1
|
Kennedy HJ, Evans MG. Conductance properties of the α9α10 nicotinic acetylcholine receptor of neonatal mouse inner and outer hair cells. Hear Res 2024; 453:109126. [PMID: 39383639 DOI: 10.1016/j.heares.2024.109126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 06/17/2024] [Accepted: 09/30/2024] [Indexed: 10/11/2024]
Abstract
In the developing cochlea, just before the onset of hearing on postnatal day 12, the medial olivocochlear efferent axons in synaptic contact with the inner hair cells (IHCs) start withdrawing and new efferent synaptic connections are formed on the outer hair cells (OHCs), thereby progressing towards the adult pattern of medial olivocochlear efferent innervation. The synapses are inhibitory, calcium influx through the α9α10 nicotinic acetylcholine receptors (nAChRs) driving opening of calcium-dependent potassium channels. The nAChRs appear to function similarly in IHCs and OHCs, although with probable kinetic differences. Our aim was to assess their functional similarity in the neonatal mouse cochlea by making whole-cell recordings from both hair cell types between postnatal day 7 and 10 when nAChRs are expressed. ACh was applied to voltage-clamped hair cells by pressure-ejection from a pipette. The cells were dialysed with a Cs+-based solution designed to eliminate calcium-dependent potassium currents. There were differences in amplitude, voltage-sensitivity and reversal potential of the nAChR currents between IHCs and OHCs. There was also some indication that IHC nAChRs have slower activation and desensitization kinetics, although the relatively slow ACh application limited interpretation of this result. These differences, particularly concerning the reversal potential, might indicate the presence of different auxiliary protein subunits of the α9α10 receptor in neonatal IHCs and OHCs.
Collapse
Affiliation(s)
- Helen J Kennedy
- School of Physiology, Pharmacology & Neuroscience, Bristol Neuroscience, University Walk, Bristol BS8 1TD, UK
| | | |
Collapse
|
2
|
Fischl M, Pederson A, Voglewede R, Cheng H, Drew J, Torres Cadenas L, Weisz CJC. Fast Inhibition Slows and Desynchronizes Mouse Auditory Efferent Neuron Activity. J Neurosci 2024; 44:e0382242024. [PMID: 38937103 PMCID: PMC11326868 DOI: 10.1523/jneurosci.0382-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/04/2024] [Accepted: 06/11/2024] [Indexed: 06/29/2024] Open
Abstract
The encoding of acoustic stimuli requires precise neuron timing. Auditory neurons in the cochlear nucleus (CN) and brainstem are well suited for accurate analysis of fast acoustic signals, given their physiological specializations of fast membrane time constants, fast axonal conduction, and reliable synaptic transmission. The medial olivocochlear (MOC) neurons that provide efferent inhibition of the cochlea reside in the ventral brainstem and participate in these fast neural circuits. However, their modulation of cochlear function occurs over time scales of a slower nature. This suggests the presence of mechanisms that reduce MOC inhibition of cochlear function. To determine how monaural excitatory and inhibitory synaptic inputs integrate to affect the timing of MOC neuron activity, we developed a novel in vitro slice preparation ("wedge-slice"). The wedge-slice maintains the ascending auditory nerve root, the entire CN and projecting axons, while preserving the ability to perform visually guided patch-clamp electrophysiology recordings from genetically identified MOC neurons. The "in vivo-like" timing of the wedge-slice demonstrates that the inhibitory pathway accelerates relative to the excitatory pathway when the ascending circuit is intact, and the CN portion of the inhibitory circuit is precise enough to compensate for reduced precision in later synapses. When combined with machine learning PSC analysis and computational modeling, we demonstrate a larger suppression of MOC neuron activity when the inhibition occurs with in vivo-like timing. This delay of MOC activity may ensure that the MOC system is only engaged by sustained background sounds, preventing a maladaptive hypersuppression of cochlear activity.
Collapse
Affiliation(s)
- Matthew Fischl
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland 20892
| | - Alia Pederson
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland 20892
| | - Rebecca Voglewede
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland 20892
| | - Hui Cheng
- NIDCD Data Science Core, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland 20892
| | - Jordan Drew
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland 20892
| | - Lester Torres Cadenas
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland 20892
| | - Catherine J C Weisz
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland 20892
| |
Collapse
|
3
|
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
|
4
|
Slika E, Fuchs PA. Genetic tools for studying cochlear inhibition. Front Cell Neurosci 2024; 18:1372948. [PMID: 38560293 PMCID: PMC10978695 DOI: 10.3389/fncel.2024.1372948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 02/28/2024] [Indexed: 04/04/2024] Open
Abstract
Efferent feedback to the mammalian cochlea includes cholinergic medial olivocochlear neurons (MOCs) that release ACh to hyperpolarize and shunt the voltage change that drives electromotility of outer hair cells (OHCs). Via brainstem connectivity, MOCs are activated by sound in a frequency- and intensity-dependent manner, thereby reducing the amplification of cochlear vibration provided by OHC electromotility. Among other roles, this efferent feedback protects the cochlea from acoustic trauma. Lesion studies, as well as a variety of genetic mouse models, support the hypothesis of efferent protection from acoustic trauma. Genetic knockout and gain-of-function knockin of the unique α9α10-containing nicotinic acetylcholine receptor (nAChR) in hair cells show that acoustic protection correlates with the efficacy of cholinergic inhibition of OHCs. This protective effect was replicated by viral transduction of the gain-of-function α9L9'T nAChR into α9-knockout mice. Continued progress with "efferent gene therapy" will require a reliable method for visualizing nAChR expression in cochlear hair cells. To that end, mice expressing HA-tagged α9 or α10 nAChRs were generated using CRISPR technology. This progress will facilitate continued study of the hair cell nAChR as a therapeutic target to prevent hearing loss and potentially to ameliorate associated pathologies such as hyperacusis.
Collapse
Affiliation(s)
| | - Paul Albert Fuchs
- The Center for Hearing and Balance, Otolaryngology-Head and Neck Surgery, Johns Hopkins, University School of Medicine Baltimore, Baltimore, MD, United States
| |
Collapse
|
5
|
Fischl M, Pederson A, Voglewede R, Cheng H, Drew J, Cadenas LT, Weisz CJ. Fast inhibition slows and desynchronizes mouse auditory efferent neuron activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.21.572886. [PMID: 38313270 PMCID: PMC10836066 DOI: 10.1101/2023.12.21.572886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
The encoding of acoustic stimuli requires precise neuron timing. Auditory neurons in the cochlear nucleus (CN) and brainstem are well-suited for accurate analysis of fast acoustic signals, given their physiological specializations of fast membrane time constants, fast axonal conduction, and reliable synaptic transmission. The medial olivocochlear (MOC) neurons that provide efferent inhibition of the cochlea reside in the ventral brainstem and participate in these fast neural circuits. However, their modulation of cochlear function occurs over time scales of a slower nature. This suggests the presence of mechanisms that restrict MOC inhibition of cochlear function. To determine how monaural excitatory and inhibitory synaptic inputs integrate to affect the timing of MOC neuron activity, we developed a novel in vitro slice preparation ('wedge-slice'). The wedge-slice maintains the ascending auditory nerve root, the entire CN and projecting axons, while preserving the ability to perform visually guided patch-clamp electrophysiology recordings from genetically identified MOC neurons. The 'in vivo-like' timing of the wedge-slice demonstrates that the inhibitory pathway accelerates relative to the excitatory pathway when the ascending circuit is intact, and the CN portion of the inhibitory circuit is precise enough to compensate for reduced precision in later synapses. When combined with machine learning PSC analysis and computational modeling, we demonstrate a larger suppression of MOC neuron activity when the inhibition occurs with in vivo-like timing. This delay of MOC activity may ensure that the MOC system is only engaged by sustained background sounds, preventing a maladaptive hyper-suppression of cochlear activity.
Collapse
Affiliation(s)
- Matthew Fischl
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, USA
- Current affiliation: Lafayette College, Neuroscience Program, Easton, PA 18042, USA
| | - Alia Pederson
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, USA
- Current affiliation: The University of Texas at Austin Dell Medical School, Austin, TX 78712, USA
| | - Rebecca Voglewede
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, USA
| | - Hui Cheng
- Bioinformatics and Biostatistics Collaboration Core, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, USA
| | - Jordan Drew
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, USA
- Current affiliation: Institute for Learning and Brain Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Lester Torres Cadenas
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, USA
| | - Catherine J.C. Weisz
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
6
|
Zhang Y, Hiel H, Vincent PF, Wood MB, Elgoyhen AB, Chien W, Lauer A, Fuchs PA. Engineering olivocochlear inhibition to reduce acoustic trauma. Mol Ther Methods Clin Dev 2023; 29:17-31. [PMID: 36941920 PMCID: PMC10023855 DOI: 10.1016/j.omtm.2023.02.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/22/2023] [Indexed: 02/27/2023]
Abstract
Efferent brain-stem neurons release acetylcholine to desensitize cochlear hair cells and can protect the inner ear from acoustic trauma. That protection is absent from knockout mice lacking efferent inhibition and is stronger in mice with a gain-of-function point mutation of the hair cell-specific nicotinic acetylcholine receptor. The present work uses viral transduction of gain-of-function receptors to restore acoustic prophylaxis to the knockout mice. Widespread postsynaptic expression of the transgene was visualized in excised tissue with a fluorophore-conjugated peptide toxin that binds selectively to hair cell acetylcholine receptors. Viral transduction into efferent knockout mice reduced the temporary hearing loss measured 1 day post acoustic trauma. The acoustic evoked-response waveform (auditory brain-stem response) recovered more rapidly in treated mice than in control mice. Thus, both cochlear amplification by outer hair cells (threshold shift) and afferent signaling (evoked-response amplitude) in knockout mice were protected by viral transduction of hair cell acetylcholine receptors. Gene therapy to strengthen efferent cochlear feedback could be complementary to existing and future therapies to prevent hearing loss, including ear coverings, hearing aids, single-gene repair, or small-molecule therapies.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- The Center for Hearing and Balance, Otolaryngology–Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hakim Hiel
- The Center for Hearing and Balance, Otolaryngology–Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Philippe F.Y. Vincent
- The Center for Hearing and Balance, Otolaryngology–Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Megan B. Wood
- The Center for Hearing and Balance, Otolaryngology–Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ana B. Elgoyhen
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular Dr. Héctor N. Torres (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428ADN CABA, Buenos Aires, Argentina
| | - Wade Chien
- The Center for Hearing and Balance, Otolaryngology–Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Inner Ear Gene Therapy Program, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, Bethesda, MD 20892, USA
| | - Amanda Lauer
- The Center for Hearing and Balance, Otolaryngology–Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Paul A. Fuchs
- The Center for Hearing and Balance, Otolaryngology–Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
7
|
Ashmore JF, Oghalai JS, Dewey JB, Olson ES, Strimbu CE, Wang Y, Shera CA, Altoè A, Abdala C, Elgoyhen AB, Eatock RA, Raphael RM. The Remarkable Outer Hair Cell: Proceedings of a Symposium in Honour of W. E. Brownell. J Assoc Res Otolaryngol 2023; 24:117-127. [PMID: 36648734 PMCID: PMC10121982 DOI: 10.1007/s10162-022-00852-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/02/2022] [Indexed: 01/18/2023] Open
Abstract
In 1985, Bill Brownell and colleagues published the remarkable observation that cochlear outer hair cells (OHCs) express voltage-driven mechanical motion: electromotility. They proposed OHC electromotility as the mechanism for the elusive "cochlear amplifier" required to explain the sensitivity of mammalian hearing. The finding and hypothesis stimulated an explosion of experiments that have transformed our understanding of cochlear mechanics and physiology, the evolution of hair cell structure and function, and audiology. Here, we bring together examples of current research that illustrate the continuing impact of the discovery of OHC electromotility.
Collapse
Affiliation(s)
| | - John S Oghalai
- Department of Otolaryngology-Head and Neck Surgery, University of Southern California, Los Angeles, USA
| | - James B Dewey
- Department of Otolaryngology-Head and Neck Surgery, University of Southern California, Los Angeles, USA
| | - Elizabeth S Olson
- Department of Otolaryngology Head and Neck Surgery, Vagelos College of Physicians and Surgeons, Columbia University, New York City, USA
| | - Clark E Strimbu
- Department of Otolaryngology Head and Neck Surgery, Vagelos College of Physicians and Surgeons, Columbia University, New York City, USA
| | - Yi Wang
- Department of Otolaryngology Head and Neck Surgery, Vagelos College of Physicians and Surgeons, Columbia University, New York City, USA
| | - Christopher A Shera
- Caruso Department of Otolaryngology and Department of Physics and Astronomy, University of Southern California, Los Angeles, USA
| | - Alessandro Altoè
- Caruso Department of Otolaryngology and Department of Physics and Astronomy, University of Southern California, Los Angeles, USA
| | - Carolina Abdala
- Caruso Department of Otolaryngology, University of Southern California, Los Angeles, USA
| | - Ana B Elgoyhen
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | | | | |
Collapse
|
8
|
Elgoyhen AB. The α9α10 acetylcholine receptor: a non-neuronal nicotinic receptor. Pharmacol Res 2023; 190:106735. [PMID: 36931539 DOI: 10.1016/j.phrs.2023.106735] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/06/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023]
Abstract
Within the superfamily of pentameric ligand-gated ion channels, cholinergic nicotinic receptors (nAChRs) were classically identified to mediate synaptic transmission in the nervous system and the neuromuscular junction. The α9 and α10 nAChR subunits were the last ones to be identified. Surprisingly, they do not fall into the dichotomic neuronal/muscle classification of nAChRs. They assemble into heteropentamers with a well-established function as canonical ion channels in inner ear hair cells, where they mediate central nervous system control of auditory and vestibular sensory processing. The present review includes expression, pharmacological, structure-function, molecular evolution and pathophysiological studies, that define receptors composed from α9 and α10 subunits as distant and distinct members within the nAChR family. Thus, although α9 and α10 were initially included within the neuronal subdivision of nAChR subunits, they form a distinct clade within the phylogeny of nAChRs. Following the classification of nAChR subunits based on their main synaptic site of action, α9 and α10 should receive a name in their own right.
Collapse
Affiliation(s)
- Ana Belén Elgoyhen
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Vuelta de Obligado 2490, Buenos Aires 1428, Argentina.
| |
Collapse
|
9
|
Cadenas LT, Cheng H, Weisz CJC. Synaptic plasticity of inhibitory synapses onto medial olivocochlear efferent neurons. J Physiol 2022; 600:2747-2763. [PMID: 35443073 PMCID: PMC9323901 DOI: 10.1113/jp282815] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/04/2022] [Indexed: 11/18/2022] Open
Abstract
Abstract The descending auditory system modulates the ascending system at every level. The final descending, or efferent, stage comprises lateral olivocochlear and medial olivocochlear (MOC) neurons. MOC somata in the ventral brainstem project axons to the cochlea to synapse onto outer hair cells (OHC), inhibiting OHC‐mediated cochlear amplification. MOC suppression of OHC function is implicated in cochlear gain control with changing sound intensity, detection of salient stimuli, attention and protection against acoustic trauma. Thus, sound excites MOC neurons to provide negative feedback of the cochlea. Sound also inhibits MOC neurons via medial nucleus of the trapezoid body (MNTB) neurons. However, MNTB–MOC synapses exhibit short‐term depression, suggesting reduced MNTB–MOC inhibition during sustained stimuli. Further, due to high rates of both baseline and sound‐evoked activity in MNTB neurons in vivo, MNTB–MOC synapses may be tonically depressed. To probe this, we characterized short‐term plasticity of MNTB–MOC synapses in mouse brain slices. We mimicked in vivo‐like temperature and extracellular calcium conditions, and in vivo‐like activity patterns of fast synaptic activation rates, sustained activation and prior tonic activity. Synaptic depression was sensitive to extracellular calcium concentration and temperature. During rapid MNTB axon stimulation, postsynaptic currents in MOC neurons summated but with concurrent depression, resulting in smaller, sustained currents, suggesting tonic inhibition of MOC neurons during rapid circuit activity. Low levels of baseline MNTB activity did not significantly reduce responses to subsequent rapid activity that mimics sound stimulation, indicating that, in vivo, MNTB inhibition of MOC neurons persists despite tonic synaptic depression. Key points Inhibitory synapses from the medial nucleus of the trapezoid body (MNTB) onto medial olivocochlear (MOC) neurons exhibit short‐term plasticity that is sensitive to calcium and temperature, with enhanced synaptic depression occurring at higher calcium concentrations and at room temperature. High rates of background synaptic activity that mimic the upper limits of spontaneous MNTB activity cause tonic synaptic depression of MNTB–MOC synapses that limits further synaptic inhibition. High rates of activity at MNTB–MOC synapses cause synaptic summation with concurrent depression to yield a response with an initial large amplitude that decays to a tonic inhibition.
Collapse
Affiliation(s)
- Lester Torres Cadenas
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD, 20892, USA
| | - Hui Cheng
- Bioinformatics and Biostatistics Collaboration Core, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD, 20892, USA
| | - Catherine J C Weisz
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD, 20892, USA
| |
Collapse
|
10
|
Elgoyhen AB. The α9α10 nicotinic acetylcholine receptor: a compelling drug target for hearing loss? Expert Opin Ther Targets 2022; 26:291-302. [PMID: 35225139 PMCID: PMC9007918 DOI: 10.1080/14728222.2022.2047931] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Hearing loss is a major health problem, impacting education, communication, interpersonal relationships, and mental health. Drugs that prevent or restore hearing are lacking and hence novel drug targets are sought. There is the possibility of targeting the α9α10 nicotinic acetylcholine receptor (nAChR) in the prevention of noise-induced, hidden hearing loss and presbycusis. This receptor mediates synaptic transmission between medial olivocochlear efferent fibers and cochlear outer hair cells. This target is key since enhanced olivocochlear activity prevents noise-induced hearing loss and delays presbycusis. AREAS COVERED The work examines the α9α10 nicotinic acetylcholine receptor (nAChR), its role in noise-induced, hidden hearing loss and presbycusis and the possibility of targeting. Data has been searched in Pubmed, the World Report on Hearing from the World Health Organization and the Global Burden of Disease Study 2019. EXPERT OPINION The design of positive allosteric modulators of α9α10 nAChRs is proposed because of the advantage of reinforcing the medial olivocochlear (MOC)-hair cell endogenous neurotransmission without directly stimulating the target receptors, therefore avoiding receptor desensitization and reduced efficacy. The time is right for the discovery and development of α9α10 nAChRs targeting agents and high throughput screening assays will support this.
Collapse
Affiliation(s)
- Ana Belén Elgoyhen
- Laboratorio de Fisiología y Genética de la Audición, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
11
|
Lipovsek M, Marcovich I, Elgoyhen AB. The Hair Cell α9α10 Nicotinic Acetylcholine Receptor: Odd Cousin in an Old Family. Front Cell Neurosci 2021; 15:785265. [PMID: 34867208 PMCID: PMC8634148 DOI: 10.3389/fncel.2021.785265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/25/2021] [Indexed: 11/13/2022] Open
Abstract
Nicotinic acetylcholine receptors (nAChRs) are a subfamily of pentameric ligand-gated ion channels with members identified in most eumetazoan clades. In vertebrates, they are divided into three subgroups, according to their main tissue of expression: neuronal, muscle and hair cell nAChRs. Each receptor subtype is composed of different subunits, encoded by paralogous genes. The latest to be identified are the α9 and α10 subunits, expressed in the mechanosensory hair cells of the inner ear and the lateral line, where they mediate efferent modulation. α9α10 nAChRs are the most divergent amongst all nicotinic receptors, showing marked differences in their degree of sequence conservation, their expression pattern, their subunit co-assembly rules and, most importantly, their functional properties. Here, we review recent advances in the understanding of the structure and evolution of nAChRs. We discuss the functional consequences of sequence divergence and conservation, with special emphasis on the hair cell α9α10 receptor, a seemingly distant cousin of neuronal and muscle nicotinic receptors. Finally, we highlight potential links between the evolution of the octavolateral system and the extreme divergence of vertebrate α9α10 receptors.
Collapse
Affiliation(s)
- Marcela Lipovsek
- Ear Institute, Faculty of Brain Sciences, University College London, London, United Kingdom
| | - Irina Marcovich
- Departments of Otolaryngology & Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Ana Belén Elgoyhen
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
12
|
Moglie MJ, Wengier DL, Elgoyhen AB, Goutman JD. Synaptic Contributions to Cochlear Outer Hair Cell Ca 2+ Dynamics. J Neurosci 2021; 41:6812-6821. [PMID: 34253627 PMCID: PMC8360681 DOI: 10.1523/jneurosci.3008-20.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 05/30/2021] [Accepted: 07/05/2021] [Indexed: 11/21/2022] Open
Abstract
For normal cochlear function, outer hair cells (OHCs) require a precise control of intracellular Ca2+ levels. In the absence of regulatory elements such as proteinaceous buffers or extrusion pumps, OHCs degenerate, leading to profound hearing impairment. Influx of Ca2+ occurs both at the stereocilia tips and the basolateral membrane. In this latter compartment, two different origins for Ca2+ influx have been poorly explored: voltage-gated L-type Ca2+ channels (VGCCs) at synapses with Type II afferent neurons, and α9α10 cholinergic nicotinic receptors at synapses with medio-olivochlear complex (MOC) neurons. Using functional imaging in mouse OHCs, we dissected Ca2+ influx individually through each of these sources, either by applying step depolarizations to activate VGCC, or stimulating MOC axons. Ca2+ ions originated in MOC synapses, but not by VGCC activation, was confined by Ca2+-ATPases most likely present in nearby synaptic cisterns. Although Ca2+ currents in OHCs are small, VGCC Ca2+ signals were comparable in size to those elicited by α9α10 receptors, and were potentiated by ryanodine receptors (RyRs). In contrast, no evidence of potentiation by RyRs was found for MOC Ca2+ signals over a wide range of presynaptic stimulation strengths. Our study shows that despite the fact that these two Ca2+ entry sites are closely positioned, they differ in their regulation by intracellular cisterns and/or organelles, suggesting the existence of well-tuned mechanisms to separate the two different OHC synaptic functions.SIGNIFICANCE STATEMENT Outer hair cells (OHCs) are sensory cells in the inner ear operating under very special constraints. Acoustic stimulation leads to fast changes both in membrane potential and in the intracellular concentration of metabolites such as Ca2+ Tight mechanisms for Ca2+ control in OHCs have been reported. Interestingly, Ca2+ is crucial for two important synaptic processes: inhibition by efferent cholinergic neurons, and glutamate release onto Type II afferent fibers. In the current study we functionally imaged Ca2+ at these two different synapses, showing close positioning within the basolateral compartment of OHCs. In addition, we show differential regulation of these two Ca2+ sources by synaptic cisterns and/or organelles, which could result crucial for functional segregation during normal hearing.
Collapse
Affiliation(s)
- Marcelo J Moglie
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI) (Consejo Nacional de Investigaciones Científicas y Tecnológicas), Ciudad Autónoma de Buenos Aires 1428, Argentina
| | - Diego L Wengier
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI) (Consejo Nacional de Investigaciones Científicas y Tecnológicas), Ciudad Autónoma de Buenos Aires 1428, Argentina
| | - A Belén Elgoyhen
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI) (Consejo Nacional de Investigaciones Científicas y Tecnológicas), Ciudad Autónoma de Buenos Aires 1428, Argentina
| | - Juan D Goutman
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI) (Consejo Nacional de Investigaciones Científicas y Tecnológicas), Ciudad Autónoma de Buenos Aires 1428, Argentina
| |
Collapse
|
13
|
Cullen KE, Wei RH. Differences in the Structure and Function of the Vestibular Efferent System Among Vertebrates. Front Neurosci 2021; 15:684800. [PMID: 34248486 PMCID: PMC8260987 DOI: 10.3389/fnins.2021.684800] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/13/2021] [Indexed: 11/13/2022] Open
Abstract
The role of the mammalian vestibular efferent system in everyday life has been a long-standing mystery. In contrast to what has been reported in lower vertebrate classes, the mammalian vestibular efferent system does not appear to relay inputs from other sensory modalities to the vestibular periphery. Furthermore, to date, the available evidence indicates that the mammalian vestibular efferent system does not relay motor-related signals to the vestibular periphery to modulate sensory coding of the voluntary self-motion generated during natural behaviors. Indeed, our recent neurophysiological studies have provided insight into how the peripheral vestibular system transmits head movement-related information to the brain in a context independent manner. The integration of vestibular and extra-vestibular information instead only occurs at next stage of the mammalian vestibular system, at the level of the vestibular nuclei. The question thus arises: what is the physiological role of the vestibular efferent system in mammals? We suggest that the mammalian vestibular efferent system does not play a significant role in short-term modulation of afferent coding, but instead plays a vital role over a longer time course, for example in calibrating and protecting the functional efficacy of vestibular circuits during development and aging in a role analogous the auditory efferent system.
Collapse
Affiliation(s)
- Kathleen E. Cullen
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
- Department of Otolaryngology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, United States
| | - Rui-Han Wei
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
14
|
Wang Y, Sanghvi M, Gribizis A, Zhang Y, Song L, Morley B, Barson DG, Santos-Sacchi J, Navaratnam D, Crair M. Efferent feedback controls bilateral auditory spontaneous activity. Nat Commun 2021; 12:2449. [PMID: 33907194 PMCID: PMC8079389 DOI: 10.1038/s41467-021-22796-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 03/24/2021] [Indexed: 12/21/2022] Open
Abstract
In the developing auditory system, spontaneous activity generated in the cochleae propagates into the central nervous system to promote circuit formation. The effects of peripheral firing patterns on spontaneous activity in the central auditory system are not well understood. Here, we describe wide-spread bilateral coupling of spontaneous activity that coincides with the period of transient efferent modulation of inner hair cells from the brainstem medial olivocochlear system. Knocking out α9/α10 nicotinic acetylcholine receptors, a requisite part of the efferent pathway, profoundly reduces bilateral correlations. Pharmacological and chemogenetic experiments confirm that the efferent system is necessary for normal bilateral coupling. Moreover, auditory sensitivity at hearing onset is reduced in the absence of pre-hearing efferent modulation. Together, these results demonstrate how afferent and efferent pathways collectively shape spontaneous activity patterns and reveal the important role of efferents in coordinating bilateral spontaneous activity and the emergence of functional responses during the prehearing period.
Collapse
Affiliation(s)
- Yixiang Wang
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Maya Sanghvi
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Alexandra Gribizis
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Max Planck Florida Institute for Neuroscience, One Max Planck Way, Jupiter, FL, USA
| | - Yueyi Zhang
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Lei Song
- Department of Surgery (Otolaryngology), Yale University School of Medicine, New Haven, CT, USA
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Barbara Morley
- Center for Sensory Neuroscience, Boys Town National Research Hospital, Omaha, NE, USA
| | - Daniel G Barson
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Joseph Santos-Sacchi
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Surgery (Otolaryngology), Yale University School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Dhasakumar Navaratnam
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Surgery (Otolaryngology), Yale University School of Medicine, New Haven, CT, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Michael Crair
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA.
- Kavli Institute for Neuroscience, Yale University, New Haven, CT, USA.
| |
Collapse
|
15
|
Moglie MJ, Marcovich I, Corradi J, Carpaneto Freixas AE, Gallino S, Plazas PV, Bouzat C, Lipovsek M, Elgoyhen AB. Loss of Choline Agonism in the Inner Ear Hair Cell Nicotinic Acetylcholine Receptor Linked to the α10 Subunit. Front Mol Neurosci 2021; 14:639720. [PMID: 33613194 PMCID: PMC7892445 DOI: 10.3389/fnmol.2021.639720] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/11/2021] [Indexed: 11/13/2022] Open
Abstract
The α9α10 nicotinic acetylcholine receptor (nAChR) plays a fundamental role in inner ear physiology. It mediates synaptic transmission between efferent olivocochlear fibers that descend from the brainstem and hair cells of the auditory sensory epithelium. The α9 and α10 subunits have undergone a distinct evolutionary history within the family of nAChRs. Predominantly in mammalian vertebrates, the α9α10 receptor has accumulated changes at the protein level that may ultimately relate to the evolutionary history of the mammalian hearing organ. In the present work, we investigated the responses of α9α10 nAChRs to choline, the metabolite of acetylcholine degradation at the synaptic cleft. Whereas choline is a full agonist of chicken α9α10 receptors it is a partial agonist of the rat receptor. Making use of the expression of α9α10 heterologous receptors, encompassing wild-type, heteromeric, homomeric, mutant, chimeric, and hybrid receptors, and in silico molecular docking, we establish that the mammalian (rat) α10 nAChR subunit underscores the reduced efficacy of choline. Moreover, we show that whereas the complementary face of the α10 subunit does not play an important role in the activation of the receptor by ACh, it is strictly required for choline responses. Thus, we propose that the evolutionary changes acquired in the mammalian α9α10 nAChR resulted in the loss of choline acting as a full agonist at the efferent synapse, without affecting the triggering of ACh responses. This may have accompanied the fine-tuning of hair cell post-synaptic responses to the high-frequency activity of efferent medial olivocochlear fibers that modulate the cochlear amplifier.
Collapse
Affiliation(s)
- Marcelo J. Moglie
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Irina Marcovich
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Jeremías Corradi
- Departamento de Biología, Bioquímica y Farmacia, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Universidad Nacional del Sur y Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Agustín E. Carpaneto Freixas
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Sofía Gallino
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Paola V. Plazas
- Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Cecilia Bouzat
- Departamento de Biología, Bioquímica y Farmacia, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Universidad Nacional del Sur y Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Marcela Lipovsek
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Centre for Developmental Neurobiology, King’s College London, Institute of Psychiatry, Psychology, and Neuroscience, Guy’s Campus, London, United Kingdom
| | - Ana Belén Elgoyhen
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
16
|
Vyas P, Wood MB, Zhang Y, Goldring AC, Chakir FZ, Fuchs PA, Hiel H. Characterization of HA-tagged α9 and α10 nAChRs in the mouse cochlea. Sci Rep 2020; 10:21814. [PMID: 33311584 PMCID: PMC7733449 DOI: 10.1038/s41598-020-78380-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 11/20/2020] [Indexed: 01/02/2023] Open
Abstract
Neurons of the medial olivary complex inhibit cochlear hair cells through the activation of α9α10-containing nicotinic acetylcholine receptors (nAChRs). Efforts to study the localization of these proteins have been hampered by the absence of reliable antibodies. To overcome this obstacle, CRISPR-Cas9 gene editing was used to generate mice in which a hemagglutinin tag (HA) was attached to the C-terminus of either α9 or α10 proteins. Immunodetection of the HA tag on either subunit in the organ of Corti of adult mice revealed immunopuncta clustered at the synaptic pole of outer hair cells. These puncta were juxtaposed to immunolabeled presynaptic efferent terminals. HA immunopuncta also occurred in inner hair cells of pre-hearing (P7) but not in adult mice. These immunolabeling patterns were similar for both homozygous and heterozygous mice. All HA-tagged genotypes had auditory brainstem responses not significantly different from those of wild type littermates. The activation of efferent neurons in heterozygous mice evoked biphasic postsynaptic currents not significantly different from those of wild type hair cells. However, efferent synaptic responses were significantly smaller and less frequent in the homozygous mice. We show that HA-tagged nAChRs introduced in the mouse by a CRISPR knock-in are regulated and expressed like the native protein, and in the heterozygous condition mediate normal synaptic function. The animals thus generated have clear advantages for localization studies.
Collapse
Affiliation(s)
- Pankhuri Vyas
- The Center for Hearing and Balance, Department of Otolaryngology Head and Neck Surgery, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 818, Baltimore, MD, 21205, USA
| | - Megan Beers Wood
- The Center for Hearing and Balance, Department of Otolaryngology Head and Neck Surgery, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 818, Baltimore, MD, 21205, USA
| | - Yuanyuan Zhang
- The Center for Hearing and Balance, Department of Otolaryngology Head and Neck Surgery, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 818, Baltimore, MD, 21205, USA.,Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Adam C Goldring
- The Center for Hearing and Balance, Department of Otolaryngology Head and Neck Surgery, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 818, Baltimore, MD, 21205, USA.,Sutter Instrument Company, 1 Digital Drive, Novato, CA, 94949, USA
| | - Fatima-Zahra Chakir
- The Center for Hearing and Balance, Department of Otolaryngology Head and Neck Surgery, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 818, Baltimore, MD, 21205, USA
| | - Paul Albert Fuchs
- The Center for Hearing and Balance, Department of Otolaryngology Head and Neck Surgery, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 818, Baltimore, MD, 21205, USA
| | - Hakim Hiel
- The Center for Hearing and Balance, Department of Otolaryngology Head and Neck Surgery, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 818, Baltimore, MD, 21205, USA.
| |
Collapse
|
17
|
|
18
|
Unraveling the Molecular Players at the Cholinergic Efferent Synapse of the Zebrafish Lateral Line. J Neurosci 2020; 41:47-60. [PMID: 33203744 DOI: 10.1523/jneurosci.1772-20.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/25/2020] [Accepted: 10/30/2020] [Indexed: 12/26/2022] Open
Abstract
The lateral line (LL) is a sensory system that allows fish and amphibians to detect water currents. LL responsiveness is modulated by efferent neurons that aid in distinguishing between external and self-generated stimuli, maintaining sensitivity to relevant cues. One component of the efferent system is cholinergic, the activation of which inhibits afferent activity. LL hair cells (HCs) share structural, functional, and molecular similarities with those of the cochlea, making them a popular model for studying human hearing and balance disorders. Because of these commonalities, one could propose that the receptor at the LL efferent synapse is a α9α10 nicotinic acetylcholine receptor (nAChR). However, the identities of the molecular players underlying ACh-mediated inhibition in the LL remain unknown. Surprisingly, through the analysis of single-cell expression studies and in situ hybridization, we describe that α9, but not the α10, subunits are enriched in zebrafish HCs. Moreover, the heterologous expression of zebrafish α9 subunits indicates that homomeric receptors are functional and exhibit robust ACh-gated currents blocked by α-bungarotoxin and strychnine. In addition, in vivo Ca2+ imaging on mechanically stimulated zebrafish LL HCs show that ACh elicits a decrease in evoked Ca2+ signals, regardless of HC polarity. This effect is blocked by both α-bungarotoxin and apamin, indicating coupling of ACh-mediated effects to small-conductance Ca2+-activated potassium (SKs) channels. Our results indicate that an α9-containing (α9*) nAChR operates at the zebrafish LL efferent synapse. Moreover, the activation of α9* nAChRs most likely leads to LL HC hyperpolarization served by SK channels.SIGNIFICANCE STATEMENT The fish lateral line (LL) mechanosensory system shares structural, functional, and molecular similarities with those of the mammalian cochlea. Thus, it has become an accessible model for studying human hearing and balance disorders. However, the molecular players serving efferent control of LL hair cell (HC) activity have not been identified. Here we demonstrate that, different from the hearing organ of vertebrate species, a nicotinic acetylcholine receptor composed only of α9 subunits operates at the LL efferent synapse. Activation of α9-containing receptors leads to LL HC hyperpolarization because of the opening of small-conductance Ca2+-activated potassium channels. These results will further aid in the interpretation of data obtained from LL HCs as a model for cochlear HCs.
Collapse
|
19
|
Zhang Y, Glowatzki E, Roux I, Fuchs PA. Nicotine evoked efferent transmitter release onto immature cochlear inner hair cells. J Neurophysiol 2020; 124:1377-1387. [PMID: 32845208 DOI: 10.1152/jn.00097.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Olivocochlear neurons make temporary cholinergic synapses on inner hair cells of the rodent cochlea in the first 2 to 3 wk after birth. Repetitive stimulation of these efferent neurons causes facilitation of evoked release and increased spontaneous release that continues for seconds to minutes. Presynaptic nicotinic acetylcholine receptors (nAChRs) are known to modulate neurotransmitter release from brain neurons. The present study explores the hypothesis that presynaptic nAChRs help to increase spontaneous release from efferent terminals on cochlear hair cells. Direct application of nicotine (which does not activate the hair cells' α9α10-containing nAChRs) produces sustained efferent transmitter release, implicating presynaptic nAChRs in this response. The effect of nicotine was reduced by application of ryanodine that reduces release of calcium from intraterminal stores.NEW & NOTEWORTHY Sensory organs exhibit spontaneous activity before the onset of response to external stimuli. Such activity in the cochlea is subject to modulation by cholinergic efferent neurons that directly inhibit sensory hair cells (inner hair cells). Those efferent neurons are themselves subject to various modulatory mechanisms. One such mechanism is positive feedback by released acetylcholine onto presynaptic nicotinic acetylcholine receptors causing further release of acetylcholine.
Collapse
Affiliation(s)
- Y Zhang
- The Center for Hearing and Balance, Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - E Glowatzki
- The Center for Hearing and Balance, Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - I Roux
- The Center for Hearing and Balance, Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Otolaryngology Branch, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, Porter Neuroscience Research Center, Bethesda, Maryland
| | - P A Fuchs
- The Center for Hearing and Balance, Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
20
|
Yu Z, McIntosh JM, Sadeghi SG, Glowatzki E. Efferent synaptic transmission at the vestibular type II hair cell synapse. J Neurophysiol 2020; 124:360-374. [PMID: 32609559 DOI: 10.1152/jn.00143.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In the vestibular peripheral organs, type I and type II hair cells (HCs) transmit incoming signals via glutamatergic quantal transmission onto afferent nerve fibers. Additionally, type I HCs transmit via "non-quantal" transmission to calyx afferent fibers, by accumulation of glutamate and potassium in the synaptic cleft. Vestibular efferent inputs originating in the brainstem contact type II HCs and vestibular afferents. Here, synaptic inputs to type II HCs were characterized by using electrical and optogenetic stimulation of efferent fibers combined with in vitro whole cell patch-clamp recording from type II HCs in the rodent vestibular crista. Properties of efferent synaptic currents in type II HCs were similar to those found in cochlear HCs and mediated by activation of α9-containing nicotinic acetylcholine receptors (nAChRs) and small-conductance calcium-activated potassium (SK) channels. While efferents showed a low probability of release at low frequencies of stimulation, repetitive stimulation resulted in facilitation and increased probability of release. Notably, the membrane potential of type II HCs during optogenetic stimulation of efferents showed a strong hyperpolarization in response to single pulses and was further enhanced by repetitive stimulation. Such efferent-mediated inhibition of type II HCs can provide a mechanism to adjust the contribution of signals from type I and type II HCs to vestibular nerve fibers, with a shift of the response to be more like that of calyx-only afferents with faster non-quantal responses.NEW & NOTEWORTHY Type II vestibular hair cells (HCs) receive inputs from efferent neurons in the brain stem. We used in vitro optogenetic and electrical stimulation of vestibular efferent fibers to study their synaptic inputs to type II HCs. Stimulation of efferents inhibited type II HCs, similar to efferent effects on cochlear HCs. We propose that efferent inputs adjust the contribution of signals from type I and II HCs to vestibular nerve fibers.
Collapse
Affiliation(s)
- Zhou Yu
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Otolaryngology-Head and Neck Surgery, The Center for Hearing and Balance, and The Center for Sensory Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Soroush G Sadeghi
- Department of Communicative Disorders and Sciences, and Center for Hearing and Deafness, State University of New York at Buffalo, Buffalo, New York.,Neuroscience Program, State University of New York at Buffalo, Buffalo, New York
| | - Elisabeth Glowatzki
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Otolaryngology-Head and Neck Surgery, The Center for Hearing and Balance, and The Center for Sensory Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
21
|
Functional Postnatal Maturation of the Medial Olivocochlear Efferent-Outer Hair Cell Synapse. J Neurosci 2020; 40:4842-4857. [PMID: 32430293 DOI: 10.1523/jneurosci.2409-19.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 04/19/2020] [Accepted: 05/11/2020] [Indexed: 01/07/2023] Open
Abstract
The organ of Corti, the auditory mammalian sensory epithelium, contains two types of mechanotransducer cells, inner hair cells (IHCs) and outer hair cells (OHCs). IHCs are involved in conveying acoustic stimuli to the CNS, while OHCs are implicated in the fine tuning and amplification of sounds. OHCs are innervated by medial olivocochlear (MOC) cholinergic efferent fibers. The functional characteristics of the MOC-OHC synapse during maturation were assessed by electrophysiological and pharmacological methods in mouse organs of Corti at postnatal day 11 (P11)-P13, hearing onset in altricial rodents, and at P20-P22 when the OHCs are morphologically and functionally mature. Synaptic currents were recorded in whole-cell voltage-clamped OHCs while electrically stimulating the MOC fibers. A progressive increase in the number of functional MOC-OHC synapses, as well as in their strength and efficacy, was observed between P11-13 and P20-22. At hearing onset, the MOC-OHC synapse presented facilitation during MOC fibers high-frequency stimulation that disappeared at mature stages. In addition, important changes were found in the VGCC that are coupled to transmitter release. Ca2+ flowing in through L-type VGCCs contribute to trigger ACh release together with P/Q- and R-type VGCCs at P11-P13, but not at P20-P22. Interestingly, N-type VGCCs were found to be involved in this process at P20-P22, but not at hearing onset. Moreover, the degree of compartmentalization of calcium channels with respect to BK channels and presynaptic release components significantly increased from P11-P13 to P20-P22. These results suggest that the MOC-OHC synapse is immature at the onset of hearing.SIGNIFICANCE STATEMENT The functional expression of both VGCCs and BK channels, as well as their localization with respect to the presynaptic components involved in transmitter release, are key elements in determining synaptic efficacy. In this work, we show dynamic changes in the expression of VGCCs and Ca2+-dependent BK K+ channels coupled to ACh release at the MOC-OHC synapse and their shift in compartmentalization during postnatal maturation. These processes most likely set the short-term plasticity pattern and reliability of the MOC-OHC synapse on high-frequency activity.
Collapse
|
22
|
Preventing presbycusis in mice with enhanced medial olivocochlear feedback. Proc Natl Acad Sci U S A 2020; 117:11811-11819. [PMID: 32393641 DOI: 10.1073/pnas.2000760117] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
"Growing old" is the most common cause of hearing loss. Age-related hearing loss (ARHL) (presbycusis) first affects the ability to understand speech in background noise, even when auditory thresholds in quiet are normal. It has been suggested that cochlear denervation ("synaptopathy") is an early contributor to age-related auditory decline. In the present work, we characterized age-related cochlear synaptic degeneration and hair cell loss in mice with enhanced α9α10 cholinergic nicotinic receptors gating kinetics ("gain of function" nAChRs). These mediate inhibitory olivocochlear feedback through the activation of associated calcium-gated potassium channels. Cochlear function was assessed via distortion product otoacoustic emissions and auditory brainstem responses. Cochlear structure was characterized in immunolabeled organ of Corti whole mounts using confocal microscopy to quantify hair cells, auditory neurons, presynaptic ribbons, and postsynaptic glutamate receptors. Aged wild-type mice had elevated acoustic thresholds and synaptic loss. Afferent synapses were lost from inner hair cells throughout the aged cochlea, together with some loss of outer hair cells. In contrast, cochlear structure and function were preserved in aged mice with gain-of-function nAChRs that provide enhanced olivocochlear inhibition, suggesting that efferent feedback is important for long-term maintenance of inner ear function. Our work provides evidence that olivocochlear-mediated resistance to presbycusis-ARHL occurs via the α9α10 nAChR complexes on outer hair cells. Thus, enhancement of the medial olivocochlear system could be a viable strategy to prevent age-related hearing loss.
Collapse
|
23
|
Effects of Efferent Activity on Hair Bundle Mechanics. J Neurosci 2020; 40:2390-2402. [PMID: 32086256 DOI: 10.1523/jneurosci.1312-19.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 01/31/2020] [Accepted: 02/09/2020] [Indexed: 11/21/2022] Open
Abstract
Hair cells in both the auditory and vestibular systems receive efferent innervation. A number of prior studies have indicated that efferent regulation serves to diminish the overall sensitivity of the auditory system. The efferent pathway is believed to affect the sensitivity and frequency selectivity of the hair cell by modulating its membrane potential. However, its effect on the mechanical response of the hair cell has not been established. We explored how stimulation of the efferent neurons affects the mechanical responsiveness of an individual hair bundle. We tested this effect on in vitro preparations of hair cells in the sacculi of American bullfrogs of both genders. Efferent stimulation routinely resulted in an immediate increase of the frequency of hair bundle spontaneous oscillations for the duration of the stimulus. Enlarging the stimulus amplitude and pulse length, or conversely, decreasing the interpulse interval led to oscillation suppression. Additionally, we tested the effects of efference on the hair bundle response to mechanical stimulation. The receptive field maps of hair cells undergoing efferent actuation demonstrated an overall desensitization with respect to those of unstimulated cells.SIGNIFICANCE STATEMENT The efferent system is an important aide for the performance of the auditory system. It has been seen to contribute to sound detection and localization, ototoxicity prevention, and speech comprehension. Although measurements have demonstrated that efference suppresses basilar membrane movement, there is still much unknown about how efferent activity affects hearing mechanics. Here, we explore the mechanical basis for the efferent system's capabilities at the level of the hair bundle. We present optical recordings, receptive field maps, and sensitivity curves that show a hair bundle is desensitized by efferent stimulation. This supports the hypothesis that efferent regulation may be a biological control parameter for tuning the hair bundle's mechanical sensitivity.
Collapse
|
24
|
Torres Cadenas L, Fischl MJ, Weisz CJC. Synaptic Inhibition of Medial Olivocochlear Efferent Neurons by Neurons of the Medial Nucleus of the Trapezoid Body. J Neurosci 2020; 40:509-525. [PMID: 31719165 PMCID: PMC6961997 DOI: 10.1523/jneurosci.1288-19.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 10/29/2019] [Accepted: 11/04/2019] [Indexed: 02/08/2023] Open
Abstract
Medial olivocochlear (MOC) efferent neurons in the brainstem comprise the final stage of descending control of the mammalian peripheral auditory system through axon projections to the cochlea. MOC activity adjusts cochlear gain and frequency tuning, and protects the ear from acoustic trauma. The neuronal pathways that activate and modulate the MOC somata in the brainstem to drive these cochlear effects are poorly understood. Evidence suggests that MOC neurons are primarily excited by sound stimuli in a three-neuron activation loop from the auditory nerve via an intermediate neuron in the cochlear nucleus. Anatomical studies suggest that MOC neurons receive diverse synaptic inputs, but the functional effect of additional synaptic influences on MOC neuron responses is unknown. Here we use patch-clamp electrophysiological recordings from identified MOC neurons in brainstem slices from mice of either sex to demonstrate that in addition to excitatory glutamatergic synapses, MOC neurons receive inhibitory GABAergic and glycinergic synaptic inputs. These synapses are activated by electrical stimulation of axons near the medial nucleus of the trapezoid body (MNTB). Focal glutamate uncaging confirms MNTB neurons as a source of inhibitory synapses onto MOC neurons. MNTB neurons inhibit MOC action potentials, but this effect depresses with repeat activation. This work identifies a new pathway of connectivity between brainstem auditory neurons and indicates that MOC neurons are both excited and inhibited by sound stimuli received at the same ear. The pathway depression suggests that the effect of MNTB inhibition of MOC neurons diminishes over the course of a sustained sound.SIGNIFICANCE STATEMENT Medial olivocochlear (MOC) neurons are the final stage of descending control of the mammalian auditory system and exert influence on cochlear mechanics to modulate perception of acoustic stimuli. The brainstem pathways that drive MOC function are poorly understood. Here we show for the first time that MOC neurons are inhibited by neurons of the MNTB, which may suppress the effects of MOC activity on the cochlea.
Collapse
Affiliation(s)
- Lester Torres Cadenas
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland 20892
| | - Matthew J Fischl
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland 20892
| | - Catherine J C Weisz
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland 20892
| |
Collapse
|
25
|
Efferent Inputs Are Required for Normal Function of Vestibular Nerve Afferents. J Neurosci 2019; 39:6922-6935. [PMID: 31285300 DOI: 10.1523/jneurosci.0237-19.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 06/28/2019] [Accepted: 07/01/2019] [Indexed: 01/09/2023] Open
Abstract
A group of vestibular afferent nerve fibers with irregular-firing resting discharges are thought to play a prominent role in responses to fast head movements and vestibular plasticity. We show that, in C57BL/6 mice (either sex, 4-5 weeks old), normal activity in the efferent vestibular pathway is required for function of these irregular afferents. Thermal inhibition of efferent fibers results in a profound inhibition of irregular afferents' resting discharges, rendering them inadequate for signaling head movements. In this way, efferent inputs adjust the contribution of the peripheral irregular afferent pathway that plays a critical role in peripheral vestibular signaling and plasticity.SIGNIFICANCE STATEMENT Vestibular end organs in the inner ear receive efferent inputs from the brainstem. Previously, electrical stimulation of efferents was linked to an increase in resting discharges of afferents and a decrease in their sensitivities. Here, we show that localized thermal inhibition of unmyelinated efferents results in a significant decrease in the activity of afferent nerve fibers, particularly those with irregular resting discharges implicated in responses to fast head movements and vestibular compensation. Thus, by upregulating and downregulating of afferent firing, particularly irregular afferents, efferents adjust neural activity sensitive to rapid head movements. These findings support the notion that peripheral vestibular end organs are not passive transducers of head movements and their sensory signal transmission is modulated by efferent inputs.
Collapse
|
26
|
Abstract
Cholinergic efferent neurons originating in the brainstem innervate the acoustico-lateralis organs (inner ear, lateral line) of vertebrates. These release acetylcholine (ACh) to inhibit hair cells through activation of calcium-dependent potassium channels. In the mammalian cochlea, ACh shunts and suppresses outer hair cell (OHC) electromotility, reducing the essential amplification of basilar membrane motion. Consequently, medial olivocochlear neurons that inhibit OHCs reduce the sensitivity and frequency selectivity of afferent neurons driven by cochlear vibration of inner hair cells (IHCs). The cholinergic synapse on hair cells involves an unusual ionotropic ACh receptor, and a near-membrane postsynaptic cistern. Lateral olivocochlear (LOC) neurons modulate type I afferents by still-to-be-defined synaptic mechanisms. Olivocochlear neurons can be activated by a reflex arc that includes the auditory nerve and projections from the cochlear nucleus. They are also subject to modulation by higher-order central auditory interneurons. Through its actions on cochlear hair cells, afferent neurons, and higher centers, the olivocochlear system protects against age-related and noise-induced hearing loss, improves signal coding in noise under certain conditions, modulates selective attention to sensory stimuli, and influences sound localization.
Collapse
Affiliation(s)
- Paul Albert Fuchs
- The Center for Hearing and Balance, Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205-2195
| | - Amanda M Lauer
- The Center for Hearing and Balance, Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205-2195
| |
Collapse
|
27
|
Kearney G, Zorrilla de San Martín J, Vattino LG, Elgoyhen AB, Wedemeyer C, Katz E. Developmental Synaptic Changes at the Transient Olivocochlear-Inner Hair Cell Synapse. J Neurosci 2019; 39:3360-3375. [PMID: 30755493 PMCID: PMC6495135 DOI: 10.1523/jneurosci.2746-18.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/04/2019] [Accepted: 01/30/2019] [Indexed: 12/18/2022] Open
Abstract
In the mature mammalian cochlea, inner hair cells (IHCs) are mainly innervated by afferent fibers that convey sound information to the CNS. During postnatal development, however, medial olivocochlear (MOC) efferent fibers transiently innervate the IHCs. The MOC-IHC synapse, functional from postnatal day 0 (P0) to hearing onset (P12), undergoes dramatic changes in the sensitivity to acetylcholine (ACh) and in the expression of key postsynaptic proteins. To evaluate whether there are associated changes in the properties of ACh release during this period, we used a cochlear preparation from mice of either sex at P4, P6-P7, and P9-P11 and monitored transmitter release from MOC terminals in voltage-clamped IHCs in the whole-cell configuration. The quantum content increased 5.6× from P4 to P9-P11 due to increases in the size and replenishment rate of the readily releasable pool of synaptic vesicles without changes in their probability of release or quantum size. This strengthening in transmission was accompanied by changes in short-term plasticity properties, which switched from facilitation at P4 to depression at P9-P11. We have previously shown that at P9-P11, ACh release is supported by P/Q- and N-type voltage-gated calcium channels (VGCCs) and negatively regulated by BK potassium channels activated by Ca2+ influx through L-type VGCCs. We now show that at P4 and P6-P7, release is mediated by P/Q-, R- and L-type VGCCs. Interestingly, L-type VGCCs have a dual role: they both support release and fuel BK channels, suggesting that at immature stages presynaptic proteins involved in release are less compartmentalized.SIGNIFICANCE STATEMENT During postnatal development before the onset of hearing, cochlear inner hair cells (IHCs) present spontaneous Ca2+ action potentials that release glutamate at the first auditory synapse in the absence of sound stimulation. The IHC Ca2+ action potential frequency pattern, which is crucial for the correct establishment and function of the auditory system, is regulated by the efferent medial olivocochlear (MOC) system that transiently innervates IHCs during this period. We show here that developmental changes in synaptic strength and synaptic plasticity properties at the MOC-IHC synapse upon MOC fiber activation at different frequencies might be crucial for tightly shaping the pattern of afferent activity during this critical period.
Collapse
Affiliation(s)
- Graciela Kearney
- 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
| | - Javier Zorrilla de San Martín
- 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
| | - 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
| | - Ana Belén Elgoyhen
- 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
- Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, 1121 Ciudad Autónoma de Buenos Aires, Argentina, and
| | - 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
| | - 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. Ciudad Universitaria, C1428EGA Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
28
|
Vyas P, Wu JS, Jimenez A, Glowatzki E, Fuchs PA. Characterization of transgenic mouse lines for labeling type I and type II afferent neurons in the cochlea. Sci Rep 2019; 9:5549. [PMID: 30944354 PMCID: PMC6447598 DOI: 10.1038/s41598-019-41770-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 03/14/2019] [Indexed: 11/09/2022] Open
Abstract
The cochlea is innervated by type I and type II afferent neurons. Type I afferents are myelinated, larger diameter neurons that send a single dendrite to contact a single inner hair cell, whereas unmyelinated type II afferents are fewer in number and receive input from many outer hair cells. This strikingly differentiated innervation pattern strongly suggests specialized functions. Those functions could be investigated with specific genetic markers that enable labeling and manipulating each afferent class without significantly affecting the other. Here three mouse models were characterized and tested for specific labeling of either type I or type II cochlear afferents. Nos1CreER mice showed selective labeling of type I afferent fibers, Slc6a4-GFP mice labeled type II fibers with a slight preference for the apical cochlea, and Drd2-Cre mice selectively labeled type II afferent neurons nearer the cochlear base. In conjunction with the Th2A-CreER and CGRPα-EGFP lines described previously for labeling type II fibers, the mouse lines reported here comprise a promising toolkit for genetic manipulations of type I and type II cochlear afferent fibers.
Collapse
Affiliation(s)
- Pankhuri Vyas
- The Center for Hearing and Balance, Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Jingjing Sherry Wu
- The Center for Hearing and Balance, Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Adrian Jimenez
- The Center for Hearing and Balance, Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Elisabeth Glowatzki
- The Center for Hearing and Balance, Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Paul Albert Fuchs
- The Center for Hearing and Balance, Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
29
|
Enhancement of the Medial Olivocochlear System Prevents Hidden Hearing Loss. J Neurosci 2018; 38:7440-7451. [PMID: 30030403 DOI: 10.1523/jneurosci.0363-18.2018] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 07/02/2018] [Accepted: 07/09/2018] [Indexed: 01/15/2023] Open
Abstract
Cochlear synaptopathy produced by exposure to noise levels that cause only transient auditory threshold elevations is a condition that affects many people and is believed to contribute to poor speech discrimination in noisy environments. These functional deficits in hearing, without changes in sensitivity, have been called hidden hearing loss (HHL). It has been proposed that activity of the medial olivocochlear (MOC) system can ameliorate acoustic trauma effects. Here we explore the role of the MOC system in HHL by comparing the performance of two different mouse models: an α9 nicotinic receptor subunit knock-out (KO; Chrna9 KO), which lacks cholinergic transmission between efferent neurons and hair cells; and a gain-of-function knock-in (KI; Chrna9L9'T KI) carrying an α9 point mutation that leads to enhanced cholinergic activity. Animals of either sex were exposed to sound pressure levels that in wild-type produced transient cochlear threshold shifts and a decrease in neural response amplitudes, together with the loss of ribbon synapses, which is indicative of cochlear synaptopathy. Moreover, a reduction in the number of efferent contacts to outer hair cells was observed. In Chrna9 KO ears, noise exposure produced permanent auditory threshold elevations together with cochlear synaptopathy. In contrast, the Chrna9L9'T KI was completely resistant to the same acoustic exposure protocol. These results show a positive correlation between the degree of HHL prevention and the level of cholinergic activity. Notably, enhancement of the MOC feedback promoted new afferent synapse formation, suggesting that it can trigger cellular and molecular mechanisms to protect and/or repair the inner ear sensory epithelium.SIGNIFICANCE STATEMENT Noise overexposure is a major cause of a variety of perceptual disabilities, including speech-in-noise difficulties, tinnitus, and hyperacusis. Here we show that exposure to noise levels that do not cause permanent threshold elevations or hair cell death can produce a loss of cochlear nerve synapses to inner hair cells as well as degeneration of medial olivocochlear (MOC) terminals contacting the outer hair cells. Enhancement of the MOC reflex can prevent both types of neuropathy, highlighting the potential use of drugs that increase α9α10 nicotinic cholinergic receptor activity as a pharmacotherapeutic strategy to avoid hidden hearing loss.
Collapse
|
30
|
Voltage-Gated Calcium Influx Modifies Cholinergic Inhibition of Inner Hair Cells in the Immature Rat Cochlea. J Neurosci 2018; 38:5677-5687. [PMID: 29789373 DOI: 10.1523/jneurosci.0230-18.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 05/14/2018] [Accepted: 05/16/2018] [Indexed: 11/21/2022] Open
Abstract
Until postnatal day (P) 12, inner hair cells of the rat cochlea are invested with both afferent and efferent synaptic connections. With the onset of hearing at P12, the efferent synapses disappear, and afferent (ribbon) synapses operate with greater efficiency. This change coincides with increased expression of voltage-gated potassium channels, the loss of calcium-dependent electrogenesis, and the onset of graded receptor potentials driven by sound. The transient efferent synapses include near-membrane postsynaptic cisterns thought to regulate calcium influx through the hair cell's α9-containing and α10-containing nicotinic acetylcholine receptors. This influx activates small-conductance Ca2+-activated K+ (SK) channels. Serial-section electron microscopy of inner hair cells from two 9-d-old (male) rat pups revealed many postsynaptic efferent cisterns and presynaptic afferent ribbons whose average minimal separation in five cells ranged from 1.1 to 1.7 μm. Efferent synaptic function was studied in rat pups (age, 7-9 d) of either sex. The duration of these SK channel-mediated IPSCs was increased by enhanced calcium influx through L-type voltage-gated channels, combined with ryanodine-sensitive release from internal stores-presumably the near-membrane postsynaptic cistern. These data support the possibility that inner hair cell calcium electrogenesis modulates the efficacy of efferent inhibition during the maturation of inner hair cell synapses.SIGNIFICANCE STATEMENT Strict calcium buffering is essential for cellular function. This problem is especially acute for compact hair cells where increasing cytoplasmic calcium promotes the opposing functions of closely adjoining afferent and efferent synapses. The near-membrane postsynaptic cistern at efferent synapses segregates synaptic calcium signals by acting as a dynamic calcium store. The hair cell serves as an informative model for synapses with postsynaptic cisterns (C synapses) found in central neurons.
Collapse
|
31
|
A Gain-of-Function Mutation in the α9 Nicotinic Acetylcholine Receptor Alters Medial Olivocochlear Efferent Short-Term Synaptic Plasticity. J Neurosci 2018; 38:3939-3954. [PMID: 29572431 DOI: 10.1523/jneurosci.2528-17.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 02/21/2018] [Accepted: 03/08/2018] [Indexed: 01/01/2023] Open
Abstract
Gain control of the auditory system operates at multiple levels. Cholinergic medial olivocochlear (MOC) fibers originate in the brainstem and make synaptic contacts at the base of the outer hair cells (OHCs), the final targets of several feedback loops from the periphery and higher-processing centers. Efferent activation inhibits OHC active amplification within the mammalian cochlea, through the activation of a calcium-permeable α9α10 ionotropic cholinergic nicotinic receptor (nAChR), functionally coupled to calcium activated SK2 potassium channels. Correct operation of this feedback requires careful matching of acoustic input with the strength of cochlear inhibition (Galambos, 1956; Wiederhold and Kiang, 1970; Gifford and Guinan, 1987), which is driven by the rate of MOC activity and short-term facilitation at the MOC-OHC synapse (Ballestero et al., 2011; Katz and Elgoyhen, 2014). The present work shows (in mice of either sex) that a mutation in the α9α10 nAChR with increased duration of channel gating (Taranda et al., 2009) greatly elongates hair cell-evoked IPSCs and Ca2+ signals. Interestingly, MOC-OHC synapses of L9'T mice presented reduced quantum content and increased presynaptic facilitation. These phenotypic changes lead to enhanced and sustained synaptic responses and OHC hyperpolarization upon high-frequency stimulation of MOC terminals. At the cochlear physiology level these changes were matched by a longer time course of efferent MOC suppression. This indicates that the properties of the MOC-OHC synapse directly determine the efficacy of the MOC feedback to the cochlea being a main player in the "gain control" of the auditory periphery.SIGNIFICANCE STATEMENT Plasticity can involve reciprocal signaling across chemical synapses. An opportunity to study this phenomenon occurs in the mammalian cochlea whose sensitivity is regulated by efferent olivocochlear neurons. These release acetylcholine to inhibit sensory hair cells. A point mutation in the hair cell's acetylcholine receptor that leads to increased gating of the receptor greatly elongates IPSCs. Interestingly, efferent terminals from mutant mice present a reduced resting release probability. However, upon high-frequency stimulation transmitter release facilitates strongly to produce stronger and far longer-lasting inhibition of cochlear function. Thus, central neuronal feedback on cochlear hair cells provides an opportunity to define plasticity mechanisms in cholinergic synapses other than the highly studied neuromuscular junction.
Collapse
|
32
|
Sinclair JL, Barnes-Davies M, Kopp-Scheinpflug C, Forsythe ID. Strain-specific differences in the development of neuronal excitability in the mouse ventral nucleus of the trapezoid body. Hear Res 2017; 354:28-37. [PMID: 28843833 DOI: 10.1016/j.heares.2017.08.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 08/10/2017] [Accepted: 08/15/2017] [Indexed: 02/06/2023]
Abstract
This investigation compared the development of neuronal excitability in the ventral nucleus of the trapezoid body (VNTB) between two strains of mice with differing progression rates for age-related hearing loss. In contrast to CBA/Ca (CBA) mice, the C57BL/6J (C57) strain are subject to hearing loss from a younger age and are more prone to damage from sound over-exposure. Higher firing rates in the medial olivocochlear system (MOC) are associated with protection from loud sounds and these cells are located in the VNTB. We postulated that reduced neuronal firing of the MOC in C57 mice could contribute to hearing loss in this strain by reducing efferent protection. Whole cell patch clamp was used to compare the electrical properties of VNTB neurons from the two strains initially in two age groups: before and after hearing onset at ∼ P9 and ∼P16, respectively. Prior to hearing onset VNTB neurons electrophysiological properties were identical in both strains, but started to diverge after hearing onset. One week after hearing onset VNTB neurons of C57 mice had larger amplitude action potentials but in contrast to CBA mice, their waveform failed to accelerate with increasing age, consistent with the faster inactivation of voltage-gated potassium currents in C57 VNTB neurons. The lower frequency action potential firing of C57 VNTB neurons at P16 was maintained to P28, indicating that this change was not a developmental delay. We conclude that C57 VNTB neurons fire at lower frequencies than in the CBA strain, supporting the hypothesis that reduced MOC firing could contribute to the greater hearing loss of the C57 strain.
Collapse
Affiliation(s)
- James L Sinclair
- MRC Toxicology Unit, University of Leicester, Leicester, LE1 9HN, UK; Department of Neuroscience, Psychology & Behaviour, University of Leicester, Leicester, LE1 9HN, UK
| | - Margaret Barnes-Davies
- Department of Neuroscience, Psychology & Behaviour, University of Leicester, Leicester, LE1 9HN, UK
| | | | - Ian D Forsythe
- MRC Toxicology Unit, University of Leicester, Leicester, LE1 9HN, UK; Department of Neuroscience, Psychology & Behaviour, University of Leicester, Leicester, LE1 9HN, UK.
| |
Collapse
|
33
|
Jackman SL, Regehr WG. The Mechanisms and Functions of Synaptic Facilitation. Neuron 2017; 94:447-464. [PMID: 28472650 DOI: 10.1016/j.neuron.2017.02.047] [Citation(s) in RCA: 241] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 02/23/2017] [Accepted: 02/28/2017] [Indexed: 12/22/2022]
Abstract
The ability of the brain to store and process information relies on changing the strength of connections between neurons. Synaptic facilitation is a form of short-term plasticity that enhances synaptic transmission for less than a second. Facilitation is a ubiquitous phenomenon thought to play critical roles in information transfer and neural processing. Yet our understanding of the function of facilitation remains largely theoretical. Here we review proposed roles for facilitation and discuss how recent progress in uncovering the underlying molecular mechanisms could enable experiments that elucidate how facilitation, and short-term plasticity in general, contributes to circuit function and animal behavior.
Collapse
Affiliation(s)
- Skyler L Jackman
- Department of Neurobiology, Harvard Medical School, Boston, MA 02118, USA
| | - Wade G Regehr
- Department of Neurobiology, Harvard Medical School, Boston, MA 02118, USA.
| |
Collapse
|
34
|
Clause A, Lauer AM, Kandler K. Mice Lacking the Alpha9 Subunit of the Nicotinic Acetylcholine Receptor Exhibit Deficits in Frequency Difference Limens and Sound Localization. Front Cell Neurosci 2017; 11:167. [PMID: 28663725 PMCID: PMC5471293 DOI: 10.3389/fncel.2017.00167] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 05/31/2017] [Indexed: 01/16/2023] Open
Abstract
Sound processing in the cochlea is modulated by cholinergic efferent axons arising from medial olivocochlear neurons in the brainstem. These axons contact outer hair cells in the mature cochlea and inner hair cells during development and activate nicotinic acetylcholine receptors composed of α9 and α10 subunits. The α9 subunit is necessary for mediating the effects of acetylcholine on hair cells as genetic deletion of the α9 subunit results in functional cholinergic de-efferentation of the cochlea. Cholinergic modulation of spontaneous cochlear activity before hearing onset is important for the maturation of central auditory circuits. In α9KO mice, the developmental refinement of inhibitory afferents to the lateral superior olive is disturbed, resulting in decreased tonotopic organization of this sound localization nucleus. In this study, we used behavioral tests to investigate whether the circuit anomalies in α9KO mice correlate with sound localization or sound frequency processing. Using a conditioned lick suppression task to measure sound localization, we found that three out of four α9KO mice showed impaired minimum audible angles. Using a prepulse inhibition of the acoustic startle response paradigm, we found that the ability of α9KO mice to detect sound frequency changes was impaired, whereas their ability to detect sound intensity changes was not. These results demonstrate that cholinergic, nicotinic α9 subunit mediated transmission in the developing cochlear plays an important role in the maturation of hearing.
Collapse
Affiliation(s)
- Amanda Clause
- Departments of Otolaryngology and Neurobiology, University of PittsburghPittsburgh, PA, United States
- Center for the Neural Basis of Cognition, University of PittsburghPittsburgh, PA, United States
| | - Amanda M. Lauer
- Center for Hearing and Balance, David M. Rubenstein Center, Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins UniversityBaltimore, MD, United States
| | - Karl Kandler
- Departments of Otolaryngology and Neurobiology, University of PittsburghPittsburgh, PA, United States
- Center for the Neural Basis of Cognition, University of PittsburghPittsburgh, PA, United States
- Department of Bioengineering, University of PittsburghPittsburgh, PA, United States
| |
Collapse
|
35
|
Pyott SJ, Duncan RK. BK Channels in the Vertebrate Inner Ear. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 128:369-99. [PMID: 27238269 DOI: 10.1016/bs.irn.2016.03.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The perception of complex acoustic stimuli begins with the deconstruction of sound into its frequency components. This spectral processing occurs first and foremost in the inner ear. In vertebrates, two very different strategies of frequency analysis have evolved. In nonmammalian vertebrates, the sensory hair cells of the inner ear are intrinsically electrically tuned to a narrow band of acoustic frequencies. This electrical tuning relies on the interplay between BK channels and voltage-gated calcium channels. Systematic variations in BK channel density and kinetics establish a gradient in electrical resonance that enables the coding of a broad range of acoustic frequencies. In contrast, mammalian hair cells are extrinsically tuned by mechanical properties of the cochlear duct. Even so, mammalian hair cells also express BK channels. These BK channels play critical roles in various aspects of mammalian auditory signaling, from developmental maturation to protection against acoustic trauma. This review summarizes the anatomical localization, biophysical properties, and functional contributions of BK channels in vertebrate inner ears. Areas of future research, based on an updated understanding of the biology of both BK channels and the inner ear, are also highlighted. Investigation of BK channels in the inner ear continues to provide fertile research grounds for examining both BK channel biophysics and the molecular mechanisms underlying signal processing in the auditory periphery.
Collapse
Affiliation(s)
- S J Pyott
- University Medical Center Groningen, Groningen, The Netherlands.
| | - R K Duncan
- Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
36
|
Goutman JD, Elgoyhen AB, Gómez-Casati ME. Cochlear hair cells: The sound-sensing machines. FEBS Lett 2015; 589:3354-61. [PMID: 26335749 PMCID: PMC4641020 DOI: 10.1016/j.febslet.2015.08.030] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 08/18/2015] [Accepted: 08/19/2015] [Indexed: 12/20/2022]
Abstract
The sensory epithelium of the mammalian inner ear contains two types of mechanosensory cells: inner (IHC) and outer hair cells (OHC). They both transduce mechanical force generated by sound waves into electrical signals. In their apical end, these cells possess a set of stereocilia representing the mechanosensing organelles. IHC are responsible for detecting sounds and transmitting the acoustic information to the brain by converting graded depolarization into trains of action potentials in auditory nerve fibers. OHC are responsible for the active mechanical amplification process that leads to the fine tuning and high sensitivity of the mammalian inner ear. This active amplification is the consequence of the ability of OHC to alter their cell length in response to changes in membrane potential, and is controlled by an efferent inhibitory innervation. Medial olivocochlear efferent fibers, originating in the brainstem, synapse directly at the base of OHC and release acetylcholine. A very special type of nicotinic receptor, assembled by α9α10 subunits, participates in this synapse. Here we review recent knowledge and the role of both afferent and efferent synapse in the inner ear.
Collapse
Affiliation(s)
- Juan D Goutman
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, "Dr. Héctor N Torres" (CONICET-UBA), Vuelta de Obligado 2490, Buenos Aires, Argentina.
| | - A Belén Elgoyhen
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, "Dr. Héctor N Torres" (CONICET-UBA), Vuelta de Obligado 2490, Buenos Aires, Argentina; Tercera Cátedra de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, Buenos Aires, Argentina
| | - María Eugenia Gómez-Casati
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, "Dr. Héctor N Torres" (CONICET-UBA), Vuelta de Obligado 2490, Buenos Aires, Argentina; Tercera Cátedra de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, Buenos Aires, Argentina.
| |
Collapse
|
37
|
Activation of BK and SK channels by efferent synapses on outer hair cells in high-frequency regions of the rodent cochlea. J Neurosci 2015; 35:1821-30. [PMID: 25653344 DOI: 10.1523/jneurosci.2790-14.2015] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Cholinergic neurons of the brainstem olivary complex project to and inhibit outer hair cells (OHCs), refining acoustic sensitivity of the mammalian cochlea. In all vertebrate hair cells studied to date, cholinergic inhibition results from the combined action of ionotropic acetylcholine receptors and associated calcium-activated potassium channels. Although inhibition was thought to involve exclusively small conductance (SK potassium channels), recent findings have shown that BK channels also contribute to inhibition in basal, high-frequency OHCs after the onset of hearing. Here we show that the waveform of randomly timed IPSCs (evoked by high extracellular potassium) in high-frequency OHCs is altered by blockade of either SK or BK channels, with BK channels supporting faster synaptic waveforms and SK channels supporting slower synaptic waveforms. Consistent with these findings, IPSCs recorded from high-frequency OHCs that express BK channels are briefer than IPSCs recorded from low-frequency (apical) OHCs that do not express BK channels and from immature high-frequency OHCs before the developmental onset of BK channel expression. Likewise, OHCs of BKα(-/-) mice lacking the pore-forming α-subunit of BK channels have longer IPSCs than do the OHCs of BKα(+/+) littermates. Furthermore, serial reconstruction of electron micrographs showed that postsynaptic cisterns of BKα(-/-) OHCs were smaller than those of BKα(+/+) OHCs, and immunofluorescent quantification showed that efferent presynaptic terminals of BKα(-/-) OHCs were smaller than those of BKα(+/+) OHCs. Together, these findings indicate that BK channels contribute to postsynaptic function, and influence the structural maturation of efferent-OHC synapses.
Collapse
|
38
|
Katz E, Elgoyhen AB. Short-term plasticity and modulation of synaptic transmission at mammalian inhibitory cholinergic olivocochlear synapses. Front Syst Neurosci 2014; 8:224. [PMID: 25520631 PMCID: PMC4251319 DOI: 10.3389/fnsys.2014.00224] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 11/06/2014] [Indexed: 12/23/2022] Open
Abstract
The organ of Corti, the mammalian sensory epithelium of the inner ear, has two types of mechanoreceptor cells, inner hair cells (IHCs) and outer hair cells (OHCs). In this sensory epithelium, vibrations produced by sound waves are transformed into electrical signals. When depolarized by incoming sounds, IHCs release glutamate and activate auditory nerve fibers innervating them and OHCs, by virtue of their electromotile property, increase the amplification and fine tuning of sound signals. The medial olivocochlear (MOC) system, an efferent feedback system, inhibits OHC activity and thereby reduces the sensitivity and sharp tuning of cochlear afferent fibers. During neonatal development, IHCs fire Ca2+ action potentials which evoke glutamate release promoting activity in the immature auditory system in the absence of sensory stimuli. During this period, MOC fibers also innervate IHCs and are thought to modulate their firing rate. Both the MOC-OHC and the MOC-IHC synapses are cholinergic, fast and inhibitory and mediated by the α9α10 nicotinic cholinergic receptor (nAChR) coupled to the activation of calcium-activated potassium channels that hyperpolarize the hair cells. In this review we discuss the biophysical, functional and molecular data which demonstrate that at the synapses between MOC efferent fibers and cochlear hair cells, modulation of transmitter release as well as short term synaptic plasticity mechanisms, operating both at the presynaptic terminal and at the postsynaptic hair-cell, determine the efficacy of these synapses and shape the hair cell response pattern.
Collapse
Affiliation(s)
- Eleonora Katz
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Buenos Aires, Argentina ; Departamento de Fisiología, Biología Molecular y Celular "Prof. Héctor Maldonado", Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires Buenos Aires, Argentina
| | - Ana Belén Elgoyhen
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Buenos Aires, Argentina ; Tercera Cátedra de Farmacología, Facultad de Medicina, Universidad de Buenos Aires Buenos Aires, Argentina
| |
Collapse
|
39
|
Loss of α-calcitonin gene-related peptide (αCGRP) reduces the efficacy of the Vestibulo-ocular Reflex (VOR). J Neurosci 2014; 34:10453-8. [PMID: 25080603 DOI: 10.1523/jneurosci.3336-13.2014] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The neuroactive peptide calcitonin-gene related peptide (CGRP) is known to act at efferent synapses and their targets in hair cell organs, including the cochlea and lateral line. CGRP is also expressed in vestibular efferent neurons as well as a number of central vestibular neurons. Although CGRP-null (-/-) mice demonstrate a significant reduction in cochlear nerve sound-evoked activity compared with wild-type mice, it is unknown whether and how the loss of CGRP influence vestibular system function. Vestibular function was assessed by quantifying the vestibulo-ocular reflex (VOR) in alert mice. The loss of CGRP in (-/-) mice was associated with a reduction of the VOR gain of ≈50% without a concomitant change in phase. Using immunohistochemistry, we confirmed that, although CGRP staining was absent in the vestibular end-organs of null (-/-) mice, cholinergic staining appeared normal, suggesting that the overall gross development of vestibular efferent innervation was unaltered. We further confirmed that the observed deficit in vestibular function of null (-/-) mice was not the result of nontargeted effects at the level of the extraocular motor neurons and/or their innervation of extraocular muscles. Analysis of the relationship between vestibular quick phase amplitude and peak velocity revealed that extraocular motor function was unchanged, and immunohistochemistry revealed no abnormalities in motor endplates. Together, our findings show that the neurotransmitter CGRP plays a key role in ensuring VOR efficacy.
Collapse
|
40
|
Fuchs PA. A 'calcium capacitor' shapes cholinergic inhibition of cochlear hair cells. J Physiol 2014; 592:3393-401. [PMID: 24566542 PMCID: PMC4229337 DOI: 10.1113/jphysiol.2013.267914] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 02/18/2014] [Indexed: 01/07/2023] Open
Abstract
Efferent cholinergic neurons project from the brainstem to inhibit sensory hair cells of the vertebrate inner ear. This inhibitory synapse combines the activity of an unusual class of ionotropic cholinergic receptor with that of nearby calcium-dependent potassium channels to shunt and hyperpolarize the hair cell. Postsynaptic calcium signalling is constrained by a thin near-membrane cistern that is co-extensive with the efferent terminal contacts. The postsynaptic cistern may play an essential role in calcium homeostasis, serving as sink or source, depending on ongoing activity and the degree of buffer saturation. Release of calcium from postsynaptic stores leads to a process of retrograde facilitation via the synthesis of nitric oxide in the hair cell. Activity-dependent synaptic modification may contribute to changes in hair cell innervation that occur during development, and in the aged or damaged cochlea.
Collapse
Affiliation(s)
- Paul Albert Fuchs
- Center for Hearing and Balance, Otolaryngology-Head and Neck Surgery, and Center for Sensory Biology, Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
41
|
Activation of presynaptic GABA(B(1a,2)) receptors inhibits synaptic transmission at mammalian inhibitory cholinergic olivocochlear-hair cell synapses. J Neurosci 2013; 33:15477-87. [PMID: 24068816 DOI: 10.1523/jneurosci.2554-13.2013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The synapse between olivocochlear (OC) neurons and cochlear mechanosensory hair cells is cholinergic, fast, and inhibitory. The inhibitory sign of this cholinergic synapse is accounted for by the activation of Ca(2+)-permeable postsynaptic α9α10 nicotinic receptors coupled to the opening of hyperpolarizing Ca(2+)-activated small-conductance type 2 (SK2)K(+) channels. Acetylcholine (ACh) release at this synapse is supported by both P/Q- and N-type voltage-gated calcium channels (VGCCs). Although the OC synapse is cholinergic, an abundant OC GABA innervation is present along the mammalian cochlea. The role of this neurotransmitter at the OC efferent innervation, however, is for the most part unknown. We show that GABA fails to evoke fast postsynaptic inhibitory currents in apical developing inner and outer hair cells. However, electrical stimulation of OC efferent fibers activates presynaptic GABA(B(1a,2)) receptors [GABA(B(1a,2))Rs] that downregulate the amount of ACh released at the OC-hair cell synapse, by inhibiting P/Q-type VGCCs. We confirmed the expression of GABA(B)Rs at OC terminals contacting the hair cells by coimmunostaining for GFP and synaptophysin in transgenic mice expressing GABA(B1)-GFP fusion proteins. Moreover, coimmunostaining with antibodies against the GABA synthetic enzyme glutamic acid decarboxylase and synaptophysin support the idea that GABA is directly synthesized at OC terminals contacting the hair cells during development. Thus, we demonstrate for the first time a physiological role for GABA in cochlear synaptic function. In addition, our data suggest that the GABA(B1a) isoform selectively inhibits release at efferent cholinergic synapses.
Collapse
|
42
|
Protection from noise-induced hearing loss by Kv2.2 potassium currents in the central medial olivocochlear system. J Neurosci 2013; 33:9113-21. [PMID: 23699522 DOI: 10.1523/jneurosci.5043-12.2013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The central auditory brainstem provides an efferent projection known as the medial olivocochlear (MOC) system, which regulates the cochlear amplifier and mediates protection on exposure to loud sound. It arises from neurons of the ventral nucleus of the trapezoid body (VNTB), so control of neuronal excitability in this pathway has profound effects on hearing. The VNTB and the medial nucleus of the trapezoid body are the only sites of expression for the Kv2.2 voltage-gated potassium channel in the auditory brainstem, consistent with a specialized function of these channels. In the absence of unambiguous antagonists, we used recombinant and transgenic methods to examine how Kv2.2 contributes to MOC efferent function. Viral gene transfer of dominant-negative Kv2.2 in wild-type mice suppressed outward K(+) currents, increasing action potential (AP) half-width and reducing repetitive firing. Similarly, VNTB neurons from Kv2.2 knock-out mice (Kv2.2KO) also showed increased AP duration. Control experiments established that Kv2.2 was not expressed in the cochlea, so any changes in auditory function in the Kv2.2KO mouse must be of central origin. Further, in vivo recordings of auditory brainstem responses revealed that these Kv2.2KO mice were more susceptible to noise-induced hearing loss. We conclude that Kv2.2 regulates neuronal excitability in these brainstem nuclei by maintaining short APs and enhancing high-frequency firing. This safeguards efferent MOC firing during high-intensity sounds and is crucial in the mediation of protection after auditory overexposure.
Collapse
|
43
|
Kong JH, Zachary S, Rohmann KN, Fuchs PA. Retrograde facilitation of efferent synapses on cochlear hair cells. J Assoc Res Otolaryngol 2012. [PMID: 23183877 DOI: 10.1007/s10162-012-0361-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Cochlear inner hair cells (IHCs) are temporarily innervated by efferent cholinergic fibers prior to the onset of hearing. During low-frequency firing, these efferent synapses have a relatively low probability of transmitter release but facilitate strongly with repetitive stimulation. A retrograde signal from the hair cell to the efferent terminal contributes to this facilitation. When IHCs were treated with the ryanodine receptor agonist, cyclic adenosine phosphoribose (cADPR), release probability of the efferent terminal rose. This effect was quantified by computing the quantum content from a train of 100 suprathreshold stimuli to the efferent fibers. Quantum content was sevenfold higher when IHCs were treated with 100 μM cADPR (applied in the recording pipette). Since cADPR is membrane impermeant, this result implies that an extracellular messenger travels from the hair cell to the efferent terminal. cADPR is presumed to generate this messenger by increasing cytoplasmic calcium. Consistent with this presumption, voltage-gated calcium flux into the IHC also caused retrograde facilitation of efferent transmission. Retrograde facilitation was observed in IHCs of a vesicular glutamate transporter (VGlut3) null mouse and for wild-type rat hair cells subject to wide-spectrum glutamate receptor blockade, demonstrating that glutamate was unlikely to be the extracellular messenger. Rather, bath application of nitric oxide (NO) donors caused an increase in potassium-evoked efferent transmitter release while the NO scavenger carboxy-PTIO was able to prevent retrograde facilitation produced by cADPR or IHC depolarization. Thus, hair cell activity can drive retrograde facilitation of efferent input via calcium-dependent production of NO.
Collapse
MESH Headings
- Acoustic Stimulation
- Amino Acid Transport Systems, Acidic/deficiency
- Amino Acid Transport Systems, Acidic/genetics
- Amino Acid Transport Systems, Acidic/metabolism
- Animals
- Calcium/metabolism
- Calcium Channel Agonists/pharmacology
- Dose-Response Relationship, Drug
- Hair Cells, Auditory, Inner/drug effects
- Hair Cells, Auditory, Inner/physiology
- Mice
- Mice, Knockout
- Models, Animal
- Neurons, Efferent/drug effects
- Neurons, Efferent/physiology
- Nitric Oxide/metabolism
- Nitric Oxide Donors/pharmacology
- Rats
- Rats, Inbred Strains
- Synapses/drug effects
- Synapses/physiology
- Synaptic Transmission/drug effects
- Synaptic Transmission/physiology
Collapse
Affiliation(s)
- Jee-Hyun Kong
- Otolaryngology-Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | | | | | |
Collapse
|
44
|
Clark NR, Brown GJ, Jürgens T, Meddis R. A frequency-selective feedback model of auditory efferent suppression and its implications for the recognition of speech in noise. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2012; 132:1535-41. [PMID: 22978882 DOI: 10.1121/1.4742745] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
The potential contribution of the peripheral auditory efferent system to our understanding of speech in a background of competing noise was studied using a computer model of the auditory periphery and assessed using an automatic speech recognition system. A previous study had shown that a fixed efferent attenuation applied to all channels of a multi-channel model could improve the recognition of connected digit triplets in noise [G. J. Brown, R. T. Ferry, and R. Meddis, J. Acoust. Soc. Am. 127, 943-954 (2010)]. In the current study an anatomically justified feedback loop was used to automatically regulate separate attenuation values for each auditory channel. This arrangement resulted in a further enhancement of speech recognition over fixed-attenuation conditions. Comparisons between multi-talker babble and pink noise interference conditions suggest that the benefit originates from the model's ability to modify the amount of suppression in each channel separately according to the spectral shape of the interfering sounds.
Collapse
Affiliation(s)
- Nicholas R Clark
- Department of Psychology, University of Essex, Colchester CO4 3SQ, United Kingdom
| | | | | | | |
Collapse
|
45
|
Phylogenetic differences in calcium permeability of the auditory hair cell cholinergic nicotinic receptor. Proc Natl Acad Sci U S A 2012; 109:4308-13. [PMID: 22371598 DOI: 10.1073/pnas.1115488109] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The α9 and α10 cholinergic nicotinic receptor subunits assemble to form the receptor that mediates efferent inhibition of hair cell function within the auditory sensory organ, a mechanism thought to modulate the dynamic range of hearing. In contrast to all nicotinic receptors, which serve excitatory neurotransmission, the activation of α9α10 produces hyperpolarization of hair cells. An evolutionary analysis has shown that the α10 subunit exhibits signatures of positive selection only along the mammalian lineage, strongly suggesting the acquisition of a unique function. To establish whether mammalian α9α10 receptors have acquired distinct functional properties as a consequence of this evolutionary pressure, we compared the properties of rat and chicken recombinant and native α9α10 receptors. Our main finding in the present work is that, in contrast to the high (pCa(2+)/pMonovalents ∼10) Ca(2+) permeability reported for rat α9α10 receptors, recombinant and native chicken α9α10 receptors have a much lower permeability (∼2) to this cation, comparable to that of neuronal α4β2 receptors. Moreover, we show that, in contrast to α10, α7 as well as α4 and β2 nicotinic subunits are under purifying selection in vertebrates, consistent with the conserved Ca(2+) permeability reported across species. These results have important consequences for the activation of signaling cascades that lead to hyperpolarization of hair cells after α9α10 gating at the cholinergic-hair cell synapse. In addition, they suggest that high Ca(2+) permeability of the α9α10 cholinergic nicotinic receptor might have evolved together with other features that have given the mammalian ear an expanded high-frequency sensitivity.
Collapse
|