1
|
Bachman JL, Kitcher SR, Vattino LG, Beaulac HJ, Chaves MG, Rivera IH, Katz E, Wedemeyer C, Weisz CJ. GABAergic synapses between auditory efferent neurons and type II spiral ganglion afferent neurons in the mouse cochlea. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.28.587185. [PMID: 38586043 PMCID: PMC10996694 DOI: 10.1101/2024.03.28.587185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Cochlear outer hair cells (OHCs) are electromotile and are implicated in mechanisms of amplification of responses to sound that enhance sound sensitivity and frequency tuning. They send information to the brain through glutamatergic synapses onto a small subpopulation of neurons of the ascending auditory nerve, the type II spiral ganglion neurons (SGNs). The OHC synapses onto type II SGNs are sparse and weak, suggesting that type II SGNs respond primarily to loud and possibly damaging levels of sound. OHCs also receive innervation from the brain through the medial olivocochlear (MOC) efferent neurons. MOC neurons are cholinergic yet exert an inhibitory effect on auditory function as they are coupled to alpha9/alpha10 nicotinic acetylcholine receptors (nAChRs) on OHCs, which leads to calcium influx that gates SK potassium channels. The net hyperpolarization exerted by this efferent synapse reduces OHC activity-evoked electromotility and is implicated in cochlear gain control, protection against acoustic trauma, and attention. MOC neurons also label for markers of gamma-aminobutyric acid (GABA) and GABA synthesis. GABAB autoreceptor (GABABR) activation by GABA released from MOC terminals has been demonstrated to reduce ACh release, confirming important negative feedback roles for GABA. However, the full complement of GABAergic activity in the cochlea is not currently understood, including the mechanisms that regulate GABA release from MOC axon terminals, whether GABA diffuses from MOC axon terminals to other postsynaptic cells, and the location and function of GABAA receptors (GABAARs). Previous electron microscopy studies suggest that MOC neurons form contacts onto several other cell types in the cochlea, but whether these contacts form functional synapses, and what neurotransmitters are employed, are unknown. Here we use immunohistochemistry, optical neurotransmitter imaging and patch-clamp electrophysiology from hair cells, afferent dendrites, and efferent axons to demonstrate that in addition to presynaptic GABABR autoreceptor activation, MOC efferent axon terminals release GABA onto type II SGN afferent dendrites with postsynaptic activity mediated by GABAARs. This synapse may have multiple roles including developmental regulation of cochlear innervation, fine tuning of OHC activity, or providing feedback to the brain about MOC and OHC activity.
Collapse
Affiliation(s)
- Julia L. Bachman
- These authors contributed equally
- Section on Neuronal Circuitry, National Institutes of Health, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
- The National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Siân R. Kitcher
- These authors contributed equally
- Section on Neuronal Circuitry, National Institutes of Health, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Lucas G. Vattino
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Dr. Héctor N. Torres, Consejo Nacional de Investigaciones Científicas y Técnicas, 1428 Ciudad Autónoma de Buenos Aires, Argentina
- Eaton Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, Boston, MA, USA
| | - Holly J. Beaulac
- Section on Neuronal Circuitry, National Institutes of Health, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - M. Grace Chaves
- Section on Neuronal Circuitry, National Institutes of Health, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
- Eaton Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, Boston, MA, USA
- Graduate Program in Speech and Hearing Biosciences and Technology, Harvard Medical School, Boston, MA, USA
| | - Israel Hernandez Rivera
- Section on Neuronal Circuitry, National Institutes of Health, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Eleonora Katz
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Dr. Héctor N. Torres, Consejo Nacional de Investigaciones Científicas y Técnicas, 1428 Ciudad Autónoma de Buenos Aires, Argentina
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, 1428 Buenos Aires, Argentina
| | - Carolina Wedemeyer
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Dr. Héctor N. Torres, Consejo Nacional de Investigaciones Científicas y Técnicas, 1428 Ciudad Autónoma de Buenos Aires, Argentina
| | - Catherine J.C. Weisz
- Section on Neuronal Circuitry, National Institutes of Health, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| |
Collapse
|
2
|
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: 1.0] [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
|
3
|
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: 0] [Impact Index Per Article: 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
|
4
|
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: 0.5] [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]
|
5
|
Zhao HB, Liu LM, Yu N, Zhu Y, Mei L, Chen J, Liang C. Efferent neurons control hearing sensitivity and protect hearing from noise through the regulation of gap junctions between cochlear supporting cells. J Neurophysiol 2022; 127:313-327. [PMID: 34907797 PMCID: PMC8759971 DOI: 10.1152/jn.00468.2021] [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] [Indexed: 01/03/2023] Open
Abstract
It is critical for hearing that the descending cochlear efferent system provides a negative feedback to hair cells to regulate hearing sensitivity and protect hearing from noise. The medial olivocochlear (MOC) efferent nerves project to outer hair cells (OHCs) to regulate OHC electromotility, which is an active cochlear amplifier and can increase hearing sensitivity. Here, we report that the MOC efferent nerves also could innervate supporting cells (SCs) in the vicinity of OHCs to regulate hearing sensitivity. MOC nerve fibers are cholinergic, and acetylcholine (ACh) is a primary neurotransmitter. Immunofluorescent staining showed that MOC nerve endings, presynaptic vesicular acetylcholine transporters (VAChTs), and postsynaptic ACh receptors were visible at SCs and in the SC area. Application of ACh in SCs could evoke a typical inward current and reduce gap junctions (GJs) between them, which consequently enhanced the direct effect of ACh on OHCs to shift but not eliminate OHC electromotility. This indirect, GJ-mediated inhibition had a long-lasting influence. In vivo experiments further demonstrated that deficiency of this GJ-mediated efferent pathway decreased the regulation of active cochlear amplification and compromised the protection against noise. In particular, distortion product otoacoustic emission (DPOAE) showed a delayed reduction after noise exposure. Our findings reveal a new pathway for the MOC efferent system via innervating SCs to control active cochlear amplification and hearing sensitivity. These data also suggest that this SC GJ-mediated efferent pathway may play a critical role in long-term efferent inhibition and is required for protection of hearing from noise trauma.NEW & NOTEWORTHY The cochlear efferent system provides a negative feedback to control hair cell activity and hearing sensitivity and plays a critical role in noise protection. We reveal a new efferent control pathway in which medial olivocochlear efferent fibers have innervations with cochlear supporting cells to control their gap junctions, therefore regulating outer hair cell electromotility and hearing sensitivity. This supporting cell gap junction-mediated efferent control pathway is required for the protection of hearing from noise.
Collapse
|
6
|
Qi Y, Xiong W, Yu S, Du Z, Qu T, He L, Wei W, Zhang L, Liu K, Li Y, He DZ, Gong S. Deletion of C1ql1 Causes Hearing Loss and Abnormal Auditory Nerve Fibers in the Mouse Cochlea. Front Cell Neurosci 2021; 15:713651. [PMID: 34512267 PMCID: PMC8424102 DOI: 10.3389/fncel.2021.713651] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 07/21/2021] [Indexed: 01/19/2023] Open
Abstract
Complement C1q Like 1 (C1QL1), a secreted component of C1Q-related protein, is known to play an important role in synaptic maturation, regulation, and maintenance in the central nervous system. C1ql1 is expressed in adult cochlear inner and outer hair cells (IHCs and OHCs) with preferential expression in OHCs. We generated C1ql1 null mice to examine the role of C1QL1 in the auditory periphery. C1ql1-null mice exhibited progressive hearing loss with elevated thresholds of auditory brainstem response and distortion product otoacoustic emission. Confocal microscopy showed that the number of nerve fibers innervating both IHCs and OHCs was significantly reduced. However, spiral ganglion neurons appeared to be normal under electron microscopy. IHC development and survival were not affected by deletion of C1ql1. Voltage-clamp recording and immunocytochmistry combined with confocal microscopy showed C1ql1-null IHCs showed no significant reduction of pre-synaptic proteins and synaptic vesicle release. This is in contrast to significant OHC loss in the KO mice. Our study suggests that C1ql1 is essential for development of hair cell innervation and OHC survival. But maturation of presynaptic machinery in IHCs does not depend on C1QL1.
Collapse
Affiliation(s)
- Yue Qi
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wei Xiong
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Shukui Yu
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhengde Du
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Tengfei Qu
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lu He
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wei Wei
- Department of Otology, Sheng Jing Hospital, China Medical University, Shenyang, China
| | - Lingjun Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ke Liu
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yi Li
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - David Z He
- Department of Biomedical Sciences, Creighton University, Omaha, NE, United States
| | - Shusheng Gong
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
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: 2.0] [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
|
8
|
Stojkovic M, Han D, Jeong M, Stojkovic P, Stankovic KM. Human induced pluripotent stem cells and CRISPR/Cas-mediated targeted genome editing: Platforms to tackle sensorineural hearing loss. STEM CELLS (DAYTON, OHIO) 2021; 39:673-696. [PMID: 33586253 DOI: 10.1002/stem.3353] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 12/13/2020] [Indexed: 11/09/2022]
Abstract
Hearing loss (HL) is a major global health problem of pandemic proportions. The most common type of HL is sensorineural hearing loss (SNHL) which typically occurs when cells within the inner ear are damaged. Human induced pluripotent stem cells (hiPSCs) can be generated from any individual including those who suffer from different types of HL. The development of new differentiation protocols to obtain cells of the inner ear including hair cells (HCs) and spiral ganglion neurons (SGNs) promises to expedite cell-based therapy and screening of potential pharmacologic and genetic therapies using human models. Considering age-related, acoustic, ototoxic, and genetic insults which are the most frequent causes of irreversible damage of HCs and SGNs, new methods of genome editing (GE), especially the CRISPR/Cas9 technology, could bring additional opportunities to understand the pathogenesis of human SNHL and identify novel therapies. However, important challenges associated with both hiPSCs and GE need to be overcome before scientific discoveries are correctly translated to effective and patient-safe applications. The purpose of the present review is (a) to summarize the findings from published reports utilizing hiPSCs for studies of SNHL, hence complementing recent reviews focused on animal studies, and (b) to outline promising future directions for deciphering SNHL using disruptive molecular and genomic technologies.
Collapse
Affiliation(s)
- Miodrag Stojkovic
- Eaton Peabody Laboratories, Department of Otolaryngology Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Dongjun Han
- Eaton Peabody Laboratories, Department of Otolaryngology Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Minjin Jeong
- Eaton Peabody Laboratories, Department of Otolaryngology Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Petra Stojkovic
- Eaton Peabody Laboratories, Department of Otolaryngology Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Konstantina M Stankovic
- Eaton Peabody Laboratories, Department of Otolaryngology Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, USA.,Program in Speech and Hearing Bioscience and Technology, Harvard University, Cambridge, Massachusetts, USA.,Harvard Program in Therapeutic Science, Harvard Medical School, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
9
|
Webber JL, Clancy JC, Zhou Y, Yraola N, Homma K, García-Añoveros J. Axodendritic versus axosomatic cochlear efferent termination is determined by afferent type in a hierarchical logic of circuit formation. SCIENCE ADVANCES 2021; 7:7/4/eabd8637. [PMID: 33523928 PMCID: PMC7817091 DOI: 10.1126/sciadv.abd8637] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/03/2020] [Indexed: 05/09/2023]
Abstract
Hearing involves a stereotyped neural network communicating cochlea and brain. How this sensorineural circuit assembles is largely unknown. The cochlea houses two types of mechanosensory hair cells differing in function (sound transmission versus amplification) and location (inner versus outer compartments). Inner (IHCs) and outer hair cells (OHCs) are each innervated by a distinct pair of afferent and efferent neurons: IHCs are contacted by type I afferents receiving axodendritic efferent contacts; OHCs are contacted by type II afferents and axosomatically terminating efferents. Using an Insm1 mouse mutant with IHCs in the position of OHCs, we discover a hierarchical sequence of instructions in which first IHCs attract, and OHCs repel, type I afferents; second, type II afferents innervate hair cells not contacted by type I afferents; and last, afferent fiber type determines if and how efferents innervate, whether axodendritically on the afferent, axosomatically on the hair cell, or not at all.
Collapse
Affiliation(s)
- Jemma L Webber
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - John C Clancy
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Yingjie Zhou
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Natalia Yraola
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Kazuaki Homma
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- The Hugh Knowles Center for Clinical and Basic Science in Hearing and its Disorders, Northwestern University, Chicago, IL 60611, USA
| | - Jaime García-Añoveros
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
- The Hugh Knowles Center for Clinical and Basic Science in Hearing and its Disorders, Northwestern University, Chicago, IL 60611, USA
- Departments of Neurology and Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
10
|
Hua Y, Ding X, Wang H, Wang F, Lu Y, Neef J, Gao Y, Moser T, Wu H. Electron Microscopic Reconstruction of Neural Circuitry in the Cochlea. Cell Rep 2021; 34:108551. [PMID: 33406431 DOI: 10.1016/j.celrep.2020.108551] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/25/2020] [Accepted: 12/03/2020] [Indexed: 02/02/2023] Open
Abstract
Recent studies reveal great diversity in the structure, function, and efferent innervation of afferent synaptic connections between the cochlear inner hair cells (IHCs) and spiral ganglion neurons (SGNs), which likely enables audition to process a wide range of sound pressures. By performing an extensive electron microscopic (EM) reconstruction of the neural circuitry in the mature mouse organ of Corti, we demonstrate that afferent SGN dendrites differ in abundance and composition of efferent innervation in a manner dependent on their afferent synaptic connectivity with IHCs. SGNs that sample glutamate release from several presynaptic ribbons receive more efferent innervation from lateral olivocochlear projections than those driven by a single ribbon. Next to the prevailing unbranched SGN dendrites, we found branched SGN dendrites that can contact several ribbons of 1-2 IHCs. Unexpectedly, medial olivocochlear neurons provide efferent innervation of SGN dendrites, preferring those forming single-ribbon, pillar-side synapses. We propose a fine-tuning of afferent and efferent SGN innervation.
Collapse
Affiliation(s)
- 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, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Connectomics, Max Planck Institute for Brain Research, Frankfurt/Main, Germany.
| | - Xu Ding
- 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
| | - Haoyu Wang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangfang Wang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Lu
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jakob Neef
- Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany; Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Yunge Gao
- 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
| | - Tobias Moser
- Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany; Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany; Multiscale Bioimaging Cluster of Excellence, University of Göttingen, Göttingen, Germany.
| | - 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, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
11
|
Disruption of Atg7-dependent autophagy causes electromotility disturbances, outer hair cell loss, and deafness in mice. Cell Death Dis 2020; 11:913. [PMID: 33099575 PMCID: PMC7585579 DOI: 10.1038/s41419-020-03110-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/04/2020] [Accepted: 10/06/2020] [Indexed: 12/11/2022]
Abstract
Atg7 is an indispensable factor that plays a role in canonical nonselective autophagy. Here we show that genetic ablation of Atg7 in outer hair cells (OHCs) in mice caused stereocilium damage, somatic electromotility disturbances, and presynaptic ribbon degeneration over time, which led to the gradual wholesale loss of OHCs and subsequent early-onset profound hearing loss. Impaired autophagy disrupted OHC mitochondrial function and triggered the accumulation of dysfunctional mitochondria that would otherwise be eliminated in a timely manner. Atg7-independent autophagy/mitophagy processes could not compensate for Atg7 deficiency and failed to rescue the terminally differentiated, non-proliferating OHCs. Our results show that OHCs orchestrate intricate nonselective and selective autophagic/mitophagy pathways working in concert to maintain cellular homeostasis. Overall, our results demonstrate that Atg7-dependent autophagy plays a pivotal cytoprotective role in preserving OHCs and maintaining hearing function.
Collapse
|
12
|
Barone CM, Douma S, Reijntjes DOJ, Browe BM, Köppl C, Klump G, Park TJ, Pyott SJ. Altered cochlear innervation in developing and mature naked and Damaraland mole rats. J Comp Neurol 2019; 527:2302-2316. [PMID: 30861124 PMCID: PMC6767702 DOI: 10.1002/cne.24682] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 01/04/2023]
Abstract
Compared to many other rodent species, naked mole rats (Heterocephalus glaber) have elevated auditory thresholds, poor frequency selectivity, and limited ability to localize sound. Because the cochlea is responsible for encoding and relaying auditory signals to the brain, we used immunofluorescence and quantitative image analysis to examine cochlear innervation in mature and developing naked mole rats compared to mice (Mus musculus), gerbils (Meriones unguiculatus), and Damaraland mole rats (Fukomys damarensis), another subterranean rodent. In comparison to mice and gerbils, we observed alterations in afferent and efferent innervation as well as their patterns of developmental refinement in naked and Damaraland mole rats. These alterations were, however, not always shared similarly between naked and Damaraland mole rats. Most conspicuously, in both naked and Damaraland mole rats, inner hair cell (IHC) afferent ribbon density was reduced, whereas outer hair cell afferent ribbon density was increased. Naked and Damaraland mole rats also showed reduced lateral and medial efferent terminal density. Developmentally, naked mole rats showed reduced and prolonged postnatal reorganization of afferent and efferent innervation. Damaraland mole rats showed no evidence of postnatal reorganization. Differences in cochlear innervation specifically between the two subterranean rodents and more broadly among rodents provides insight into the cochlear mechanisms that enhance frequency sensitivity and sound localization, maturation of the auditory system, and the evolutionary adaptations occurring in response to subterranean environments.
Collapse
Affiliation(s)
- Catherine M Barone
- Laboratory of Integrative Neuroscience, Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Sytse Douma
- Department of Otorhinolaryngology and Head/Neck Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Daniël O J Reijntjes
- Department of Otorhinolaryngology and Head/Neck Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Brigitte M Browe
- Laboratory of Integrative Neuroscience, Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Christine Köppl
- Cluster of Excellence "Hearing4All", Department of Neuroscience, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany.,Research Center Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Georg Klump
- Cluster of Excellence "Hearing4All", Department of Neuroscience, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany.,Research Center Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Thomas J Park
- Laboratory of Integrative Neuroscience, Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Sonja J Pyott
- Department of Otorhinolaryngology and Head/Neck Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
13
|
Di Stadio A, Ralli M, Ishai R, D'Ascanio L, Trabalzini F, Della Volpe A, Babighian G, Ricci G. Nucleolus vs Nucleus Count for Identifying Spiral Ganglion in Human Temporal Bone. J Int Adv Otol 2018; 14:181-189. [PMID: 30256195 DOI: 10.5152/iao.2018.5517] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVES Spiral ganglion (SG) counting is used in experimental studies conducted on age-, noise-, and drug-induced sensorineural hearing loss, as well as in the assessment of cochlear implant performances. Different methods of counting have been reported, but no definite standardization of such procedure has been published. The aim of our study is to identify the best method to count human spiral ganglions (SGs). MATERIALS AND METHODS By identification of nuclei or nucleoli as described by Schucknect, seven researchers with different experience levels counted SGs in 123 human temporal bones (TBs). Data on time of post-mortem bone removal post-mortem, methods of specimen's fixation, decalcification, and coloration were collected to test their possible influence on human tissue. Percentage, two-tailed t-test, Spearman's test, and one-way ANOVA were used to analyze the data. RESULTS Nucleoli were identified in 61% of cases, whereas nuclei were recognized in 100% of cases (p<0.005). Nucleoli presence in all four segments in the same temporal bone (TB) was observed in 69 cases (92%), whereas nuclei were identified in all four segments in 103 cases (83.7%) (p<0.001). The junior investigators requested a double check by the seniors in 25 (20.3%) cases for identifying and counting nucleoli, whereas the senior researchers showed no doubts in their identification and count. The only parameter positively affecting nucleoli identification in tissue preparation was bone removal for <12 h with respect to longer post-mortem time (p<0.001). CONCLUSION We suggest counting nuclei, rather than nucleoli, for spiral ganglion computation because of easier recognition of nuclei, especially in case of investigator's limited experience.
Collapse
Affiliation(s)
- Arianna Di Stadio
- University of Perugia, Permanent Temporal Bone Laboratory, Perugia, Italy
| | - Massimo Ralli
- University La Sapienza, Department of Oral and Maxillofacial Sciences, Rome, Italy
| | - Reuven Ishai
- Toronto General Hospital, Otolaryngology Department, Toronto, Canada
| | - Luca D'Ascanio
- "Carlo Poma" Civil Hospital, Department of Otolaryngology-Head and Neck Surgery, Mantova, Italy
| | | | - Antonio Della Volpe
- Santobono-Posillipon Hospital, Otology and Cochlear Implant Unit, Naples, Italy
| | - Gregorio Babighian
- University of Perugia, Permanent Temporal Bone Laboratory, Perugia, Italy
| | - Giampietro Ricci
- University of Perugia, Otolaryngology Department, Perugia, Italy
| |
Collapse
|
14
|
Ibrahim LA, Huang JJ, Wang SZ, Kim YJ, Zhang LI, Tao HW. Sparse Labeling and Neural Tracing in Brain Circuits by STARS Strategy: Revealing Morphological Development of Type II Spiral Ganglion Neurons. Cereb Cortex 2018; 31:5049854. [PMID: 29982390 DOI: 10.1093/cercor/bhy154] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Indexed: 11/12/2022] Open
Abstract
Elucidating axonal and dendritic projection patterns of individual neurons is a key for understanding the cytoarchitecture of neural circuits in the brain. This requires genetic approaches to achieve Golgi-like sparse labeling of desired types of neurons. Here, we explored a novel strategy of stochastic gene activation with regulated sparseness (STARS), in which the stochastic choice between 2 competing Cre-lox recombination events is controlled by varying the lox efficiency and cassette length. In a created STARS transgenic mouse crossed with various Cre driver lines, sparse neuronal labeling with a relatively uniform level of sparseness was achieved across different brain regions and cell types in both central and peripheral nervous systems. Tracing of individual type II peripheral auditory fibers revealed for the first time that they undergo experience-dependent developmental refinement, which is impaired by attenuating external sound input. Our results suggest that STARS strategy can be applied for circuit mapping and sparse gene manipulation.
Collapse
Affiliation(s)
- Leena A Ibrahim
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Junxiang J Huang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Medical Biology Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Sheng-Zhi Wang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Young J Kim
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - L I Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - H W Tao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
15
|
Rigon F, Gasparini F, Shimeld SM, Candiani S, Manni L. Developmental signature, synaptic connectivity and neurotransmission are conserved between vertebrate hair cells and tunicate coronal cells. J Comp Neurol 2018; 526:957-971. [PMID: 29277977 DOI: 10.1002/cne.24382] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 12/15/2017] [Accepted: 12/18/2017] [Indexed: 11/08/2022]
Abstract
In tunicates, the coronal organ represents a sentinel checking particle entrance into the pharynx. The organ differentiates from an anterior embryonic area considered a proto-placode. For their embryonic origin, morphological features and function, coronal sensory cells have been hypothesized to be homologues to vertebrate hair cells. However, vertebrate hair cells derive from a posterior placode. This contradicts one of the principle historical criteria for homology, similarity of position, which could be taken as evidence against coronal cells/hair cells homology. In the tunicates Ciona intestinalis and C. robusta, we found that the coronal organ expresses genes (Atoh, Notch, Delta-like, Hairy-b, and Musashi) characterizing vertebrate neural and hair cell development. Moreover, coronal cells exhibit a complex synaptic connectivity pattern, and express neurotransmitters (Glu, ACh, GABA, 5-HT, and catecholamines), or enzymes for their synthetic machinery, involved in hair cell activity. Lastly, coronal cells express the Trpa gene, which encodes an ion channel expressed in hair cells. These data lead us to hypothesize a model in which competence to make secondary mechanoreceptors was initially broadly distributed through placode territories, but has become confined to different placodes during the evolution of the vertebrate and tunicate lineages.
Collapse
Affiliation(s)
- Francesca Rigon
- Dipartimento di Biologia, Università di Padova, Padova, Italy
| | - Fabio Gasparini
- Dipartimento di Biologia, Università di Padova, Padova, Italy
| | | | - Simona Candiani
- Dipartimento di Scienze della Terra dell'Ambiente e della Vita, Università di Genova, Genova, Italy
| | - Lucia Manni
- Dipartimento di Biologia, Università di Padova, Padova, Italy
| |
Collapse
|
16
|
Bulut E, Öztürk L. Spontaneous otoacoustic emission recordings during contralateral pure-tone activation of medial olivocochlear reflex. Physiol Int 2017. [PMID: 28648121 DOI: 10.1556/2060.104.2017.2.7] [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: 11/19/2022]
Abstract
We hypothesized that cochlear frequency discrimination occurs through medial olivocochlear efferent (MOCE)-induced alterations in outer hair cell (OHC) electromotility, which is independent from basilar membrane traveling waves. After obtaining informed consent, volunteers with normal hearing (n = 10; mean age: 20.6 ± 1.2 years) and patients with unilateral deafness (n = 10; mean age: 30.2 ± 17.9 years) or bilateral deafness (n = 8; mean age: 30.7 ± 13.8 years) underwent a complete physical and audiological examination, and audiological tests including transient evoked otoacoustic emission and spontaneous otoacoustic emission (TEOAE and SOAE, respectively). SOAE recordings were performed during contralateral pure-tone stimuli at 1 and 3 kHz. SOAE recordings in the presence of contralateral pure-tone stimuli showed frequency-specific activation out of the initial frequency range of SOAE responses. Basilar membrane motion during pure-tone stimulation results from OHC activation by means of MOCE neurons rather than from a traveling wave. Eventually, frequency-specific responses obtained from SOAEs suggested that OHC electromotility may be responsible for frequency discrimination of the cochlea independently from basilar membrane motion.
Collapse
Affiliation(s)
- E Bulut
- 1 Department of Audiology, Trakya University Faculty of Health Sciences , Edirne, Turkey.,2 Department of Physiology, Faculty of Medicine, Trakya University , Edirne, Turkey
| | - L Öztürk
- 2 Department of Physiology, Faculty of Medicine, Trakya University , Edirne, Turkey
| |
Collapse
|
17
|
Type II Cochlear Ganglion Neurons Do Not Drive the Olivocochlear Reflex: Re-Examination of the Cochlear Phenotype in Peripherin Knock-Out Mice. eNeuro 2016; 3:eN-NWR-0207-16. [PMID: 27570826 PMCID: PMC4987660 DOI: 10.1523/eneuro.0207-16.2016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 07/15/2016] [Indexed: 12/26/2022] Open
Abstract
The cochlear nerve includes a small population of unmyelinated sensory fibers connecting outer hair cells to the brain. The functional role of these type II afferent neurons is controversial, because neurophysiological data are sparse. A recent study (Froud et al., 2015) reported that targeted deletion of peripherin, a type of neurofilament, eliminated type II afferents and inactivated efferent feedback to the outer hair cells, thereby suggesting that type II afferents were the sensory drive to this sound-evoked, negative-feedback reflex, the olivocochlear pathway. Here, we re-evaluated the cochlear phenotype in mice from the peripherin knock-out line and show that (1) type II afferent terminals are present in normal number and (2) olivocochlear suppression of cochlear responses is absent even when this efferent pathway is directly activated by shocks. We conclude that type II neurons are not the sensory drive for the efferent reflex and that peripherin deletion likely causes dysfunction of synaptic transmission between olivocochlear terminals and their peripheral targets.
Collapse
|
18
|
GluA2-Containing AMPA Receptors Distinguish Ribbon-Associated from Ribbonless Afferent Contacts on Rat Cochlear Hair Cells. eNeuro 2016; 3:eN-NWR-0078-16. [PMID: 27257620 PMCID: PMC4874539 DOI: 10.1523/eneuro.0078-16.2016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 04/26/2016] [Indexed: 12/12/2022] Open
Abstract
Mechanosensory hair cells release glutamate at ribbon synapses to excite postsynaptic afferent neurons, via AMPA-type ionotropic glutamate receptors (AMPARs). However, type II afferent neurons contacting outer hair cells in the mammalian cochlea were thought to differ in this respect, failing to show GluA immunolabeling and with many “ribbonless” afferent contacts. Here it is shown that antibodies to the AMPAR subunit GluA2 labeled afferent contacts below inner and outer hair cells in the rat cochlea, and that synaptic currents in type II afferents had AMPAR-specific pharmacology. Only half the postsynaptic densities of type II afferents that labeled for PSD-95, Shank, or Homer were associated with GluA2 immunopuncta or presynaptic ribbons, the “empty slots” corresponding to ribbonless contacts described previously. These results extend the universality of AMPAergic transmission by hair cells, and support the existence of silent afferent contacts.
Collapse
|
19
|
Buniello A, Ingham NJ, Lewis MA, Huma AC, Martinez-Vega R, Varela-Nieto I, Vizcay-Barrena G, Fleck RA, Houston O, Bardhan T, Johnson SL, White JK, Yuan H, Marcotti W, Steel KP. Wbp2 is required for normal glutamatergic synapses in the cochlea and is crucial for hearing. EMBO Mol Med 2016; 8:191-207. [PMID: 26881968 PMCID: PMC4772953 DOI: 10.15252/emmm.201505523] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 12/18/2015] [Accepted: 12/21/2015] [Indexed: 01/01/2023] Open
Abstract
WBP2 encodes the WW domain-binding protein 2 that acts as a transcriptional coactivator for estrogen receptor α (ESR1) and progesterone receptor (PGR). We reported that the loss of Wbp2 expression leads to progressive high-frequency hearing loss in mouse, as well as in two deaf children, each carrying two different variants in the WBP2 gene. The earliest abnormality we detect in Wbp2-deficient mice is a primary defect at inner hair cell afferent synapses. This study defines a new gene involved in the molecular pathway linking hearing impairment to hormonal signalling and provides new therapeutic targets.
Collapse
Affiliation(s)
- Annalisa Buniello
- Wolfson Centre For Age-Related Diseases, King's College London, London, UK Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Neil J Ingham
- Wolfson Centre For Age-Related Diseases, King's College London, London, UK Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Morag A Lewis
- Wolfson Centre For Age-Related Diseases, King's College London, London, UK Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Andreea C Huma
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Raquel Martinez-Vega
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain Centre for Biomedical Network Research on Rare Diseases (CIBERER), Unit 761, Instituto de Salud Carlos III, Madrid, Spain
| | - Isabel Varela-Nieto
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain Centre for Biomedical Network Research on Rare Diseases (CIBERER), Unit 761, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Roland A Fleck
- Centre for Ultrastructural Imaging, King's College London, London, UK
| | - Oliver Houston
- Department of Biomedical Science, University of Sheffield, Sheffield, UK
| | - Tanaya Bardhan
- Department of Biomedical Science, University of Sheffield, Sheffield, UK
| | - Stuart L Johnson
- Department of Biomedical Science, University of Sheffield, Sheffield, UK
| | - Jacqueline K White
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Huijun Yuan
- Medical Genetics Center, Southwest Hospital Third Military Medical University, Chongqing, China
| | - Walter Marcotti
- Department of Biomedical Science, University of Sheffield, Sheffield, UK
| | - Karen P Steel
- Wolfson Centre For Age-Related Diseases, King's College London, London, UK Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| |
Collapse
|
20
|
Cochlear afferent innervation development. Hear Res 2015; 330:157-69. [DOI: 10.1016/j.heares.2015.07.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 06/02/2015] [Accepted: 07/21/2015] [Indexed: 01/11/2023]
|
21
|
Blosa M, Sonntag M, Jäger C, Weigel S, Seeger J, Frischknecht R, Seidenbecher CI, Matthews RT, Arendt T, Rübsamen R, Morawski M. The extracellular matrix molecule brevican is an integral component of the machinery mediating fast synaptic transmission at the calyx of Held. J Physiol 2015. [PMID: 26223835 DOI: 10.1113/jp270849] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS The proteoglycan brevican is a major component of the extracellular matrix of perineuronal nets and is highly enriched in the perisynaptic space suggesting a role for synaptic transmission. We have introduced the calyx of Held in the auditory brainstem as a model system to study the impact of brevican on dynamics and reliability of synaptic transmission. In vivo extracellular single-unit recordings at the calyx of Held in brevican-deficient mice yielded a significant increase in the action potential (AP) transmission delay and a prolongation of pre- and postsynaptic APs. The changes in dynamics of signal transmission were accompanied by the reduction of presynaptic vGlut1 and ultrastructural changes in the perisynaptic space. These data show that brevican is an important mediator of fast synaptic transmission at the calyx of Held. ABSTRACT The extracellular matrix is an integral part of the neural tissue. Its most conspicuous manifestation in the brain are the perineuronal nets (PNs) which surround somata and proximal dendrites of distinct neuron types. The chondroitin sulfate proteoglycan brevican is a major component of PNs. In contrast to other PN-comprising proteoglycans (e.g. aggrecan and neurocan), brevican is mainly expressed in the perisynaptic space closely associated with both the pre- and postsynaptic membrane. This specific localization prompted the hypothesis that brevican might play a role in synaptic transmission. In the present study we specifically investigated the role of brevican in synaptic transmission at a central synapse, the calyx of Held in the medial nucleus of the trapezoid body, by the use of in vivo electrophysiology, immunohistochemistry, biochemistry and electron microscopy. In vivo extracellular single-unit recordings were acquired in brevican-deficient mice and the dynamics and reliability of synaptic transmission were compared to wild-type littermates. In knockout mice, the speed of pre-to-postsynaptic action potential (AP) transmission was reduced and the duration of the respective pre- and postsynaptic APs increased. The reliability of signal transmission, however, was not affected by the lack of brevican. The changes in dynamics of signal transmission were accompanied by the reduction of (i) presynaptic vGlut1 and (ii) the size of subsynaptic cavities. The present results suggest an essential role of brevican for the functionality of high-speed synaptic transmission at the calyx of Held.
Collapse
Affiliation(s)
- Maren Blosa
- Paul Flechsig Institute for Brain Research, Faculty of Medicine, University of Leipzig, 04103, Leipzig, Germany
| | - Mandy Sonntag
- Paul Flechsig Institute for Brain Research, Faculty of Medicine, University of Leipzig, 04103, Leipzig, Germany.,Institute of Biology, Faculty of Biology, Pharmacy and Psychology, University of Leipzig, 04103, Leipzig, Germany
| | - Carsten Jäger
- Paul Flechsig Institute for Brain Research, Faculty of Medicine, University of Leipzig, 04103, Leipzig, Germany
| | - Solveig Weigel
- Paul Flechsig Institute for Brain Research, Faculty of Medicine, University of Leipzig, 04103, Leipzig, Germany
| | - Johannes Seeger
- Institute of Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, University of Leipzig, 04103, Leipzig, Germany
| | | | | | - Russell T Matthews
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA
| | - Thomas Arendt
- Paul Flechsig Institute for Brain Research, Faculty of Medicine, University of Leipzig, 04103, Leipzig, Germany
| | - Rudolf Rübsamen
- Institute of Biology, Faculty of Biology, Pharmacy and Psychology, University of Leipzig, 04103, Leipzig, Germany
| | - Markus Morawski
- Paul Flechsig Institute for Brain Research, Faculty of Medicine, University of Leipzig, 04103, Leipzig, Germany
| |
Collapse
|
22
|
Nouvian R, Eybalin M, Puel JL. Cochlear efferents in developing adult and pathological conditions. Cell Tissue Res 2015; 361:301-9. [DOI: 10.1007/s00441-015-2158-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 02/19/2015] [Indexed: 10/23/2022]
|
23
|
Flores EN, Duggan A, Madathany T, Hogan AK, Márquez FG, Kumar G, Seal RP, Edwards RH, Liberman MC, García-Añoveros J. A non-canonical pathway from cochlea to brain signals tissue-damaging noise. Curr Biol 2015; 25:606-12. [PMID: 25639244 DOI: 10.1016/j.cub.2015.01.009] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 11/17/2014] [Accepted: 01/02/2015] [Indexed: 10/24/2022]
Abstract
Intense noise damages the cochlear organ of Corti, particularly the outer hair cells (OHCs) [1]; however, this epithelium is not innervated by nociceptors of somatosensory ganglia, which detect damage elsewhere in the body. The only sensory neurons innervating the organ of Corti originate from the spiral ganglion, roughly 95% of which innervate exclusively inner hair cells (IHCs) [2-4]. Upon sound stimulation, IHCs release glutamate to activate AMPA-type receptors on these myelinated type-I neurons, which carry the neuronal signals to the cochlear nucleus. The remaining spiral ganglion cells (type IIs) are unmyelinated and contact OHCs [2-4]. Their function is unknown. Using immunoreactivity to cFos, we documented neuronal activation in the brainstem of Vglut3(-/-) mice, in which the canonical auditory pathway (activation of type-I afferents by glutamate released from inner hair cells) is silenced [5, 6]. In these deaf mice, we found responses to noxious noise, which damages hair cells, but not to innocuous noise, in neurons of the cochlear nucleus, but not in the vestibular or trigeminal nuclei. This response originates in the cochlea and not in other areas also stimulated by intense noise (middle ear and vestibule) as it was absent in CD1 mice with selective cochlear degeneration but normal vestibular and somatosensory function. These data imply the existence of an alternative neuronal pathway from cochlea to brainstem that is activated by tissue-damaging noise and does not require glutamate release from IHCs. This detection of noise-induced tissue damage, possibly by type-II cochlear afferents, represents a novel form of sensation that we term auditory nociception.
Collapse
Affiliation(s)
- Emma N Flores
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Anne Duggan
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Thomas Madathany
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ann K Hogan
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Freddie G Márquez
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Gagan Kumar
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Rebecca P Seal
- Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA 94158-2517, USA
| | - Robert H Edwards
- Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA 94158-2517, USA
| | - M Charles Liberman
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, and Department of Otology and Laryngology, Harvard Medical School, Boston, MA 02114, USA
| | - Jaime García-Añoveros
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Departments of Neurology and Physiology and Hugh Knowles Center for Clinical and Basic Science in Hearing and Its Disorders, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
24
|
Overexpression of X-Linked Inhibitor of Apoptotic Protein (XIAP) reduces age-related neuronal degeneration in the mouse cochlea. Gene Ther 2014; 21:967-74. [PMID: 25142138 PMCID: PMC4978537 DOI: 10.1038/gt.2014.77] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Revised: 06/18/2014] [Accepted: 06/24/2014] [Indexed: 12/05/2022]
Abstract
Previously, we showed that age-related hearing loss (AHL) was delayed in C57BL6 mice overexpressing X-Linked Inhibitor of Apoptotic Protein (XIAP), and the delayed AHL was associated with attenuated hair cell (HC) loss in XIAP-overexpressing mice. Similar to other reports, the HC loss in aged mice was restricted to the basal turn in this previous study, and occurred slightly at the apical end of the cochlea, showing considerably less spread than the frequency region of hearing loss. In the present study, we examined whether and how AHL is related to the degeneration of neuronal innervation of the cochlea and if the overexpression of XIAP exerts a protective effect against age-related degeneration in both afferent and efferent cochlear neurites. In contrast to HC loss, degeneration of both afferent and efferent neurites spread to the middle turns of the cochlea. Moreover, XIAP-overexpressing mice lost fewer HC afferent dendrites and efferent axons, as well as fewer spiral ganglion neurons (SGNs) between 3– 14 months of age in comparison to wild-type littermates. The results suggest that age-related degeneration of cochlear neurites may be independent of HC loss. Further, the inhibition of apoptosis by XIAP appears to reduce degeneration of both afferent and efferent cochlear neurites.
Collapse
|
25
|
Guinan JJ. Olivocochlear efferent function: issues regarding methods and the interpretation of results. Front Syst Neurosci 2014; 8:142. [PMID: 25161612 PMCID: PMC4129934 DOI: 10.3389/fnsys.2014.00142] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 07/23/2014] [Indexed: 11/19/2022] Open
Abstract
As studies of the olivocochlear (OC) efferent system have matured, issues have been identified that need to be taken into account in the design of new studies and in the interpretation of existing work. The need for high signal-to-noise ratios (SNRs), multiple alternations of conditions, and avoiding middle-ear-muscle activation have been previously highlighted. Less well-known issues include: Contralateral medial OC (MOC) effects may not be good proxies for ipsilateral (ipsi) MOC effects; MOC-induced changes in otoacoustic emissions (OAEs) may not accurately show MOC-induced changes in auditory-nerve (AN) responses; measuring OAE differences from before to after psychophysical trials yields the transient OAE change but not tonic MOC activation; tonic MOC activation may be measurable by several techniques including by OAE differences in trials in which the subject’s judgment was correct vs. trials that were incorrect; SNRs can be preserved by Bootstrap statistical tests; differences in task difficulty may outweigh differences in subject attention; lateral efferent effects are little understood and may be tied to MOC effects; to assess whether MOC strength predicts protection from acoustic trauma, prospective tests in humans are needed.
Collapse
Affiliation(s)
- John J Guinan
- Eaton Peabody Laboratory of Auditory Physiology, Department of Otolaryngology, Massachusetts Eye and Ear Infirmary Boston, MA, USA ; Department of Otology and Laryngology, Harvard Medical School Boston, Massachusetts, USA
| |
Collapse
|
26
|
Topographic and quantitative evaluation of gentamicin-induced damage to peripheral innervation of mouse cochleae. Neurotoxicology 2013; 40:86-96. [PMID: 24308912 DOI: 10.1016/j.neuro.2013.11.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 11/11/2013] [Accepted: 11/11/2013] [Indexed: 11/24/2022]
Abstract
Ototoxicity induced by aminoglycoside antibiotics appears to occur both in hair cells (HCs) and the cochlear nerves that innervate them. Although HC loss can be easily quantified, neuronal lesions are difficult to quantify because two types of afferent dendrites and two types of efferent axons are tangled beneath the hair cells. In the present study, ototoxicity was induced by gentamicin in combination with the diuretic agent furosemide. Neuronal lesions were quantified in cochlear whole-mount preparations combined with microsections across the habenular perforate (HP) openings to achieve a clear picture of the topographic relationship between neuronal damage and HC loss. Multiple immunostaining methods were employed to differentiate the two types of afferent dendrites and two types of efferent axons. The results show that co-administration of gentamicin and furosemide resulted in a typical dynamic pattern of HC loss that spread from the basal turn to the outer hair cells to the apex and inner hair cells, depending on the dose and survival time after drug administration. Lesions of the innervation appeared to occur at two stages. At the early stage (2-4 days), the loss of labeling of the two types of afferent dendrites was more obvious than the loss of labeled efferent axons. At the late stage (2-4 weeks), the loss of labeled efferent axons was more rapid. In the high-dose gentamicin group, the loss of outer HCs was congruent with afferent dendrite loss at the early stage and efferent axon loss at the late stage. In the low-dose gentamicin group, the loss of labeling for cochlear innervation was more severe and widespread. Thus, we hypothesize that the gentamicin-induced damage to cochlear innervation occurs independently of hair cell loss.
Collapse
|
27
|
Kumaresan R, Peddinti VK, Cariani P. Synchrony capture filterbank: auditory-inspired signal processing for tracking individual frequency components in speech. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2013; 133:4290-4310. [PMID: 23742379 DOI: 10.1121/1.4802653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
A processing scheme for speech signals is proposed that emulates synchrony capture in the auditory nerve. The role of stimulus-locked spike timing is important for representation of stimulus periodicity, low frequency spectrum, and spatial location. In synchrony capture, dominant single frequency components in each frequency region impress their time structures on temporal firing patterns of auditory nerve fibers with nearby characteristic frequencies (CFs). At low frequencies, for voiced sounds, synchrony capture divides the nerve into discrete CF territories associated with individual harmonics. An adaptive, synchrony capture filterbank (SCFB) consisting of a fixed array of traditional, passive linear (gammatone) filters cascaded with a bank of adaptively tunable, bandpass filter triplets is proposed. Differences in triplet output envelopes steer triplet center frequencies via voltage controlled oscillators (VCOs). The SCFB exhibits some cochlea-like responses, such as two-tone suppression and distortion products, and possesses many desirable properties for processing speech, music, and natural sounds. Strong signal components dominate relatively greater numbers of filter channels, thereby yielding robust encodings of relative component intensities. The VCOs precisely lock onto harmonics most important for formant tracking, pitch perception, and sound separation.
Collapse
Affiliation(s)
- Ramdas Kumaresan
- Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, Rhode Island 02881, USA.
| | | | | |
Collapse
|
28
|
Van De Water TR. Historical Aspects of Inner Ear Anatomy and Biology that Underlie the Design of Hearing and Balance Prosthetic Devices. Anat Rec (Hoboken) 2012; 295:1741-59. [PMID: 23045252 DOI: 10.1002/ar.22598] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 07/24/2012] [Indexed: 12/19/2022]
Abstract
This review presents some of the major historical events that advanced the body of knowledge of the anatomy of the inner ear and its sensory receptors as well as the biology of these receptors that underlies the sensory functions of hearing and balance. This knowledge base of the inner ear's structure/function has been an essential factor for the design and construction of prosthetic devices to aid patients with deficits in their senses of hearing and balance. Prosthetic devices are now available for severely hearing impaired and deaf patients to restore hearing and are known as cochlear implants and auditory brain stem implants. A prosthetic device for patients with balance disorders is being perfected and is in an animal model testing phase with another prosthetic device for controlling intractable dizziness in Meniere's patients currently being evaluated in clinical testing. None of this would have been possible without the pioneering studies and discoveries of the investigators mentioned in this review and with the work of many other talented investigators to numerous to be covered in this review.
Collapse
Affiliation(s)
- Thomas R Van De Water
- Cochlear Implant Research Program, University of Miami Ear Institute, Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida 33136-1015, USA.
| |
Collapse
|
29
|
Mammano F. ATP-dependent intercellular Ca2+ signaling in the developing cochlea: facts, fantasies and perspectives. Semin Cell Dev Biol 2012; 24:31-9. [PMID: 23022499 DOI: 10.1016/j.semcdb.2012.09.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 09/14/2012] [Indexed: 10/27/2022]
Abstract
Hearing relies on a sensitive mechanoelectrical transduction process in the cochlea of the inner ear. The cochlea contains sensory, secretory, neural, supporting and epithelial cells which are all essential to the sound transduction process. It is well known that a complex extracellular purinergic signaling system contributes to cochlear homeostasis, altering cochlear sensitivity and neural output via ATP-gated ion channels (P2X receptors) and G protein-coupled P2Y receptors. This review focuses on the emerging roles of ATP that are currently under investigation in the developing sensory epithelium, with particular emphasis on the link between ATP release, Ca(2+) signaling, the expression and function of gap junction proteins connexin26 and connexin30, and the acquisition of hearing.
Collapse
Affiliation(s)
- Fabio Mammano
- Dipartimento di Fisica e Astronomia G. Galilei, Università di Padova, 35131 Padova, Italy.
| |
Collapse
|
30
|
Abstract
Auditory hair cells are surrounded on their basolateral aspects by supporting cells, and these two cell types together constitute the sensory epithelium of the organ of Corti, which is the hearing apparatus of the ear. We show here that Lgr5, a marker for adult stem cells, was expressed in a subset of supporting cells in the newborn and adult murine cochlea. Lgr5-expressing supporting cells, sorted by flow cytometry and cultured in a single-cell suspension, compared with unsorted cells, displayed an enhanced capacity for self-renewing neurosphere formation in response to Wnt and were converted to hair cells at a higher (>10-fold) rate. The greater differentiation of hair cells in the neurosphere assay showed that Lgr5-positive cells had the capacity to act as cochlear progenitor cells, and lineage tracing confirmed that Lgr5-expressing cells accounted for the cells that formed neurospheres and differentiated to hair cells. The responsiveness to Wnt of cells with a capacity for division and sensory cell formation suggests a potential route to new hair cell generation in the adult cochlea.
Collapse
|
31
|
Bulankina AV, Moser T. Neural circuit development in the mammalian cochlea. Physiology (Bethesda) 2012; 27:100-12. [PMID: 22505666 DOI: 10.1152/physiol.00036.2011] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The organ of Corti, the sensory epithelium of the mammalian auditory system, uses afferent and efferent synapses for encoding auditory signals and top-down modulation of cochlear function. During development, the final precisely ordered sensorineural circuit is established following excessive formation of afferent and efferent synapses and subsequent refinement. Here, we review the development of innervation of the mouse organ of Corti and its regulation.
Collapse
Affiliation(s)
- A V Bulankina
- InnerEarLab, Department of Otolaryngology, University of Goettingen School of Medicine, Goettingen, Germany
| | | |
Collapse
|
32
|
Maison SF, Usubuchi H, Vetter DE, Elgoyhen AB, Thomas SA, Liberman MC. Contralateral-noise effects on cochlear responses in anesthetized mice are dominated by feedback from an unknown pathway. J Neurophysiol 2012; 108:491-500. [PMID: 22514298 DOI: 10.1152/jn.01050.2011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Suppression of ipsilateral distortion product otoacoustic emissions (DPOAEs) by contralateral noise is used in humans and animals to assay the strength of sound-evoked negative feedback from the medial olivocochlear (MOC) efferent pathway. However, depending on species and anesthesia, contributions of other feedback systems to the middle or inner ear can cloud the interpretation. Here, contributions of MOC and middle-ear muscle reflexes, as well as autonomic feedback, to contra-noise suppression in anesthetized mice are dissected by selectively eliminating each pathway by surgical transection, pharmacological blockade, or targeted gene deletion. When ipsilateral DPOAEs were evoked by low-level primaries, contra-noise suppression was typically ~1 dB with contra-noise levels around 95 dB SPL, and it always disappeared upon contralateral cochlear destruction. Lack of middle-ear muscle contribution was suggested by persistence of contra-noise suppression after paralysis with curare, tensor tympani cauterization, or section of the facial nerve. Contribution of cochlear sympathetics was ruled out by studying mutant mice lacking adrenergic signaling (dopamine β-hydroxylase knockouts). Surprisingly, contra-noise effects on low-level DPOAEs were also not diminished by eliminating the MOC system pharmacologically (strychnine), surgically, or by deletion of relevant cholinergic receptors (α9/α10). In contrast, when ipsilateral DPOAEs were evoked by high-level primaries, the contra-noise suppression, although comparable in magnitude, was largely eliminated by MOC blockade or section. Possible alternate pathways are discussed for the source of contra-noise-evoked effects at low ipsilateral levels.
Collapse
Affiliation(s)
- Stéphane F Maison
- Department of Otology and Laryngology, Harvard Medical School and Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, MA 02114-3096, USA.
| | | | | | | | | | | |
Collapse
|
33
|
Lilaonitkul W, Guinan JJ. Frequency tuning of medial-olivocochlear-efferent acoustic reflexes in humans as functions of probe frequency. J Neurophysiol 2011; 107:1598-611. [PMID: 22190630 DOI: 10.1152/jn.00549.2011] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The medial-olivocochlear (MOC) acoustic reflex is thought to provide frequency-specific feedback that adjusts the gain of cochlear amplification, but little is known about how frequency specific the reflex actually is. We measured human MOC tuning through changes in stimulus frequency otoacoustic emissions (SFOAEs) from 40-dB-SPL tones at probe frequencies (f(p)s) near 0.5, 1.0, and 4.0 kHz. MOC activity was elicited by 60-dB-SPL ipsilateral, contralateral, or bilateral tones or half-octave noise bands, with elicitor frequency (f(e)) varied in half-octave steps. Tone and noise elicitors produced similar results. At all probe frequencies, SFOAE changes were produced by a wide range of elicitor frequencies with elicitor frequencies near 0.7-2.0 kHz being particularly effective. MOC-induced changes in SFOAE magnitude and SFOAE phase were surprisingly different functions of f(e): magnitude inhibition largest for f(e) close to f(p), phase change largest for f(e) remote from f(p). The metric ΔSFOAE, which combines both magnitude and phase changes, provided the best match to reported (cat) MOC neural inhibition. Ipsilateral and contralateral MOC reflexes often showed dramatic differences in plots of MOC effect vs. elicitor frequency, indicating that the contralateral reflex does not give an accurate picture of ipsilateral-reflex properties. These differences in MOC effects appear to imply that ipsilateral and contralateral reflexes have different actions in the cochlea. The implication of these results for MOC function, cochlear mechanics, and the production of SFOAEs are discussed.
Collapse
Affiliation(s)
- Watjana Lilaonitkul
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | |
Collapse
|
34
|
Ida-Eto M, Oyabu A, Ohkawara T, Tashiro Y, Narita N, Narita M. Existence of manserin, a secretogranin II-derived neuropeptide, in the rat inner ear: relevance to modulation of auditory and vestibular system. J Histochem Cytochem 2011; 60:69-75. [PMID: 22034517 DOI: 10.1369/0022155411425688] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Manserin is a 40-amino acid neuropeptide derived from rat brain. Manserin has been shown to distribute in the neuroendocrine system, such as the pituitary and adrenal glands, but it has been little studied in other organs. In this study, the authors examined localization of manserin in the inner ear of the adult Wistar rat using immunohistochemical analyses. Manserin immunoreactivity was detected in the neuronal terminals of the organ of Corti and type II spiral ganglion cells. In addition to being identified in the auditory system, manserin was detected at the synapses of the vestibular system, such as saccule, utricle, and semicircular canal. These results suggest that inner ear manserin may be involved in the function of peripheral auditory and vestibular systems.
Collapse
Affiliation(s)
- Michiru Ida-Eto
- Department of Anatomy II, Mie University, Graduate School of Medicine, Mie, Japan.
| | | | | | | | | | | |
Collapse
|
35
|
Nayagam BA, Muniak MA, Ryugo DK. The spiral ganglion: connecting the peripheral and central auditory systems. Hear Res 2011; 278:2-20. [PMID: 21530629 PMCID: PMC3152679 DOI: 10.1016/j.heares.2011.04.003] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Revised: 03/26/2011] [Accepted: 04/03/2011] [Indexed: 12/15/2022]
Abstract
In mammals, the initial bridge between the physical world of sound and perception of that sound is established by neurons of the spiral ganglion. The cell bodies of these neurons give rise to peripheral processes that contact acoustic receptors in the organ of Corti, and the central processes collect together to form the auditory nerve that projects into the brain. In order to better understand hearing at this initial stage, we need to know the following about spiral ganglion neurons: (1) their cell biology including cytoplasmic, cytoskeletal, and membrane properties, (2) their peripheral and central connections including synaptic structure; (3) the nature of their neural signaling; and (4) their capacity for plasticity and rehabilitation. In this report, we will update the progress on these topics and indicate important issues still awaiting resolution.
Collapse
Affiliation(s)
- Bryony A Nayagam
- Department of Otolaryngology, University of Melbourne, Melbourne, VIC Australia
| | - Michael A Muniak
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD USA
| | - David K Ryugo
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD USA
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, Baltimore, MD USA
- Garvan Institute, Darlinghurst, NSW Australia
| |
Collapse
|
36
|
Defourny J, Lallemend F, Malgrange B. Structure and development of cochlear afferent innervation in mammals. Am J Physiol Cell Physiol 2011; 301:C750-61. [PMID: 21753183 DOI: 10.1152/ajpcell.00516.2010] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In mammals, sensorineural deafness results from damage to the auditory receptors of the inner ear, the nerve pathways to the brain or the cortical area that receives sound information. In this review, we first focused on the cellular and molecular events taking part to spiral ganglion axon growth, extension to the organ of Corti, and refinement. In the second half, we considered the functional maturation of synaptic contacts between sensory hair cells and their afferent projections. A better understanding of all these processes could open insights into novel therapeutic strategies aimed to re-establish primary connections from sound transducers to the ascending auditory nerve pathways.
Collapse
|
37
|
Guinan JJ. Physiology of the Medial and Lateral Olivocochlear Systems. AUDITORY AND VESTIBULAR EFFERENTS 2011. [DOI: 10.1007/978-1-4419-7070-1_3] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
38
|
Wersinger E, Fuchs PA. Modulation of hair cell efferents. Hear Res 2010; 279:1-12. [PMID: 21187136 DOI: 10.1016/j.heares.2010.12.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Revised: 12/10/2010] [Accepted: 12/20/2010] [Indexed: 02/01/2023]
Abstract
Outer hair cells (OHCs) amplify the sound-evoked motion of the basilar membrane to enhance acoustic sensitivity and frequency selectivity. Medial olivocochlear (MOC) efferents inhibit OHCs to reduce the sound-evoked response of cochlear afferent neurons. OHC inhibition occurs through the activation of postsynaptic α9α10 nicotinic receptors tightly coupled to calcium-dependent SK2 channels that hyperpolarize the hair cell. MOC neurons are cholinergic but a number of other neurotransmitters and neuromodulators have been proposed to participate in efferent transmission, with emerging evidence for both pre- and postsynaptic effects. Cochlear inhibition in vivo is maximized by repetitive activation of the efferents, reflecting facilitation and summation of transmitter release onto outer hair cells. This review summarizes recent studies on cellular and molecular mechanisms of cholinergic inhibition and the regulation of those molecular components, in particular the involvement of intracellular calcium. Facilitation at the efferent synapse is compared in a variety of animals, as well as other possible mechanisms of modulation of ACh release. These results suggest that short-term plasticity contributes to effective cholinergic inhibition of hair cells.
Collapse
Affiliation(s)
- Eric Wersinger
- The Center for Hearing and Balance, Department of Otolaryngology Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | |
Collapse
|
39
|
Salt AN, Hullar TE. Responses of the ear to low frequency sounds, infrasound and wind turbines. Hear Res 2010; 268:12-21. [PMID: 20561575 PMCID: PMC2923251 DOI: 10.1016/j.heares.2010.06.007] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Revised: 06/07/2010] [Accepted: 06/09/2010] [Indexed: 01/12/2023]
Abstract
Infrasonic sounds are generated internally in the body (by respiration, heartbeat, coughing, etc) and by external sources, such as air conditioning systems, inside vehicles, some industrial processes and, now becoming increasingly prevalent, wind turbines. It is widely assumed that infrasound presented at an amplitude below what is audible has no influence on the ear. In this review, we consider possible ways that low frequency sounds, at levels that may or may not be heard, could influence the function of the ear. The inner ear has elaborate mechanisms to attenuate low frequency sound components before they are transmitted to the brain. The auditory portion of the ear, the cochlea, has two types of sensory cells, inner hair cells (IHC) and outer hair cells (OHC), of which the IHC are coupled to the afferent fibers that transmit "hearing" to the brain. The sensory stereocilia ("hairs") on the IHC are "fluid coupled" to mechanical stimuli, so their responses depend on stimulus velocity and their sensitivity decreases as sound frequency is lowered. In contrast, the OHC are directly coupled to mechanical stimuli, so their input remains greater than for IHC at low frequencies. At very low frequencies the OHC are stimulated by sounds at levels below those that are heard. Although the hair cells in other sensory structures such as the saccule may be tuned to infrasonic frequencies, auditory stimulus coupling to these structures is inefficient so that they are unlikely to be influenced by airborne infrasound. Structures that are involved in endolymph volume regulation are also known to be influenced by infrasound, but their sensitivity is also thought to be low. There are, however, abnormal states in which the ear becomes hypersensitive to infrasound. In most cases, the inner ear's responses to infrasound can be considered normal, but they could be associated with unfamiliar sensations or subtle changes in physiology. This raises the possibility that exposure to the infrasound component of wind turbine noise could influence the physiology of the ear.
Collapse
Affiliation(s)
- Alec N Salt
- Department of Otolaryngology, Washington University School of Medicine, Box 8115, 660 South Euclid Avenue, St Louis, MO 63110, USA.
| | | |
Collapse
|
40
|
Muscarinic signaling in the cochlea: presynaptic and postsynaptic effects on efferent feedback and afferent excitability. J Neurosci 2010; 30:6751-62. [PMID: 20463237 DOI: 10.1523/jneurosci.5080-09.2010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Acetylcholine is the major neurotransmitter of the olivocochlear efferent system, which provides feedback to cochlear hair cells and sensory neurons. To study the role of cochlear muscarinic receptors, we studied receptor localization with immunohistochemistry and reverse transcription-PCR and measured olivocochlear function, cochlear responses, and histopathology in mice with targeted deletion of each of the five receptor subtypes. M2, M4, and M5 were detected in microdissected immature (postnatal days 10-13) inner hair cells and spiral ganglion cells but not outer hair cells. In the adult (6 weeks), the same transcripts were found in microdissected organ of Corti and spiral ganglion samples. M2 protein was found, by immunohistochemistry, in olivocochlear fibers in both outer and inner hair cell areas. M3 mRNA was amplified only from whole cochleas, and M1 message was never seen in wild-type ears. Auditory brainstem responses (ABRs) and distortion product otoacoustic emissions (DPOAEs) were unaffected by loss of Gq-coupled receptors (M1, M3, or M5), as were shock-evoked olivocochlear effects and vulnerability to acoustic injury. In contrast, loss of Gi-coupled receptors (M2 and/or M4) decreased neural responses without affecting DPOAEs (at low frequencies). This phenotype and the expression pattern are consistent with excitatory muscarinic signaling in cochlear sensory neurons. At high frequencies, both ABRs and DPOAEs were attenuated by loss of M2 and/or M4, and the vulnerability to acoustic injury was dramatically decreased. This aspect of the phenotype and the expression pattern are consistent with a presynaptic role for muscarinic autoreceptors in decreasing ACh release from olivocochlear terminals during high-level acoustic stimulation and suggest that muscarinic antagonists could enhance the resistance of the inner ear to noise-induced hearing loss.
Collapse
|
41
|
Vater M, Kössl M. Comparative aspects of cochlear functional organization in mammals. Hear Res 2010; 273:89-99. [PMID: 20630478 DOI: 10.1016/j.heares.2010.05.018] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2009] [Revised: 05/02/2010] [Accepted: 05/25/2010] [Indexed: 01/10/2023]
Abstract
This review addresses the functional organization of the mammalian cochlea under a comparative and evolutionary perspective. A comparison of the monotreme cochlea with that of marsupial and placental mammals highlights important evolutionary steps towards a hearing organ dedicated to process higher frequencies and a larger frequency range than found in non-mammalian vertebrates. Among placental mammals, there are numerous cochlear specializations which relate to hearing range in adaptation to specific habitats that are superimposed on a common basic design. These are illustrated by examples of specialist ears which evolved excellent high frequency hearing and echolocation (bats and dolphins) and by the example of subterranean rodents with ears devoted to processing low frequencies. Furthermore, structural functional correlations important for tonotopic cochlear organization and predictions of hearing capabilities are discussed.
Collapse
Affiliation(s)
- Marianne Vater
- Institut Biochemie und Biologie, Allgemeine Zoologie, Universität Potsdam, Karl Liebknecht Str. 26, 14476 Golm, Germany.
| | | |
Collapse
|
42
|
Xia A, Gao SS, Yuan T, Osborn A, Bress A, Pfister M, Maricich SM, Pereira FA, Oghalai JS. Deficient forward transduction and enhanced reverse transduction in the alpha tectorin C1509G human hearing loss mutation. Dis Model Mech 2010; 3:209-23. [PMID: 20142329 DOI: 10.1242/dmm.004135] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Most forms of hearing loss are associated with loss of cochlear outer hair cells (OHCs). OHCs require the tectorial membrane (TM) for stereociliary bundle stimulation (forward transduction) and active feedback (reverse transduction). Alpha tectorin is a protein constituent of the TM and the C1509G mutation in alpha tectorin in humans results in autosomal dominant hearing loss. We engineered and validated this mutation in mice and found that the TM was shortened in heterozygous Tecta(C1509G/+) mice, reaching only the first row of OHCs. Thus, deficient forward transduction renders OHCs within the second and third rows non-functional, producing partial hearing loss. Surprisingly, both Tecta(C1509G/+) and Tecta(C1509G/C1509G) mice were found to have increased reverse transduction as assessed by sound- and electrically-evoked otoacoustic emissions. We show that an increase in prestin, a protein necessary for electromotility, in all three rows of OHCs underlies this phenomenon. This mouse model demonstrates a human hearing loss mutation in which OHC function is altered through a non-cell-autonomous variation in prestin.
Collapse
Affiliation(s)
- Anping Xia
- The Bobby R. Alford Department of Otolaryngology - Head and Neck Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
The mammalian cochlea is innervated by two classes of sensory neurons. Type I neurons make up 90-95% of the cochlear nerve and contact single inner hair cells (IHCs) to provide acoustic analysis as we know it. In contrast, the far less numerous Type II neurons arborize extensively among outer hair cells (OHCs) 1,2 and supporting cells3,4. Their scarcity, and smaller caliber axons, have made them the subject of much speculation, but little experimental progress for the past 50 years. Here we record from Type II fibers near their terminal arbors under OHCs to show that these receive excitatory glutamatergic synaptic input. The Type II peripheral arbor conducts action potentials, but the small and infrequent glutamatergic excitation implies a requirement for strong acoustic stimulation. Further, we show that Type II neurons are excited by adenosine tri-phosphate (ATP). Exogenous ATP depolarized Type II neurons both directly, and by evoking glutamatergic synaptic input 5. The present results prove that Type II neurons function as cochlear afferents, and can be modulated by ATP. The lesser magnitude of synaptic drive dictates a fundamentally different role in auditory signaling from that of Type I afferents.
Collapse
|
44
|
Current world literature. Curr Opin Otolaryngol Head Neck Surg 2009; 17:412-8. [PMID: 19755872 DOI: 10.1097/moo.0b013e3283318f24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
45
|
Complexin-I is required for high-fidelity transmission at the endbulb of Held auditory synapse. J Neurosci 2009; 29:7991-8004. [PMID: 19553439 DOI: 10.1523/jneurosci.0632-09.2009] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Complexins (CPXs I-IV) presumably act as regulators of the SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) complex, but their function in the intact mammalian nervous system is not well established. Here, we explored the role of CPXs in the mouse auditory system. Hearing was impaired in CPX I knock-out mice but normal in knock-out mice for CPXs II, III, IV, and III/IV as measured by auditory brainstem responses. Complexins were not detectable in cochlear hair cells but CPX I was expressed in spiral ganglion neurons (SGNs) that give rise to the auditory nerve. Ca(2+)-dependent exocytosis of inner hair cells and sound encoding by SGNs were unaffected in CPX I knock-out mice. In the absence of CPX I, the resting release probability in the endbulb of Held synapses of the auditory nerve fibers with bushy cells in the cochlear nucleus was reduced. As predicted by computational modeling, bushy cells had decreased spike rates at sound onset as well as longer and more variable first spike latencies explaining the abnormal auditory brainstem responses. In addition, we found synaptic transmission to outlast the stimulus at many endbulb of Held synapses in vitro and in vivo, suggesting impaired synchronization of release to stimulus offset. Although sound encoding in the cochlea proceeds in the absence of complexins, CPX I is required for faithful processing of sound onset and offset in the cochlear nucleus.
Collapse
|
46
|
|
47
|
Human medial olivocochlear reflex: effects as functions of contralateral, ipsilateral, and bilateral elicitor bandwidths. J Assoc Res Otolaryngol 2009; 10:459-70. [PMID: 19263165 DOI: 10.1007/s10162-009-0163-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2008] [Accepted: 02/09/2009] [Indexed: 10/21/2022] Open
Abstract
Animal studies have led to the view that the acoustic medial olivocochlear (MOC) efferent reflex provides sharply tuned frequency-specific feedback that inhibits cochlear amplification. To determine if MOC activation is indeed narrow band, we measured the MOC effects in humans elicited by 60-dB sound pressure level (SPL) contralateral, ipsilateral, and bilateral noise bands as a function of noise bandwidth from 1/2 to 6.7 octaves. MOC effects were quantified by the change in stimulus frequency otoacoustic emissions from 40 dB SPL probe tones near 0.5, 1, and 4 kHz. In a second experiment, the noise bands were centered 2 octaves below probe frequencies near 1 and 4 kHz. In all cases, the MOC effects increased as elicitor bandwidth increased, with the effect saturating at about 4 octaves. Generally, the MOC effects increased as the probe frequency decreased, opposite expectations based on MOC innervation density in the cochlea. Bilateral-elicitor effects were always the largest. The ratio of ipsilateral/contralateral effects depended on elicitor bandwidth; the ratio was large for narrow-band probe-centered elicitors and approximately unity for wide-band elicitors. In another experiment, the MOC effects from increasing elicitor bandwidths were calculated from measurements of the MOC effects from adjacent half-octave noise bands. The predicted bandwidth function agreed well with the measured bandwidth function for contralateral elicitors, but overestimated it for ipsilateral and bilateral elicitors. Overall, the results indicate that (1) the MOC reflexes integrate excitation from almost the entire cochlear length, (2) as elicitor bandwidth is increased, the excitation from newly stimulated cochlear regions more than overcomes the reduced excitation at frequencies in the center of the elicitor band, and (3) contralateral, ipsilateral, and bilateral elicitors show MOC reflex spatial summation over most of the cochlea, but ipsilateral spatial summation is less additive than contralateral.
Collapse
|
48
|
Lilaonitkul W, Guinan JJ. Reflex control of the human inner ear: a half-octave offset in medial efferent feedback that is consistent with an efferent role in the control of masking. J Neurophysiol 2009; 101:1394-406. [PMID: 19118109 PMCID: PMC2666406 DOI: 10.1152/jn.90925.2008] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2008] [Accepted: 12/29/2008] [Indexed: 11/22/2022] Open
Abstract
The high sensitivity and frequency selectivity of the mammalian cochlea is due to amplification produced by outer hair cells (OHCs) and controlled by medial olivocochlear (MOC) efferents. Data from animals led to the view that MOC fibers provide frequency-specific inhibitory feedback; however, these studies did not measure intact MOC reflexes. To test whether MOC inhibition is primarily at the frequency that elicits the MOC activity, acoustically elicited MOC effects were quantified in humans by the change in otoacoustic emissions produced by 60-dB SPL tone and half-octave-band noise elicitors at different frequencies relative to a 40-dB SPL, 1-kHz probe tone. On average, all elicitors produced MOC effects that were skewed (elicitor frequencies -1 octave below the probe produced larger effects than those -1 octave above). The largest MOC effects were from elicitors below the probe frequency for contra- and bilateral elicitors but were from elicitors centered at the probe frequency for ipsilateral elicitors. Typically, ipsilateral elicitors produced larger effects than contralateral elicitors and bilateral elicitors produced effects near the ipsi+contra sum. Elicitors at levels down to 30-dB SPL produced similar patterns. Tuning curves (TCs) interpolated from these data were V-shaped with Q10s approximately 2. These are sharper than MOC-fiber TCs found near 1 kHz in cats and guinea pigs. Because cochlear amplification is skewed (more below the best frequency of a cochlear region), these data are consistent with an anti-masking role of MOC efferents that reduces masking by reducing the cochlear amplification seen at 1 kHz.
Collapse
Affiliation(s)
- Watjana Lilaonitkul
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | |
Collapse
|
49
|
Maison SF, Casanova E, Holstein GR, Bettler B, Liberman MC. Loss of GABAB receptors in cochlear neurons: threshold elevation suggests modulation of outer hair cell function by type II afferent fibers. J Assoc Res Otolaryngol 2008; 10:50-63. [PMID: 18925381 DOI: 10.1007/s10162-008-0138-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2008] [Accepted: 09/02/2008] [Indexed: 01/25/2023] Open
Abstract
Despite pharmacological and immunohistochemical evidence for GABA as a neurotransmitter in the olivocochlear efferent bundle, a clear functional role of GABA in the inner ear has not emerged. To explore the role of metabotropic GABA(B) receptors, we characterized the cochlear phenotype of mice with targeted deletion of the GABA(B1) subunit and determined its tissue localization using a mouse line expressing a GFP-tagged GABA(B1) subunit under the endogenous promoter. Immunostaining revealed GABA(B1) expression in both type I and type II ganglion cells and in their synaptic terminals under inner and outer hair cells, respectively. No GABA(B1) expression was observed in hair cells. Mean cochlear thresholds, measured via both auditory brainstem responses and distortion product otoacoustic emissions (DPOAEs), were elevated by approximately 10 dB in GABA(B1)-deficient mice, consistent with outer hair cell dysfunction. Olivocochlear efferent function, assessed via DPOAE suppression during efferent electrical stimulation, was unaffected by GABA(B1) deletion. GABA(B1)-deficient mice showed increased resistance to permanent acoustic injury, with mean threshold shifts approximately 25 dB smaller than wild-types after exposure to 8-16-kHz noise at 100 dB for 2 h. In contrast, there was no vulnerability difference to temporary acoustic injury following exposure to the same noise at 94 dB for 15 min. Our results suggest that GABAergic signaling in type II afferent neurons may be required for normal outer hair cell amplifier function at low sound levels and may also modulate outer hair cell responses to high-level sound.
Collapse
Affiliation(s)
- Stéphane F Maison
- Department of Otology and Laryngology, Harvard Medical School and Eaton-Peabody Laboratory, Massachusetts Eye & Ear Infirmary, 243 Charles St., Boston, MA 02114-3096, USA.
| | | | | | | | | |
Collapse
|
50
|
Glowatzki E, Grant L, Fuchs P. Hair cell afferent synapses. Curr Opin Neurobiol 2008; 18:389-95. [PMID: 18824101 DOI: 10.1016/j.conb.2008.09.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2008] [Revised: 09/08/2008] [Accepted: 09/15/2008] [Indexed: 12/30/2022]
Abstract
This review will cover advances in the study of hair cell afferent synaptic function occurring between 2005 and 2008. During this time, capacitance measurements of vesicular fusion have continued to be refined, optical methods have added insights regarding vesicle trafficking, and paired intracellular recordings have established the transfer function of the afferent synapse at high resolution. Further, genes have been identified with forms of deafness known as auditory neuropathy, and their role in afferent signaling explored in mouse models. With these advances, our view of the hair cell afferent synapse has continued to be refined, and surprising properties have been revealed that emphasize the unique role of this structure in neural function.
Collapse
Affiliation(s)
- Elisabeth Glowatzki
- Department of Otolaryngology, Johns Hopkins School of Medicine, 720 Rutland Avenue, Ross 824, Baltimore, MD 21205, United States.
| | | | | |
Collapse
|