1
|
Hosoya M, Ueno M, Shimanuki MN, Nishiyama T, Oishi N, Ozawa H. A primate model animal revealed the inter-species differences and similarities in the subtype specifications of the spiral ganglion neurons. Sci Rep 2024; 14:25166. [PMID: 39448766 PMCID: PMC11502759 DOI: 10.1038/s41598-024-76892-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024] Open
Abstract
Type I spiral ganglion neurons are peripheral neurons essential for hearing perception. While they can be subdivided in mice based on characteristic gene expression patterns, detailed examinations of these subtypes in primates and humans are lacking. In this study, we investigated the developmental subtypes of spiral ganglion neurons in the common marmoset (Callithrix jacchus). We confirmed that Type I spiral ganglion can be divided based on the characteristic gene expression patterns of several marker genes. However, some combinations of these genes differ from those in rodents, suggesting common marmoset's suitability for advancing our understanding of human cochlear development. Additionally, identifying the essential time points for subtype specifications and subsequent maturation will aid in studying the primate-specific developmental biology of the inner ear. This could lead to new treatment strategies for hearing loss in humans and be valuable for studying age-related hearing loss, as well as designing regenerative therapies.
Collapse
Affiliation(s)
- Makoto Hosoya
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Masafumi Ueno
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Marie N Shimanuki
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Takanori Nishiyama
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Naoki Oishi
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hiroyuki Ozawa
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| |
Collapse
|
2
|
Conrad LJ, Grandi FC, Carlton AJ, Jeng JY, de Tomasi L, Zarecki P, Marcotti W, Johnson SL, Mustapha M. The upregulation of K + and HCN channels in developing spiral ganglion neurons is mediated by cochlear inner hair cells. J Physiol 2024; 602:5329-5351. [PMID: 39324853 DOI: 10.1113/jp286134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 09/02/2024] [Indexed: 09/27/2024] Open
Abstract
Spiral ganglion neurons (SGNs) are primary sensory afferent neurons that relay acoustic information from the cochlear inner hair cells (IHCs) to the brainstem. The response properties of different SGNs diverge to represent a wide range of sound intensities in an action-potential code. This biophysical heterogeneity is established during pre-hearing stages of development, a time when IHCs fire spontaneous Ca2+ action potentials that drive glutamate release from their ribbon synapses onto the SGN terminals. The role of spontaneous IHC activity in the refinement of SGN characteristics is still largely unknown. Using pre-hearing otoferlin knockout mice (Otof-/-), in which Ca2+-dependent exocytosis in IHCs is abolished, we found that developing SGNs fail to upregulate low-voltage-activated K+-channels and hyperpolarisation-activated cyclic-nucleotide-gated channels. This delayed maturation resulted in hyperexcitable SGNs with immature firing characteristics. We have also shown that SGNs that synapse with the pillar side of the IHCs selectively express a resurgent K+ current, highlighting a novel biophysical marker for these neurons. RNA-sequencing showed that several K+ channels are downregulated in Otof-/- mice, further supporting the electrophysiological recordings. Our data demonstrate that spontaneous Ca2+-dependent activity in pre-hearing IHCs regulates some of the key biophysical and molecular features of the developing SGNs. KEY POINTS: Ca2+-dependent exocytosis in inner hair cells (IHCs) is otoferlin-dependent as early as postnatal day 1. A lack of otoferlin in IHCs affects potassium channel expression in SGNs. The absence of otoferlin is associated with SGN hyperexcitability. We propose that type I spiral ganglion neuron functional maturation depends on IHC exocytosis.
Collapse
Affiliation(s)
- Linus J Conrad
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Fiorella C Grandi
- INSERM, Institute de Myologie, Centre de Recherche en Myologie F-75013, Sorbonne Université, Paris, France
| | - Adam J Carlton
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Jing-Yi Jeng
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Lara de Tomasi
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Patryk Zarecki
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Walter Marcotti
- School of Biosciences, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Stuart L Johnson
- School of Biosciences, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Mirna Mustapha
- School of Biosciences, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| |
Collapse
|
3
|
Capshaw G, Diebold CA, Sterbing SJ, Lauer AM, Moss CF. Echolocating bats show species-specific variation in susceptibility to acoustic forward masking. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2024; 156:511-523. [PMID: 39013168 PMCID: PMC11254387 DOI: 10.1121/10.0026624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 06/06/2024] [Accepted: 06/21/2024] [Indexed: 07/18/2024]
Abstract
Echolocating bats rely on precise auditory temporal processing to detect echoes generated by calls that may be emitted at rates reaching 150-200 Hz. High call rates can introduce forward masking perceptual effects that interfere with echo detection; however, bats may have evolved specializations to prevent repetition suppression of auditory responses and facilitate detection of sounds separated by brief intervals. Recovery of the auditory brainstem response (ABR) was assessed in two species that differ in the temporal characteristics of their echolocation behaviors: Eptesicus fuscus, which uses high call rates to capture prey, and Carollia perspicillata, which uses lower call rates to avoid obstacles and forage for fruit. We observed significant species differences in the effects of forward masking on ABR wave 1, in which E. fuscus maintained comparable ABR wave 1 amplitudes when stimulated at intervals of <3 ms, whereas post-stimulus recovery in C. perspicillata required 12 ms. When the intensity of the second stimulus was reduced by 20-30 dB relative to the first, however, C. perspicillata showed greater recovery of wave 1 amplitudes. The results demonstrate that species differences in temporal resolution are established at early levels of the auditory pathway and that these differences reflect auditory processing requirements of species-specific echolocation behaviors.
Collapse
Affiliation(s)
- Grace Capshaw
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Clarice A Diebold
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Susanne J Sterbing
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Amanda M Lauer
- Department of Otolaryngology-Head & Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Cynthia F Moss
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, Maryland 21218, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
4
|
Bovee S, Klump GM, Pyott SJ, Sielaff C, Köppl C. Cochlear Ribbon Synapses in Aged Gerbils. Int J Mol Sci 2024; 25:2738. [PMID: 38473985 DOI: 10.3390/ijms25052738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/15/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
In mammalian hearing, type-I afferent auditory nerve fibers comprise the basis of the afferent auditory pathway. They are connected to inner hair cells of the cochlea via specialized ribbon synapses. Auditory nerve fibers of different physiological types differ subtly in their synaptic location and morphology. Low-spontaneous-rate auditory nerve fibers typically connect on the modiolar side of the inner hair cell, while high-spontaneous-rate fibers are typically found on the pillar side. In aging and noise-damaged ears, this fine-tuned balance between auditory nerve fiber populations can be disrupted and the functional consequences are currently unclear. Here, using immunofluorescent labeling of presynaptic ribbons and postsynaptic glutamate receptor patches, we investigated changes in synaptic morphology at three different tonotopic locations along the cochlea of aging gerbils compared to those of young adults. Quiet-aged gerbils showed about 20% loss of afferent ribbon synapses. While the loss was random at apical, low-frequency cochlear locations, at the basal, high-frequency location it almost exclusively affected the modiolar-located synapses. The subtle differences in volumes of pre- and postsynaptic elements located on the inner hair cell's modiolar versus pillar side were unaffected by age. This is consistent with known physiology and suggests a predominant, age-related loss in the low-spontaneous-rate auditory nerve population in the cochlear base, but not the apex.
Collapse
Affiliation(s)
- Sonny Bovee
- Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany
| | - Georg M Klump
- Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany
- Cluster of Excellence "Hearing4all", Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany
- Research Centre Neurosensory Science, Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany
| | - Sonja J Pyott
- Department of Otorhinolaryngology/Head and Neck Surgery, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
| | - Charlotte Sielaff
- Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), 30625 Hannover, Germany
| | - Christine Köppl
- Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany
- Cluster of Excellence "Hearing4all", Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany
- Research Centre Neurosensory Science, Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany
| |
Collapse
|
5
|
Carr CE, Wang T, Kraemer I, Capshaw G, Ashida G, Köppl C, Kempter R, Kuokkanen PT. Experience-Dependent Plasticity in Nucleus Laminaris of the Barn Owl. J Neurosci 2024; 44:e0940232023. [PMID: 37989591 PMCID: PMC10851688 DOI: 10.1523/jneurosci.0940-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 10/12/2023] [Accepted: 11/01/2023] [Indexed: 11/23/2023] Open
Abstract
Interaural time differences (ITDs) are a major cue for sound localization and change with increasing head size. Since the barn owl's head width more than doubles in the month after hatching, we hypothesized that the development of their ITD detection circuit might be modified by experience. To test this, we raised owls with unilateral ear inserts that delayed and attenuated the acoustic signal, and then measured the ITD representation in the brainstem nucleus laminaris (NL) when they were adults. The ITD circuit is composed of delay line inputs to coincidence detectors, and we predicted that plastic changes would lead to shorter delays in the axons from the manipulated ear, and complementary shifts in ITD representation on the two sides. In owls that received ear inserts starting around P14, the maps of ITD shifted in the predicted direction, but only on the ipsilateral side, and only in those tonotopic regions that had not experienced auditory stimulation prior to insertion. The contralateral map did not change. Thus, experience-dependent plasticity of the ITD circuit occurs in NL, and our data suggest that ipsilateral and contralateral delays are independently regulated. As a result, altered auditory input during development leads to long-lasting changes in the representation of ITD.Significance Statement The early life of barn owls is marked by increasing sensitivity to sound, and by increasing ITDs. Their prolonged post-hatch development allowed us to examine the role of altered auditory experience in the development of ITD detection circuits. We raised owls with a unilateral ear insert and found that their maps of ITD were altered by experience, but only in those tonotopic regions ipsilateral to the occluded ear that had not experienced auditory stimulation prior to insertion. This experience-induced plasticity allows the sound localization circuits to be customized to individual characteristics, such as the size of the head, and potentially to compensate for imbalanced hearing sensitivities between the left and right ears.
Collapse
Affiliation(s)
- Catherine E Carr
- Department of Biology, University of Maryland College Park, College Park, MD 20742
| | - Tiffany Wang
- Department of Biology, University of Maryland College Park, College Park, MD 20742
| | - Ira Kraemer
- Department of Biology, University of Maryland College Park, College Park, MD 20742
| | - Grace Capshaw
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD 21218
| | - Go Ashida
- Department of Neuroscience, School of Medicine and Health Sciences, Research Center for Neurosensory Sciences and Cluster of Excellence "Hearing4all" Carl von Ossietzky University, 26129 Oldenburg, Germany
| | - Christine Köppl
- Department of Neuroscience, School of Medicine and Health Sciences, Research Center for Neurosensory Sciences and Cluster of Excellence "Hearing4all" Carl von Ossietzky University, 26129 Oldenburg, Germany
| | - Richard Kempter
- Institute for Theoretical Biology, Department of Biology, Humboldt-Universität zu Berlin, 10115 Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, 10115 Berlin, Germany
- Einstein Center for Neurosciences Berlin, 10117 Berlin, Germany
| | - Paula T Kuokkanen
- Department of Biology, University of Maryland College Park, College Park, MD 20742
| |
Collapse
|
6
|
Wong W. A Fundamental Inequality Governing the Rate Coding Response of Sensory Neurons. BIOLOGICAL CYBERNETICS 2023; 117:285-295. [PMID: 37597017 DOI: 10.1007/s00422-023-00971-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 07/30/2023] [Indexed: 08/21/2023]
Abstract
A fundamental inequality governing the spike activity of peripheral neurons is derived and tested against auditory data. This inequality states that the steady-state firing rate must lie between the arithmetic and geometric means of the spontaneous and peak activities during adaptation. Implications towards the development of auditory mechanistic models are explored.
Collapse
Affiliation(s)
- Willy Wong
- Department of Electrical and Computer Engineering and Institute of Biomedical Engineering, University of Toronto, Toronto, M5S3G4, Canada.
| |
Collapse
|
7
|
Heeringa AN, Teske F, Ashida G, Köppl C. Cochlear aging disrupts the correlation between spontaneous rate- and sound-level coding in auditory nerve fibers. J Neurophysiol 2023; 130:736-750. [PMID: 37584075 DOI: 10.1152/jn.00090.2023] [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: 02/28/2023] [Revised: 07/12/2023] [Accepted: 08/10/2023] [Indexed: 08/17/2023] Open
Abstract
The spiking activity of auditory nerve fibers (ANFs) transmits information about the acoustic environment from the cochlea to the central auditory system. Increasing age leads to degeneration of cochlear tissues, including the sensory hair cells and stria vascularis. Here, we aim to identify the functional effects of such age-related cochlear pathologies of ANFs. Rate-level functions (RLFs) were recorded from single-unit ANFs of young adult (n = 52, 3-12 months) and quiet-aged (n = 24, >36 months) Mongolian gerbils of either sex. RLFs were used to determine sensitivity and spontaneous rates (SRs) and were classified into flat-saturating, sloping-saturating, and straight categories, as previously established. A physiologically based cochlear model, adapted for the gerbil, was used to simulate the effects of cochlear degeneration on ANF physiology. In ANFs tuned to low frequencies (<3.5 kHz), SR was lower in those of aged gerbils, while an age-related loss of low-SR fibers was evident in ANFs tuned to high frequencies. These changes in SR distribution did not affect the typical SR versus sensitivity correlation. The distribution of RLF types among low-SR fibers, however, shifted toward that of high-SR fibers, specifically showing more fast-saturating and fewer sloping-saturating RLFs. A modeled striatal degeneration, which affects the combined inner hair cell and synaptic output, reduced SR but left RLF type unchanged. An additional reduced basilar membrane gain, which decreased sensitivity, explained the changed RLF types. Overall, the data indicated age-related changes in the characteristics of single ANFs that blurred the established relationships between SR and RLF types.NEW & NOTEWORTHY Auditory nerve fibers, which connect the cochlea to the central auditory system, change their encoding of sound level in aged gerbils. In addition to a general shift to higher levels, indicative of decreased sensitivity, level coding was also differentially affected in fibers with low- and high-spontaneous rates. Loss of low-spontaneous rate fibers, combined with a general decrease of spontaneous rate, further blurs the categorization of auditory nerve fiber types in the aged gerbil.
Collapse
Affiliation(s)
- Amarins N Heeringa
- Cluster of Excellence "Hearing4all," Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
- Research Centre Neurosensory Science, Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Fiona Teske
- Research Centre Neurosensory Science, Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Go Ashida
- Cluster of Excellence "Hearing4all," Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
- Research Centre Neurosensory Science, Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Christine Köppl
- Cluster of Excellence "Hearing4all," Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
- Research Centre Neurosensory Science, Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
8
|
Siebald C, Vincent PFY, Bottom RT, Sun S, Reijntjes DOJ, Manca M, Glowatzki E, Müller U. Molecular signatures define subtypes of auditory afferents with distinct peripheral projection patterns and physiological properties. Proc Natl Acad Sci U S A 2023; 120:e2217033120. [PMID: 37487063 PMCID: PMC10400978 DOI: 10.1073/pnas.2217033120] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 06/06/2023] [Indexed: 07/26/2023] Open
Abstract
Type I spiral ganglion neurons (SGNs) are the auditory afferents that transmit sound information from cochlear inner hair cells (IHCs) to the brainstem. These afferents consist of physiological subtypes that differ in their spontaneous firing rate (SR), activation threshold, and dynamic range and have been described as low, medium, and high SR fibers. Lately, single-cell RNA sequencing experiments have revealed three molecularly defined type I SGN subtypes. The extent to which physiological type I SGN subtypes correspond to molecularly defined subtypes is unclear. To address this question, we have generated mouse lines expressing CreERT2 in SGN subtypes that allow for a physiological assessment of molecular subtypes. We show that Lypd1-CreERT2 expressing SGNs represent a well-defined group of neurons that preferentially innervate the IHC modiolar side and exhibit a narrow range of low SRs. In contrast, Calb2-CreERT2 expressing SGNs preferentially innervate the IHC pillar side and exhibit a wider range of SRs, thus suggesting that a strict stratification of all SGNs into three molecular subclasses is not obvious, at least not with the CreERT2 tools used here. Genetically marked neuronal subtypes refine their innervation specificity onto IHCs postnatally during the time when activity is required to refine their molecular phenotype. Type I SGNs thus consist of genetically defined subtypes with distinct physiological properties and innervation patterns. The molecular subtype-specific lines characterized here will provide important tools for investigating the role of the physiologically distinct type I SGNs in encoding sound signals.
Collapse
Affiliation(s)
- Caroline Siebald
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Philippe F. Y. Vincent
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Riley T. Bottom
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Shuohao Sun
- National Institute of Biological Science, Beijing102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing100084, China
| | - Daniel O. J. Reijntjes
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Marco Manca
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Elisabeth Glowatzki
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Ulrich Müller
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
| |
Collapse
|
9
|
Carr CE, Wang T, Kraemer I, Capshaw G, Ashida G, Koeppl C, Kempter R, Kuokkanen PT. Experience-Dependent Plasticity in Nucleus Laminaris of the Barn Owl. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.02.526884. [PMID: 36778252 PMCID: PMC9915572 DOI: 10.1101/2023.02.02.526884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Barn owls experience increasing interaural time differences (ITDs) during development, because their head width more than doubles in the month after hatching. We therefore hypothesized that their ITD detection circuit might be modified by experience. To test this, we raised owls with unilateral ear inserts that delayed and attenuated the acoustic signal, then measured the ITD representation in the brainstem nucleus laminaris (NL) when they were adult. The ITD circuit is composed of delay line inputs to coincidence detectors, and we predicted that plastic changes would lead to shorter delays in the axons from the manipulated ear, and complementary shifts in ITD representation on the two sides. In owls that received ear inserts starting around P14, the maps of ITD shifted in the predicted direction, but only on the ipsilateral side, and only in those tonotopic regions that had not experienced auditory stimulation prior to insertion. The contralateral map did not change. Experience-dependent plasticity of the ITD circuit occurs in NL, and our data suggest that ipsilateral and contralateral delays are independently regulated. Thus, altered auditory input during development leads to long-lasting changes in the representation of ITD.
Collapse
|
10
|
Sun G, Tang M, Wang X, Li D, Liu W, Qi J, Wang H, Hu B. Generation of human otic neuronal organoids using pluripotent stem cells. Cell Prolif 2023; 56:e13434. [PMID: 36825797 DOI: 10.1111/cpr.13434] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
Otic neurons, also known as spiral ganglion neurons (SGNs) in mammalian cochlea, transmit electrical signals from sensory hair cells to cochlear nuclei of the auditory system. SGNs are sensitive to toxic insults, vulnerable to get irreversible damaged and hardly regenerate after damage, causing persistent sensorineural hearing loss. Yet, to get authentic SGNs for research or therapeutic purpose remains challenging. Here we developed a protocol to generate human otic neuronal organoids (hONOs) from human pluripotent stem cells (hESCs), in which hESCs were step-wisely induced to SGNs of the corresponding stages according to their developmental trajectory. The hONOs were enriched for SGN-like cells at early stage, and for both neurons and astrocytes, Schwann cells or supporting cells thereafter. In these hONOs, we also determined the existence of typical Type I and Type II SGNs. Mature hONOs (at differentiation Day 60) formed neural network, featured by giant depolarizing potential (GDP)-like events and rosette-organized regions-elicited calcium traces. Electrophysiological analysis confirmed the existence of glutamate-responsive neurons in these hONOs. The otic neuronal organoids generated in this study provide an ideal model to study SGNs and related disorders, facilitating therapeutic development for sensorineural hearing loss.
Collapse
Affiliation(s)
- Gaoying Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Mingming Tang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Xinyue Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Da Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Wenwen Liu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jianhuan Qi
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Haibo Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Baoyang Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
11
|
Qiu X, Müller U. Sensing sound: Cellular specializations and molecular force sensors. Neuron 2022; 110:3667-3687. [PMID: 36223766 PMCID: PMC9671866 DOI: 10.1016/j.neuron.2022.09.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/03/2022] [Accepted: 09/14/2022] [Indexed: 11/08/2022]
Abstract
Organisms of all phyla express mechanosensitive ion channels with a wide range of physiological functions. In recent years, several classes of mechanically gated ion channels have been identified. Some of these ion channels are intrinsically mechanosensitive. Others depend on accessory proteins to regulate their response to mechanical force. The mechanotransduction machinery of cochlear hair cells provides a particularly striking example of a complex force-sensing machine. This molecular ensemble is embedded into a specialized cellular compartment that is crucial for its function. Notably, mechanotransduction channels of cochlear hair cells are not only critical for auditory perception. They also shape their cellular environment and regulate the development of auditory circuitry. Here, we summarize recent discoveries that have shed light on the composition of the mechanotransduction machinery of cochlear hair cells and how this machinery contributes to the development and function of the auditory system.
Collapse
Affiliation(s)
- Xufeng Qiu
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ulrich Müller
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
12
|
Mansuri J, Aleem H, Grzywacz NM. Systematic errors in the perception of rhythm. Front Hum Neurosci 2022; 16:1009219. [DOI: 10.3389/fnhum.2022.1009219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/14/2022] [Indexed: 11/11/2022] Open
Abstract
One hypothesis for why humans enjoy musical rhythms relates to their prediction of when each beat should occur. The ability to predict the timing of an event is important from an evolutionary perspective. Therefore, our brains have evolved internal mechanisms for processing the progression of time. However, due to inherent noise in neural signals, this prediction is not always accurate. Theoretical considerations of optimal estimates suggest the occurrence of certain systematic errors made by the brain when estimating the timing of beats in rhythms. Here, we tested psychophysically whether these systematic errors exist and if so, how they depend on stimulus parameters. Our experimental data revealed two main types of systematic errors. First, observers perceived the time of the last beat of a rhythmic pattern as happening earlier than actual when the inter-beat interval was short. Second, the perceived time of the last beat was later than the actual when the inter-beat interval was long. The magnitude of these systematic errors fell as the number of beats increased. However, with many beats, the errors due to long inter-beat intervals became more apparent. We propose a Bayesian model for these systematic errors. The model fits these data well, allowing us to offer possible explanations for how these errors occurred. For instance, neural processes possibly contributing to the errors include noisy and temporally asymmetric impulse responses, priors preferring certain time intervals, and better-early-than-late loss functions. We finish this article with brief discussions of both the implications of systematic errors for the appreciation of rhythm and the possible compensation by the brain’s motor system during a musical performance.
Collapse
|
13
|
Wolf BJ, Kusch K, Hunniford V, Vona B, Kühler R, Keppeler D, Strenzke N, Moser T. Is there an unmet medical need for improved hearing restoration? EMBO Mol Med 2022; 14:e15798. [PMID: 35833443 PMCID: PMC9358394 DOI: 10.15252/emmm.202215798] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/12/2022] [Accepted: 06/02/2022] [Indexed: 12/26/2022] Open
Abstract
Hearing impairment, the most prevalent sensory deficit, affects more than 466 million people worldwide (WHO). We presently lack causative treatment for the most common form, sensorineural hearing impairment; hearing aids and cochlear implants (CI) remain the only means of hearing restoration. We engaged with CI users to learn about their expectations and their willingness to collaborate with health care professionals on establishing novel therapies. We summarize upcoming CI innovations, gene therapies, and regenerative approaches and evaluate the chances for clinical translation of these novel strategies. We conclude that there remains an unmet medical need for improving hearing restoration and that we are likely to witness the clinical translation of gene therapy and major CI innovations within this decade.
Collapse
Affiliation(s)
- Bettina Julia Wolf
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Auditory Neuroscience and Optogenetics Laboratory, German Primate Center, Göttingen, Germany.,Auditory Neuroscience & Synaptic Nanophysiology Group, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Kathrin Kusch
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Functional Auditory Genomics Group, Auditory Neuroscience and Optogenetics Laboratory, German Primate Center, Göttingen, Germany
| | - Victoria Hunniford
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Sensory and Motor Neuroscience PhD Program, Göttingen Graduate Center for Neurosciences, Biophysics, and Molecular Biosciences, Göttingen, Germany
| | - Barbara Vona
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Robert Kühler
- Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
| | - Daniel Keppeler
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Auditory Neuroscience & Synaptic Nanophysiology Group, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Nicola Strenzke
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany.,Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Auditory Neuroscience and Optogenetics Laboratory, German Primate Center, Göttingen, Germany.,Auditory Neuroscience & Synaptic Nanophysiology Group, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.,Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
| |
Collapse
|
14
|
Kessler D, Carr CE, Kretzberg J, Ashida G. Theoretical Relationship Between Two Measures of Spike Synchrony: Correlation Index and Vector Strength. Front Neurosci 2022; 15:761826. [PMID: 34987357 PMCID: PMC8721039 DOI: 10.3389/fnins.2021.761826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/09/2021] [Indexed: 11/24/2022] Open
Abstract
Information processing in the nervous system critically relies on temporally precise spiking activity. In the auditory system, various degrees of phase-locking can be observed from the auditory nerve to cortical neurons. The classical metric for quantifying phase-locking is the vector strength (VS), which captures the periodicity in neuronal spiking. More recently, another metric, called the correlation index (CI), was proposed to quantify the temporally reproducible response characteristics of a neuron. The CI is defined as the peak value of a normalized shuffled autocorrelogram (SAC). Both VS and CI have been used to investigate how temporal information is processed and propagated along the auditory pathways. While previous analyses of physiological data in cats suggested covariation of these two metrics, general characterization of their connection has never been performed. In the present study, we derive a rigorous relationship between VS and CI. To model phase-locking, we assume Poissonian spike trains with a temporally changing intensity function following a von Mises distribution. We demonstrate that VS and CI are mutually related via the so-called concentration parameter that determines the degree of phase-locking. We confirm that these theoretical results are largely consistent with physiological data recorded in the auditory brainstem of various animals. In addition, we generate artificial phase-locked spike sequences, for which recording and analysis parameters can be systematically manipulated. Our analysis results suggest that mismatches between empirical data and the theoretical prediction can often be explained with deviations from the von Mises distribution, including skewed or multimodal period histograms. Furthermore, temporal relations of spike trains across trials can contribute to higher CI values than predicted mathematically based on the VS. We find that, for most applications, a SAC bin width of 50 ms seems to be a favorable choice, leading to an estimated error below 2.5% for physiologically plausible conditions. Overall, our results provide general relations between the two measures of phase-locking and will aid future analyses of different physiological datasets that are characterized with these metrics.
Collapse
Affiliation(s)
- Dominik Kessler
- Computational Neuroscience, Department of Neuroscience, Faculty VI, University of Oldenburg, Oldenburg, Germany
| | - Catherine E Carr
- Department of Biology, University of Maryland, College Park, MD, United States
| | - Jutta Kretzberg
- Computational Neuroscience, Department of Neuroscience, Faculty VI, University of Oldenburg, Oldenburg, Germany.,Cluster of Excellence Hearing4all, Department of Neuroscience, Faculty VI, University of Oldenburg, Oldenburg, Germany
| | - Go Ashida
- Computational Neuroscience, Department of Neuroscience, Faculty VI, University of Oldenburg, Oldenburg, Germany.,Cluster of Excellence Hearing4all, Department of Neuroscience, Faculty VI, University of Oldenburg, Oldenburg, Germany
| |
Collapse
|
15
|
Suthakar K, Liberman MC. Auditory-nerve responses in mice with noise-induced cochlear synaptopathy. J Neurophysiol 2021; 126:2027-2038. [PMID: 34788179 DOI: 10.1152/jn.00342.2021] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Cochlear synaptopathy is the noise-induced or age-related loss of ribbon synapses between inner hair cells (IHCs) and auditory-nerve fibers (ANFs), first reported in CBA/CaJ mice. Recordings from single ANFs in anesthetized, noise-exposed guinea pigs suggested that neurons with low spontaneous rates (SRs) and high thresholds are more vulnerable than low-threshold, high-SR fibers. However, there is extensive postexposure regeneration of ANFs in guinea pigs but not in mice. Here, we exposed CBA/CaJ mice to octave-band noise and recorded sound-evoked and spontaneous activity from single ANFs at least 2 wk later. Confocal analysis of cochleae immunostained for pre- and postsynaptic markers confirmed the expected loss of 40%-50% of ANF synapses in the basal half of the cochlea; however, our data were not consistent with a selective loss of low-SR fibers. Rather they suggested a loss of both SR groups in synaptopathic regions. Single-fiber thresholds and frequency tuning recovered to pre-exposure levels; however, response to tone bursts showed increased peak and steady-state firing rates, as well as decreased jitter in first-spike latencies. This apparent gain-of-function increased the robustness of tone-burst responses in the presence of continuous masking noise. This study suggests that the nature of noise-induced synaptic damage varies between different species and that, in mouse, the noise-induced hyperexcitability seen in central auditory circuits is also observed at the level of the auditory nerve.NEW & NOTEWORTHY Noise-induced damage to synapses between inner hair cells and auditory-nerve fibers (ANFs) can occur without permanent hair cell damage, resulting in pathophysiology that "hides" behind normal thresholds. Prior single-fiber neurophysiology in guinea pig suggested that noise selectively targets high-threshold ANFs. Here, we show that the lingering pathophysiology differs in mouse, with both ANF groups affected and a paradoxical gain-of-function in surviving low-threshold fibers, including increased onset rate, decreased onset jitter, and reduced maskability.
Collapse
Affiliation(s)
- Kirupa Suthakar
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, Massachusetts.,Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts
| | - M Charles Liberman
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, Massachusetts.,Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
16
|
Liu J, Wang S, Lu Y, Wang H, Wang F, Qiu M, Xie Q, Han H, Hua Y. Aligned Organization of Synapses and Mitochondria in Auditory Hair Cells. Neurosci Bull 2021; 38:235-248. [PMID: 34837647 PMCID: PMC8975952 DOI: 10.1007/s12264-021-00801-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/25/2021] [Indexed: 10/19/2022] Open
Abstract
Recent studies have revealed great functional and structural heterogeneity in the ribbon-type synapses at the basolateral pole of the isopotential inner hair cell (IHC). This feature is believed to be critical for audition over a wide dynamic range, but whether the spatial gradient of ribbon morphology is fine-tuned in each IHC and how the mitochondrial network is organized to meet local energy demands of synaptic transmission remain unclear. By means of three-dimensional electron microscopy and artificial intelligence-based algorithms, we demonstrated the cell-wide structural quantification of ribbons and mitochondria in mature mid-cochlear IHCs of mice. We found that adjacent IHCs in staggered pairs differ substantially in cell body shape and ribbon morphology gradient as well as mitochondrial organization. Moreover, our analysis argues for a location-specific arrangement of correlated ribbon and mitochondrial function at the basolateral IHC pole.
Collapse
Affiliation(s)
- Jing Liu
- grid.9227.e0000000119573309National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190 China ,grid.410726.60000 0004 1797 8419School of Artificial Intelligence, School of Future Technology, University of Chinese Academy of Sciences, Beijing, 101408 China ,grid.507732.4CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai, 200031 China
| | - Shengxiong Wang
- grid.24516.340000000123704535Putuo People’s Hospital, Tongji University, Shanghai, 200060 China ,grid.16821.3c0000 0004 0368 8293Shanghai Institute of Precision Medicine, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125 China
| | - Yan Lu
- grid.16821.3c0000 0004 0368 8293Shanghai Institute of Precision Medicine, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125 China ,grid.412523.3Department of Otolaryngology–Head and Neck Surgery, Shanghai Ninth People’s Hospital, Shanghai, 200125 China ,grid.16821.3c0000 0004 0368 8293Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125 China ,grid.412987.10000 0004 0630 1330Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200125 China
| | - Haoyu Wang
- grid.16821.3c0000 0004 0368 8293Shanghai Institute of Precision Medicine, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125 China ,grid.412523.3Department of Otolaryngology–Head and Neck Surgery, Shanghai Ninth People’s Hospital, Shanghai, 200125 China ,grid.16821.3c0000 0004 0368 8293Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125 China ,grid.412987.10000 0004 0630 1330Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200125 China
| | - Fangfang Wang
- grid.16821.3c0000 0004 0368 8293Shanghai Institute of Precision Medicine, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125 China
| | - Miaoxin Qiu
- grid.24516.340000000123704535Putuo People’s Hospital, Tongji University, Shanghai, 200060 China
| | - Qiwei Xie
- grid.28703.3e0000 0000 9040 3743Research Base of Beijing Modern Manufacturing Development, Beijing University of Technology, Beijing, 100124 China
| | - Hua Han
- National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China. .,School of Artificial Intelligence, School of Future Technology, University of Chinese Academy of Sciences, Beijing, 101408, China. .,CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai, 200031, China.
| | - Yunfeng Hua
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China. .,Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai, 200125, China. .,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China. .,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200125, China.
| |
Collapse
|
17
|
Gonzalez-Velez V, Gil A, Castaneda-Villa N. Simulation of the Physiological Characteristics of Pillar and Modiolar Fibers of the Auditory Nerve. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2021; 2021:4196-4199. [PMID: 34892149 DOI: 10.1109/embc46164.2021.9630647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The study of the physiological characteristics of the auditory nerve fibers is fundamental to understand their capability to encode sounds. These characteristics include their spontaneous firing rate, their threshold, and their dynamic range. Although it is possible to perform in vitro recordings of these characteristics in different cell models, it is complicated to obtain in vivo measurements of them directly from the cochlea. For example, the apex of the cochlea since it is an unreachable region which is vulnerable to surgical trauma that could result in altered recordings. In this paper, the behavior of Pillar and Modiolar fibers of the auditory nerve were simulated in response to tone bursts of different frequencies and intensities. The proposed model allowed us to associate the basal firing rates with the physiological characteristics of the different auditory nerve fibers. This is especially important since some noise-associated hearing losses, such as acoustic trauma, have been explained as selective fiber damages.Clinical Relevance- Models that describe the properties of auditory nerve fibers are important to study specific aspects of maturation as well as the causes of sensorineural hearing loss in humans.
Collapse
|
18
|
Mackey C, Tarabillo A, Ramachandran R. Three psychophysical metrics of auditory temporal integration in macaques. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2021; 150:3176. [PMID: 34717465 PMCID: PMC8556002 DOI: 10.1121/10.0006658] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
The relationship between sound duration and detection threshold has long been thought to reflect temporal integration. Reports of species differences in this relationship are equivocal: some meta-analyses report no species differences, whereas others report substantial differences, particularly between humans and their close phylogenetic relatives, macaques. This renders translational work in macaques problematic. To reevaluate this difference, tone detection performance was measured in macaques using a go/no-go reaction time (RT) task at various tone durations and in the presence of broadband noise (BBN). Detection thresholds, RTs, and the dynamic range (DR) of the psychometric function decreased as the tone duration increased. The threshold by duration trends suggest macaques integrate at a similar rate to humans. The RT trends also resemble human data and are the first reported in animals. Whereas the BBN did not affect how the threshold or RT changed with the duration, it substantially reduced the DR at short durations. A probabilistic Poisson model replicated the effects of duration on threshold and DR and required integration from multiple simulated auditory nerve fibers to explain the performance at shorter durations. These data suggest that, contrary to previous studies, macaques are uniquely well-suited to model human temporal integration and form the baseline for future neurophysiological studies.
Collapse
Affiliation(s)
- Chase Mackey
- Neuroscience Graduate Program, Vanderbilt University, Nashville, Tennessee 37240, USA
| | - Alejandro Tarabillo
- Department of Hearing and Speech Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - Ramnarayan Ramachandran
- Department of Hearing and Speech Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW We review recent progress in the characterization of spiral ganglion neurons (SGNs), the afferent neurons that transmit sound information from mechanosensory hair cells in the inner ear to the central nervous system. RECENT FINDINGS Single-cell ribonucleic acid sequencing studies of murine SGNs have demonstrated that SGNs consist of molecularly distinct subtypes. The molecularly defined SGN subtypes likely correspond to SGN subtypes previously identified on the basis of physiological properties, although this has not been experimentally demonstrated. Subtype maturation is completed postnatally in an activity-dependent manner and is impaired in several models of hearing loss. SUMMARY The recent molecular studies open new avenues to rigorously test whether SGN subtypes are important for the encoding of different sound features and if they show differential vulnerability to genetic factors and environmental insults. This could have important implications for the development of therapeutic strategies to treat hearing loss.
Collapse
Affiliation(s)
- Shuohao Sun
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | |
Collapse
|
20
|
Curthoys IS, Grant JW, Pastras CJ, Fröhlich L, Brown DJ. Similarities and Differences Between Vestibular and Cochlear Systems - A Review of Clinical and Physiological Evidence. Front Neurosci 2021; 15:695179. [PMID: 34456671 PMCID: PMC8397526 DOI: 10.3389/fnins.2021.695179] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/12/2021] [Indexed: 12/04/2022] Open
Abstract
The evoked response to repeated brief stimuli, such as clicks or short tone bursts, is used for clinical evaluation of the function of both the auditory and vestibular systems. One auditory response is a neural potential - the Auditory Brainstem Response (ABR) - recorded by surface electrodes on the head. The clinical analogue for testing the otolithic response to abrupt sounds and vibration is the myogenic potential recorded from tensed muscles - the vestibular evoked myogenic potential (VEMP). VEMPs have provided clinicians with a long sought-after tool - a simple, clinically realistic indicator of the function of each of the 4 otolithic sensory regions. We review the basic neural evidence for VEMPs and discuss the similarities and differences between otolithic and cochlear receptors and afferents. VEMPs are probably initiated by sound or vibration selectively activating afferent neurons with irregular resting discharge originating from the unique type I receptors at a specialized region of the otolithic maculae (the striola). We review how changes in VEMP responses indicate the functional state of peripheral vestibular function and the likely transduction mechanisms allowing otolithic receptors and afferents to trigger such very short latency responses. In section "ELECTROPHYSIOLOGY" we show how cochlear and vestibular receptors and afferents have many similar electrophysiological characteristics [e.g., both generate microphonics, summating potentials, and compound action potentials (the vestibular evoked potential, VsEP)]. Recent electrophysiological evidence shows that the hydrodynamic changes in the labyrinth caused by increased fluid volume (endolymphatic hydrops), change the responses of utricular receptors and afferents in a way which mimics the changes in vestibular function attributed to endolymphatic hydrops in human patients. In section "MECHANICS OF OTOLITHS IN VEMPS TESTING" we show how the major VEMP results (latency and frequency response) follow from modeling the physical characteristics of the macula (dimensions, stiffness etc.). In particular, the structure and mechanical operation of the utricular macula explains the very fast response of the type I receptors and irregular afferents which is the very basis of VEMPs and these structural changes of the macula in Menière's Disease (MD) predict the upward shift of VEMP tuning in these patients.
Collapse
Affiliation(s)
- Ian S. Curthoys
- Vestibular Research Laboratory, School of Psychology, The University of Sydney, Sydney, NSW, Australia
| | - John Wally Grant
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, United States
| | - Christopher J. Pastras
- The Menière’s Research Laboratory, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Laura Fröhlich
- Department of Otorhinolaryngology, Head and Neck Surgery, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Daniel J. Brown
- School of Pharmacy and Biomedical Sciences, Curtin University, Bentley, WA, Australia
| |
Collapse
|
21
|
Peterson AJ, Heil P. A simplified physiological model of rate-level functions of auditory-nerve fibers. Hear Res 2021; 406:108258. [PMID: 34010767 DOI: 10.1016/j.heares.2021.108258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/09/2021] [Accepted: 04/23/2021] [Indexed: 12/24/2022]
Abstract
Several approaches have been used to describe the rate-level functions of auditory-nerve fibers (ANFs). One approach uses descriptive models that can be fitted easily to data. Another derives rate-level functions from comprehensive physiological models of auditory peripheral processing. Here, we seek to identify the minimal set of components needed to provide a physiologically plausible account of rate-level functions. Our model consists of a first-order Boltzmann mechanoelectrical transducer function relating the instantaneous stimulus pressure to an instantaneous output, followed by a lowpass filter that eliminates the AC component, followed by an exponential synaptic transfer function relating the DC component to the mean spike rate. This is perhaps the simplest physiologically plausible model capable of accounting for rate-level functions under the assumption that the model parameters for a given ANF and stimulus frequency are level-independent. We find that the model typically accounts well for rate-level functions from cat ANFs for all stimulus frequencies. More complicated model variants having saturating synaptic transfer functions do not perform significantly better, implying the system operates far away from synaptic saturation. Rate saturation in the model is caused by saturation of the DC component of the filter output (e.g., the receptor potential), which in turn is due to the saturation of the transducer function. The maximum mean spike rate is approximately constant across ANFs, such that the slope parameter of the exponential synaptic transfer function decreases with increasing spontaneous rate. If the synaptic parameters for a given ANF are assumed to be constant across stimulus frequencies, then frequency- and level-dependent input nonlinearities are derived that are qualitatively similar to those reported in the literature. Contrary to assumptions in the literature, such nonlinearities are obtained even for ANFs having high spontaneous rates. Finally, spike-rate adaptation is examined and found to be accounted for by a decrease in the slope parameter of the synaptic transfer function over time following stimulus onset.
Collapse
Affiliation(s)
- Adam J Peterson
- Department of Systems Physiology of Learning, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Peter Heil
- Department of Systems Physiology of Learning, Leibniz Institute for Neurobiology, Magdeburg, Germany; Center for Behavioral Brain Sciences, Magdeburg, Germany.
| |
Collapse
|
22
|
Bhardwaj A, Khyam MO, Müller R. Biomimetic detection of dynamic signatures in foliage echoes. BIOINSPIRATION & BIOMIMETICS 2021; 16:046026. [PMID: 33862609 DOI: 10.1088/1748-3190/abf910] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 04/16/2021] [Indexed: 06/12/2023]
Abstract
Certain bat species (familyRhinolophidae) dynamically deform their emission baffles (noseleaves) and reception baffles (pinnae) during echolocation. Prior research using numerical models, laboratory characterizations, and experiments with simple targets have suggested that this dynamics may manifest itself in time-variant echo signatures. Since the pronounced random nature of echoes from natural targets such as foliage has not been reflected in these experiments, we have collected a large number (>55 000) of foliage echoes outdoors with a sonar head that mimics the dynamic periphery in bats. The echo data was processed with a custom auditory processing model to create spike-based echo representations. Deep-learning classifiers were able to estimate the dynamic state of the periphery, i.e., static or dynamic, based on single echoes with accuracies of up to 80%. This suggests that the effects of the peripheral dynamics are present in the bat brains and could hence be used by the animals. The best classification performances were obtained for data obtained within a spatially confined area. Hence, if the bat brains suffer from the same generalization issues, they would have to have a way to adapt their neural echo processing to such local fluctuations to exploit the dynamic effects successfully.
Collapse
Affiliation(s)
- Ananya Bhardwaj
- Department of Mechanical Engineering, Virginia Tech, Blacksburg 24060, United States of America
| | - M Omar Khyam
- Department of Mechanical Engineering, Virginia Tech, Blacksburg 24060, United States of America
| | - Rolf Müller
- Department of Mechanical Engineering, Virginia Tech, Blacksburg 24060, United States of America
| |
Collapse
|
23
|
Rutherford MA, von Gersdorff H, Goutman JD. Encoding sound in the cochlea: from receptor potential to afferent discharge. J Physiol 2021; 599:2527-2557. [PMID: 33644871 PMCID: PMC8127127 DOI: 10.1113/jp279189] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 02/22/2021] [Indexed: 12/17/2022] Open
Abstract
Ribbon-class synapses in the ear achieve analog to digital transformation of a continuously graded membrane potential to all-or-none spikes. In mammals, several auditory nerve fibres (ANFs) carry information from each inner hair cell (IHC) to the brain in parallel. Heterogeneity of transmission among synapses contributes to the diversity of ANF sound-response properties. In addition to the place code for sound frequency and the rate code for sound level, there is also a temporal code. In series with cochlear amplification and frequency tuning, neural representation of temporal cues over a broad range of sound levels enables auditory comprehension in noisy multi-speaker settings. The IHC membrane time constant introduces a low-pass filter that attenuates fluctuations of the receptor potential above 1-2 kHz. The ANF spike generator adds a high-pass filter via its depolarization-rate threshold that rejects slow changes in the postsynaptic potential and its phasic response property that ensures one spike per depolarization. Synaptic transmission involves several stochastic subcellular processes between IHC depolarization and ANF spike generation, introducing delay and jitter that limits the speed and precision of spike timing. ANFs spike at a preferred phase of periodic sounds in a process called phase-locking that is limited to frequencies below a few kilohertz by both the IHC receptor potential and the jitter in synaptic transmission. During phase-locking to periodic sounds of increasing intensity, faster and facilitated activation of synaptic transmission and spike generation may be offset by presynaptic depletion of synaptic vesicles, resulting in relatively small changes in response phase. Here we review encoding of spike-timing at cochlear ribbon synapses.
Collapse
Affiliation(s)
- Mark A. Rutherford
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Henrique von Gersdorff
- Vollum Institute, Oregon Hearing Research Center, Oregon Health and Sciences University, Portland, Oregon 97239
| | | |
Collapse
|
24
|
Huet AT, Dombrowski T, Rankovic V, Thirumalai A, Moser T. Developing Fast, Red-Light Optogenetic Stimulation of Spiral Ganglion Neurons for Future Optical Cochlear Implants. Front Mol Neurosci 2021; 14:635897. [PMID: 33776648 PMCID: PMC7991399 DOI: 10.3389/fnmol.2021.635897] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/26/2021] [Indexed: 01/19/2023] Open
Abstract
Optogenetic stimulation of type I spiral ganglion neurons (SGNs) promises an alternative to the electrical stimulation by current cochlear implants (CIs) for improved hearing restoration by future optical CIs (oCIs). Most of the efforts in using optogenetic stimulation in the cochlea so far used early postnatal injection of viral vectors carrying blue-light activated channelrhodopsins (ChRs) into the cochlea of mice. However, preparing clinical translation of the oCI requires (i) reliable and safe transduction of mature SGNs of further species and (ii) use of long-wavelength light to avoid phototoxicity. Here, we employed a fast variant of the red-light activated channelrhodopsin Chrimson (f-Chrimson) and different AAV variants to implement optogenetic SGN stimulation in Mongolian gerbils. We compared early postnatal (p8) and adult (>8 weeks) AAV administration, employing different protocols for injection of AAV-PHP.B and AAV2/6 into the adult cochlea. Success of the optogenetic manipulation was analyzed by optically evoked auditory brainstem response (oABR) and immunohistochemistry of mid-modiolar cryosections of the cochlea. In order to most efficiently evaluate the immunohistochemical results a semi-automatic procedure to identify transduced cells in confocal images was developed. Our results indicate that the rate of SGN transduction is significantly lower for AAV administration into the adult cochlea compared to early postnatal injection. SGN transduction upon AAV administration into the adult cochlea was largely independent of the chosen viral vector and injection approach. The higher the rate of SGN transduction, the lower were oABR thresholds and the larger were oABR amplitudes. Our results highlight the need to optimize viral vectors and virus administration for efficient optogenetic manipulation of SGNs in the adult cochlea for successful clinical translation of SGN-targeting gene therapy and of the oCI.
Collapse
Affiliation(s)
- Antoine Tarquin Huet
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience and Optogenetics Laboratory, German Primate Center, Göttingen, Germany
| | - Tobias Dombrowski
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Department of Otolaryngology, Head and Neck Surgery, St. Elisabeth Hospital, Ruhr University Bochum, Bochum, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
| | - Vladan Rankovic
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience and Optogenetics Laboratory, German Primate Center, Göttingen, Germany
- Restorative Cochlear Genomics Group, Auditory Neuroscience and Optogenetics Laboratory, German Primate Center, Göttingen, Germany
| | - Anupriya Thirumalai
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Göttingen Graduate School for Neurosciences and Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience and Optogenetics Laboratory, German Primate Center, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
- Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
25
|
Gómez-Casati ME, Goutman JD. Divide and conquer acoustic diversity. EMBO J 2021; 40:e107531. [PMID: 33555064 DOI: 10.15252/embj.2020107531] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 01/07/2021] [Indexed: 12/12/2022] Open
Abstract
Humans can recognize differences in sound intensity of up to 6 orders of magnitude. However, it is not clear how this is achieved and what enables our auditory systems to encode such a gradient. Özçete & Moser (2021) report in this issue that the key to this lies in the synaptic heterogeneity within individual sensory cells in the inner ear.
Collapse
Affiliation(s)
- Maria E Gómez-Casati
- Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, CA Buenos Aires, Argentina
| | - Juan D Goutman
- Instituto de Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (CONICET), CA Buenos Aires, Argentina
| |
Collapse
|
26
|
Salicylate decreases the spontaneous firing rate of guinea pig auditory nerve fibres. Neurosci Lett 2021; 747:135705. [PMID: 33548408 PMCID: PMC7957321 DOI: 10.1016/j.neulet.2021.135705] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/26/2021] [Accepted: 01/29/2021] [Indexed: 02/03/2023]
Abstract
Spontaneous firing rates were recorded from single auditory fibres in vivo. Salicylate was injected at 350 mg/kg by the subcutaneous route. Median firing rate decreased by 32 spikes/s in nerve fibres after salicylate injection. The high spontaneous rate fibres (type 1A) showed the main reduction.
Tinnitus has similarities to chronic neuropathic pain where there are changes in the firing rate of different types of afferent neurons. We postulated that one possible cause of tinnitus is a change in the distribution of spontaneous firing rates in at least one type of afferent auditory nerve fibre in anaesthetised guinea pigs. In control animals there was a bimodal distribution of spontaneous rates, but the position of the second mode was different depending upon whether the fibres responded best to high (> 4 kHz) or low (≤4 kHz) frequency tonal stimulation. The simplest and most reliable way of inducing tinnitus in experimental animals is to administer a high dose of sodium salicylate. The distribution of the spontaneous firing rates was different when salicylate (350 mg/kg) was administered, even when the sample was matched for the distribution of characteristic frequencies in the control population. The proportion of medium spontaneous rate fibres (MSR, 1≤ spikes/s ≤20) increased while the proportion of the highest, high spontaneous firing rate fibres (HSR, > 80 spikes/s) decreased following salicylate. The median rate fell from 64.7 spikes/s (control) to 35.4 spikes/s (salicylate); a highly significant change (Kruskal-Wallis test p < 0.001). When the changes were compared with various models of statistical probability, the most accurate model was one where most HSR fibres decreased their firing rate by 32 spikes/s. Thus, we have shown a reduction in the firing rate of HSR fibres that may be related to tinnitus.
Collapse
|
27
|
Ming C, Haro S, Simmons AM, Simmons JA. A comprehensive computational model of animal biosonar signal processing. PLoS Comput Biol 2021; 17:e1008677. [PMID: 33596199 PMCID: PMC7888678 DOI: 10.1371/journal.pcbi.1008677] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 01/07/2021] [Indexed: 11/23/2022] Open
Abstract
Computational models of animal biosonar seek to identify critical aspects of echo processing responsible for the superior, real-time performance of echolocating bats and dolphins in target tracking and clutter rejection. The Spectrogram Correlation and Transformation (SCAT) model replicates aspects of biosonar imaging in both species by processing wideband biosonar sounds and echoes with auditory mechanisms identified from experiments with bats. The model acquires broadband biosonar broadcasts and echoes, represents them as time-frequency spectrograms using parallel bandpass filters, translates the filtered signals into ten parallel amplitude threshold levels, and then operates on the resulting time-of-occurrence values at each frequency to estimate overall echo range delay. It uses the structure of the echo spectrum by depicting it as a series of local frequency nulls arranged regularly along the frequency axis of the spectrograms after dechirping them relative to the broadcast. Computations take place entirely on the timing of threshold-crossing events for each echo relative to threshold-events for the broadcast. Threshold-crossing times take into account amplitude-latency trading, a physiological feature absent from conventional digital signal processing. Amplitude-latency trading transposes the profile of amplitudes across frequencies into a profile of time-registrations across frequencies. Target shape is extracted from the spacing of the object's individual acoustic reflecting points, or glints, using the mutual interference pattern of peaks and nulls in the echo spectrum. These are merged with the overall range-delay estimate to produce a delay-based reconstruction of the object's distance as well as its glints. Clutter echoes indiscriminately activate multiple parts in the null-detecting system, which then produces the equivalent glint-delay spacings in images, thus blurring the overall echo-delay estimates by adding spurious glint delays to the image. Blurring acts as an anticorrelation process that rejects clutter intrusion into perceptions.
Collapse
Affiliation(s)
- Chen Ming
- Department of Neuroscience and Carney Institute for Brain Science, Brown University Providence, United States of America
| | - Stephanie Haro
- Speech and Hearing Biosciences and Technology, Harvard University, Boston, United States of America
| | - Andrea Megela Simmons
- Department of Cognitive, Linguistic and Psychological Sciences and Carney Institute for Brain Science, Brown University Providence, United States of America
| | - James A. Simmons
- Department of Neuroscience and Carney Institute for Brain Science, Brown University Providence, United States of America
| |
Collapse
|
28
|
Parida S, Bharadwaj H, Heinz MG. Spectrally specific temporal analyses of spike-train responses to complex sounds: A unifying framework. PLoS Comput Biol 2021; 17:e1008155. [PMID: 33617548 PMCID: PMC7932515 DOI: 10.1371/journal.pcbi.1008155] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 03/04/2021] [Accepted: 02/04/2021] [Indexed: 11/24/2022] Open
Abstract
Significant scientific and translational questions remain in auditory neuroscience surrounding the neural correlates of perception. Relating perceptual and neural data collected from humans can be useful; however, human-based neural data are typically limited to evoked far-field responses, which lack anatomical and physiological specificity. Laboratory-controlled preclinical animal models offer the advantage of comparing single-unit and evoked responses from the same animals. This ability provides opportunities to develop invaluable insight into proper interpretations of evoked responses, which benefits both basic-science studies of neural mechanisms and translational applications, e.g., diagnostic development. However, these comparisons have been limited by a disconnect between the types of spectrotemporal analyses used with single-unit spike trains and evoked responses, which results because these response types are fundamentally different (point-process versus continuous-valued signals) even though the responses themselves are related. Here, we describe a unifying framework to study temporal coding of complex sounds that allows spike-train and evoked-response data to be analyzed and compared using the same advanced signal-processing techniques. The framework uses a set of peristimulus-time histograms computed from single-unit spike trains in response to polarity-alternating stimuli to allow advanced spectral analyses of both slow (envelope) and rapid (temporal fine structure) response components. Demonstrated benefits include: (1) novel spectrally specific temporal-coding measures that are less confounded by distortions due to hair-cell transduction, synaptic rectification, and neural stochasticity compared to previous metrics, e.g., the correlogram peak-height, (2) spectrally specific analyses of spike-train modulation coding (magnitude and phase), which can be directly compared to modern perceptually based models of speech intelligibility (e.g., that depend on modulation filter banks), and (3) superior spectral resolution in analyzing the neural representation of nonstationary sounds, such as speech and music. This unifying framework significantly expands the potential of preclinical animal models to advance our understanding of the physiological correlates of perceptual deficits in real-world listening following sensorineural hearing loss.
Collapse
Affiliation(s)
- Satyabrata Parida
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
| | - Hari Bharadwaj
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
- Department of Speech, Language, and Hearing Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - Michael G. Heinz
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
- Department of Speech, Language, and Hearing Sciences, Purdue University, West Lafayette, Indiana, United States of America
| |
Collapse
|
29
|
Hua Y, Ding X, Wang H, Wang F, Lu Y, Neef J, Gao Y, Moser T, Wu H. Electron Microscopic Reconstruction of Neural Circuitry in the Cochlea. Cell Rep 2021; 34:108551. [PMID: 33406431 DOI: 10.1016/j.celrep.2020.108551] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/25/2020] [Accepted: 12/03/2020] [Indexed: 02/02/2023] Open
Abstract
Recent studies reveal great diversity in the structure, function, and efferent innervation of afferent synaptic connections between the cochlear inner hair cells (IHCs) and spiral ganglion neurons (SGNs), which likely enables audition to process a wide range of sound pressures. By performing an extensive electron microscopic (EM) reconstruction of the neural circuitry in the mature mouse organ of Corti, we demonstrate that afferent SGN dendrites differ in abundance and composition of efferent innervation in a manner dependent on their afferent synaptic connectivity with IHCs. SGNs that sample glutamate release from several presynaptic ribbons receive more efferent innervation from lateral olivocochlear projections than those driven by a single ribbon. Next to the prevailing unbranched SGN dendrites, we found branched SGN dendrites that can contact several ribbons of 1-2 IHCs. Unexpectedly, medial olivocochlear neurons provide efferent innervation of SGN dendrites, preferring those forming single-ribbon, pillar-side synapses. We propose a fine-tuning of afferent and efferent SGN innervation.
Collapse
Affiliation(s)
- Yunfeng Hua
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai, China; Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China; Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Connectomics, Max Planck Institute for Brain Research, Frankfurt/Main, Germany.
| | - Xu Ding
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai, China; Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Haoyu Wang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangfang Wang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Lu
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jakob Neef
- Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany; Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Yunge Gao
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai, China; Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Tobias Moser
- Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany; Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany; Multiscale Bioimaging Cluster of Excellence, University of Göttingen, Göttingen, Germany.
| | - Hao Wu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai, China; Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China; Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
30
|
Özçete ÖD, Moser T. A sensory cell diversifies its output by varying Ca 2+ influx-release coupling among active zones. EMBO J 2020; 40:e106010. [PMID: 33346936 PMCID: PMC7917556 DOI: 10.15252/embj.2020106010] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 02/06/2023] Open
Abstract
The cochlea encodes sound pressures varying over six orders of magnitude by collective operation of functionally diverse spiral ganglion neurons (SGNs). The mechanisms enabling this functional diversity remain elusive. Here, we asked whether the sound intensity information, contained in the receptor potential of the presynaptic inner hair cell (IHC), is fractionated via heterogeneous synapses. We studied the transfer function of individual IHC synapses by combining patch‐clamp recordings with dual‐color Rhod‐FF and iGluSnFR imaging of presynaptic Ca2+ signals and glutamate release. Synapses differed in the voltage dependence of release: Those residing at the IHC' pillar side activated at more hyperpolarized potentials and typically showed tight control of release by few Ca2+ channels. We conclude that heterogeneity of voltage dependence and release site coupling of Ca2+ channels among the synapses varies synaptic transfer within individual IHCs and, thereby, likely contributes to the functional diversity of SGNs. The mechanism reported here might serve sensory cells and neurons more generally to diversify signaling even in close‐by synapses.
Collapse
Affiliation(s)
- Özge D Özçete
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Collaborative Research Center 889, University of Göttingen, Göttingen, Germany.,Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Göttingen Graduate Center for Neurosciences, Biophysics and Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Collaborative Research Center 889, University of Göttingen, Göttingen, Germany.,Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Göttingen Graduate Center for Neurosciences, Biophysics and Molecular Biosciences, University of Göttingen, Göttingen, Germany.,Synaptic Nanophysiology Group, Max Planck Institute of Biophysical Chemistry, Göttingen, Germany.,Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
31
|
McClaskey CM, Panganiban CH, Noble KV, Dias JW, Lang H, Harris KC. A multi-metric approach to characterizing mouse peripheral auditory nerve function using the auditory brainstem response. J Neurosci Methods 2020; 346:108937. [PMID: 32910925 PMCID: PMC7957964 DOI: 10.1016/j.jneumeth.2020.108937] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 09/03/2020] [Accepted: 09/03/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND The auditory brainstem response (ABR), specifically wave I, is widely used to noninvasively measure auditory nerve (AN) function. Recent work in humans has introduced novel electrocochleographic measures to comprehensively characterize AN function that emphasize suprathreshold processing and estimate neural synchrony. NEW METHOD This study establishes new tools for evaluating AN function in vivo in adult mice using tone-evoked ABRs obtained from young-adult CBA/CaJ mice, adapting the approach previously introduced in humans. Six metrics are obtained from ABR wave I at suprathreshold stimulus levels. RESULTS Change-point analyses show that the metrics' rate of change with stimulus level differs between moderate and high suprathreshold levels, suggesting that this approach can potentially characterize the presence of heterogeneous AN fiber types. COMPARISON WITH EXISTING METHODS Traditional ABR approaches focus on response thresholds and averaged amplitudes/latencies. In contrast, our multi-metric approach, which uses single-trial data and suprathreshold stimuli, provides novel information and identifies evidence of neural synchrony deficits and changes in the heterogeneity of AN fibers underlying AN behavior. CONCLUSION The techniques reported here provide a novel tool to assess changes in AN function in vivo in a commonly used animal model. A benchmark of most current hearing research is the transition from animal to human studies. Here we established a translational objective approach, applying methods that were first developed in humans to animals. This approach enables researchers to identify changes in AN function arising from the animal models with well-characterized pathology, and predict similar pathological changes in human AN dysfunction and hearing loss.
Collapse
Affiliation(s)
- Carolyn M McClaskey
- Department of Otolaryngology, Head & Neck Surgery, Medical University of South Carolina, 135 Rutledge Ave, MSC 550, Charleston, SC, 29425, United States.
| | - Clarisse H Panganiban
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 171 Ashley Avenue, MSC 908, Charleston, SC, 29425, United States.
| | - Kenyaria V Noble
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 171 Ashley Avenue, MSC 908, Charleston, SC, 29425, United States.
| | - James W Dias
- Department of Otolaryngology, Head & Neck Surgery, Medical University of South Carolina, 135 Rutledge Ave, MSC 550, Charleston, SC, 29425, United States.
| | - Hainan Lang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 171 Ashley Avenue, MSC 908, Charleston, SC, 29425, United States.
| | - Kelly C Harris
- Department of Otolaryngology, Head & Neck Surgery, Medical University of South Carolina, 135 Rutledge Ave, MSC 550, Charleston, SC, 29425, United States.
| |
Collapse
|
32
|
Nesse WH, Maler L, Longtin A. Enhanced Signal Detection by Adaptive Decorrelation of Interspike Intervals. Neural Comput 2020; 33:341-375. [PMID: 33253034 DOI: 10.1162/neco_a_01347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Spike trains with negative interspike interval (ISI) correlations, in which long/short ISIs are more likely followed by short/long ISIs, are common in many neurons. They can be described by stochastic models with a spike-triggered adaptation variable. We analyze a phenomenon in these models where such statistically dependent ISI sequences arise in tandem with quasi-statistically independent and identically distributed (quasi-IID) adaptation variable sequences. The sequences of adaptation states and resulting ISIs are linked by a nonlinear decorrelating transformation. We establish general conditions on a family of stochastic spiking models that guarantee this quasi-IID property and establish bounds on the resulting baseline ISI correlations. Inputs that elicit weak firing rate changes in samples with many spikes are known to be more detectible when negative ISI correlations are present because they reduce spike count variance; this defines a variance-reduced firing rate coding benchmark. We performed a Fisher information analysis on these adapting models exhibiting ISI correlations to show that a spike pattern code based on the quasi-IID property achieves the upper bound of detection performance, surpassing rate codes with the same mean rate-including the variance-reduced rate code benchmark-by 20% to 30%. The information loss in rate codes arises because the benefits of reduced spike count variance cannot compensate for the lower firing rate gain due to adaptation. Since adaptation states have similar dynamics to synaptic responses, the quasi-IID decorrelation transformation of the spike train is plausibly implemented by downstream neurons through matched postsynaptic kinetics. This provides an explanation for observed coding performance in sensory systems that cannot be accounted for by rate coding, for example, at the detection threshold where rate changes can be insignificant.
Collapse
Affiliation(s)
- William H Nesse
- Department of Mathematics, University of Utah, Salt Lake City, UT 84112, U.S.A.
| | - Leonard Maler
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - André Longtin
- Department of Physics, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
33
|
Gault R, McGinnity TM, Coleman S. Perceptual Modeling of Tinnitus Pitch and Loudness. IEEE Trans Cogn Dev Syst 2020. [DOI: 10.1109/tcds.2020.2964841] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
34
|
Phase Locking of Auditory Nerve Fibers: The Role of Lowpass Filtering by Hair Cells. J Neurosci 2020; 40:4700-4714. [PMID: 32376778 DOI: 10.1523/jneurosci.2269-19.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 02/13/2020] [Accepted: 04/22/2020] [Indexed: 11/21/2022] Open
Abstract
Phase locking of auditory-nerve-fiber (ANF) responses to the temporal fine structure of acoustic stimuli, a hallmark of the auditory system's temporal precision, is important for many aspects of hearing. Previous work has shown that phase-locked period histograms are often well described by exponential transfer functions relating instantaneous stimulus pressure to instantaneous spike rate, with no observed clipping of the histograms. The operating points and slopes of these functions change with stimulus level. The mechanism underlying this apparent gain control is unclear but is distinct from mechanical compression, is independent of refractoriness and spike-rate adaptation, and is apparently instantaneous. Here we show that these findings can be accounted for by a model consisting of a static Boltzmann transducer function yielding a clipped output, followed by a lowpass filter and a static exponential transfer function. Using responses to tones of ANFs from cats of both sexes, we show that, for a given ANF, the period histograms obtained at all stimulus levels for a given stimulus frequency can be described using one set of level-independent model parameters. The model also accounts for changes in the maximum and minimum instantaneous spike rates with changes in stimulus level. Notably, the estimated cutoff frequency is lower for low- than for high-spontaneous-rate ANFs, implying a synapse-specific contribution to lowpass filtering. These findings advance our understanding of ANF phase locking by highlighting the role of peripheral filtering mechanisms in shaping responses of individual ANFs.SIGNIFICANCE STATEMENT Phase locking of auditory-nerve-fiber responses to the temporal fine structure of acoustic stimuli is important for many aspects of hearing. Period histograms typically retain an approximately sinusoidal shape across stimulus levels, with the peripheral auditory system operating as though its overall transfer function is an exponential function whose slope decreases with increasing stimulus level. This apparent gain control can be accounted for by a static saturating transducer function followed by a lowpass filter. In addition to attenuating the AC component, the filter approximately recovers the sinusoidal waveform of the stimulus. The estimated cutoff frequency varies with spontaneous rate, revealing a synaptic contribution to lowpass filtering. These findings highlight the significant impact of peripheral filtering mechanisms on phase locking.
Collapse
|
35
|
Dieter A, Keppeler D, Moser T. Towards the optical cochlear implant: optogenetic approaches for hearing restoration. EMBO Mol Med 2020; 12:e11618. [PMID: 32227585 PMCID: PMC7136966 DOI: 10.15252/emmm.201911618] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/08/2020] [Accepted: 01/28/2020] [Indexed: 12/30/2022] Open
Abstract
Cochlear implants (CIs) are considered the most successful neuroprosthesis as they enable speech comprehension in the majority of half a million CI users suffering from sensorineural hearing loss. By electrically stimulating the auditory nerve, CIs constitute an interface re-connecting the brain and the auditory scene, providing the patient with information regarding the latter. However, since electric current is hard to focus in conductive environments such as the cochlea, the precision of electrical sound encoding-and thus quality of artificial hearing-is limited. Recently, optogenetic stimulation of the cochlea has been suggested as an alternative approach for hearing restoration. Cochlear optogenetics promises increased spectral selectivity of artificial sound encoding, hence improved hearing, as light can conveniently be confined in space to activate the auditory nerve within smaller tonotopic ranges. In this review, we discuss the latest experimental and technological developments of cochlear optogenetics and outline the remaining challenges on the way to clinical translation.
Collapse
Affiliation(s)
- Alexander Dieter
- Institute for Auditory Neuroscience and InnerEarLabUniversity Medical Center GöttingenGöttingenGermany
- Göttingen Graduate School for NeurosciencesBiophysics and Molecular BiosciencesUniversity of GöttingenGöttingenGermany
| | - Daniel Keppeler
- Institute for Auditory Neuroscience and InnerEarLabUniversity Medical Center GöttingenGöttingenGermany
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLabUniversity Medical Center GöttingenGöttingenGermany
- Auditory Neuroscience and Optogenetics LaboratoryGerman Primate CenterGöttingenGermany
- Auditory Neuroscience GroupMax Planck Institute of Experimental MedicineGöttingenGermany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC)University of GöttingenGöttingenGermany
| |
Collapse
|
36
|
Ross JM, Hamm JP. Cortical Microcircuit Mechanisms of Mismatch Negativity and Its Underlying Subcomponents. Front Neural Circuits 2020; 14:13. [PMID: 32296311 PMCID: PMC7137737 DOI: 10.3389/fncir.2020.00013] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 03/17/2020] [Indexed: 12/11/2022] Open
Abstract
In the neocortex, neuronal processing of sensory events is significantly influenced by context. For instance, responses in sensory cortices are suppressed to repetitive or redundant stimuli, a phenomenon termed “stimulus-specific adaptation” (SSA). However, in a context in which that same stimulus is novel, or deviates from expectations, neuronal responses are augmented. This augmentation is termed “deviance detection” (DD). This contextual modulation of neural responses is fundamental for how the brain efficiently processes the sensory world to guide immediate and future behaviors. Notably, context modulation is deficient in some neuropsychiatric disorders such as schizophrenia (SZ), as quantified by reduced “mismatch negativity” (MMN), an electroencephalography waveform reflecting a combination of SSA and DD in sensory cortex. Although the role of NMDA-receptor function and other neuromodulatory systems on MMN is established, the precise microcircuit mechanisms of MMN and its underlying components, SSA and DD, remain unknown. When coupled with animal models, the development of powerful precision neurotechnologies over the past decade carries significant promise for making new progress into understanding the neurobiology of MMN with previously unreachable spatial resolution. Currently, rodent models represent the best tool for mechanistic study due to the vast genetic tools available. While quantifying human-like MMN waveforms in rodents is not straightforward, the “oddball” paradigms used to study it in humans and its underlying subcomponents (SSA/DD) are highly translatable across species. Here we summarize efforts published so far, with a focus on cortically measured SSA and DD in animals to maintain relevance to the classically measured MMN, which has cortical origins. While mechanistic studies that measure and contrast both components are sparse, we synthesize a potential set of microcircuit mechanisms from the existing rodent, primate, and human literature. While MMN and its subcomponents likely reflect several mechanisms across multiple brain regions, understanding fundamental microcircuit mechanisms is an important step to understand MMN as a whole. We hypothesize that SSA reflects adaptations occurring at synapses along the sensory-thalamocortical pathways, while DD depends on both SSA inherited from afferent inputs and resulting disinhibition of non-adapted neurons arising from the distinct physiology and wiring properties of local interneuronal subpopulations and NMDA-receptor function.
Collapse
Affiliation(s)
- Jordan M Ross
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States.,Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, United States
| | - Jordan P Hamm
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States.,Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, United States.,Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
37
|
Reijntjes DOJ, Köppl C, Pyott SJ. Volume gradients in inner hair cell-auditory nerve fiber pre- and postsynaptic proteins differ across mouse strains. Hear Res 2020; 390:107933. [PMID: 32203820 DOI: 10.1016/j.heares.2020.107933] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 02/12/2020] [Accepted: 02/26/2020] [Indexed: 02/04/2023]
Abstract
In different animal models, auditory nerve fibers display variation in spontaneous activity and response threshold. Functional and structural differences among inner hair cell ribbon synapses are believed to contribute to this variation. The relative volumes of synaptic proteins at individual synapses might be one such difference. This idea is based on the observation of opposing volume gradients of the presynaptic ribbons and associated postsynaptic glutamate receptor patches in mice along the pillar modiolar axis of the inner hair cell, the same axis along which fibers were shown to vary in their physiological properties. However, it is unclear whether these opposing gradients are expressed consistently across animal models. In addition, such volume gradients observed for separate populations of presynaptic ribbons and postsynaptic glutamate receptor patches suggest different relative volumes of these synaptic structures at individual synapses; however, these differences have not been examined in mice. Furthermore, it is unclear whether such gradients are limited to these synaptic proteins. Therefore, we analyzed organs of Corti isolated from CBA/CaJ, C57BL/6, and FVB/NJ mice using immunofluorescence, confocal microscopy, and quantitative image analysis. We find consistent expression of presynaptic volume gradients across strains of mice and inconsistent expression of postsynaptic volume gradients. We find differences in the relative volume of synaptic proteins, but these are different between CBA/CaJ mice, and C57BL/6 and FVB/NJ mice. We find similar results in C57BL/6 and FVB/NJ mice when using other postsynaptic density proteins (Shank1, Homer, and PSD95). These results have implications for the mechanisms by which volumes of synaptic proteins contribute to variations in the physiology of individual auditory nerve fibers and their vulnerability to excitotoxicity.
Collapse
Affiliation(s)
- Daniël O J Reijntjes
- University of Groningen, University Medical Center Groningen, Groningen, Department of Otorhinolaryngology and Head/Neck Surgery, 9713 GZ, Groningen, the Netherlands.
| | - Christine Köppl
- Cluster of Excellence "Hearing4all" and Research Centre Neurosensory Science, Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky University Oldenburg, Germany
| | - Sonja J Pyott
- University of Groningen, University Medical Center Groningen, Groningen, Department of Otorhinolaryngology and Head/Neck Surgery, 9713 GZ, Groningen, the Netherlands
| |
Collapse
|
38
|
Heeringa AN, Zhang L, Ashida G, Beutelmann R, Steenken F, Köppl C. Temporal Coding of Single Auditory Nerve Fibers Is Not Degraded in Aging Gerbils. J Neurosci 2020. [PMID: 31719164 DOI: 10.1101/2020.02.10.942011] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2023] Open
Abstract
People suffering from age-related hearing loss typically present with deficits in temporal processing tasks. Temporal processing deficits have also been shown in single-unit studies at the level of the auditory brainstem, midbrain, and cortex of aged animals. In this study, we explored whether temporal coding is already affected at the level of the input to the central auditory system. Single-unit auditory nerve fiber recordings were obtained from 41 Mongolian gerbils of either sex, divided between young, middle-aged, and old gerbils. Temporal coding quality was evaluated as vector strength in response to tones at best frequency, and by constructing shuffled and cross-stimulus autocorrelograms, and reverse correlations, from responses to 1 s noise bursts at 10-30 dB sensation level (dB above threshold). At comparable sensation levels, all measures showed that temporal coding was not altered in auditory nerve fibers of aging gerbils. Furthermore, both temporal fine structure and envelope coding remained unaffected. However, spontaneous rates were decreased in aging gerbils. Importantly, despite elevated pure tone thresholds, the frequency tuning of auditory nerve fibers was not affected. These results suggest that age-related temporal coding deficits arise more centrally, possibly due to a loss of auditory nerve fibers (or their peripheral synapses) but not due to qualitative changes in the responses of remaining auditory nerve fibers. The reduced spontaneous rate and elevated thresholds, but normal frequency tuning, of aged auditory nerve fibers can be explained by the well known reduction of endocochlear potential due to strial dysfunction in aged gerbils.SIGNIFICANCE STATEMENT As our society ages, age-related hearing deficits become ever more prevalent. Apart from decreased hearing sensitivity, elderly people often suffer from a reduced ability to communicate in daily settings, which is thought to be caused by known age-related deficits in auditory temporal processing. The current study demonstrated, using several different stimuli and analysis techniques, that these putative temporal processing deficits are not apparent in responses of single-unit auditory nerve fibers of quiet-aged gerbils. This suggests that age-related temporal processing deficits may develop more central to the auditory nerve, possibly due to a reduced population of active auditory nerve fibers, which will be of importance for the development of treatments for age-related hearing disorders.
Collapse
Affiliation(s)
- Amarins N Heeringa
- Cluster of Excellence "Hearing4all" and Research Centre Neurosensory Science, Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| | - Lichun Zhang
- Cluster of Excellence "Hearing4all" and Research Centre Neurosensory Science, Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| | - Go Ashida
- Cluster of Excellence "Hearing4all" and Research Centre Neurosensory Science, Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| | - Rainer Beutelmann
- Cluster of Excellence "Hearing4all" and Research Centre Neurosensory Science, Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| | - Friederike Steenken
- Cluster of Excellence "Hearing4all" and Research Centre Neurosensory Science, Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| | - Christine Köppl
- Cluster of Excellence "Hearing4all" and Research Centre Neurosensory Science, Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| |
Collapse
|
39
|
Temporal Coding of Single Auditory Nerve Fibers Is Not Degraded in Aging Gerbils. J Neurosci 2019; 40:343-354. [PMID: 31719164 DOI: 10.1523/jneurosci.2784-18.2019] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 10/25/2019] [Accepted: 11/04/2019] [Indexed: 02/03/2023] Open
Abstract
People suffering from age-related hearing loss typically present with deficits in temporal processing tasks. Temporal processing deficits have also been shown in single-unit studies at the level of the auditory brainstem, midbrain, and cortex of aged animals. In this study, we explored whether temporal coding is already affected at the level of the input to the central auditory system. Single-unit auditory nerve fiber recordings were obtained from 41 Mongolian gerbils of either sex, divided between young, middle-aged, and old gerbils. Temporal coding quality was evaluated as vector strength in response to tones at best frequency, and by constructing shuffled and cross-stimulus autocorrelograms, and reverse correlations, from responses to 1 s noise bursts at 10-30 dB sensation level (dB above threshold). At comparable sensation levels, all measures showed that temporal coding was not altered in auditory nerve fibers of aging gerbils. Furthermore, both temporal fine structure and envelope coding remained unaffected. However, spontaneous rates were decreased in aging gerbils. Importantly, despite elevated pure tone thresholds, the frequency tuning of auditory nerve fibers was not affected. These results suggest that age-related temporal coding deficits arise more centrally, possibly due to a loss of auditory nerve fibers (or their peripheral synapses) but not due to qualitative changes in the responses of remaining auditory nerve fibers. The reduced spontaneous rate and elevated thresholds, but normal frequency tuning, of aged auditory nerve fibers can be explained by the well known reduction of endocochlear potential due to strial dysfunction in aged gerbils.SIGNIFICANCE STATEMENT As our society ages, age-related hearing deficits become ever more prevalent. Apart from decreased hearing sensitivity, elderly people often suffer from a reduced ability to communicate in daily settings, which is thought to be caused by known age-related deficits in auditory temporal processing. The current study demonstrated, using several different stimuli and analysis techniques, that these putative temporal processing deficits are not apparent in responses of single-unit auditory nerve fibers of quiet-aged gerbils. This suggests that age-related temporal processing deficits may develop more central to the auditory nerve, possibly due to a reduced population of active auditory nerve fibers, which will be of importance for the development of treatments for age-related hearing disorders.
Collapse
|
40
|
Deiters KK, Flamme GA, Tasko SM, Murphy WJ, Greene NT, Jones HG, Ahroon WA. Generalizability of clinically measured acoustic reflexes to brief sounds. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2019; 146:3993. [PMID: 31795698 PMCID: PMC7043895 DOI: 10.1121/1.5132705] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 08/20/2019] [Accepted: 08/22/2019] [Indexed: 05/08/2023]
Abstract
Middle ear muscle contractions (MEMC) can be elicited in response to high-level sounds, and have been used clinically as acoustic reflexes (ARs) during evaluations of auditory system integrity. The results of clinical AR evaluations do not necessarily generalize to different signal types or durations. The purpose of this study was to evaluate the likelihood of observing MEMC in response to brief sound stimuli (tones, recorded gunshots, noise) in adult participants (N = 190) exhibiting clinical ARs and excellent hearing sensitivity. Results revealed that the presence of clinical ARs was not a sufficient indication that listeners will also exhibit MEMC for brief sounds. Detection rates varied across stimulus types between approximately 20% and 80%. Probabilities of observing MEMC also differed by clinical AR magnitude and latency, and declined over the period of minutes during the course of the MEMC measurement series. These results provide no support for the inclusion of MEMC as a protective factor in damage-risk criteria for impulsive noises, and the limited predictability of whether a given individual will exhibit MEMC in response to a brief sound indicates a need to measure and control for MEMC in studies evaluating pharmaceutical interventions for hearing loss.
Collapse
Affiliation(s)
- Kristy K Deiters
- Stephenson and Stephenson Research and Consulting (SASRAC), Forest Grove, Oregon 97116, USA
| | - Gregory A Flamme
- Stephenson and Stephenson Research and Consulting (SASRAC), Forest Grove, Oregon 97116, USA
| | - Stephen M Tasko
- Stephenson and Stephenson Research and Consulting (SASRAC), Forest Grove, Oregon 97116, USA
| | - William J Murphy
- National Institute for Occupational Safety and Health (NIOSH), Cincinnati, Ohio 45226, USA
| | - Nathaniel T Greene
- United States (U.S.) Army Aeromedical Research Lab (USAARL), Fort Rucker, Alabama 36362, USA
| | - Heath G Jones
- United States (U.S.) Army Aeromedical Research Lab (USAARL), Fort Rucker, Alabama 36362, USA
| | - William A Ahroon
- United States (U.S.) Army Aeromedical Research Lab (USAARL), Fort Rucker, Alabama 36362, USA
| |
Collapse
|
41
|
Heil P, Peterson AJ. Nelson's notch in the rate-level functions of auditory-nerve fibers might be caused by PIEZO2-mediated reverse-polarity currents in hair cells. Hear Res 2019; 381:107783. [DOI: 10.1016/j.heares.2019.107783] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/17/2019] [Accepted: 08/06/2019] [Indexed: 11/30/2022]
|
42
|
Phase Locking of Auditory-Nerve Fibers Reveals Stereotyped Distortions and an Exponential Transfer Function with a Level-Dependent Slope. J Neurosci 2019; 39:4077-4099. [PMID: 30867259 DOI: 10.1523/jneurosci.1801-18.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 02/28/2019] [Accepted: 03/07/2019] [Indexed: 12/16/2022] Open
Abstract
Phase locking of auditory-nerve-fiber (ANF) responses to the fine structure of acoustic stimuli is a hallmark of the auditory system's temporal precision and is important for many aspects of hearing. Period histograms from phase-locked ANF responses to low-frequency tones exhibit spike-rate and temporal asymmetries, but otherwise retain an approximately sinusoidal shape as stimulus level increases, even beyond the level at which the mean spike rate saturates. This is intriguing because apical cochlear mechanical vibrations show little compression, and mechanoelectrical transduction in the receptor cells is thought to obey a static sigmoidal nonlinearity, which might be expected to produce peak clipping at moderate and high stimulus levels. Here we analyze phase-locked responses of ANFs from cats of both sexes. We show that the lack of peak clipping is due neither to ANF refractoriness nor to spike-rate adaptation on time scales longer than the stimulus period. We demonstrate that the relationship between instantaneous pressure and instantaneous rate is well described by an exponential function whose slope decreases with increasing stimulus level. Relatively stereotyped harmonic distortions in the input to the exponential can account for the temporal asymmetry of the period histograms, including peak splitting. We show that the model accounts for published membrane-potential waveforms when assuming a power-of-three, but not a power-of-one, relationship to exocytosis. Finally, we demonstrate the relationship between the exponential transfer functions and the sigmoidal pseudotransducer functions obtained in the literature by plotting the maxima and minima of the voltage responses against the maxima and minima of the stimuli.SIGNIFICANCE STATEMENT Phase locking of auditory-nerve-fiber responses to the temporal fine structure of acoustic stimuli is important for many aspects of hearing, but the mechanisms underlying phase locking are not fully understood. Intriguingly, period histograms retain an approximately sinusoidal shape across sound levels, even when the mean rate has saturated. We find that neither refractoriness nor spike-rate adaptation is responsible for this behavior. Instead, the peripheral auditory system operates as though it contains an exponential transfer function whose slope changes with stimulus level. The underlying mechanism is distinct from the comparatively weak cochlear mechanical compression in the cochlear apex, and likely resides in the receptor cells.
Collapse
|
43
|
Vercammen C, Goossens T, Undurraga J, Wouters J, van Wieringen A. Electrophysiological and Behavioral Evidence of Reduced Binaural Temporal Processing in the Aging and Hearing Impaired Human Auditory System. Trends Hear 2019; 22:2331216518785733. [PMID: 30022734 PMCID: PMC6053861 DOI: 10.1177/2331216518785733] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
A person’s ability to process temporal fine structure information is
indispensable for speech understanding. As speech understanding typically
deteriorates throughout adult life, this study aimed to disentangle age and
hearing impairment (HI)-related changes in binaural temporal processing. This
was achieved by examining neural and behavioral processing of interaural phase
differences (IPDs). Neural IPD processing was studied electrophysiologically
through steady-state activity in the electroencephalogram evoked by periodic
changes in IPDs over time, embedded in the temporal fine structure of acoustic
stimulation. In addition, behavioral IPD discrimination thresholds were
determined for the same stimuli. To disentangle potential effects of age from
those of HI, both measures were applied to six participant groups: young,
middle-aged, and older persons, with either normal hearing or sensorineural HI.
All participants passed a cognitive screening, and stimulus audibility was
controlled for in participants with HI. The results demonstrated that HI changes
neural processing of binaural temporal information for all age-groups included
in this study. These outcomes were revealed, superimposed on age-related changes
that emerge between young adulthood and middle age. Poorer neural outcomes were
also associated with poorer behavioral performance, even though the behavioral
IPD discrimination thresholds were affected by age rather than by HI. The neural
outcomes of this study are the first to evidence and disentangle the dual load
of age and HI on binaural temporal processing. These results could be a valuable
first step toward future research on rehabilitation.
Collapse
Affiliation(s)
- Charlotte Vercammen
- 1 Department of Neurosciences, Research Group Experimental Oto-Rhino-Laryngology, KU Leuven-University of Leuven, Belgium
| | - Tine Goossens
- 1 Department of Neurosciences, Research Group Experimental Oto-Rhino-Laryngology, KU Leuven-University of Leuven, Belgium
| | - Jaime Undurraga
- 2 Department of Linguistics, The Australian Hearing Hub, Macquarie University, Sydney, Australia.,3 Ear Institute, University College London, London, UK
| | - Jan Wouters
- 1 Department of Neurosciences, Research Group Experimental Oto-Rhino-Laryngology, KU Leuven-University of Leuven, Belgium
| | - Astrid van Wieringen
- 1 Department of Neurosciences, Research Group Experimental Oto-Rhino-Laryngology, KU Leuven-University of Leuven, Belgium
| |
Collapse
|
44
|
Wang J, Yin S, Chen H, Shi L. Noise-Induced Cochlear Synaptopathy and Ribbon Synapse Regeneration: Repair Process and Therapeutic Target. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1130:37-57. [PMID: 30915700 DOI: 10.1007/978-981-13-6123-4_3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The synapse between the inner hair cells (IHCs) and the spiral ganglion neurons (SGNs) in mammalian cochleae is characterized as having presynaptic ribbons and therefore is called ribbon synapse. The special molecular organization is reviewed in this chapter in association with the functional feature of this synapse in signal processing. This is followed by the review on noise-induced damage to this synapse with a focus on recent reports in animal models in which the effect of brief noise exposures is observed without causing significant permanent threshold shift (PTS). In this regard, the potential mechanism of the synaptic damage by noise and the impact of this damage on hearing are summarized to clarify the concept of noise-induced hidden hearing loss, which is defined as the functional deficits in hearing without threshold elevation. A controversial issue is addressed in this review as whether the disrupted synapses can be regenerated. Moreover, the review summarizes the work of therapeutic research to protect the synapses or to promote the regeneration of the synapse after initial disruption. Lastly, several unresolved issues are raised for investigation in the future.
Collapse
Affiliation(s)
- Jian Wang
- School of Communication Science and Disorders, Dalhousie University, Halifax, NS, Canada.
| | - Shankai Yin
- Otolaryngology Research Institute, 6th Affiliated Hospital, Shanghai Jiao-Tong University, Shanghai, China
| | - Hengchao Chen
- Otolaryngology Research Institute, 6th Affiliated Hospital, Shanghai Jiao-Tong University, Shanghai, China
| | - Lijuan Shi
- Department of Physiology, Medical College of Southeast University, Nanjing, China
| |
Collapse
|
45
|
Jittered echo-delay resolution in bottlenose dolphins (Tursiops truncatus). J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2018; 205:125-137. [PMID: 30588550 PMCID: PMC6394490 DOI: 10.1007/s00359-018-1309-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 12/04/2018] [Indexed: 11/24/2022]
Abstract
Psychophysical methods similar to those employed with bats were used to examine jittered echo-delay resolution in bottlenose dolphins (Tursiops truncatus). Two dolphins were trained to produce echolocation clicks and report a change from electronic echoes with a fixed delay of ~ 12.6 ms (~ 9.4 m simulated range) to echoes with delays that alternated (jittered) between successive emitted signals. Jitter delays varied from 0 to 50 µs. Jittered echo-delay thresholds were between 1 and 2 µs—the lowest achievable (non-zero) values with the hardware configuration. Error functions matched the click autocorrelation function near zero jitter delay, and were well within the envelope of the autocorrelation function; however, measured jitter delay thresholds were larger than predictions for a coherent or semicoherent receiver at comparable signal-to-noise ratios. When one of the two alternating jittered echoes was inverted in polarity, both dolphins reliably discriminated echoes at all jittered echo delays, including 0 µs (i.e., only jittering in polarity, not delay). Finally, both dolphins used unusual patterns of click emissions, where groups of echolocation clicks were interspersed with silent gaps. Further tests with sub-microsecond jitter values and various echo signal-to-noise ratios would be necessary for proper direct comparison with jitter detection values obtained for bats.
Collapse
|
46
|
Peterson AJ, Huet A, Bourien J, Puel JL, Heil P. Recovery of auditory-nerve-fiber spike amplitude under natural excitation conditions. Hear Res 2018; 370:248-263. [DOI: 10.1016/j.heares.2018.08.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 08/20/2018] [Accepted: 08/22/2018] [Indexed: 12/23/2022]
|
47
|
Huet A, Batrel C, Wang J, Desmadryl G, Nouvian R, Puel JL, Bourien J. Sound Coding in the Auditory Nerve: From Single Fiber Activity to Cochlear Mass Potentials in Gerbils. Neuroscience 2018; 407:83-92. [PMID: 30342201 DOI: 10.1016/j.neuroscience.2018.10.010] [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: 07/01/2018] [Revised: 10/01/2018] [Accepted: 10/08/2018] [Indexed: 12/21/2022]
Abstract
Auditory nerve fibers (ANFs) convey acoustic information from the sensory cells to the brainstem using an elaborated neural code based on both spike timing and rate. As the stimulus tone frequency increases, time coding fades and ceases, resulting in high-frequency tone encoding that relies mostly on the spike discharge rate. Here, we recapitulated our recent single-unit data from gerbil's auditory nerve to highlight the most relevant mode of coding (spike timing versus spike rate) in tone-in-noise. We report that high-spontaneous rate (SR) fibers driven by low-frequency tones in noise are able to phase lock ∼30 dB below the level that evoked a significant elevation of the discharge rate, whereas medium- and low-SR fibers switch their preferential mode of coding from rate coding in quiet, to time coding in noise. For high-frequency tone, the low-threshold/high-SR fibers reach their maximum discharge rate in noise and do not respond to tones, whereas medium- and low-SR fibers are still able to respond to tones making them more resistant to background noise. Based on these findings, we first discuss the ecological function of the ANF distribution according to their spontaneous discharge rate. Then, we point out the poor synchronization of the low-SR ANFs, accounting for the discrepancy between ANF number and the amplitude of the compound action potential of the of the auditory nerve. Finally, we proposed a new diagnostic tool to assess low-SR fibers, which does not rely on the onset response of the ANFs.
Collapse
Affiliation(s)
- A Huet
- INM, Inserm, Univ Montpellier, Montpellier, France
| | - C Batrel
- INM, Inserm, Univ Montpellier, Montpellier, France
| | - J Wang
- INM, Inserm, Univ Montpellier, Montpellier, France
| | - G Desmadryl
- INM, Inserm, Univ Montpellier, Montpellier, France
| | - R Nouvian
- INM, Inserm, Univ Montpellier, Montpellier, France
| | - J L Puel
- INM, Inserm, Univ Montpellier, Montpellier, France.
| | - J Bourien
- INM, Inserm, Univ Montpellier, Montpellier, France
| |
Collapse
|
48
|
Fischer BJ, Wydick JL, Köppl C, Peña JL. Multidimensional stimulus encoding in the auditory nerve of the barn owl. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2018; 144:2116. [PMID: 30404459 PMCID: PMC6185867 DOI: 10.1121/1.5056171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 09/07/2018] [Accepted: 09/10/2018] [Indexed: 06/08/2023]
Abstract
Auditory perception depends on multi-dimensional information in acoustic signals that must be encoded by auditory nerve fibers (ANF). These dimensions are represented by filters with different frequency selectivities. Multiple models have been suggested; however, the identification of relevant filters and type of interactions has been elusive, limiting progress in modeling the cochlear output. Spike-triggered covariance analysis of barn owl ANF responses was used to determine the number of relevant stimulus filters and estimate the nonlinearity that produces responses from filter outputs. This confirmed that ANF responses depend on multiple filters. The first, most dominant filter was the spike-triggered average, which was excitatory for all neurons. The second and third filters could be either suppressive or excitatory with center frequencies above or below that of the first filter. The nonlinear function mapping the first two filter outputs to the spiking probability ranged from restricted to nearly circular-symmetric, reflecting different modes of interaction between stimulus dimensions across the sample. This shows that stimulus encoding in ANFs of the barn owl is multidimensional and exhibits diversity over the population, suggesting that models must allow for variable numbers of filters and types of interactions between filters to describe how sound is encoded in ANFs.
Collapse
Affiliation(s)
- Brian J Fischer
- Department of Mathematics, Seattle University, Seattle, Washington 98122, USA
| | - Jacob L Wydick
- Department of Mathematics, Seattle University, Seattle, Washington 98122, USA
| | - Christine Köppl
- Cluster of Excellence "Hearing4all" and Research Centre Neurosensory Science, Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky University, Oldenburg, Germany
| | - José L Peña
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, New York 10461, USA
| |
Collapse
|
49
|
Chen H, Shi L, Liu L, Yin S, Aiken S, Wang J. Noise-induced Cochlear Synaptopathy and Signal Processing Disorders. Neuroscience 2018; 407:41-52. [PMID: 30267832 DOI: 10.1016/j.neuroscience.2018.09.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 01/18/2023]
Abstract
Noise-induced hidden hearing loss (NIHHL) has attracted great attention in hearing research and clinical audiology since the discovery of significant noise-induced synaptic damage in the absence of permanent threshold shifts (PTS) in animal models. Although the extant evidence for this damage is based on animal models, NIHHL likely occurs in humans as well. This review focuses on three issues concerning NIHHL that are somewhat controversial: (1) whether disrupted synapses can be re-established; (2) whether synaptic damage and repair are responsible for the initial temporal threshold shifts (TTS) and subsequent recovery; and (3) the relationship between the synaptic damage and repair processes and neural coding deficits. We conclude that, after a single, brief noise exposure, (1) the damaged and the totally destroyed synapses can be partially repaired, but the repaired synapses are functionally abnormal; (2) While deficits are observed in some aspects of neural responses related to temporal and intensity coding in the auditory nerve, we did not find strong evidence for hypothesized coding-in-noise deficits; (3) the sensitivity and the usefulness of the envelope following responses to amplitude modulation signals in detecting cochlear synaptopathy is questionable.
Collapse
Affiliation(s)
- Hengchao Chen
- Otolaryngology Research Institute, 6th Affiliated Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Lijuan Shi
- Department of Physiology, Medical College of Southeast University, Nanjing, China
| | - Lijie Liu
- Department of Physiology, Medical College of Southeast University, Nanjing, China
| | - Shankai Yin
- Otolaryngology Research Institute, 6th Affiliated Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Steven Aiken
- School of Communication Sciences and Disorders, Dalhousie University, Halfiax, Canada
| | - Jian Wang
- Otolaryngology Research Institute, 6th Affiliated Hospital, Shanghai Jiao Tong University, Shanghai, China; School of Communication Sciences and Disorders, Dalhousie University, Halfiax, Canada.
| |
Collapse
|
50
|
Sun S, Babola T, Pregernig G, So KS, Nguyen M, Su SSM, Palermo AT, Bergles DE, Burns JC, Müller U. Hair Cell Mechanotransduction Regulates Spontaneous Activity and Spiral Ganglion Subtype Specification in the Auditory System. Cell 2018; 174:1247-1263.e15. [PMID: 30078710 PMCID: PMC6429032 DOI: 10.1016/j.cell.2018.07.008] [Citation(s) in RCA: 190] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/23/2018] [Accepted: 07/02/2018] [Indexed: 01/06/2023]
Abstract
Type I spiral ganglion neurons (SGNs) transmit sound information from cochlear hair cells to the CNS. Using transcriptome analysis of thousands of single neurons, we demonstrate that murine type I SGNs consist of subclasses that are defined by the expression of subsets of transcription factors, cell adhesion molecules, ion channels, and neurotransmitter receptors. Subtype specification is initiated prior to the onset of hearing during the time period when auditory circuits mature. Gene mutations linked to deafness that disrupt hair cell mechanotransduction or glutamatergic signaling perturb the firing behavior of SGNs prior to hearing onset and disrupt SGN subtype specification. We thus conclude that an intact hair cell mechanotransduction machinery is critical during the pre-hearing period to regulate the firing behavior of SGNs and their segregation into subtypes. Because deafness is frequently caused by defects in hair cells, our findings have significant ramifications for the etiology of hearing loss and its treatment.
Collapse
Affiliation(s)
- Shuohao Sun
- The Solomon Snyder Department of Neuroscience and Department of Otolaryngology, Head and Neck Surgery, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Travis Babola
- The Solomon Snyder Department of Neuroscience and Department of Otolaryngology, Head and Neck Surgery, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Gabriela Pregernig
- Decibel Therapeutics, 1325 Boylston Street, Suite 500, Boston, MA 02215, USA
| | - Kathy S So
- Decibel Therapeutics, 1325 Boylston Street, Suite 500, Boston, MA 02215, USA
| | - Matthew Nguyen
- Decibel Therapeutics, 1325 Boylston Street, Suite 500, Boston, MA 02215, USA
| | - Shin-San M Su
- Decibel Therapeutics, 1325 Boylston Street, Suite 500, Boston, MA 02215, USA
| | - Adam T Palermo
- Decibel Therapeutics, 1325 Boylston Street, Suite 500, Boston, MA 02215, USA
| | - Dwight E Bergles
- The Solomon Snyder Department of Neuroscience and Department of Otolaryngology, Head and Neck Surgery, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Joseph C Burns
- Decibel Therapeutics, 1325 Boylston Street, Suite 500, Boston, MA 02215, USA.
| | - Ulrich Müller
- The Solomon Snyder Department of Neuroscience and Department of Otolaryngology, Head and Neck Surgery, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA.
| |
Collapse
|