
- Mitchell L. Heuermann
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Sophia Matos
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, United States
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Deborah Hamilton
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Brandon C. Cox
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, United States
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
- *Correspondence: Brandon C. Cox,
| |
Collapse
|
25
|
Jahn KN. Clinical and investigational tools for monitoring noise-induced hyperacusis. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2022; 152:553. [PMID: 35931527 PMCID: PMC9448410 DOI: 10.1121/10.0012684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Hyperacusis is a recognized perceptual consequence of acoustic overexposure that can lead to debilitating psychosocial effects. Despite the profound impact of hyperacusis on quality of life, clinicians and researchers lack objective biomarkers and standardized protocols for its assessment. Outcomes of conventional audiologic tests are highly variable in the hyperacusis population and do not adequately capture the multifaceted nature of the condition on an individual level. This presents challenges for the differential diagnosis of hyperacusis, its clinical surveillance, and evaluation of new treatment options. Multiple behavioral and objective assays are emerging as contenders for inclusion in hyperacusis assessment protocols but most still await rigorous validation. There remains a pressing need to develop tools to quantify common nonauditory symptoms, including annoyance, fear, and pain. This review describes the current literature on clinical and investigational tools that have been used to diagnose and monitor hyperacusis, as well as those that hold promise for inclusion in future trials.
Collapse
Affiliation(s)
- Kelly N Jahn
- Department of Speech, Language, and Hearing, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080, USA
| |
Collapse
|
26
|
Deans MR. Planar cell polarity signaling guides cochlear innervation. Dev Biol 2022; 486:1-4. [DOI: 10.1016/j.ydbio.2022.03.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 03/07/2022] [Accepted: 03/13/2022] [Indexed: 01/24/2023]
|
27
|
Immediate Early Gene c-fos in the Brain: Focus on Glial Cells. Brain Sci 2022; 12:brainsci12060687. [PMID: 35741573 PMCID: PMC9221432 DOI: 10.3390/brainsci12060687] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 12/13/2022] Open
Abstract
The c-fos gene was first described as a proto-oncogene responsible for the induction of bone tumors. A few decades ago, activation of the protein product c-fos was reported in the brain after seizures and other noxious stimuli. Since then, multiple studies have used c-fos as a brain activity marker. Although it has been attributed to neurons, growing evidence demonstrates that c-fos expression in the brain may also include glial cells. In this review, we collect data showing that glial cells also express this proto-oncogene. We present evidence demonstrating that at least astrocytes, oligodendrocytes, and microglia express this immediate early gene (IEG). Unlike neurons, whose expression changes used to be associated with depolarization, glial cells seem to express the c-fos proto-oncogene under the influence of proliferation, differentiation, growth, inflammation, repair, damage, plasticity, and other conditions. The collected evidence provides a complementary view of c-fos as an activity marker and urges the introduction of the glial cell perspective into brain activity studies. This glial cell view may provide additional information related to the brain microenvironment that is difficult to obtain from the isolated neuron paradigm. Thus, it is highly recommended that detection techniques are improved in order to better differentiate the phenotypes expressing c-fos in the brain and to elucidate the specific roles of c-fos expression in glial cells.
Collapse
|
28
|
Bigras C, Villatte B, Duda V, Hébert S. The electrophysiological markers of hyperacusis: a scoping review. Int J Audiol 2022:1-11. [PMID: 35549972 DOI: 10.1080/14992027.2022.2070083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVE Hyperacusis is known as a reduced tolerance to sounds perceived as normal to the majority of the population. There is currently no agreed definition, diagnostic tool, or objective measure of its occurrence. The purpose of this review is to catalogue the research to date on the use of auditory evoked potentials (AEP) to assess hyperacusis. DESIGN A step-by-step methodology was conducted following guidelines. Four databases were searched. A total of 3343 papers were identified. A final yield of 35 articles were retained for analysis. RESULTS The analysis identified four types of aetiologies to describe the hyperacusic population in AEP studies; developmental disorders (n = 19), neurological disorders (n = 3), induced hearing damage (n = 8) and idiopathic aetiology (n = 5). Electrophysiological measures were of short (n = 16), middle (n = 13) and long (n = 19) latencies, believed to reflect the activity of the ascending and descending pathways of the auditory system from periphery to cortex. CONCLUSIONS The results of this review revealed the potential use of electrophysiological measures for further understanding the mechanisms of hyperacusis. However, according to the disparity of concepts to define hyperacusis, definitions and populations need to be clarified before biomarkers specific to hyperacusis can be identified.
Collapse
Affiliation(s)
- Charlotte Bigras
- School of Speech-Language Pathology and Audiology, Université de Montréal, Montreal, Canada.,Center of Research on Brain, Language and Music (CRBLM), Montreal, Canada
| | - Bérangère Villatte
- School of Speech-Language Pathology and Audiology, Université de Montréal, Montreal, Canada.,Center of Research on Brain, Language and Music (CRBLM), Montreal, Canada
| | - Victoria Duda
- School of Speech-Language Pathology and Audiology, Université de Montréal, Montreal, Canada.,Centre de recherche interdisciplinaire en réadaptation (CRIR), Montreal, Canada
| | - Sylvie Hébert
- School of Speech-Language Pathology and Audiology, Université de Montréal, Montreal, Canada.,Center of Research on Brain, Language and Music (CRBLM), Montreal, Canada
| |
Collapse
|
29
|
Sex Differences in the Expression of c-fos in a Rat Brain after Exposure to Environmental Noise. SUSTAINABILITY 2022. [DOI: 10.3390/su14052798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Noise is an inarticulate stimulus that threatens health and well-being. It compromises audition and induces a strong stress response that activates the brain at several levels. In the present study, we expose male and female rats to environmental noise in order to investigate if acute or chronic stimulation produces differential brain activation patterns. The animals were exposed to a rat’s audiogram-fitted adaptation of a noisy environment and later sacrificed to quantify the expression of the brain activity marker c-fos. Additionally, the serum corticosterone (CORT) levels were measured to elucidate possible the stress-related effects of noise. It was found that environmental noise differentially increased the serum CORT levels in male and female rats. We identified 17 brain regions outside the classical auditory circuits with a high expression of c-fos, including the hypothalamus, prefrontal cortex, habenular complex, septum, cingulate cortex, nucleus accumbens, insular cortex, amygdala, and hippocampus. Overall, we evidenced that females exhibit less intense c-fos expression in most of the examined areas. We concluded that females might be less affected by the changes produced by environmental noise.
Collapse
|
30
|
Zheng J, Takahashi S, Zhou Y, Cheatham MA. Prestin and electromotility may serve multiple roles in cochlear outer hair cells. Hear Res 2021; 423:108428. [PMID: 34987016 DOI: 10.1016/j.heares.2021.108428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/16/2021] [Accepted: 12/23/2021] [Indexed: 11/04/2022]
Abstract
Outer hair cells (OHCs) are innervated by both medial olivocochlear (MOC) efferents and type II afferents, which also innervate supporting cells to form a local neural network. It has also been demonstrated that prestin provides the molecular basis for OHC somatic electromotility, amplifying movements within the organ of Corti. Although not anticipated, early-onset OHC loss was found in two prestin transgenic mouse models that either lack prestin protein or lack electromotility. To uncover the molecular pathways that evoke OHC death, we profiled the coding transcriptome of OHCs from wildtype (WT), prestin-knockout (KO), and 499-knockin (KI) mice using single-cell RNA sequencing (scRNA-seq). scRNA-Seq transcriptomics and pathway analyses did not reveal common pathways associated with OHC loss observed in prestin-KO and 499-KI mice. Clustering enrichment analysis showed that increased gene expression in OHCs from prestin-KO mice was associated with lipid metabolic processes and cell death pathways. These mRNA profiles likely contribute to the OHC loss observed in prestin-KO mice and support the notion that prestin is also a structural protein, important for the normal plasma membrane compartmentalization that is essential to establish MOC efferent synapses. In contrast, the mRNA profile of OHCs from 499-KI mice did not provide a rational explanation of the early-onset OHC loss in this mutant. OHCs from 499-KI mice have normal plasma membrane compartmentalization and normal OHC-MOC contacts. However, 499 prestin lacks electromotility and appears to change the local neural network around OHCs, as more synaptic markers were found near neighboring supporting cells when compared to WT and prestin-KO mice. Thus, OHCs in prestin-KOs (no prestin protein, no electromotility) and 499-KIs (prestin protein present, no electromotility) may influence local neuronal networks in different ways. Collectively, our data suggest that prestin and its motile properties are important for OHC survival and the maintenance of local afferent/efferent circuits, as well as for its role in cochlear amplification.
Collapse
Affiliation(s)
- Jing Zheng
- Departments of Otolaryngology, Feinberg School of Medicine, Northwestern University, Chicago, IL; Communication Sciences and Disorders, School of Communication; The Knowles Hearing Center, Northwestern University, Evanston, IL.
| | - Satoe Takahashi
- Departments of Otolaryngology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Yingjie Zhou
- Communication Sciences and Disorders, School of Communication
| | - Mary Ann Cheatham
- Communication Sciences and Disorders, School of Communication; The Knowles Hearing Center, Northwestern University, Evanston, IL
| |
Collapse
|
31
|
The Impact of Occupational Noise Exposure on Hyperacusis: a Longitudinal Population Study of Female Workers in Sweden. Ear Hear 2021; 43:1366-1377. [PMID: 34966161 PMCID: PMC9197140 DOI: 10.1097/aud.0000000000001194] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Objectives: The aim was to assess the risk of hyperacusis in relation to occupational noise exposure among female workers in general, and among women working in preschool specifically. Design: A retrospective longitudinal study was performed. Survey data were collected in 2013 and 2014 from two cohorts: randomly selected women from the population in region Västra Götaland, Sweden, and women selected based on having received a preschool teacher degree from universities in the same region. The final study sample included n = 8328 women born between 1948 and 1989. Occupational noise exposure was objectively assigned to all time periods from the first to the last reported occupation throughout working life, using the Swedish Job-Exposure Matrix (JEM) with three exposure intervals: <75 dB(A), 75 to 85 dB(A), and >85 dB(A). The JEM assigns preschool teachers to the 75 to 85 dB(A) exposure interval. The outcome hyperacusis was assessed by self-report using one question addressing discomfort or pain from everyday sounds. In the main analysis, a hyperacusis event was defined by the reported year of onset, if reported to occur at least a few times each week. Additional sensitivity analyses were performed using more strict definitions: (a) at least several times each week and (b) every day. The risk (hazard ratio, HR) of hyperacusis was analyzed in relation to years of occupational noise exposure, using survival analysis with frailty regression modeling accounting for individual variation in survival times which reflect, for example, noise exposure during years prior to onset. Occupational noise exposure was defined by the occupation held at year of hyperacusis onset, or the occupation held at the survey year if no event occurred. Models were adjusted for confounders including age, education, income, family history of hearing loss, and change of jobs due to noise. Results: In total, n = 1966 hyperacusis events between 1960 and 2014 were analyzed in the main analysis. A significantly increased risk of hyperacusis was found among women working in any occupation assigned to the 75 to 85 dB(A) noise exposure group [HR: 2.6, 95% confidence interval (CI): 2.4–2.9], compared with the reference group <75 dB(A). The risk was tripled among preschool teachers specifically (HR: 3.4, 95% CI: 3.0–3.7), with the crude Kaplan-Meier curve showing a higher rate of onset early in the working life in preschool teachers compared with all the other exposure groups. The risk was increased, but not statistically significant in the main analysis, for the highest exposure group >85 dB(A), where only six hyperacusis events were identified (HR: 1.4, 95% CI: 0.6–3.1). In the sensitivity analysis, where hyperacusis was defined as occurring every day, the HR was significant also in the highest exposure group (HR: 3.8, 95% CI: 1.4–10.3), and generally slightly higher in the other exposure groups compared to the main analysis. Conclusions: This study indicates increased risk of hyperacusis already below the permissible occupational noise exposure limit in Sweden (85 dB LAeq,8h) among female workers in general, and in particular among preschool teachers. Prospective studies and less wide exposure intervals could confirm causal effects and assess dose–response relationships, respectively, although this study at present suggest a need for risk assessment, improved hearing prevention measures, and noise abatement measures in occupations with noise levels from 75 dB(A). The results could also have implications for management of occupational disability claims.
Collapse
|
32
|
Pagella S, Deussing JM, Kopp-Scheinpflug C. Expression Patterns of the Neuropeptide Urocortin 3 and Its Receptor CRFR2 in the Mouse Central Auditory System. Front Neural Circuits 2021; 15:747472. [PMID: 34867212 PMCID: PMC8633543 DOI: 10.3389/fncir.2021.747472] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/18/2021] [Indexed: 11/16/2022] Open
Abstract
Sensory systems have to be malleable to context-dependent modulations occurring over different time scales, in order to serve their evolutionary function of informing about the external world while also eliciting survival-promoting behaviors. Stress is a major context-dependent signal that can have fast and delayed effects on sensory systems, especially on the auditory system. Urocortin 3 (UCN3) is a member of the corticotropin-releasing factor family. As a neuropeptide, UCN3 regulates synaptic activity much faster than the classic steroid hormones of the hypothalamic-pituitary-adrenal axis. Moreover, due to the lack of synaptic re-uptake mechanisms, UCN3 can have more long-lasting and far-reaching effects. To date, a modest number of studies have reported the presence of UCN3 or its receptor CRFR2 in the auditory system, particularly in the cochlea and the superior olivary complex, and have highlighted the importance of this stress neuropeptide for protecting auditory function. However, a comprehensive map of all neurons synthesizing UCN3 or CRFR2 within the auditory pathway is lacking. Here, we utilize two reporter mouse lines to elucidate the expression patterns of UCN3 and CRFR2 in the auditory system. Additional immunolabelling enables further characterization of the neurons that synthesize UCN3 or CRFR2. Surprisingly, our results indicate that within the auditory system, UCN3 is expressed predominantly in principal cells, whereas CRFR2 expression is strongest in non-principal, presumably multisensory, cell types. Based on the presence or absence of overlap between UCN3 and CRFR2 labeling, our data suggest unusual modes of neuromodulation by UCN3, involving volume transmission and autocrine signaling.
Collapse
Affiliation(s)
- Sara Pagella
- Division of Neurobiology, Faculty of Biology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jan M Deussing
- Research Group Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Conny Kopp-Scheinpflug
- Division of Neurobiology, Faculty of Biology, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
33
|
Prior Acoustic Trauma Alters Type II Afferent Activity in the Mouse Cochlea. eNeuro 2021; 8:ENEURO.0383-21.2021. [PMID: 34607806 PMCID: PMC8589282 DOI: 10.1523/eneuro.0383-21.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 09/23/2021] [Indexed: 01/11/2023] Open
Abstract
Auditory stimuli travel from the cochlea to the brainstem through type I and type II cochlear afferents. While type I afferents convey information about the frequency, intensity, and timing of sounds, the role of type II afferents remains unresolved. Limited recordings of type II afferents from cochlear apex of prehearing rats reveal they are activated by widespread outer hair cell stimulation, ATP, and by the rupture of nearby outer hair cells. Altogether, these lines of evidence suggest that type II afferents sense loud, potentially damaging levels of sound. To explore this hypothesis further, calcium imaging was used to determine the impact of acoustic trauma on the activity of type II cochlear afferents of young adult mice of both sexes. Two known marker genes (Th, Drd2) and one new marker gene (Tac1), expressed in type II afferents and some other cochlear cell types, drove GCaMP6f expression to reveal calcium transients in response to focal damage in the organ of Corti in all turns of the cochlea. Mature type II afferents responded to acute photoablation damage less often but at greater length compared with prehearing neurons. In addition, days after acoustic trauma, acute photoablation triggered a novel response pattern in type II afferents and surrounding epithelial cells, delayed bursts of activity occurring minutes after the initial response subsided. Overall, calcium imaging can report type II afferent responses to damage even in mature and noise-exposed animals and reveals previously unknown tissue hyperactivity subsequent to acoustic trauma.
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW We review recent progress in the characterization of spiral ganglion neurons (SGNs), the afferent neurons that transmit sound information from mechanosensory hair cells in the inner ear to the central nervous system. RECENT FINDINGS Single-cell ribonucleic acid sequencing studies of murine SGNs have demonstrated that SGNs consist of molecularly distinct subtypes. The molecularly defined SGN subtypes likely correspond to SGN subtypes previously identified on the basis of physiological properties, although this has not been experimentally demonstrated. Subtype maturation is completed postnatally in an activity-dependent manner and is impaired in several models of hearing loss. SUMMARY The recent molecular studies open new avenues to rigorously test whether SGN subtypes are important for the encoding of different sound features and if they show differential vulnerability to genetic factors and environmental insults. This could have important implications for the development of therapeutic strategies to treat hearing loss.
Collapse
Affiliation(s)
- Shuohao Sun
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | |
Collapse
|
35
|
Möhrle D, Wang W, Whitehead SN, Schmid S. GABA B Receptor Agonist R-Baclofen Reverses Altered Auditory Reactivity and Filtering in the Cntnap2 Knock-Out Rat. Front Integr Neurosci 2021; 15:710593. [PMID: 34489651 PMCID: PMC8417788 DOI: 10.3389/fnint.2021.710593] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 07/27/2021] [Indexed: 12/23/2022] Open
Abstract
Altered sensory information processing, and auditory processing, in particular, is a common impairment in individuals with autism spectrum disorder (ASD). One prominent hypothesis for the etiology of ASD is an imbalance between neuronal excitation and inhibition. The selective GABAB receptor agonist R-Baclofen has been shown previously to improve social deficits and repetitive behaviors in several mouse models for neurodevelopmental disorders including ASD, and its formulation Arbaclofen has been shown to ameliorate social avoidance symptoms in some individuals with ASD. The present study investigated whether R-Baclofen can remediate ASD-related altered sensory processing reliant on excitation/inhibition imbalance in the auditory brainstem. To assess a possible excitation/inhibition imbalance in the startle-mediating brainstem underlying ASD-like auditory-evoked behaviors, we detected and quantified brain amino acid levels in the nucleus reticularis pontis caudalis (PnC) of rats with a homozygous loss-of-function mutation in the ASD-linked gene Contactin-associated protein-like 2 (Cntnap2) and their wildtype (WT) littermates using Matrix-Assisted Laser Desorption Ionization Mass Spectrometry (MALDI MS). Abnormal behavioral read-outs of brainstem auditory signaling in Cntnap2 KO rats were accompanied by increased levels of GABA, glutamate, and glutamine in the PnC. We then compared the effect of R-Baclofen on behavioral read-outs of brainstem auditory signaling in Cntnap2 KO and WT rats. Auditory reactivity, sensory filtering, and sensorimotor gating were tested in form of acoustic startle response input-output functions, short-term habituation, and prepulse inhibition before and after acute administration of R-Baclofen (0.75, 1.5, and 3 mg/kg). Systemic R-Baclofen treatment improved disruptions in sensory filtering in Cntnap2 KO rats and suppressed exaggerated auditory startle responses, in particular to moderately loud sounds. Lower ASR thresholds in Cntnap2 KO rats were increased in a dose-dependent fashion, with the two higher doses bringing thresholds close to controls, whereas shorter ASR peak latencies at the threshold were further exacerbated. Impaired prepulse inhibition increased across various acoustic prepulse conditions after administration of R-Baclofen in Cntnap2 KO rats, whereas R-Baclofen did not affect prepulse inhibition in WT rats. Our findings suggest that GABAB receptor agonists may be useful for pharmacologically targeting multiple aspects of sensory processing disruptions involving neuronal excitation/inhibition imbalances in ASD.
Collapse
Affiliation(s)
- Dorit Möhrle
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Wenxuan Wang
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Shawn N Whitehead
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Susanne Schmid
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
36
|
Moglie MJ, Wengier DL, Elgoyhen AB, Goutman JD. Synaptic Contributions to Cochlear Outer Hair Cell Ca 2+ Dynamics. J Neurosci 2021; 41:6812-6821. [PMID: 34253627 PMCID: PMC8360681 DOI: 10.1523/jneurosci.3008-20.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 05/30/2021] [Accepted: 07/05/2021] [Indexed: 11/21/2022] Open
Abstract
For normal cochlear function, outer hair cells (OHCs) require a precise control of intracellular Ca2+ levels. In the absence of regulatory elements such as proteinaceous buffers or extrusion pumps, OHCs degenerate, leading to profound hearing impairment. Influx of Ca2+ occurs both at the stereocilia tips and the basolateral membrane. In this latter compartment, two different origins for Ca2+ influx have been poorly explored: voltage-gated L-type Ca2+ channels (VGCCs) at synapses with Type II afferent neurons, and α9α10 cholinergic nicotinic receptors at synapses with medio-olivochlear complex (MOC) neurons. Using functional imaging in mouse OHCs, we dissected Ca2+ influx individually through each of these sources, either by applying step depolarizations to activate VGCC, or stimulating MOC axons. Ca2+ ions originated in MOC synapses, but not by VGCC activation, was confined by Ca2+-ATPases most likely present in nearby synaptic cisterns. Although Ca2+ currents in OHCs are small, VGCC Ca2+ signals were comparable in size to those elicited by α9α10 receptors, and were potentiated by ryanodine receptors (RyRs). In contrast, no evidence of potentiation by RyRs was found for MOC Ca2+ signals over a wide range of presynaptic stimulation strengths. Our study shows that despite the fact that these two Ca2+ entry sites are closely positioned, they differ in their regulation by intracellular cisterns and/or organelles, suggesting the existence of well-tuned mechanisms to separate the two different OHC synaptic functions.SIGNIFICANCE STATEMENT Outer hair cells (OHCs) are sensory cells in the inner ear operating under very special constraints. Acoustic stimulation leads to fast changes both in membrane potential and in the intracellular concentration of metabolites such as Ca2+ Tight mechanisms for Ca2+ control in OHCs have been reported. Interestingly, Ca2+ is crucial for two important synaptic processes: inhibition by efferent cholinergic neurons, and glutamate release onto Type II afferent fibers. In the current study we functionally imaged Ca2+ at these two different synapses, showing close positioning within the basolateral compartment of OHCs. In addition, we show differential regulation of these two Ca2+ sources by synaptic cisterns and/or organelles, which could result crucial for functional segregation during normal hearing.
Collapse
Affiliation(s)
- Marcelo J Moglie
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI) (Consejo Nacional de Investigaciones Científicas y Tecnológicas), Ciudad Autónoma de Buenos Aires 1428, Argentina
| | - Diego L Wengier
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI) (Consejo Nacional de Investigaciones Científicas y Tecnológicas), Ciudad Autónoma de Buenos Aires 1428, Argentina
| | - A Belén Elgoyhen
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI) (Consejo Nacional de Investigaciones Científicas y Tecnológicas), Ciudad Autónoma de Buenos Aires 1428, Argentina
| | - Juan D Goutman
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI) (Consejo Nacional de Investigaciones Científicas y Tecnológicas), Ciudad Autónoma de Buenos Aires 1428, Argentina
| |
Collapse
|
37
|
Williams ZJ, Suzman E, Woynaroski TG. A Phenotypic Comparison of Loudness and Pain Hyperacusis: Symptoms, Comorbidity, and Associated Features in a Multinational Patient Registry. Am J Audiol 2021; 30:341-358. [PMID: 33877881 PMCID: PMC8642094 DOI: 10.1044/2021_aja-20-00209] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/23/2021] [Accepted: 01/26/2021] [Indexed: 11/09/2022] Open
Abstract
Purpose Hyperacusis is a complex and poorly understood auditory disorder characterized by decreased tolerance to sound at levels that would not trouble most individuals. Recently, it has been suggested that individuals who experience otalgia in response to everyday sounds (termed pain hyperacusis) may differ clinically from those whose primary symptom is the perception of everyday sounds as excessively loud (termed loudness hyperacusis). Despite this theoretical distinction, there have been no empirical studies directly comparing these two populations of hyperacusis patients. Method Using data from a multinational patient registry (the Coordination of Rare Diseases at Sanford Registry), we examined self-reported demographics, symptoms, comorbidity, and response to treatment in a sample of 243 adults with hyperacusis, 152 of whom were classified as having pain hyperacusis based on reported symptoms. Bayesian statistical tests were used to investigate both the presence and absence of group differences between patients with loudness and pain hyperacusis. Results Individuals with pain hyperacusis presented with a more severe clinical phenotype, reporting a higher frequency of temporary symptom exacerbations (i.e., "setbacks"), less perceived symptom improvement over time, more severe comorbid headache disorders, and reduced benefit from sound therapy. However, the two hypothesized hyperacusis subtypes exhibited more similarities than differences, with the majority of symptoms and comorbidities being equally prevalent across groups. Multiple comorbidities were commonly observed, including tinnitus, primary headache disorders, psychiatric disorders, and functional somatic syndromes. Intolerance of sensory stimuli in other modalities was also frequently reported. Conclusion Although this study provides little evidence that loudness and pain hyperacusis are pathophysiologically distinct conditions, our findings indicate that a pain-predominant phenotype may be a meaningful prognostic marker in patients with hyperacusis.
Collapse
Affiliation(s)
- Zachary J. Williams
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN
- Department of Hearing and Speech Sciences, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN
- Frist Center for Autism and Innovation, Vanderbilt University, Nashville, TN
| | - Evan Suzman
- Graduate Program in Biomedical Sciences, Vanderbilt University, Nashville, TN
| | - Tiffany G. Woynaroski
- Department of Hearing and Speech Sciences, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN
- Frist Center for Autism and Innovation, Vanderbilt University, Nashville, TN
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
38
|
Kitcher SR, Pederson AM, Weisz CJC. Diverse identities and sites of action of cochlear neurotransmitters. Hear Res 2021; 419:108278. [PMID: 34108087 DOI: 10.1016/j.heares.2021.108278] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 04/30/2021] [Accepted: 05/18/2021] [Indexed: 11/18/2022]
Abstract
Accurate encoding of acoustic stimuli requires temporally precise responses to sound integrated with cellular mechanisms that encode the complexity of stimuli over varying timescales and orders of magnitude of intensity. Sound in mammals is initially encoded in the cochlea, the peripheral hearing organ, which contains functionally specialized cells (including hair cells, afferent and efferent neurons, and a multitude of supporting cells) to allow faithful acoustic perception. To accomplish the demanding physiological requirements of hearing, the cochlea has developed synaptic arrangements that operate over different timescales, with varied strengths, and with the ability to adjust function in dynamic hearing conditions. Multiple neurotransmitters interact to support the precision and complexity of hearing. Here, we review the location of release, action, and function of neurotransmitters in the mammalian cochlea with an emphasis on recent work describing the complexity of signaling.
Collapse
Affiliation(s)
- Siân R Kitcher
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, United States
| | - Alia M Pederson
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, United States
| | - Catherine J C Weisz
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, United States.
| |
Collapse
|
39
|
Biswas J, Pijewski RS, Makol R, Miramontes TG, Thompson BL, Kresic LC, Burghard AL, Oliver DL, Martinelli DC. C1ql1 is expressed in adult outer hair cells of the cochlea in a tonotopic gradient. PLoS One 2021; 16:e0251412. [PMID: 33979385 PMCID: PMC8115824 DOI: 10.1371/journal.pone.0251412] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/26/2021] [Indexed: 01/08/2023] Open
Abstract
Hearing depends on the transduction of sounds into neural signals by the inner hair cells of the cochlea. Cochleae also have outer hair cells with unique electromotile properties that increase auditory sensitivity, but they are particularly susceptible to damage by intense noise exposure, ototoxic drugs, and aging. Although the outer hair cells have synapses on afferent neurons that project to the brain, the function of this neuronal circuit is unclear. Here, we created a novel mouse allele that inserts a fluorescent reporter at the C1ql1 locus which revealed gene expression in the outer hair cells and allowed creation of outer hair cell-specific C1ql1 knockout mice. We found that C1ql1 expression in outer hair cells corresponds to areas with the most sensitive frequencies of the mouse audiogram, and that it has an unexpected adolescence-onset developmental timing. No expression was observed in the inner hair cells. Since C1QL1 in the brain is made by neurons, transported anterogradely in axons, and functions in the synaptic cleft, C1QL1 may serve a similar function at the outer hair cell afferent synapse. Histological analyses revealed that C1ql1 conditional knockout cochleae may have reduced outer hair cell afferent synapse maintenance. However, auditory behavioral and physiological assays did not reveal a compelling phenotype. Nonetheless, this study identifies a potentially useful gene expressed in the cochlea and opens the door for future studies aimed at elucidating the function of C1QL1 and the function of the outer hair cell and its afferent neurons.
Collapse
Affiliation(s)
- Joyshree Biswas
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, United States of America
| | - Robert S. Pijewski
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, United States of America
| | - Rohit Makol
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, United States of America
- The Connecticut Institute for the Brain and Cognitive Sciences (IBACS), Storrs, CT, United States of America
| | - Tania G. Miramontes
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, United States of America
| | - Brianna L. Thompson
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, United States of America
| | - Lyndsay C. Kresic
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, United States of America
| | - Alice L. Burghard
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, United States of America
| | - Douglas L. Oliver
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, United States of America
| | - David C. Martinelli
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, United States of America
- The Connecticut Institute for the Brain and Cognitive Sciences (IBACS), Storrs, CT, United States of America
- * E-mail:
| |
Collapse
|
40
|
Henton A, Tzounopoulos T. What's the buzz? The neuroscience and the treatment of tinnitus. Physiol Rev 2021; 101:1609-1632. [PMID: 33769102 DOI: 10.1152/physrev.00029.2020] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Tinnitus is a pervasive public health issue that affects ∼15% of the United States population. Similar estimates have also been shown on a global scale, with similar prevalence found in Europe, Asia, and Africa. The severity of tinnitus is heterogeneous, ranging from mildly bothersome to extremely disruptive. In the United States, ∼10-20% of individuals who experience tinnitus report symptoms that severely reduce their quality of life. Due to the huge personal and societal burden, in the last 20 yr a concerted effort on basic and clinical research has significantly advanced our understanding and treatment of this disorder. Yet, neither full understanding, nor cure exists. We know that tinnitus is the persistent involuntary phantom percept of internally generated nonverbal indistinct noises and tones, which in most cases is initiated by acquired hearing loss and maintained only when this loss is coupled with distinct neuronal changes in auditory and extra-auditory brain networks. Yet, the exact mechanisms and patterns of neural activity that are necessary and sufficient for the perceptual generation and maintenance of tinnitus remain incompletely understood. Combinations of animal model and human research will be essential in filling these gaps. Nevertheless, the existing progress in investigating the neurophysiological mechanisms has improved current treatment and highlighted novel targets for drug development and clinical trials. The aim of this review is to thoroughly discuss the current state of human and animal tinnitus research, outline current challenges, and highlight new and exciting research opportunities.
Collapse
Affiliation(s)
- A Henton
- Pittsburgh Hearing Research Center and Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - T Tzounopoulos
- Pittsburgh Hearing Research Center and Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
41
|
Wang H, Wang S, Lu Y, Chen Y, Huang W, Qiu M, Wu H, Hua Y. Cytoarchitecture and innervation of the mouse cochlear amplifier revealed by large-scale volume electron microscopy. J Comp Neurol 2021; 529:2958-2969. [PMID: 33719053 PMCID: PMC8252425 DOI: 10.1002/cne.25137] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/31/2022]
Abstract
In mammalian cochlea, sound‐induced vibration is amplified by a three‐row lattice of Y‐shaped microstructures consisting of electromotile outer hair cell and supporting Deiters cell. This highly organized structure is thought to be essential for hearing of low‐level sounds. Prior studies reported differences in geometry and synaptic innervation of the outer hair cells between rows, but how these fine features are achieved at subcellular level still remains unclear. Using serial block‐face electron microscopy, we acquired few‐hundred‐micron‐sized cytoarchitecture of mouse organ of Corti at nanometer resolution. Structural quantifications were performed on the Y‐shapes as well as afferent and efferent projections to outer hair cells (OHCs). Several new features, which support the previously observed inter‐row heterogeneity, are described. Our result provides structural bases for the gradient of mechanical properties and diverse centrifugal regulation of OHC rows.
Collapse
Affiliation(s)
- Haoyu Wang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai, China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China.,Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai, China
| | - Shengxiong Wang
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai, China.,Putuo People's Hospital, Tongji University, Shanghai, China
| | - Yan Lu
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai, China
| | - Ying Chen
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai, China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Wenqing Huang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai, China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China.,Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai, China
| | - Miaoxin Qiu
- Putuo People's Hospital, Tongji University, Shanghai, China
| | - Hao Wu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai, China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China.,Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai, China
| | - Yunfeng Hua
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai, China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China.,Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai, China
| |
Collapse
|
42
|
Refat F, Wertz J, Hinrichs P, Klose U, Samy H, Abdelkader RM, Saemisch J, Hofmeier B, Singer W, Rüttiger L, Knipper M, Wolpert S. Co-occurrence of Hyperacusis Accelerates With Tinnitus Burden Over Time and Requires Medical Care. Front Neurol 2021; 12:627522. [PMID: 33815254 PMCID: PMC8012887 DOI: 10.3389/fneur.2021.627522] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/22/2021] [Indexed: 12/14/2022] Open
Abstract
Although tinnitus represents a major global burden, no causal therapy has yet been established. Ongoing controversies about the neuronal pathophysiology of tinnitus hamper efforts in developing advanced therapies. Hypothesizing that the unnoticed co-occurrence of hyperacusis and differences in the duration of tinnitus may possibly differentially influence the neural correlate of tinnitus, we analyzed 33 tinnitus patients without (T-group) and 20 tinnitus patients with hyperacusis (TH-group). We found crucial differences between the T-group and the TH-group in the increase of annoyance, complaints, tinnitus loudness, and central neural gain as a function of tinnitus duration. Hearing thresholds did not differ between T-group and TH-group. In the TH-group, the tinnitus complaints (total tinnitus score) were significantly greater from early on and the tinnitus intensity distinctly increased over time from ca. 12 to 17 dB when tinnitus persisted more than 5 years, while annoyance responses to normal sound remained nearly constant. In contrast, in the T-group tinnitus complaints remained constant, although the tinnitus intensity declined over time from ca. 27 down to 15 dB beyond 5 years of tinnitus persistence. This was explained through a gradually increased annoyance to normal sound over time, shown by a hyperacusis questionnaire. Parallel a shift from a mainly unilateral (only 17% bilateral) to a completely bilateral (100%) tinnitus percept occurred in the T-group, while bilateral tinnitus dominated in the TH-group from the start (75%). Over time in the T-group, ABR wave V amplitudes (and V/I ratios) remained reduced and delayed. By contrast, in the TH-group especially the ABR wave III and V (and III/I ratio) continued to be enhanced and shortened in response to high-level sound stimuli. Interestingly, in line with signs of an increased co-occurrence of hyperacusis in the T-group over time, ABR wave III also slightly increased in the T-group. The findings disclose an undiagnosed co-occurrence of hyperacusis in tinnitus patients as a main cause of distress and the cause of complaints about tinnitus over time. To achieve urgently needed and personalized therapies, possibly using the objective tools offered here, a systematic sub-classification of tinnitus and the co-occurrence of hyperacusis is recommended.
Collapse
Affiliation(s)
- Fatma Refat
- Audio-Vestibular Unit, Department of Ear Nose Throat, Minia University, Minia, Egypt.,Tübingen Hearing Research Centre, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany
| | - Jakob Wertz
- Tübingen Hearing Research Centre, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany
| | - Pauline Hinrichs
- Tübingen Hearing Research Centre, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany
| | - Uwe Klose
- Department of Diagnostic and Interventional Neuroradiology, University of Tübingen, Tübingen, Germany
| | - Hesham Samy
- Audio-Vestibular Unit, Department of Ear Nose Throat, Minia University, Minia, Egypt
| | | | - Jörg Saemisch
- Tübingen Hearing Research Centre, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany
| | - Benedikt Hofmeier
- Tübingen Hearing Research Centre, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany
| | - Wibke Singer
- Tübingen Hearing Research Centre, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany
| | - Lukas Rüttiger
- Tübingen Hearing Research Centre, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany
| | - Marlies Knipper
- Tübingen Hearing Research Centre, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany
| | - Stephan Wolpert
- Tübingen Hearing Research Centre, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany
| |
Collapse
|
43
|
Huang G, Eckrich S. Quantitative Fluorescent in situ Hybridization Reveals Differential Transcription Profile Sharpening of Endocytic Proteins in Cochlear Hair Cells Upon Maturation. Front Cell Neurosci 2021; 15:643517. [PMID: 33716676 PMCID: PMC7952526 DOI: 10.3389/fncel.2021.643517] [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: 12/18/2020] [Accepted: 02/09/2021] [Indexed: 12/04/2022] Open
Abstract
The organ of Corti (OC) comprises two types of sensory cells: outer hair cells (OHCs) and inner hair cells (IHCs). While both are mechanotransducers, OHCs serve as cochlear amplifiers, whereas IHCs transform sound into transmitter release. Reliable sound encoding is ensured by indefatigable exocytosis of synaptic vesicles associated with efficient replenishment of the vesicle pool. Vesicle reformation requires retrieval of vesicle membrane from the hair cell’s membrane via endocytosis. So far, the protein machinery for endocytosis in pre-mature and terminally differentiated hair cells has only partially been deciphered. Here, we studied three endocytic proteins, dynamin-1, dynamin-3, and endophilin-A1, by assessing their transcription profiles in pre-mature and mature mouse OCs. State-of-the-art RNAscope® fluorescent in situ hybridization (FISH) of whole-mount OCs was used for quantification of target mRNAs on single-cell level. We found that pre-mature IHCs contained more mRNA transcripts of dnm1 (encoding dynamin-1) and sh3gl2 (endophilin-A1), but less of dnm3 (dynamin-3) than OHCs. These differential transcription profiles between OHCs and IHCs were sharpened upon maturation. It is noteworthy that low but heterogeneous signal numbers were found between individual negative controls, which highlights the importance of corresponding analyses in RNAscope® assays. Complementary immunolabeling revealed strong expression of dynamin-1 in the soma of mature IHCs, which was much weaker in pre-mature IHCs. By contrast, dynamin-3 was predominantly found in the soma and at the border of the cuticular plates of pre-mature and mature OHCs. In summary, using quantitative RNAscope® FISH and immunohistochemistry on whole-mount tissue of both pre-mature and mature OCs, we disclosed the cellular upregulation of endocytic proteins at the level of transcription/translation during terminal differentiation of the OC. Dynamin-1 and endophilin-A1 likely contribute to the strengthening of the endocytic machinery in IHCs after the onset of hearing, whereas expression of dynamin-3 at the cuticular plate of pre-mature and mature OHCs suggests its possible involvement in activity-independent apical endocytosis.
Collapse
Affiliation(s)
- Guobin Huang
- Center for Integrative Physiology and Molecular Medicine, School of Medicine, Department of Biophysics, Saarland University, Homburg, Germany
| | - Stephanie Eckrich
- Center for Integrative Physiology and Molecular Medicine, School of Medicine, Department of Biophysics, Saarland University, Homburg, Germany
| |
Collapse
|
44
|
Outer Hair Cell Glutamate Signaling through Type II Spiral Ganglion Afferents Activates Neurons in the Cochlear Nucleus in Response to Nondamaging Sounds. J Neurosci 2021; 41:2930-2943. [PMID: 33574178 DOI: 10.1523/jneurosci.0619-20.2021] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 01/24/2021] [Accepted: 02/04/2021] [Indexed: 11/21/2022] Open
Abstract
Cochlear outer hair cells (OHCs) are known to uniquely participate in auditory processing through their electromotility, and like inner hair cells, are also capable of releasing vesicular glutamate onto spiral ganglion (SG) neurons: in this case, onto the sparse Type II SG neurons. However, unlike glutamate signaling at the inner hair cell-Type I SG neuron synapse, which is robust across a wide spectrum of sound intensities, glutamate signaling at the OHC-Type II SG neuron synapse is weaker and has been hypothesized to occur only at intense, possibly damaging sound levels. Here, we tested the ability of the OHC-Type II SG pathway to signal to the brain in response to moderate, nondamaging sound (80 dB SPL) as well as to intense sound (115 dB SPL). First, we determined the VGluTs associated with OHC signaling and then confirmed the loss of glutamatergic synaptic transmission from OHCs to Type II SG neurons in KO mice using dendritic patch-clamp recordings. Next, we generated genetic mouse lines in which vesicular glutamate release occurs selectively from OHCs, and then assessed c-Fos expression in the cochlear nucleus in response to sound. From these analyses, we show, for the first time, that glutamatergic signaling at the OHC-Type II SG neuron synapse is capable of activating cochlear nucleus neurons, even at moderate sound levels.SIGNIFICANCE STATEMENT Evidence suggests that cochlear outer hair cells (OHCs) release glutamate onto Type II spiral ganglion neurons only when exposed to loud sound, and that Type II neurons are activated by tissue damage. Knowing whether moderate level sound, without tissue damage, activates this pathway has functional implications for this fundamental auditory pathway. We first determined that OHCs rely largely on VGluT3 for synaptic glutamate release. We then used a genetically modified mouse line in which OHCs, but not inner hair cells, release vesicular glutamate to demonstrate that moderate sound exposure activates cochlear nucleus neurons via the OHC-Type II spiral ganglion pathway. Together, these data indicate that glutamate signaling at the OHC-Type II afferent synapse participates in auditory function at moderate sound levels.
Collapse
|
45
|
Williams ZJ, He JL, Cascio CJ, Woynaroski TG. A review of decreased sound tolerance in autism: Definitions, phenomenology, and potential mechanisms. Neurosci Biobehav Rev 2021; 121:1-17. [PMID: 33285160 PMCID: PMC7855558 DOI: 10.1016/j.neubiorev.2020.11.030] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 12/23/2022]
Abstract
Atypical behavioral responses to environmental sounds are common in autistic children and adults, with 50-70 % of this population exhibiting decreased sound tolerance (DST) at some point in their lives. This symptom is a source of significant distress and impairment across the lifespan, contributing to anxiety, challenging behaviors, reduced community participation, and school/workplace difficulties. However, relatively little is known about its phenomenology or neurocognitive underpinnings. The present article synthesizes a large body of literature on the phenomenology and pathophysiology of DST-related conditions to generate a comprehensive theoretical account of DST in autism. Notably, we argue against conceptualizing DST as a unified construct, suggesting that it be separated into three phenomenologically distinct conditions: hyperacusis (the perception of everyday sounds as excessively loud or painful), misophonia (an acquired aversive reaction to specific sounds), and phonophobia (a specific phobia of sound), each responsible for a portion of observed DST behaviors. We further elaborate our framework by proposing preliminary neurocognitive models of hyperacusis, misophonia, and phonophobia that incorporate neurophysiologic findings from studies of autism.
Collapse
Affiliation(s)
- Zachary J Williams
- Medical Scientist Training Program, Vanderbilt University School of Medicine, 221 Eskind Biomedical Library and Learning Center, 2209 Garland Ave., Nashville, TN, 37240, United States; Department of Hearing and Speech Sciences, Vanderbilt University Medical Center, 1215 21st Avenue South, Medical Center East, Room 8310, Nashville, TN, 37232, United States; Vanderbilt Brain Institute, Vanderbilt University, 7203 Medical Research Building III, 465 21st Avenue South, Nashville, TN, 37232, United States; Frist Center for Autism and Innovation, Vanderbilt University, 2414 Highland Avenue, Suite 115, Nashville, TN, 37212, United States.
| | - Jason L He
- Department of Forensic and Neurodevelopmental Sciences, Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Strand Building, Strand Campus, Strand, London, WC2R 2LS, London, United Kingdom.
| | - Carissa J Cascio
- Vanderbilt Brain Institute, Vanderbilt University, 7203 Medical Research Building III, 465 21st Avenue South, Nashville, TN, 37232, United States; Frist Center for Autism and Innovation, Vanderbilt University, 2414 Highland Avenue, Suite 115, Nashville, TN, 37212, United States; Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, 2254 Village at Vanderbilt, 1500 21st Ave South, Nashville, TN, 37212, United States; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, 110 Magnolia Cir, Nashville, TN, 37203, United States.
| | - Tiffany G Woynaroski
- Department of Hearing and Speech Sciences, Vanderbilt University Medical Center, 1215 21st Avenue South, Medical Center East, Room 8310, Nashville, TN, 37232, United States; Vanderbilt Brain Institute, Vanderbilt University, 7203 Medical Research Building III, 465 21st Avenue South, Nashville, TN, 37232, United States; Frist Center for Autism and Innovation, Vanderbilt University, 2414 Highland Avenue, Suite 115, Nashville, TN, 37212, United States; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, 110 Magnolia Cir, Nashville, TN, 37203, United States.
| |
Collapse
|
46
|
Pavlinkova G. Molecular Aspects of the Development and Function of Auditory Neurons. Int J Mol Sci 2020; 22:ijms22010131. [PMID: 33374462 PMCID: PMC7796308 DOI: 10.3390/ijms22010131] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 01/08/2023] Open
Abstract
This review provides an up-to-date source of information on the primary auditory neurons or spiral ganglion neurons in the cochlea. These neurons transmit auditory information in the form of electric signals from sensory hair cells to the first auditory nuclei of the brain stem, the cochlear nuclei. Congenital and acquired neurosensory hearing loss affects millions of people worldwide. An increasing body of evidence suggest that the primary auditory neurons degenerate due to noise exposure and aging more readily than sensory cells, and thus, auditory neurons are a primary target for regenerative therapy. A better understanding of the development and function of these neurons is the ultimate goal for long-term maintenance, regeneration, and stem cell replacement therapy. In this review, we provide an overview of the key molecular factors responsible for the function and neurogenesis of the primary auditory neurons, as well as a brief introduction to stem cell research focused on the replacement and generation of auditory neurons.
Collapse
Affiliation(s)
- Gabriela Pavlinkova
- BIOCEV, Institute of Biotechnology of the Czech Academy of Sciences, 25250 Vestec, Czech Republic
| |
Collapse
|
47
|
Cochlear homeostasis: a molecular physiological perspective on maintenance of sound transduction and auditory neurotransmission with noise and ageing. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.09.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
48
|
Petitpré C, Bourien J, Wu H, Diuba A, Puel JL, Lallemend F. Genetic and functional diversity of primary auditory afferents. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
49
|
Neuronal processes and glial precursors form a scaffold for wiring the developing mouse cochlea. Nat Commun 2020; 11:5866. [PMID: 33203842 PMCID: PMC7672226 DOI: 10.1038/s41467-020-19521-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 10/02/2020] [Indexed: 01/20/2023] Open
Abstract
In the developing nervous system, axons navigate through complex terrains that change depending on when and where outgrowth begins. For instance, in the developing cochlea, spiral ganglion neurons extend their peripheral processes through a growing and heterogeneous environment en route to their final targets, the hair cells. Although the basic principles of axon guidance are well established, it remains unclear how axons adjust strategies over time and space. Here, we show that neurons with different positions in the spiral ganglion employ different guidance mechanisms, with evidence for both glia-guided growth and fasciculation along a neuronal scaffold. Processes from neurons in the rear of the ganglion are more directed and grow faster than those from neurons at the border of the ganglion. Further, processes at the wavefront grow more efficiently when in contact with glial precursors growing ahead of them. These findings suggest a tiered mechanism for reliable axon guidance.
Collapse
|
50
|
Wood MB, Nowak N, Mull K, Goldring A, Lehar M, Fuchs PA. Acoustic Trauma Increases Ribbon Number and Size in Outer Hair Cells of the Mouse Cochlea. J Assoc Res Otolaryngol 2020; 22:19-31. [PMID: 33151428 PMCID: PMC7822997 DOI: 10.1007/s10162-020-00777-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 10/19/2020] [Indexed: 01/01/2023] Open
Abstract
Outer hair cells (OHCs) in the mouse cochlea are contacted by up to three type II afferent boutons. On average, only half of these are postsynaptic to presynaptic ribbons. Mice of both sexes were subjected to acoustic trauma that produced a threshold shift of 44.2 ± 9.1 dB 7 days after exposure. Ribbon synapses of OHCs were quantified in post-trauma and littermate controls using immunolabeling of CtBP2. Visualization with virtual reality was used to determine 3-D cytoplasmic localization of CtBP2 puncta to the synaptic pole of OHCs. Acoustic trauma was associated with a statistically significant increase in the number of synaptic ribbons per OHC. Serial section TEM was carried out on similarly treated mice. This also showed a significant increase in the number of ribbons in post-trauma OHCs, as well as a significant increase in ribbon volume compared to ribbons in control OHCs. An increase in OHC ribbon synapses after acoustic trauma is a novel observation that has implications for OHC:type II afferent signaling. A mathematical model showed that the observed increase in OHC ribbons considered alone could produce a significant increase in action potentials among type II afferent neurons during strong acoustic stimulation.
Collapse
Affiliation(s)
- Megan B Wood
- Department of Otolaryngology - Head and Neck, Surgery, Johns Hopkins University School of Medicine, 820 Richard Starr Ross Research Building, 720 Rutland Ave, Baltimore, MD, 21205, USA.
| | - Nathaniel Nowak
- Department of Otolaryngology - Head and Neck, Surgery, Johns Hopkins University School of Medicine, 820 Richard Starr Ross Research Building, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Keira Mull
- Department of Otolaryngology - Head and Neck, Surgery, Johns Hopkins University School of Medicine, 820 Richard Starr Ross Research Building, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Adam Goldring
- Department of Otolaryngology - Head and Neck, Surgery, Johns Hopkins University School of Medicine, 820 Richard Starr Ross Research Building, 720 Rutland Ave, Baltimore, MD, 21205, USA.,Sutter Instrument, Co. 1 Digital Drive, Novato, CA, 94949, USA
| | - Mohamed Lehar
- Department of Otolaryngology - Head and Neck, Surgery, Johns Hopkins University School of Medicine, 820 Richard Starr Ross Research Building, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Paul Albert Fuchs
- Department of Otolaryngology - Head and Neck, Surgery, Johns Hopkins University School of Medicine, 820 Richard Starr Ross Research Building, 720 Rutland Ave, Baltimore, MD, 21205, USA
| |
Collapse
|