1
|
Oestreicher D, Chepurwar S, Kusch K, Rankovic V, Jung S, Strenzke N, Pangrsic T. CaBP1 and 2 enable sustained Ca V1.3 calcium currents and synaptic transmission in inner hair cells. eLife 2024; 13:RP93646. [PMID: 39718549 DOI: 10.7554/elife.93646] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024] Open
Abstract
To encode continuous sound stimuli, the inner hair cell (IHC) ribbon synapses utilize calcium-binding proteins (CaBPs), which reduce the inactivation of their CaV1.3 calcium channels. Mutations in the CABP2 gene underlie non-syndromic autosomal recessive hearing loss DFNB93. Besides CaBP2, the structurally related CaBP1 is highly abundant in the IHCs. Here, we investigated how the two CaBPs cooperatively regulate IHC synaptic function. In Cabp1/2 double-knockout mice, we find strongly enhanced CaV1.3 inactivation, slowed recovery from inactivation and impaired sustained exocytosis. Already mild IHC activation further reduces the availability of channels to trigger synaptic transmission and may effectively silence synapses. Spontaneous and sound-evoked responses of spiral ganglion neurons in vivo are strikingly reduced and strongly depend on stimulation rates. Transgenic expression of CaBP2 leads to substantial recovery of IHC synaptic function and hearing sensitivity. We conclude that CaBP1 and 2 act together to suppress voltage- and calcium-dependent inactivation of IHC CaV1.3 channels in order to support sufficient rate of exocytosis and enable fast, temporally precise and indefatigable sound encoding.
Collapse
Affiliation(s)
- David Oestreicher
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Shashank Chepurwar
- Auditory Systems Physiology Group, Institute for Auditory Neuroscience, InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
| | - Kathrin Kusch
- Functional Auditory Genomics, Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Vladan Rankovic
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Restorative Cochlear Genomics Group, Auditory Neuroscience and Optogenetics Laboratory, German Primate Cente, Göttingen, Germany
| | - Sangyong Jung
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Nicola Strenzke
- Auditory Systems Physiology Group, Institute for Auditory Neuroscience, InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
| | - Tina Pangrsic
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
- Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
2
|
Jaime Tobón LM, Moser T. Bridging the gap between presynaptic hair cell function and neural sound encoding. eLife 2024; 12:RP93749. [PMID: 39718472 DOI: 10.7554/elife.93749] [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] [Indexed: 12/25/2024] Open
Abstract
Neural diversity can expand the encoding capacity of a circuitry. A striking example of diverse structure and function is presented by the afferent synapses between inner hair cells (IHCs) and spiral ganglion neurons (SGNs) in the cochlea. Presynaptic active zones at the pillar IHC side activate at lower IHC potentials than those of the modiolar side that have more presynaptic Ca2+ channels. The postsynaptic SGNs differ in their spontaneous firing rates, sound thresholds, and operating ranges. While a causal relationship between synaptic heterogeneity and neural response diversity seems likely, experimental evidence linking synaptic and SGN physiology has remained difficult to obtain. Here, we aimed at bridging this gap by ex vivo paired recordings of murine IHCs and postsynaptic SGN boutons with stimuli and conditions aimed to mimic those of in vivo SGN characterization. Synapses with high spontaneous rate of release (SR) were found predominantly on the pillar side of the IHC. These high SR synapses had larger and more temporally compact spontaneous EPSCs, lower voltage thresholds, tighter coupling of Ca2+ channels and vesicular release sites, shorter response latencies, and higher initial release rates. This study indicates that synaptic heterogeneity in IHCs directly contributes to the diversity of spontaneous and sound-evoked firing of SGNs.
Collapse
Affiliation(s)
- Lina María Jaime Tobón
- Auditory Neuroscience and Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany
- Collaborative Research Center, University of Göttingen, Göttingen, Germany
- Cluster of Excellence 'Multiscale Bioimaging of Excitable Cells', Göttingen, Germany
| | - Tobias Moser
- Auditory Neuroscience and Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany
- Collaborative Research Center, University of Göttingen, Göttingen, Germany
- Cluster of Excellence 'Multiscale Bioimaging of Excitable Cells', Göttingen, Germany
| |
Collapse
|
3
|
Kostrikov S, Hjortkjaer J, Dau T, Corfas G, Liberman LD, Liberman MC. A modiolar-pillar gradient in auditory-nerve dendritic length: A novel post-synaptic contribution to dynamic range? Hear Res 2024; 456:109172. [PMID: 39708764 DOI: 10.1016/j.heares.2024.109172] [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: 09/27/2024] [Revised: 12/06/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Auditory-nerve fibers (ANFs) from a given cochlear region can vary in threshold sensitivity by up to 60 dB, corresponding to a 1000-fold difference in stimulus level, although each fiber innervates a single inner hair cell (IHC) via a single synapse. ANFs with high-thresholds also have low spontaneous rates (SRs) and synapse on the side of the IHC closer to the modiolus, whereas the low-threshold, high-SR fibers synapse on the side closer to the pillar cells. Prior biophysical work has identified modiolar-pillar differences in both pre- and post-synaptic properties, but a comprehensive explanation for the wide range of sensitivities remains elusive. Here, in guinea pigs, we used immunostaining for several neuronal markers, including Caspr, a key protein in nodes of Ranvier, to reveal a novel modiolar-pillar gradient in the location of the first ANF heminodes, presumed to be the site of the spike generator, just outside the sensory epithelium. Along the cochlea, from apex to base, the unmyelinated terminal dendrites of modiolar ANFs were 2-4 times longer than those of pillar ANFs. This modiolar-pillar gradient in dendritic length, coupled with the 2-4 fold smaller caliber of modiolar dendrites seen in prior single-fiber labeling studies, suggests there could be a large difference in the number of length constants between the synapse and the spike initiation zone for low- vs high-SR fibers. The resultant differences in attenuation of post-synaptic potentials propagating along these unmyelinated dendrites could be a key contributor to the observed range of threshold sensitivities among ANFs.
Collapse
Affiliation(s)
- Serhii Kostrikov
- Department of Health Technology, Centre for Auditory Neuroscience, Hearing Systems, Technical University of Denmark, Lyngby, Denmark
| | - Jens Hjortkjaer
- Department of Health Technology, Centre for Auditory Neuroscience, Hearing Systems, Technical University of Denmark, Lyngby, Denmark
| | - Torsten Dau
- Department of Health Technology, Centre for Auditory Neuroscience, Hearing Systems, Technical University of Denmark, Lyngby, Denmark
| | - Gabriel Corfas
- Department of Otolaryngology-Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI, United States
| | - Leslie D Liberman
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA, United States
| | - M Charles Liberman
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA, United States; Department of Otolaryngology-Head & Neck Surgery, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
4
|
Kostrikov S, Hjortkjaer J, Dau T, Corfas G, Liberman LD, Liberman MC. A modiolar-pillar gradient in auditory-nerve dendritic length: a novel post-synaptic contribution to dynamic range? BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.621861. [PMID: 39574647 PMCID: PMC11580876 DOI: 10.1101/2024.11.04.621861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
Auditory-nerve fibers (ANFs) from a given cochlear region can vary in threshold sensitivity by up to 60 dB, corresponding to a 1000-fold difference in stimulus level, although each fiber innervates a single inner hair cell (IHC) via a single synapse. ANFs with high-thresholds also have low spontaneous rates (SRs) and synapse on the side of the IHC closer to the modiolus, whereas the low-threshold, high-SR fibers synapse on the side closer to the pillar cells. Prior biophysical work has identified modiolar-pillar differences in both pre- and post-synaptic properties, but a comprehensive explanation for the wide range of sensitivities remains elusive. Here, in guinea pigs, we used immunostaining for several neuronal markers, including Caspr, a key protein in nodes of Ranvier, to reveal a novel modiolar-pillar gradient in the location of the first ANF heminodes, presumed to be the site of the spike generator, just outside the sensory epithelium. Along the cochlea, from apex to base, the unmyelinated terminal dendrites of modiolar ANFs were 2 - 4 times longer than those of pillar ANFs. This modiolar-pillar gradient in dendritic length, coupled with the 2 - 4 fold smaller caliber of modiolar dendrites seen in prior single-fiber labeling studies, suggests there could be a large difference in the number of length constants between the synapse and the spike initiation zone for low- vs high-SR fibers. The resultant differences in attenuation of post-synaptic potentials propagating along these unmyelinated dendrites could be a key contributor to the observed range of threshold sensitivities among ANFs.
Collapse
|
5
|
Yeo XY, Kwon S, Rinai KR, Lee S, Jung S, Park R. A Consolidated Understanding of the Contribution of Redox Dysregulation in the Development of Hearing Impairment. Antioxidants (Basel) 2024; 13:598. [PMID: 38790703 PMCID: PMC11118506 DOI: 10.3390/antiox13050598] [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: 02/22/2024] [Revised: 04/26/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
The etiology of hearing impairment is multifactorial, with contributions from both genetic and environmental factors. Although genetic studies have yielded valuable insights into the development and function of the auditory system, the contribution of gene products and their interaction with alternate environmental factors for the maintenance and development of auditory function requires further elaboration. In this review, we provide an overview of the current knowledge on the role of redox dysregulation as the converging factor between genetic and environmental factor-dependent development of hearing loss, with a focus on understanding the interaction of oxidative stress with the physical components of the peripheral auditory system in auditory disfunction. The potential involvement of molecular factors linked to auditory function in driving redox imbalance is an important promoter of the development of hearing loss over time.
Collapse
Affiliation(s)
- Xin Yi Yeo
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
- Department of Medical Science, College of Medicine, CHA University, Seongnam 13488, Republic of Korea;
| | - Soohyun Kwon
- Department of Medical Science, College of Medicine, CHA University, Seongnam 13488, Republic of Korea;
- Department of BioNanotechnology, Gachon University, Seongnam 13120, Republic of Korea
| | - Kimberley R. Rinai
- Department of Life Science, College of Medicine, CHA University, Seongnam 13488, Republic of Korea;
| | - Sungsu Lee
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Hospital and Medical School, Gwangju 61469, Republic of Korea;
| | - Sangyong Jung
- Department of Medical Science, College of Medicine, CHA University, Seongnam 13488, Republic of Korea;
| | - Raekil Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science & Technology (GIST), Gwangju 61005, Republic of Korea
| |
Collapse
|
6
|
Pal I, Bhattacharyya A, V-Ghaffari B, Williams ED, Xiao M, Rutherford MA, Rubio ME. Female GluA3-KO mice show early onset hearing loss and afferent swellings in ambient sound levels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.21.581467. [PMID: 38659964 PMCID: PMC11042237 DOI: 10.1101/2024.02.21.581467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
AMPA-type glutamate receptors (AMPAR) mediate excitatory cochlear transmission. However, the unique roles of AMPAR subunits are unresolved. Lack of subunit GluA3 (Gria3KO) in male mice reduced cochlear output by 8-weeks of age. Since Gria3 is X-linked and considering sex differences in hearing vulnerability, we hypothesized accelerated presbycusis in Gria3KO females. Here, auditory brainstem responses (ABR) were similar in 3-week-old female Gria3WT and Gria3KO mice. However, when raised in ambient sound, ABR thresholds were elevated and wave-1 amplitudes were diminished at 5-weeks and older in Gria3KO. In contrast, these metrics were similar between genotypes when raised in quiet. Paired synapses were similar in number, but lone ribbons and ribbonless synapses were increased in female Gria3KO mice in ambient sound compared to Gria3WT or to either genotype raised in quiet. Synaptic GluA4:GluA2 ratios increased relative to Gria3WT, particularly in ambient sound, suggesting an activity-dependent increase in calcium-permeable AMPARs in Gria3KO. Swollen afferent terminals were observed by 5-weeks only in Gria3KO females reared in ambient sound. We propose that lack of GluA3 induces sex-dependent vulnerability to AMPAR-mediated excitotoxicity.
Collapse
Affiliation(s)
- Indra Pal
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Atri Bhattacharyya
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO 63110
| | - Babak V-Ghaffari
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO 63110
| | - Essence D. Williams
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Maolei Xiao
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO 63110
| | - Mark A. Rutherford
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO 63110
| | - María Eulalia Rubio
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| |
Collapse
|
7
|
Hong J, Dai P, Liang H, Sun G, Qi W, Bi Y. Extrasynaptic distribution of NMDA receptors in cochlear inner hair cell afferent signaling complex. J Chem Neuroanat 2024; 137:102417. [PMID: 38570170 DOI: 10.1016/j.jchemneu.2024.102417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/30/2024] [Accepted: 03/30/2024] [Indexed: 04/05/2024]
Abstract
OBJECTIVE The distribution and role of NMDA receptors is unclear in the afferent signaling complex of the cochlea. The present study aimed to examine the distribution of NMDA receptors in cochlear afferent signaling complex of the adult mouse, and their relationship with ribbon synapses of inner hair cells (IHCs) and GABAergic efferent terminals of the lateral olivocochlear (LOC). METHODS Immunofluorescence staining in combination with confocal microscopy was used to investigate the distribution of glutamatergic NMDA and AMPA receptors in afferent terminals of SGNs, and their relationship with ribbon synapses of IHCs and GABAergic efferent terminals of LOC. RESULTS Terminals with AMPA receptors along with Ribbons of IHC formed afferent synapses in the basal pole of IHCs, and those with NMDA receptors were mainly distributed longitudinally in the IHCs nuclei region. Significant difference was found in the distribution of NMDA and AMPA receptors in IHC afferent signaling complex (P<0.05). Some GABAergic terminals colocalized with NMDA receptors at the IHC nucleus region (P>0.05). CONCLUSION There is significant difference in the distribution of NMDA and AMPA receptors in cochlear afferent signaling complex. NMDA receptors are present in the extra-synaptic region of ribbon synapses of IHCs, and they are related to GABA efferent terminals of the afferent signaling complex.
Collapse
Affiliation(s)
- Juan Hong
- Department of Otorhinolaryngology, Huashan Hospital of Fudan University, Shanghai 200040, China.
| | - Peidong Dai
- ENT Institute, Eye & ENT Hospital of Fudan University; NHC Hearing Medicine Key Laboratory (Fudan University), Shanghai 200031, China
| | - Huazheng Liang
- Monash Suzhou Research Institute, Suzhou Industrial Park, Jiangsu, China
| | - Guangbin Sun
- Department of Otorhinolaryngology, Huashan Hospital of Fudan University, Shanghai 200040, China
| | - Weidong Qi
- Department of Otorhinolaryngology, Huashan Hospital of Fudan University, Shanghai 200040, China
| | - Yong Bi
- Department of Neurology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China.
| |
Collapse
|
8
|
Cepeda AP, Ninov M, Neef J, Parfentev I, Kusch K, Reisinger E, Jahn R, Moser T, Urlaub H. Proteomic Analysis Reveals the Composition of Glutamatergic Organelles of Auditory Inner Hair Cells. Mol Cell Proteomics 2024; 23:100704. [PMID: 38128648 PMCID: PMC10832297 DOI: 10.1016/j.mcpro.2023.100704] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/08/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023] Open
Abstract
In the ear, inner hair cells (IHCs) employ sophisticated glutamatergic ribbon synapses with afferent neurons to transmit auditory information to the brain. The presynaptic machinery responsible for neurotransmitter release in IHC synapses includes proteins such as the multi-C2-domain protein otoferlin and the vesicular glutamate transporter 3 (VGluT3). Yet, much of this likely unique molecular machinery remains to be deciphered. The scarcity of material has so far hampered biochemical studies which require large amounts of purified samples. We developed a subcellular fractionation workflow combined with immunoisolation of VGluT3-containing membrane vesicles, allowing for the enrichment of glutamatergic organelles that are likely dominated by synaptic vesicles (SVs) of IHCs. We have characterized their protein composition in mice before and after hearing onset using mass spectrometry and confocal imaging and provide a fully annotated proteome with hitherto unidentified proteins. Despite the prevalence of IHC marker proteins across IHC maturation, the profiles of trafficking proteins differed markedly before and after hearing onset. Among the proteins enriched after hearing onset were VAMP-7, syntaxin-7, syntaxin-8, syntaxin-12/13, SCAMP1, V-ATPase, SV2, and PKCα. Our study provides an inventory of the machinery associated with synaptic vesicle-mediated trafficking and presynaptic activity at IHC ribbon synapses and serves as a foundation for future functional studies.
Collapse
Affiliation(s)
- Andreia P Cepeda
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Momchil Ninov
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany; Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Jakob Neef
- 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
| | - Iwan Parfentev
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Kathrin Kusch
- Functional Auditory Genomics Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Ellen Reisinger
- Gene Therapy for Hearing Impairment and Deafness, Department for Otolaryngology, Head & Neck Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Reinhard Jahn
- Laboratory of Neurobiology, 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.
| | - Tobias Moser
- 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; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany; Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
9
|
Jaime Tobón LM, Moser T. Ca 2+ regulation of glutamate release from inner hair cells of hearing mice. Proc Natl Acad Sci U S A 2023; 120:e2311539120. [PMID: 38019860 DOI: 10.1073/pnas.2311539120] [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: 07/07/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
In our hearing organ, sound is encoded at ribbon synapses formed by inner hair cells (IHCs) and spiral ganglion neurons (SGNs). How the underlying synaptic vesicle (SV) release is controlled by Ca2+ in IHCs of hearing animals remained to be investigated. Here, we performed patch-clamp SGN recordings of the initial rate of release evoked by brief IHC Ca2+-influx in an ex vivo cochlear preparation from hearing mice. We aimed to closely mimic physiological conditions by perforated-patch recordings from IHCs kept at the physiological resting potential and at body temperature. We found release to relate supralinearly to Ca2+-influx (power, m: 4.3) when manipulating the [Ca2+] available for SV release by Zn2+-flicker-blocking of the single Ca2+-channel current. In contrast, a near linear Ca2+ dependence (m: 1.2 to 1.5) was observed when varying the number of open Ca2+-channels during deactivating Ca2+-currents and by dihydropyridine channel-inhibition. Concurrent changes of number and current of open Ca2+-channels over the range of physiological depolarizations revealed m: 1.8. These findings indicate that SV release requires ~4 Ca2+-ions to bind to their Ca2+-sensor of fusion. We interpret the near linear Ca2+-dependence of release during manipulations that change the number of open Ca2+-channels to reflect control of SV release by the high [Ca2+] in the Ca2+-nanodomain of one or few nearby Ca2+-channels. We propose that a combination of Ca2+ nanodomain control and supralinear intrinsic Ca2+-dependence of fusion optimally links SV release to the timing and amplitude of the IHC receptor potential and separates it from other IHC Ca2+-signals unrelated to afferent synaptic transmission.
Collapse
Affiliation(s)
- Lina María Jaime Tobón
- Auditory Neuroscience and Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
- Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen 37075, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen 37075, Germany
- Multiscale Bioimaging of Excitable Cells, Cluster of Excellence, Göttingen 37075, Germany
| | - Tobias Moser
- Auditory Neuroscience and Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
- Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen 37075, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen 37075, Germany
- Multiscale Bioimaging of Excitable Cells, Cluster of Excellence, Göttingen 37075, Germany
| |
Collapse
|
10
|
Tichacek O, Mistrík P, Jungwirth P. From the outer ear to the nerve: A complete computer model of the peripheral auditory system. Hear Res 2023; 440:108900. [PMID: 37944408 DOI: 10.1016/j.heares.2023.108900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 10/03/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023]
Abstract
Computer models of the individual components of the peripheral auditory system - the outer, middle, and inner ears and the auditory nerve - have been developed in the past, with varying level of detail, breadth, and faithfulness of the underlying parameters. Building on previous work, we advance the modeling of the ear by presenting a complete, physiologically justified, bottom-up computer model based on up-to-date experimental data that integrates all of these parts together seamlessly. The detailed bottom-up design of the present model allows for the investigation of partial hearing mechanisms and their defects, including genetic, molecular, and microscopic factors. Also, thanks to the completeness of the model, one can study microscopic effects in the context of their implications on hearing as a whole, enabling the correlation with neural recordings and non-invasive psychoacoustic methods. Such a model is instrumental for advancing quantitative understanding of the mechanism of hearing, for investigating various forms of hearing impairment, as well as for devising next generation hearing aids and cochlear implants.
Collapse
Affiliation(s)
- Ondrej Tichacek
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nam. 2, 160 00 Prague 6, Czech Republic.
| | | | - Pavel Jungwirth
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nam. 2, 160 00 Prague 6, Czech Republic.
| |
Collapse
|
11
|
Moser T, Karagulyan N, Neef J, Jaime Tobón LM. Diversity matters - extending sound intensity coding by inner hair cells via heterogeneous synapses. EMBO J 2023; 42:e114587. [PMID: 37800695 PMCID: PMC10690447 DOI: 10.15252/embj.2023114587] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/26/2023] [Accepted: 08/07/2023] [Indexed: 10/07/2023] Open
Abstract
Our sense of hearing enables the processing of stimuli that differ in sound pressure by more than six orders of magnitude. How to process a wide range of stimulus intensities with temporal precision is an enigmatic phenomenon of the auditory system. Downstream of dynamic range compression by active cochlear micromechanics, the inner hair cells (IHCs) cover the full intensity range of sound input. Yet, the firing rate in each of their postsynaptic spiral ganglion neurons (SGNs) encodes only a fraction of it. As a population, spiral ganglion neurons with their respective individual coding fractions cover the entire audible range. How such "dynamic range fractionation" arises is a topic of current research and the focus of this review. Here, we discuss mechanisms for generating the diverse functional properties of SGNs and formulate testable hypotheses. We postulate that an interplay of synaptic heterogeneity, molecularly distinct subtypes of SGNs, and efferent modulation serves the neural decomposition of sound information and thus contributes to a population code for sound intensity.
Collapse
Affiliation(s)
- Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLabUniversity Medical Center GöttingenGöttingenGermany
- Auditory Neuroscience and Synaptic Nanophysiology GroupMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
- Cluster of Excellence “Multiscale Bioimaging of Excitable Cells”GöttingenGermany
| | - Nare Karagulyan
- Institute for Auditory Neuroscience and InnerEarLabUniversity Medical Center GöttingenGöttingenGermany
- Auditory Neuroscience and Synaptic Nanophysiology GroupMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
- Hertha Sponer CollegeCluster of Excellence “Multiscale Bioimaging of Excitable Cells” Cluster of ExcellenceGöttingenGermany
| | - Jakob Neef
- Institute for Auditory Neuroscience and InnerEarLabUniversity Medical Center GöttingenGöttingenGermany
- Auditory Neuroscience and Synaptic Nanophysiology GroupMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Lina María Jaime Tobón
- Institute for Auditory Neuroscience and InnerEarLabUniversity Medical Center GöttingenGöttingenGermany
- Auditory Neuroscience and Synaptic Nanophysiology GroupMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
- Hertha Sponer CollegeCluster of Excellence “Multiscale Bioimaging of Excitable Cells” Cluster of ExcellenceGöttingenGermany
| |
Collapse
|
12
|
Michanski S, Kapoor R, Steyer AM, Möbius W, Früholz I, Ackermann F, Gültas M, Garner CC, Hamra FK, Neef J, Strenzke N, Moser T, Wichmann C. Piccolino is required for ribbon architecture at cochlear inner hair cell synapses and for hearing. EMBO Rep 2023; 24:e56702. [PMID: 37477166 PMCID: PMC10481675 DOI: 10.15252/embr.202256702] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 06/30/2023] [Accepted: 07/06/2023] [Indexed: 07/22/2023] Open
Abstract
Cochlear inner hair cells (IHCs) form specialized ribbon synapses with spiral ganglion neurons that tirelessly transmit sound information at high rates over long time periods with extreme temporal precision. This functional specialization is essential for sound encoding and is attributed to a distinct molecular machinery with unique players or splice variants compared to conventional neuronal synapses. Among these is the active zone (AZ) scaffold protein piccolo/aczonin, which is represented by its short splice variant piccolino at cochlear and retinal ribbon synapses. While the function of piccolo at synapses of the central nervous system has been intensively investigated, the role of piccolino at IHC synapses remains unclear. In this study, we characterize the structure and function of IHC synapses in piccolo gene-trap mutant rats (Pclogt/gt ). We find a mild hearing deficit with elevated thresholds and reduced amplitudes of auditory brainstem responses. Ca2+ channel distribution and ribbon morphology are altered in apical IHCs, while their presynaptic function seems to be unchanged. We conclude that piccolino contributes to the AZ organization in IHCs and is essential for normal hearing.
Collapse
Affiliation(s)
- Susann Michanski
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLabUniversity Medical Center GöttingenGöttingenGermany
- Center for Biostructural Imaging of NeurodegenerationUniversity Medical Center GöttingenGöttingenGermany
- Collaborative Research Center 889 “Cellular Mechanisms of Sensory Processing”GöttingenGermany
- Multiscale Bioimaging of Excitable Cells, Cluster of ExcellenceGöttingenGermany
| | - Rohan Kapoor
- Institute for Auditory Neuroscience and InnerEarLabUniversity Medical Center GöttingenGöttingenGermany
- Auditory Neuroscience and Synaptic Nanophysiology GroupMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
- IMPRS Molecular Biology, Göttingen Graduate School for Neuroscience and Molecular BiosciencesUniversity of GöttingenGöttingenGermany
| | - Anna M Steyer
- Electron Microscopy Core Unit, Department of NeurogeneticsMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Wiebke Möbius
- Multiscale Bioimaging of Excitable Cells, Cluster of ExcellenceGöttingenGermany
- Electron Microscopy Core Unit, Department of NeurogeneticsMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Iris Früholz
- Developmental, Neural, and Behavioral Biology Master ProgramUniversity of GöttingenGöttingenGermany
| | | | - Mehmet Gültas
- Faculty of AgricultureSouth Westphalia University of Applied SciencesSoestGermany
| | - Craig C Garner
- German Center for Neurodegenerative DiseasesBerlinGermany
- NeuroCureCluster of ExcellenceCharité – UniversitätsmedizinBerlinGermany
| | - F Kent Hamra
- Department of Obstetrics and GynecologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Jakob Neef
- Collaborative Research Center 889 “Cellular Mechanisms of Sensory Processing”GöttingenGermany
- Institute for Auditory Neuroscience and InnerEarLabUniversity Medical Center GöttingenGöttingenGermany
- Auditory Neuroscience and Synaptic Nanophysiology GroupMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Nicola Strenzke
- Collaborative Research Center 889 “Cellular Mechanisms of Sensory Processing”GöttingenGermany
- Auditory Systems Physiology Group, Institute for Auditory Neuroscience and InnerEarLabUniversity Medical Center GöttingenGöttingenGermany
| | - Tobias Moser
- Collaborative Research Center 889 “Cellular Mechanisms of Sensory Processing”GöttingenGermany
- Multiscale Bioimaging of Excitable Cells, Cluster of ExcellenceGöttingenGermany
- Institute for Auditory Neuroscience and InnerEarLabUniversity Medical Center GöttingenGöttingenGermany
- Auditory Neuroscience and Synaptic Nanophysiology GroupMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLabUniversity Medical Center GöttingenGöttingenGermany
- Center for Biostructural Imaging of NeurodegenerationUniversity Medical Center GöttingenGöttingenGermany
- Collaborative Research Center 889 “Cellular Mechanisms of Sensory Processing”GöttingenGermany
- Multiscale Bioimaging of Excitable Cells, Cluster of ExcellenceGöttingenGermany
| |
Collapse
|
13
|
Karagulyan N, Moser T. Synaptic activity is not required for establishing heterogeneity of inner hair cell ribbon synapses. Front Mol Neurosci 2023; 16:1248941. [PMID: 37745283 PMCID: PMC10512025 DOI: 10.3389/fnmol.2023.1248941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/17/2023] [Indexed: 09/26/2023] Open
Abstract
Neural sound encoding in the mammalian cochlea faces the challenge of representing audible sound pressures that vary over six orders of magnitude. The cochlea meets this demand through the use of active micromechanics as well as the diversity and adaptation of afferent neurons and their synapses. Mechanisms underlying neural diversity likely include heterogeneous presynaptic input from inner hair cells (IHCs) to spiral ganglion neurons (SGNs) as well as differences in the molecular profile of SGNs and in their efferent control. Here, we tested whether glutamate release from IHCs, previously found to be critical for maintaining different molecular SGN profiles, is required for establishing heterogeneity of active zones (AZs) in IHCs. We analyzed structural and functional heterogeneity of IHC AZs in mouse mutants with disrupted glutamate release from IHCs due to lack of a vesicular glutamate transporter (Vglut3) or impaired exocytosis due to defective otoferlin. We found the variance of the voltage-dependence of presynaptic Ca2+ influx to be reduced in exocytosis-deficient IHCs of otoferlin mutants. Yet, the spatial gradients of maximal amplitude and voltage-dependence of Ca2+ influx along the pillar-modiolar IHC axis were maintained in both mutants. Further immunohistochemical analysis showed an intact spatial gradient of ribbon size in Vglut3-/- mice. These results indicate that IHC exocytosis and glutamate release are not strictly required for establishing the heterogeneity of IHC AZs.
Collapse
Affiliation(s)
- Nare Karagulyan
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience and Nanophysiology Group, Max Planck Institute of Multidisciplinary Sciences, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
- Göttingen Graduate School for Neurosciences and Molecular Biosciences, University of Göttingen, Göttingen, Germany
- Hertha Sponer College, Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, Göttingen, Germany
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience and Nanophysiology Group, Max Planck Institute of Multidisciplinary Sciences, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
- Göttingen Graduate School for Neurosciences and Molecular Biosciences, University of Göttingen, Göttingen, Germany
- Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
14
|
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: 5] [Impact Index Per Article: 5.0] [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
|
15
|
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: 12] [Impact Index Per Article: 12.0] [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
|
16
|
Grabner CP, Futagi D, Shi J, Bindokas V, Kitano K, Schwartz EA, DeVries SH. Mechanisms of simultaneous linear and nonlinear computations at the mammalian cone photoreceptor synapse. Nat Commun 2023; 14:3486. [PMID: 37328451 PMCID: PMC10276006 DOI: 10.1038/s41467-023-38943-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 05/22/2023] [Indexed: 06/18/2023] Open
Abstract
Neurons enhance their computational power by combining linear and nonlinear transformations in extended dendritic trees. Rich, spatially distributed processing is rarely associated with individual synapses, but the cone photoreceptor synapse may be an exception. Graded voltages temporally modulate vesicle fusion at a cone's ~20 ribbon active zones. Transmitter then flows into a common, glia-free volume where bipolar cell dendrites are organized by type in successive tiers. Using super-resolution microscopy and tracking vesicle fusion and postsynaptic responses at the quantal level in the thirteen-lined ground squirrel, Ictidomys tridecemlineatus, we show that certain bipolar cell types respond to individual fusion events in the vesicle stream while other types respond to degrees of locally coincident events, creating a gradient across tiers that are increasingly nonlinear. Nonlinearities emerge from a combination of factors specific to each bipolar cell type including diffusion distance, contact number, receptor affinity, and proximity to glutamate transporters. Complex computations related to feature detection begin within the first visual synapse.
Collapse
Affiliation(s)
- Chad P Grabner
- Institute for Auditory Neuroscience, University Medical Center Göttingen, 37075, Göttingen, Germany
- Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary Sciences, 37077, Göttingen, Germany
| | - Daiki Futagi
- College of Information Science and Engineering, Ritsumeikan University, Shiga, Japan
- Center for Systems Visual Science, Organization of Science and Technology, Ritsumeikan University, Shiga, Japan
- Ritsumeikan Global Innovation Research Organisation, Ritsumeikan University, Shiga, Japan
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Jun Shi
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Vytas Bindokas
- Dept of Pharmacological and Physiological Sciences, The University of Chicago, Chicago, IL, 60637, USA
| | - Katsunori Kitano
- College of Information Science and Engineering, Ritsumeikan University, Shiga, Japan
- Center for Systems Visual Science, Organization of Science and Technology, Ritsumeikan University, Shiga, Japan
| | - Eric A Schwartz
- Dept of Pharmacological and Physiological Sciences, The University of Chicago, Chicago, IL, 60637, USA
| | - Steven H DeVries
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
17
|
Kaczmarek LK. Modulation of potassium conductances optimizes fidelity of auditory information. Proc Natl Acad Sci U S A 2023; 120:e2216440120. [PMID: 36930599 PMCID: PMC10041146 DOI: 10.1073/pnas.2216440120] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 02/08/2023] [Indexed: 03/18/2023] Open
Abstract
Potassium channels in auditory neurons are rapidly modified by changes in the auditory environment. In response to elevated auditory stimulation, short-term mechanisms such as protein phosphorylation and longer-term mechanisms such as accelerated channel synthesis increase the amplitude of currents that promote high-frequency firing. It has been suggested that this allows neurons to fire at high rates in response to high sound levels. We have carried out simple simulations of the response to postsynaptic neurons to patterns of neurotransmitter release triggered by auditory stimuli. These demonstrate that the amplitudes of potassium currents required for optimal encoding of a low-amplitude auditory signal differ from those for louder sounds. Specifically, the cross-correlation of the output of a neuron with an auditory stimulus is improved by increasing potassium currents as sound amplitude increases. Temporal fidelity for low-frequency stimuli is improved by increasing potassium currents that activate at negative potentials, while that for high-frequency stimuli requires increases in currents that activate at positive membrane potentials. These effects are independent of the firing rate. Moreover, levels of potassium currents that maximize the fidelity of the output of an ensemble of neurons differ from those that maximize fidelity for a single neuron. This suggests that the modulatory mechanisms must coordinate channel activity in groups of neurons or an entire nucleus. The simulations provide an explanation for the modulation of the intrinsic excitability of auditory brainstem neurons by changes in environmental sound levels, and the results may extend to information processing in other neural systems.
Collapse
Affiliation(s)
- Leonard K. Kaczmarek
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT06520
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT06520
| |
Collapse
|
18
|
Shrestha BR, Wu L, Goodrich LV. Runx1 controls auditory sensory neuron diversity in mice. Dev Cell 2023; 58:306-319.e5. [PMID: 36800995 PMCID: PMC10202259 DOI: 10.1016/j.devcel.2023.01.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/28/2022] [Accepted: 01/26/2023] [Indexed: 02/18/2023]
Abstract
Sound stimulus is encoded in mice by three molecularly and physiologically diverse subtypes of sensory neurons, called Ia, Ib, and Ic spiral ganglion neurons (SGNs). Here, we show that the transcription factor Runx1 controls SGN subtype composition in the murine cochlea. Runx1 is enriched in Ib/Ic precursors by late embryogenesis. Upon the loss of Runx1 from embryonic SGNs, more SGNs take on Ia rather than Ib or Ic identities. This conversion was more complete for genes linked to neuronal function than to connectivity. Accordingly, synapses in the Ib/Ic location acquired Ia properties. Suprathreshold SGN responses to sound were enhanced in Runx1CKO mice, confirming the expansion of neurons with Ia-like functional properties. Runx1 deletion after birth also redirected Ib/Ic SGNs toward Ia identity, indicating that SGN identities are plastic postnatally. Altogether, these findings show that diverse neuronal identities essential for normal auditory stimulus coding arise hierarchically and remain malleable during postnatal development.
Collapse
Affiliation(s)
- Brikha R Shrestha
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA 02114, USA; Department of Otolaryngology, Harvard Medical School, Boston, MA 02114, USA.
| | - Lorna Wu
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Lisa V Goodrich
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
19
|
Mittring A, Moser T, Huet AT. Graded optogenetic activation of the auditory pathway for hearing restoration. Brain Stimul 2023; 16:466-483. [PMID: 36702442 PMCID: PMC10159867 DOI: 10.1016/j.brs.2023.01.1671] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
Optogenetic control of neural activity enables innovative approaches to improve functional restoration of diseased sensory and motor systems. For clinical translation to succeed, optogenetic stimulation needs to closely match the coding properties of the targeted neuronal population and employ optimally operating emitters. This requires the customization of channelrhodopsins, emitters and coding strategies. Here, we provide a framework to parametrize optogenetic neural control and apply it to the auditory pathway that requires high temporal fidelity of stimulation. We used a viral gene transfer of ultrafast targeting-optimized Chronos into spiral ganglion neurons (SGNs) of the cochlea of mice. We characterized the light-evoked response by in vivo recordings from individual SGNs and neurons of the anteroventral cochlear nucleus (AVCN) that detect coincident SGN inputs. Our recordings from single SGNs demonstrated that their spike probability can be graded by adjusting the duration of light pulses at constant intensity, which optimally serves efficient laser diode operation. We identified an effective pulse width of 1.6 ms to maximize encoding in SGNs at the maximal light intensity employed here (∼35 mW). Alternatively, SGNs were activated at lower energy thresholds using short light pulses (<1 ms). An upper boundary of optical stimulation rates was identified at 316 Hz, inducing a robust spike rate adaptation that required a few tens of milliseconds to recover. We developed a semi-stochastic stimulation paradigm to rapidly (within minutes) estimate the input/output function from light to SGN firing and approximate the time constant of neuronal integration in the AVCN. By that, our data pave the way to design the sound coding strategies of future optical cochlear implants.
Collapse
Affiliation(s)
- Artur Mittring
- Auditory Circuit Lab, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany; Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany; Auditory Neuroscience and Optogenetics Laboratory, German Primate Center, Göttingen, Germany; Auditory Neuroscience and Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
| | - Antoine Tarquin Huet
- Auditory Circuit Lab, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany; Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany; Auditory Neuroscience and Optogenetics Laboratory, German Primate Center, Göttingen, Germany.
| |
Collapse
|
20
|
Rutherford MA, Bhattacharyya A, Xiao M, Cai HM, Pal I, Rubio ME. GluA3 subunits are required for appropriate assembly of AMPAR GluA2 and GluA4 subunits on cochlear afferent synapses and for presynaptic ribbon modiolar-pillar morphology. eLife 2023; 12:e80950. [PMID: 36648432 PMCID: PMC9891727 DOI: 10.7554/elife.80950] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 01/16/2023] [Indexed: 01/18/2023] Open
Abstract
Cochlear sound encoding depends on α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPARs), but reliance on specific pore-forming subunits is unknown. With 5-week-old male C57BL/6J Gria3-knockout mice (i.e., subunit GluA3KO) we determined cochlear function, synapse ultrastructure, and AMPAR molecular anatomy at ribbon synapses between inner hair cells (IHCs) and spiral ganglion neurons. GluA3KO and wild-type (GluA3WT) mice reared in ambient sound pressure level (SPL) of 55-75 dB had similar auditory brainstem response (ABR) thresholds, wave-1 amplitudes, and latencies. Postsynaptic densities (PSDs), presynaptic ribbons, and synaptic vesicle sizes were all larger on the modiolar side of the IHCs from GluA3WT, but not GluA3KO, demonstrating GluA3 is required for modiolar-pillar synapse differentiation. Presynaptic ribbons juxtaposed with postsynaptic GluA2/4 subunits were similar in quantity, however, lone ribbons were more frequent in GluA3KO and GluA2-lacking synapses were observed only in GluA3KO. GluA2 and GluA4 immunofluorescence volumes were smaller on the pillar side than the modiolar side in GluA3KO, despite increased pillar-side PSD size. Overall, the fluorescent puncta volumes of GluA2 and GluA4 were smaller in GluA3KO than GluA3WT. However, GluA3KO contained less GluA2 and greater GluA4 immunofluorescence intensity relative to GluA3WT (threefold greater mean GluA4:GluA2 ratio). Thus, GluA3 is essential in development, as germline disruption of Gria3 caused anatomical synapse pathology before cochlear output became symptomatic by ABR. We propose the hearing loss in older male GluA3KO mice results from progressive synaptopathy evident in 5-week-old mice as decreased abundance of GluA2 subunits and an increase in GluA2-lacking, GluA4-monomeric Ca2+-permeable AMPARs.
Collapse
Affiliation(s)
- Mark A Rutherford
- Department of Otolaryngology, Washington University School of MedicineSt LouisUnited States
| | - Atri Bhattacharyya
- Department of Otolaryngology, Washington University School of MedicineSt LouisUnited States
| | - Maolei Xiao
- Department of Otolaryngology, Washington University School of MedicineSt LouisUnited States
| | - Hou-Ming Cai
- Department of Neurobiology, University of Pittsburgh School of MedicinePittsburghUnited States
| | - Indra Pal
- Department of Neurobiology, University of Pittsburgh School of MedicinePittsburghUnited States
| | - Maria Eulalia Rubio
- Department of Neurobiology, University of Pittsburgh School of MedicinePittsburghUnited States
- Department of Otolaryngology, University of Pittsburgh School of MedicinePittsburghUnited States
| |
Collapse
|
21
|
Li X, Shi L, Wang L. A review of the mechanisms underlying the role of the GIPC3 gene in hereditary deafness. Front Synaptic Neurosci 2023; 14:1101587. [PMID: 36704659 PMCID: PMC9872657 DOI: 10.3389/fnsyn.2022.1101587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 12/16/2022] [Indexed: 01/09/2023] Open
Abstract
The GAIP interacting protein c terminus (GIPC) genes encode a small family of proteins characterized by centrally located PDZ domains. GIPC3 encodes a 312 amino acid protein. Variants of human GIPC3 are associated with non-syndromic hearing loss. GIPC3 is one of over a hundred different genes with variants causing human deafness. Screening for variants of GIPC3 is essential for early detection of hearing loss in children and eventually treatment of deafness. Accordingly, this paper assesses the status of research developments on the role of GIPC3 in hereditary deafness and the effects of pathogenic variants on the auditory system.
Collapse
Affiliation(s)
- Xinxin Li
- Department of Otolaryngology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Lin Shi
- Department of Otolaryngology, First Affiliated Hospital of Dalian Medical University, Dalian, China,*Correspondence: Lin Shi,
| | - Liang Wang
- National Joint Engineering Laboratory, Stem Cell Clinical Research Center, Regenerative Medicine Center, First Affiliated Hospital of Dalian Medical University, Dalian, China,Liang Wang,
| |
Collapse
|
22
|
Cantu-Guerra HL, Papazian MR, Gorsky AL, Alekos NS, Caccavano A, Karagulyan N, Neef J, Vicini S, Moser T, Coate TM. Cochlear hair cell innervation is dependent on a modulatory function of Semaphorin-3A. Dev Dyn 2023; 252:124-144. [PMID: 36284453 PMCID: PMC9812910 DOI: 10.1002/dvdy.548] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/20/2022] [Accepted: 10/06/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Proper connectivity between type I spiral ganglion neurons (SGNs) and inner hair cells (IHCs) in the cochlea is necessary for conveying sound information to the brain in mammals. Previous studies have shown that type I SGNs are heterogeneous in form, function and synaptic location on IHCs, but factors controlling their patterns of connectivity are not well understood. RESULTS During development, cochlear supporting cells and SGNs express Semaphorin-3A (SEMA3A), a known axon guidance factor. Mice homozygous for a point mutation that attenuates normal SEMA3A repulsive activity (Sema3aK108N ) show cochleae with grossly normal patterns of IHC innervation. However, genetic sparse labeling and three-dimensional reconstructions of individual SGNs show that cochleae from Sema3aK108N mice lacked the normal synaptic distribution of type I SGNs. Additionally, Sema3aK108N cochleae show a disrupted distribution of GLUA2 postsynaptic patches around the IHCs. The addition of SEMA3A-Fc to postnatal cochleae led to increases in SGN branching, similar to the effects of inhibiting glutamate receptors. Ca2+ imaging studies show that SEMA3A-Fc decreases SGN activity. CONCLUSIONS Contrary to the canonical view of SEMA3A as a guidance ligand, our results suggest SEMA3A may regulate SGN excitability in the cochlea, which may influence the morphology and synaptic arrangement of type I SGNs.
Collapse
Affiliation(s)
- Homero L. Cantu-Guerra
- Department of Biology, Georgetown University, Washington,
District of Columbia, USA
- Interdisciplinary Program in Neuroscience, Georgetown
University, Washington, District of Columbia, USA
| | - Michael R. Papazian
- Department of Biology, Georgetown University, Washington,
District of Columbia, USA
| | - Anna L. Gorsky
- Department of Biology, Georgetown University, Washington,
District of Columbia, USA
| | - Nathalie S. Alekos
- Department of Biology, Georgetown University, Washington,
District of Columbia, USA
| | - Adam Caccavano
- Interdisciplinary Program in Neuroscience, Georgetown
University, Washington, District of Columbia, USA
- Department of Pharmacology, Georgetown University School of
Medicine, Washington, District of Columbia, USA
| | - Nare Karagulyan
- Institute for Auditory Neuroscience and InnerEarLab,
University Medical Center, and Auditory Neuroscience & Synaptic Nanophysiology
Group, Max Planck Institute for Multidisciplinary Sciences, and Cluster of
Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of
Excitable Cells” (MBExC), Göttingen, Germany
| | - Jakob Neef
- Institute for Auditory Neuroscience and InnerEarLab,
University Medical Center, and Auditory Neuroscience & Synaptic Nanophysiology
Group, Max Planck Institute for Multidisciplinary Sciences, and Cluster of
Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of
Excitable Cells” (MBExC), Göttingen, Germany
| | - Stefano Vicini
- Interdisciplinary Program in Neuroscience, Georgetown
University, Washington, District of Columbia, USA
- Department of Pharmacology, Georgetown University School of
Medicine, Washington, District of Columbia, USA
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab,
University Medical Center, and Auditory Neuroscience & Synaptic Nanophysiology
Group, Max Planck Institute for Multidisciplinary Sciences, and Cluster of
Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of
Excitable Cells” (MBExC), Göttingen, Germany
| | - Thomas M. Coate
- Department of Biology, Georgetown University, Washington,
District of Columbia, USA
- Interdisciplinary Program in Neuroscience, Georgetown
University, Washington, District of Columbia, USA
| |
Collapse
|
23
|
Liao M, Hu Y, Zhang Y, Wang K, Fang Q, Qi Y, Shen Y, Cheng H, Fu X, Tang M, Sun S, Gao X, Chai R. 3D Ti 3C 2T x MXene-Matrigel with Electroacoustic Stimulation to Promote the Growth of Spiral Ganglion Neurons. ACS NANO 2022; 16:16744-16756. [PMID: 36222600 PMCID: PMC9620407 DOI: 10.1021/acsnano.2c06306] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Cochlear implantation has become the most effective treatment method for patients with profound and total hearing loss. However, its therapeutic efficacy is dependent on the number and normal physiological function of cochlear implant-targeted spiral ganglion neurons (SGNs). Electrical stimulation can be used as an effective cue to regulate the morphology and function of excitatory cells. Therefore, it is important to develop an efficient cochlear implant electroacoustic stimulation (EAS) system to study the behavior of SGNs. In this work, we present an electrical stimulation system constructed by combining a cochlear implant and a conductive Ti3C2Tx MXene-matrigel hydrogel. SGNs were cultured in the Ti3C2Tx MXene-matrigel hydrogel and exposed to electrical stimulation transduced by the cochlear implant. It was demonstrated that low-frequency stimulation promoted the growth cone development and neurite outgrowth of SGNs as well as signal transmission between cells. This work may have potential value for the clinical application of the Ti3C2Tx MXene hydrogel to optimize the postoperative listening effect of cochlear implantation and benefit people with sensorineural hearing loss.
Collapse
Affiliation(s)
- Menghui Liao
- State
Key Laboratory of Bioelectronics, Department of Otolaryngology Head
and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology,
Advanced Institute for Life and Health, Jiangsu Province High-Tech
Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, Jiangsu 210096, China
- Department
of Otorhinolaryngology−Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, China
| | - Yangnan Hu
- State
Key Laboratory of Bioelectronics, Department of Otolaryngology Head
and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology,
Advanced Institute for Life and Health, Jiangsu Province High-Tech
Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, Jiangsu 210096, China
- Department
of Otorhinolaryngology−Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, China
| | - Yuhua Zhang
- State
Key Laboratory of Bioelectronics, Department of Otolaryngology Head
and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology,
Advanced Institute for Life and Health, Jiangsu Province High-Tech
Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, Jiangsu 210096, China
| | - Kaichen Wang
- Chien-Shiung
Wu College, Southeast University, Nanjing, Jiangsu 210096, China
| | - Qiaojun Fang
- State
Key Laboratory of Bioelectronics, Department of Otolaryngology Head
and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology,
Advanced Institute for Life and Health, Jiangsu Province High-Tech
Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, Jiangsu 210096, China
| | - Yanru Qi
- State
Key Laboratory of Bioelectronics, Department of Otolaryngology Head
and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology,
Advanced Institute for Life and Health, Jiangsu Province High-Tech
Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, Jiangsu 210096, China
| | - Yingbo Shen
- Chien-Shiung
Wu College, Southeast University, Nanjing, Jiangsu 210096, China
| | - Hong Cheng
- State
Key Laboratory of Bioelectronics, Department of Otolaryngology Head
and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology,
Advanced Institute for Life and Health, Jiangsu Province High-Tech
Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, Jiangsu 210096, China
| | - Xiaolong Fu
- State
Key Laboratory of Bioelectronics, Department of Otolaryngology Head
and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology,
Advanced Institute for Life and Health, Jiangsu Province High-Tech
Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, Jiangsu 210096, China
| | - Mingliang Tang
- State
Key Laboratory of Bioelectronics, Department of Otolaryngology Head
and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology,
Advanced Institute for Life and Health, Jiangsu Province High-Tech
Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, Jiangsu 210096, China
- Institute for Cardiovascular Science and Department of Cardiovascular Surgery of the First Affiliated
Hospital, Medical College, Soochow University, Suzhou, Jiangsu 215000, China
- Co-Innovation
Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Shan Sun
- ENT
Institute and Department
of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory
of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
| | - Xia Gao
- Department
of Otorhinolaryngology−Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, China
| | - Renjie Chai
- State
Key Laboratory of Bioelectronics, Department of Otolaryngology Head
and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology,
Advanced Institute for Life and Health, Jiangsu Province High-Tech
Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, Jiangsu 210096, China
- Co-Innovation
Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
- Department
of Otolaryngology−Head and Neck Surgery, Sichuan Provincial
People’s Hospital, University of
Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
- Institute
for Stem Cell and Regeneration, Chinese
Academy of Science, Beijing 100101, China
- Beijing
Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing 100069, China
| |
Collapse
|
24
|
Grierson KE, Hickman TT, Liberman MC. Dopaminergic and cholinergic innervation in the mouse cochlea after noise-induced or age-related synaptopathy. Hear Res 2022; 422:108533. [PMID: 35671600 PMCID: PMC11195664 DOI: 10.1016/j.heares.2022.108533] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/11/2022] [Accepted: 05/20/2022] [Indexed: 11/17/2022]
Abstract
Cochlear synaptopathy, the loss of or damage to connections between auditory-nerve fibers (ANFs) and inner hair cells (IHCs), is a prominent pathology in noise-induced and age-related hearing loss. Here, we investigated if degeneration of the olivocochlear (OC) efferent innervation is also a major aspect of the synaptopathic ear, by quantifying the volume and spatial organization of its cholinergic and dopaminergic components, using antibodies to vesicular acetylcholine transporter (VAT) and tyrosine hydroxylase (TH), respectively. CBA/CaJ male mice were examined 1 day to 8 months after a synaptopathic noise exposure, and compared to unexposed age-matched controls and unexposed aged mice at 24-28 months. In normal ears, cholinergic lateral (L)OC terminals were denser in the apical half of the cochlea and on the modiolar side of the inner hair cells (IHCs), where ANFs of low-spontaneous rate are typically found, while dopaminergic terminals were more common in the basal third of the cochlea and, re the IHC axes, were offset towards the habenula with respect to cholinergic terminals. The noise had only small and transient effects on the density of LOC innervation, its spatial organization around the IHC axes, or the extent to which TH and VAT signal were colocalized. The synaptopathic noise also had relatively small and transient effects on cholinergic innervation density in the outer hair cell (OHC) area, which normally peaks in the 16 kHz region and falls monotonically towards higher and lower frequencies. In contrast, in the aged ears, there was massive degeneration of OHC efferents, especially in the apical half of the cochlea, where there was also significant loss of OHCs. In the IHC area, there was significant loss of cholinergic terminals in both apical and basal regions and of dopaminergic innervation in the basal half. Furthermore, the cholinergic terminals in the aged ears spread from their normal clustering near the IHC basolateral pole, where the ANF synapses are found, to positions up and down the IHC somata and regions of the neuropil closer to the habenula. This apparent migration was most striking in the apex, where the hair cell pathology was greatest, and may be a harbinger of impending hair cell death.
Collapse
Affiliation(s)
- Kiera E Grierson
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA, 02114 USA; Dept of Otolaryngology-Head & Neck Surgery, Harvard Medical School, Boston, MA, 02115 USA; Hearing Research Lab, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, AUS
| | - Tyler T Hickman
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA, 02114 USA; Dept of Otolaryngology-Head & Neck Surgery, Harvard Medical School, Boston, MA, 02115 USA.
| | - M Charles Liberman
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA, 02114 USA; Dept of Otolaryngology-Head & Neck Surgery, Harvard Medical School, Boston, MA, 02115 USA
| |
Collapse
|
25
|
Hou S, Zhang J, Wu Y, Junmin C, Yuyu H, He B, Yang Y, Hong Y, Chen J, Yang J, Li S. FGF22 deletion causes hidden hearing loss by affecting the function of inner hair cell ribbon synapses. Front Mol Neurosci 2022; 15:922665. [PMID: 35966010 PMCID: PMC9366910 DOI: 10.3389/fnmol.2022.922665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 07/04/2022] [Indexed: 12/04/2022] Open
Abstract
Ribbon synapses are important structures in transmitting auditory signals from the inner hair cells (IHCs) to their corresponding spiral ganglion neurons (SGNs). Over the last few decades, deafness has been primarily attributed to the deterioration of cochlear hair cells rather than ribbon synapses. Hearing dysfunction that cannot be detected by the hearing threshold is defined as hidden hearing loss (HHL). The relationship between ribbon synapses and FGF22 deletion remains unknown. In this study, we used a 6-week-old FGF22 knockout mice model (Fgf22–/–) and mainly focused on alteration in ribbon synapses by applying the auditory brainstem response (ABR) test, the immunofluorescence staining, the patch-clamp recording, and quantitative real-time PCR. In Fgf22–/– mice, we found the decreased amplitude of ABR wave I, the reduced vesicles of ribbon synapses, and the decreased efficiency of exocytosis, which was suggested by a decrease in the capacitance change. Quantitative real-time PCR revealed that Fgf22–/– led to dysfunction in ribbon synapses by downregulating SNAP-25 and Gipc3 and upregulating MEF2D expression, which was important for the maintenance of ribbon synapses’ function. Our research concluded that FGF22 deletion caused HHL by affecting the function of IHC ribbon synapses and may offer a novel therapeutic target to meet an ever-growing demand for deafness treatment.
Collapse
Affiliation(s)
- Shule Hou
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Jifang Zhang
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Yan Wu
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Chen Junmin
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Huang Yuyu
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Baihui He
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Yan Yang
- Liaoning Medical Device Test Institute, Shenyang, China
| | - Yuren Hong
- Laboratory of Electron Microscope Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiarui Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Shanghai Children’s Hospital, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Jiarui Chen,
| | - Jun Yang
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
- Jun Yang,
| | - Shuna Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
- Shuna Li,
| |
Collapse
|
26
|
Signatures of cochlear processing in neuronal coding of auditory information. Mol Cell Neurosci 2022; 120:103732. [PMID: 35489636 DOI: 10.1016/j.mcn.2022.103732] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 11/22/2022] Open
Abstract
The vertebrate ear is endowed with remarkable perceptual capabilities. The faintest sounds produce vibrations of magnitudes comparable to those generated by thermal noise and can nonetheless be detected through efficient amplification of small acoustic stimuli. Two mechanisms have been proposed to underlie such sound amplification in the mammalian cochlea: somatic electromotility and active hair-bundle motility. These biomechanical mechanisms may work in concert to tune auditory sensitivity. In addition to amplitude sensitivity, the hearing system shows exceptional frequency discrimination allowing mammals to distinguish complex sounds with great accuracy. For instance, although the wide hearing range of humans encompasses frequencies from 20 Hz to 20 kHz, our frequency resolution extends to one-thirtieth of the interval between successive keys on a piano. In this article, we review the different cochlear mechanisms underlying sound encoding in the auditory system, with a particular focus on the frequency decomposition of sounds. The relation between peak frequency of activation and location along the cochlea - known as tonotopy - arises from multiple gradients in biophysical properties of the sensory epithelium. Tonotopic mapping represents a major organizational principle both in the peripheral hearing system and in higher processing levels and permits the spectral decomposition of complex tones. The ribbon synapses connecting sensory hair cells to auditory afferents and the downstream spiral ganglion neurons are also tuned to process periodic stimuli according to their preferred frequency. Though sensory hair cells and neurons necessarily filter signals beyond a few kHz, many animals can hear well beyond this range. We finally describe how the cochlear structure shapes the neural code for further processing in order to send meaningful information to the brain. Both the phase-locked response of auditory nerve fibers and tonotopy are key to decode sound frequency information and place specific constraints on the downstream neuronal network.
Collapse
|
27
|
Metabotropic Glutamate Receptors at Ribbon Synapses in the Retina and Cochlea. Cells 2022; 11:cells11071097. [PMID: 35406660 PMCID: PMC8998116 DOI: 10.3390/cells11071097] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/17/2022] [Accepted: 03/22/2022] [Indexed: 02/01/2023] Open
Abstract
Our senses define our view of the world. They allow us to adapt to environmental stimuli and are essential for communication and social behaviour. For most humans, seeing and hearing are central senses for their daily life. Our eyes and ears respond to an extraordinary broad range of stimuli covering about 12 log units of light intensity or acoustic power, respectively. The cellular basis is represented by sensory cells (photoreceptors in the retina and inner hair cells in the cochlea) that convert sensory inputs into electrical signals. Photoreceptors and inner hair cells have developed a specific pre-synaptic structure, termed synaptic ribbon, that is decorated with numerous vesicles filled with the excitatory neurotransmitter glutamate. At these ribbon synapses, glutamatergic signal transduction is guided by distinct sets of metabotropic glutamate receptors (mGluRs). MGluRs belong to group II and III of the receptor classification can inhibit neuronal activity, thus protecting neurons from overstimulation and subsequent degeneration. Consequently, dysfunction of mGluRs is associated with vision and hearing disorders. In this review, we introduce the principle characteristics of ribbon synapses and describe group II and III mGluRs in these fascinating structures in the retina and cochlea.
Collapse
|
28
|
Vogl C, Neef J, Wichmann C. Methods for multiscale structural and functional analysis of the mammalian cochlea. Mol Cell Neurosci 2022; 120:103720. [DOI: 10.1016/j.mcn.2022.103720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 02/13/2022] [Accepted: 03/08/2022] [Indexed: 01/11/2023] Open
|
29
|
Wichmann C, Kuner T. Heterogeneity of glutamatergic synapses: cellular mechanisms and network consequences. Physiol Rev 2022; 102:269-318. [PMID: 34727002 DOI: 10.1152/physrev.00039.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chemical synapses are commonly known as a structurally and functionally highly diverse class of cell-cell contacts specialized to mediate communication between neurons. They represent the smallest "computational" unit of the brain and are typically divided into excitatory and inhibitory as well as modulatory categories. These categories are subdivided into diverse types, each representing a different structure-function repertoire that in turn are thought to endow neuronal networks with distinct computational properties. The diversity of structure and function found among a given category of synapses is referred to as heterogeneity. The main building blocks for this heterogeneity are synaptic vesicles, the active zone, the synaptic cleft, the postsynaptic density, and glial processes associated with the synapse. Each of these five structural modules entails a distinct repertoire of functions, and their combination specifies the range of functional heterogeneity at mammalian excitatory synapses, which are the focus of this review. We describe synapse heterogeneity that is manifested on different levels of complexity ranging from the cellular morphology of the pre- and postsynaptic cells toward the expression of different protein isoforms at individual release sites. We attempt to define the range of structural building blocks that are used to vary the basic functional repertoire of excitatory synaptic contacts and discuss sources and general mechanisms of synapse heterogeneity. Finally, we explore the possible impact of synapse heterogeneity on neuronal network function.
Collapse
Affiliation(s)
- Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, InnerEarLab and Institute for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Thomas Kuner
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg, Germany
| |
Collapse
|
30
|
Chakrabarti R, Jaime Tobón LM, Slitin L, Redondo Canales M, Hoch G, Slashcheva M, Fritsch E, Bodensiek K, Özçete ÖD, Gültas M, Michanski S, Opazo F, Neef J, Pangrsic T, Moser T, Wichmann C. Optogenetics and electron tomography for structure-function analysis of cochlear ribbon synapses. eLife 2022; 11:79494. [PMID: 36562477 PMCID: PMC9908081 DOI: 10.7554/elife.79494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
Ribbon synapses of cochlear inner hair cells (IHCs) are specialized to indefatigably transmit sound information at high rates. To understand the underlying mechanisms, structure-function analysis of the active zone (AZ) of these synapses is essential. Previous electron microscopy studies of synaptic vesicle (SV) dynamics at the IHC AZ used potassium stimulation, which limited the temporal resolution to minutes. Here, we established optogenetic IHC stimulation followed by quick freezing within milliseconds and electron tomography to study the ultrastructure of functional synapse states with good temporal resolution in mice. We characterized optogenetic IHC stimulation by patch-clamp recordings from IHCs and postsynaptic boutons revealing robust IHC depolarization and neurotransmitter release. Ultrastructurally, the number of docked SVs increased upon short (17-25 ms) and long (48-76 ms) light stimulation paradigms. We did not observe enlarged SVs or other morphological correlates of homotypic fusion events. Our results indicate a rapid recruitment of SVs to the docked state upon stimulation and suggest that univesicular release prevails as the quantal mechanism of exocytosis at IHC ribbon synapses.
Collapse
Affiliation(s)
- Rituparna Chakrabarti
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Center for Biostructural Imaging of Neurodegeneration, University Medical Center GöttingenGöttingenGermany,Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany
| | - Lina María Jaime Tobón
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany,Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Auditory Neuroscience & Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Loujin Slitin
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Center for Biostructural Imaging of Neurodegeneration, University Medical Center GöttingenGöttingenGermany,Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany
| | - Magdalena Redondo Canales
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Center for Biostructural Imaging of Neurodegeneration, University Medical Center GöttingenGöttingenGermany,Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany
| | - Gerhard Hoch
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Auditory Neuroscience & Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Marina Slashcheva
- Göttingen Graduate School for Neuroscience and Molecular Biosciences, University of GöttingenGöttingenGermany
| | - Elisabeth Fritsch
- Göttingen Graduate School for Neuroscience and Molecular Biosciences, University of GöttingenGöttingenGermany
| | - Kai Bodensiek
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany
| | - Özge Demet Özçete
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany,Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Auditory Neuroscience & Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Mehmet Gültas
- Faculty of Agriculture, South Westphalia University of Applied SciencesSoestGermany
| | - Susann Michanski
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Center for Biostructural Imaging of Neurodegeneration, University Medical Center GöttingenGöttingenGermany,Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany
| | - Felipe Opazo
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center GöttingenGöttingenGermany,NanoTag Biotechnologies GmbHGöttingenGermany,Institute of Neuro- and Sensory Physiology, University Medical Center GöttingenGöttingenGermany
| | - Jakob Neef
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany,Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Auditory Neuroscience & Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Tina Pangrsic
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany,Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Auditory Neuroscience & Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany,Multiscale Bioimaging: from Molecular Machines to Networks of Excitable CellsGöttingenGermany,Synaptic Physiology of Mammalian Vestibular Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany
| | - Tobias Moser
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany,Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Auditory Neuroscience & Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany,Multiscale Bioimaging: from Molecular Machines to Networks of Excitable CellsGöttingenGermany
| | - Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center GöttingenGöttingenGermany,Center for Biostructural Imaging of Neurodegeneration, University Medical Center GöttingenGöttingenGermany,Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing"GöttingenGermany,Multiscale Bioimaging: from Molecular Machines to Networks of Excitable CellsGöttingenGermany
| |
Collapse
|
31
|
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: 36] [Impact Index Per Article: 12.0] [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
|
32
|
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
|
33
|
Ikäheimo K, Herranen A, Iivanainen V, Lankinen T, Aarnisalo AA, Sivonen V, Patel KA, Demir K, Saarma M, Lindahl M, Pirvola U. MANF supports the inner hair cell synapse and the outer hair cell stereocilia bundle in the cochlea. Life Sci Alliance 2021; 5:5/2/e202101068. [PMID: 34815294 PMCID: PMC8616558 DOI: 10.26508/lsa.202101068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 11/08/2021] [Accepted: 11/08/2021] [Indexed: 11/29/2022] Open
Abstract
The authors show in the mouse how the auditory hair cell structural maintenance is perturbed by the inactivation of Manf and the concomitant ER stress, causing early-onset, progressive hearing loss. Failure in the structural maintenance of the hair cell stereocilia bundle and ribbon synapse causes hearing loss. Here, we have studied how ER stress elicits hair cell pathology, using mouse models with inactivation of Manf (mesencephalic astrocyte-derived neurotrophic factor), encoding an ER-homeostasis-promoting protein. From hearing onset, Manf deficiency caused disarray of the outer hair cell stereocilia bundle and reduced cochlear sound amplification capability throughout the tonotopic axis. In high-frequency outer hair cells, the pathology ended in molecular changes in the stereocilia taper region and in strong stereocilia fusion. In high-frequency inner hair cells, Manf deficiency degraded ribbon synapses. The altered phenotype strongly depended on the mouse genetic background. Altogether, the failure in the ER homeostasis maintenance induced early-onset stereociliopathy and synaptopathy and accelerated the effect of genetic causes driving age-related hearing loss. Correspondingly, MANF mutation in a human patient induced severe sensorineural hearing loss from a young age onward. Thus, we present MANF as a novel protein and ER stress as a mechanism that regulate auditory hair cell maintenance in both mice and humans.
Collapse
Affiliation(s)
- Kuu Ikäheimo
- Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, Finland
| | - Anni Herranen
- Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, Finland
| | - Vilma Iivanainen
- Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, Finland
| | - Tuuli Lankinen
- Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, Finland
| | - Antti A Aarnisalo
- Department of Otorhinolaryngology-Head and Neck Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Ville Sivonen
- Department of Otorhinolaryngology-Head and Neck Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Kashyap A Patel
- Institute of Biomedical and Clinical Science, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Korcan Demir
- Department of Paediatric Endocrinology, Dokuz Eylul University, Izmir, Turkey
| | - Mart Saarma
- Institute of Biotechnology, HILIFE Unit, University of Helsinki, Helsinki, Finland
| | - Maria Lindahl
- Institute of Biotechnology, HILIFE Unit, University of Helsinki, Helsinki, Finland
| | - Ulla Pirvola
- Molecular and Integrative Biosciences Research Programme, University of Helsinki, Helsinki, Finland
| |
Collapse
|
34
|
Joshi Y, Petit CP, Miot S, Guillet M, Sendin G, Bourien J, Wang J, Pujol R, El Mestikawy S, Puel JL, Nouvian R. VGLUT3-p.A211V variant fuses stereocilia bundles and elongates synaptic ribbons. J Physiol 2021; 599:5397-5416. [PMID: 34783032 PMCID: PMC9299590 DOI: 10.1113/jp282181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/01/2021] [Indexed: 11/16/2022] Open
Abstract
Abstract DFNA25 is an autosomal‐dominant and progressive form of human deafness caused by mutations in the SLC17A8 gene, which encodes the vesicular glutamate transporter type 3 (VGLUT3). To resolve the mechanisms underlying DFNA25, we studied phenotypes of mice harbouring the p.A221V mutation in humans (corresponding to p.A224V in mice). Using auditory brainstem response and distortion product otoacoustic emissions, we showed progressive hearing loss with intact cochlear amplification in the VGLUT3A224V/A224V mouse. The summating potential was reduced, indicating the alteration of inner hair cell (IHC) receptor potential. Scanning electron microscopy examinations demonstrated the collapse of stereocilia bundles in IHCs, leaving those from outer hair cells unaffected. In addition, IHC ribbon synapses underwent structural and functional modifications at later stages. Using super‐resolution microscopy, we observed oversized synaptic ribbons and patch‐clamp membrane capacitance measurements showed an increase in the rate of the sustained releasable pool exocytosis. These results suggest that DFNA25 stems from a failure in the mechano‐transduction followed by a change in synaptic transfer. The VGLUT3A224V/A224V mouse model opens the way to a deeper understanding and to a potential treatment for DFNA25. Key points The vesicular glutamate transporter type 3 (VGLUT3) loads glutamate into the synaptic vesicles of auditory sensory cells, the inner hair cells (IHCs). The VGLUT3‐p.A211V variant is associated with human deafness DFNA25. Mutant mice carrying the VGLUT3‐p.A211V variant show progressive hearing loss. IHCs from mutant mice harbour distorted stereocilary bundles, which detect incoming sound stimulation, followed by oversized synaptic ribbons, which release glutamate onto the afferent nerve fibres. These results suggest that DFNA25 stems from the failure of auditory sensory cells to faithfully transduce acoustic cues into neural messages.
Collapse
Affiliation(s)
- Yuvraj Joshi
- INM, Univ Montpellier, INSERM, Montpellier, France
| | | | - Stéphanie Miot
- INM, Univ Montpellier, INSERM, Montpellier, France.,Sorbonne Universités, Université Pierre et Marie Curie UM 119, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Paris, France
| | | | | | | | - Jing Wang
- INM, Univ Montpellier, INSERM, Montpellier, France
| | - Rémy Pujol
- INM, Univ Montpellier, INSERM, Montpellier, France
| | - Salah El Mestikawy
- Sorbonne Universités, Université Pierre et Marie Curie UM 119, Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Paris, France.,Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada
| | | | - Régis Nouvian
- INM, Univ Montpellier, INSERM, Montpellier, France.,INM, Univ Montpellier, INSERM, CNRS, Montpellier, France
| |
Collapse
|
35
|
Ingham NJ, Banafshe N, Panganiban C, Crunden JL, Chen J, Lewis MA, Steel KP. Inner hair cell dysfunction in Klhl18 mutant mice leads to low frequency progressive hearing loss. PLoS One 2021; 16:e0258158. [PMID: 34597341 PMCID: PMC8486144 DOI: 10.1371/journal.pone.0258158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/19/2021] [Indexed: 12/30/2022] Open
Abstract
Age-related hearing loss in humans (presbycusis) typically involves impairment of high frequency sensitivity before becoming progressively more severe at lower frequencies. Pathologies initially affecting lower frequency regions of hearing are less common. Here we describe a progressive, predominantly low-frequency recessive hearing impairment in two mutant mouse lines carrying different mutant alleles of the Klhl18 gene: a spontaneous missense mutation (Klhl18lowf) and a targeted mutation (Klhl18tm1a(KOMP)Wtsi). Both males and females were studied, and the two mutant lines showed similar phenotypes. Threshold for auditory brainstem responses (ABR; a measure of auditory nerve and brainstem neural activity) were normal at 3 weeks old but showed progressive increases from 4 weeks onwards. In contrast, distortion product otoacoustic emission (DPOAE) sensitivity and amplitudes (a reflection of cochlear outer hair cell function) remained normal in mutants. Electrophysiological recordings from the round window of Klhl18lowf mutants at 6 weeks old revealed 1) raised compound action potential thresholds that were similar to ABR thresholds, 2) cochlear microphonic potentials that were normal compared with wildtype and heterozygous control mice and 3) summating potentials that were reduced in amplitude compared to control mice. Scanning electron microscopy showed that Klhl18lowf mutant mice had abnormally tapering of the tips of inner hair cell stereocilia in the apical half of the cochlea while their synapses appeared normal. These results suggest that Klhl18 is necessary to maintain inner hair cell stereocilia and normal inner hair cell function at low frequencies.
Collapse
Affiliation(s)
- Neil J. Ingham
- Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom
| | - Navid Banafshe
- Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom
| | - Clarisse Panganiban
- Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom
| | - Julia L. Crunden
- Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom
| | - Jing Chen
- Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom
| | - Morag A. Lewis
- Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom
| | - Karen P. Steel
- Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom
| |
Collapse
|
36
|
Caus Capdevila MQ, Sienknecht UJ, Köppl C. Developmental maturation of presynaptic ribbon numbers in chicken basilar-papilla hair cells and its perturbation by long-term overexpression of Wnt9a. Dev Neurobiol 2021; 81:817-832. [PMID: 34309221 DOI: 10.1002/dneu.22845] [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: 03/25/2021] [Revised: 06/20/2021] [Accepted: 07/15/2021] [Indexed: 11/07/2022]
Abstract
The avian basilar papilla is a valuable model system for exploring the developmental determination and differentiation of sensory hair cells and their innervation. In the mature basilar papilla, hair cells form a well-known continuum between two extreme types-tall and short hair cells-that differ strikingly in their innervation. Previous work identified Wnt9a as a crucial factor in this differentiation. Here, we quantified the number and volume of immunolabelled presynaptic ribbons in tall and short hair cells of chickens, from developmental stages shortly after ribbons first appear to the mature posthatching condition. Two longitudinal locations were sampled, responding to best frequencies of approximately 1 kHz and approximately 5.5 kHz when mature. We found significant reductions of ribbon number during normal development in the tall-hair-cell domains, but stable, low numbers in the short-hair-cell domains. Exposing developing hair cells to continuous, excessive Wnt9a levels (through virus-mediated overexpression) led to transiently abnormal high numbers of ribbons and a delayed reduction of ribbon numbers at all sampled locations. Thus, (normally) short-hair-cell domains also showed tall-hair-cell like behaviour, confirming previous findings (Munnamalai et al., 2017). However, at 3 weeks posthatching, ribbon numbers had decreased to the location-specific typical values of control hair cells at all sampled locations. Furthermore, as shown previously, mature hair cells at the basal, high-frequency location harboured larger ribbons than more apically located hair cells. This was true for both normal and Wnt9a-overexposed basilar papillae.
Collapse
Affiliation(s)
- M Queralt Caus Capdevila
- Cluster of Excellence "Hearing4all" and Research Centre Neurosensory Science, Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Ulrike J Sienknecht
- Cluster of Excellence "Hearing4all" and 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" and Research Centre Neurosensory Science, Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
37
|
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
|
38
|
Payne SA, Joens MS, Chung H, Skigen N, Frank A, Gattani S, Vaughn K, Schwed A, Nester M, Bhattacharyya A, Iyer G, Davis B, Carlquist J, Patel H, Fitzpatrick JAJ, Rutherford MA. Maturation of Heterogeneity in Afferent Synapse Ultrastructure in the Mouse Cochlea. Front Synaptic Neurosci 2021; 13:678575. [PMID: 34220482 PMCID: PMC8248813 DOI: 10.3389/fnsyn.2021.678575] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/11/2021] [Indexed: 12/11/2022] Open
Abstract
Auditory nerve fibers (ANFs) innervating the same inner hair cell (IHC) may have identical frequency tuning but different sound response properties. In cat and guinea pig, ANF response properties correlate with afferent synapse morphology and position on the IHC, suggesting a causal structure-function relationship. In mice, this relationship has not been fully characterized. Here we measured the emergence of synaptic morphological heterogeneities during maturation of the C57BL/6J mouse cochlea by comparing postnatal day 17 (p17, ∼3 days after hearing onset) with p34, when the mouse cochlea is mature. Using serial block face scanning electron microscopy and three-dimensional reconstruction we measured the size, shape, vesicle content, and position of 70 ribbon synapses from the mid-cochlea. Several features matured over late postnatal development. From p17 to p34, presynaptic densities (PDs) and post-synaptic densities (PSDs) became smaller on average (PDs: 0.75 to 0.33; PSDs: 0.58 to 0.31 μm2) and less round as their short axes shortened predominantly on the modiolar side, from 770 to 360 nm. Membrane-associated synaptic vesicles decreased in number from 53 to 30 per synapse from p17 to p34. Anatomical coupling, measured as PSD to ribbon distance, tightened predominantly on the pillar side. Ribbons became less spherical as long-axes lengthened only on the modiolar side of the IHC, from 372 to 541 nm. A decreasing gradient of synaptic ribbon size along the modiolar-pillar axis was detected only at p34 after aligning synapses of adjacent IHCs to a common reference frame (median volumes in nm3 × 106: modiolar 4.87; pillar 2.38). The number of ribbon-associated synaptic vesicles scaled with ribbon size (range 67 to 346 per synapse at p34), thus acquiring a modiolar-pillar gradient at p34, but overall medians were similar at p17 (120) and p34 (127), like ribbon surface area (0.36 vs. 0.34 μm2). PD and PSD morphologies were tightly correlated to each other at individual synapses, more so at p34 than p17, but not to ribbon morphology. These observations suggest that PDs and PSDs mature according to different cues than ribbons, and that ribbon size may be more influenced by cues from the IHC than the surrounding tissue.
Collapse
Affiliation(s)
- Shelby A. Payne
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States
| | - Matthew S. Joens
- Center for Cellular Imaging, Washington University in St. Louis, St. Louis, MO, United States
- TESCAN USA, Inc., Warrendale, PA, United States
| | - Heather Chung
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States
- Department of Biology, Washington University in St. Louis, St. Louis, MO, United States
| | - Natalie Skigen
- Department of Biology, Washington University in St. Louis, St. Louis, MO, United States
| | - Adam Frank
- Department of Biology, Washington University in St. Louis, St. Louis, MO, United States
| | - Sonali Gattani
- Department of Biology, Washington University in St. Louis, St. Louis, MO, United States
| | - Kya Vaughn
- Department of Biology, Washington University in St. Louis, St. Louis, MO, United States
| | - Allison Schwed
- Graduate Program in Audiology and Communications Sciences, Washington University School of Medicine, St. Louis, MO, United States
| | - Matt Nester
- Department of Biology, Washington University in St. Louis, St. Louis, MO, United States
| | - Atri Bhattacharyya
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States
- Department of Biology, Washington University in St. Louis, St. Louis, MO, United States
| | - Guhan Iyer
- Department of Biology, Washington University in St. Louis, St. Louis, MO, United States
| | - Bethany Davis
- Graduate Program in Audiology and Communications Sciences, Washington University School of Medicine, St. Louis, MO, United States
| | - Jason Carlquist
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States
- Department of Biology, Washington University in St. Louis, St. Louis, MO, United States
| | - Honey Patel
- Department of Biology, Washington University in St. Louis, St. Louis, MO, United States
| | - James A. J. Fitzpatrick
- Center for Cellular Imaging, Washington University in St. Louis, St. Louis, MO, United States
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, United States
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, United States
- Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, MO, United States
| | - Mark A. Rutherford
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
39
|
Niwa M, Young ED, Glowatzki E, Ricci AJ. Functional subgroups of cochlear inner hair cell ribbon synapses differently modulate their EPSC properties in response to stimulation. J Neurophysiol 2021; 125:2461-2479. [PMID: 33949873 PMCID: PMC8285665 DOI: 10.1152/jn.00452.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Spiral ganglion neurons (SGNs) form single synapses on inner hair cells (IHCs), transforming sound-induced IHC receptor potentials into trains of action potentials. SGN neurons are classified by spontaneous firing rates as well as their threshold response to sound intensity levels. We investigated the hypothesis that synaptic specializations underlie mouse SGN response properties and vary with pillar versus modiloar synapse location around the hair cell. Depolarizing hair cells with 40 mM K+ increased the rate of postsynaptic responses. Pillar synapses matured later than modiolar synapses. Excitatory postsynaptic current (EPSC) amplitude, area, and number of underlying events per EPSC were similar between synapse locations at steady state. However, modiolar synapses produced larger monophasic EPSCs when EPSC rates were low and EPSCs became more multiphasic and smaller in amplitude when rates were higher, while pillar synapses produced more monophasic and larger EPSCs when the release rates were higher. We propose that pillar and modiolar synapses have different operating points. Our data provide insight into underlying mechanisms regulating EPSC generation. NEW & NOTEWORTHY Data presented here provide the first direct functional evidence of late synaptic maturation of the hair cell- spiral ganglion neuron synapse, where pillar synapses mature after postnatal day 20. Data identify a presynaptic difference in release during stimulation. This difference may in part drive afferent firing properties.
Collapse
Affiliation(s)
- Mamiko Niwa
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California.,Center for Hearing and Balance, Johns Hopkins School of Medicine, Baltimore, Maryland.,Department of Otolaryngology-Head, and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Eric D Young
- Center for Hearing and Balance, Johns Hopkins School of Medicine, Baltimore, Maryland.,Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, Maryland.,Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Elisabeth Glowatzki
- Center for Hearing and Balance, Johns Hopkins School of Medicine, Baltimore, Maryland.,Department of Otolaryngology-Head, and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland.,Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Anthony J Ricci
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California.,Department of Molecular and Cellular Physiology, Stanford University, Stanford, California
| |
Collapse
|
40
|
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
|
41
|
Piccolo is essential for the maintenance of mouse retina but not cochlear hair cell function. Aging (Albany NY) 2021; 13:11678-11695. [PMID: 33882456 PMCID: PMC8109093 DOI: 10.18632/aging.202861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/16/2021] [Indexed: 12/14/2022]
Abstract
Piccolo is a presynaptic protein with high conservation among different species, and the expression of Piccolo is extensive in vertebrates. Recently, a small fragment of Piccolo (Piccolino), arising due to the incomplete splicing of intron 5/6, was found to be present in the synapses of retinas and cochleae. However, the comprehensive function of Piccolo in the retina and cochlea remains unclear. In this study, we generated Piccolo knockout mice using CRISPR-Cas9 technology to explore the function of Piccolo. Unexpectedly, whereas no abnormalities were found in the cochlear hair cells of the mutant mice, significant differences were found in the retinas, in which two layers (the outer nuclear layer and the outer plexiform layer) were absent. Additionally, the amplitudes of electroretinograms were significantly reduced and pigmentation was observed in the fundoscopy of the mutant mouse retinas. The expression levels of Bassoon, a homolog of Piccolo, as well as synapse-associated proteins CtBP1, CtBP2, Kif3A, and Rim1 were down-regulated. The numbers of ribbon synapses in the retinas of the mutant mice were also reduced. Altogether, the phenotype of Piccolo-/- mice resembled the symptoms of retinitis pigmentosa (RP) in humans, suggesting Piccolo might be a candidate gene of RP and indicates Piccolo knockout mice are a good model for elucidating the molecular mechanisms of RP.
Collapse
|
42
|
Colón-Cruz L, Rodriguez-Morales R, Santana-Cruz A, Cantres-Velez J, Torrado-Tapias A, Lin SJ, Yudowski G, Kensler R, Marie B, Burgess SM, Renaud O, Varshney GK, Behra M. Cnr2 Is Important for Ribbon Synapse Maturation and Function in Hair Cells and Photoreceptors. Front Mol Neurosci 2021; 14:624265. [PMID: 33958989 PMCID: PMC8093779 DOI: 10.3389/fnmol.2021.624265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/24/2021] [Indexed: 02/04/2023] Open
Abstract
The role of the cannabinoid receptor 2 (CNR2) is still poorly described in sensory epithelia. We found strong cnr2 expression in hair cells (HCs) of the inner ear and the lateral line (LL), a superficial sensory structure in fish. Next, we demonstrated that sensory synapses in HCs were severely perturbed in larvae lacking cnr2. Appearance and distribution of presynaptic ribbons and calcium channels (Cav1.3) were profoundly altered in mutant animals. Clustering of membrane-associated guanylate kinase (MAGUK) in post-synaptic densities (PSDs) was also heavily affected, suggesting a role for cnr2 for maintaining the sensory synapse. Furthermore, vesicular trafficking in HCs was strongly perturbed suggesting a retrograde action of the endocannabinoid system (ECs) via cnr2 that was modulating HC mechanotransduction. We found similar perturbations in retinal ribbon synapses. Finally, we showed that larval swimming behaviors after sound and light stimulations were significantly different in mutant animals. Thus, we propose that cnr2 is critical for the processing of sensory information in the developing larva.
Collapse
Affiliation(s)
- Luis Colón-Cruz
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | - Roberto Rodriguez-Morales
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | - Alexis Santana-Cruz
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | - Juan Cantres-Velez
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | - Aranza Torrado-Tapias
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | - Sheng-Jia Lin
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Guillermo Yudowski
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico.,School of Medicine, Institute of Neurobiology, University of Puerto Rico, San Juan, Puerto Rico
| | - Robert Kensler
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | - Bruno Marie
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico.,School of Medicine, Institute of Neurobiology, University of Puerto Rico, San Juan, Puerto Rico
| | - Shawn M Burgess
- Developmental Genomics Section, Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Olivier Renaud
- Cell and Tissue Imaging Facility (PICT-IBiSA, FranceBioImaging), Institut Curie, PSL Research University, U934/UMR3215, Paris, France
| | - Gaurav K Varshney
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Martine Behra
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| |
Collapse
|
43
|
Effertz T, Moser T, Oliver D. Recent advances in cochlear hair cell nanophysiology: subcellular compartmentalization of electrical signaling in compact sensory cells. Fac Rev 2021; 9:24. [PMID: 33659956 PMCID: PMC7886071 DOI: 10.12703/r/9-24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In recent years, genetics, physiology, and structural biology have advanced into the molecular details of the sensory physiology of auditory hair cells. Inner hair cells (IHCs) and outer hair cells (OHCs) mediate two key functions: active amplification and non-linear compression of cochlear vibrations by OHCs and sound encoding by IHCs at their afferent synapses with the spiral ganglion neurons. OHCs and IHCs share some molecular physiology, e.g. mechanotransduction at the apical hair bundles, ribbon-type presynaptic active zones, and ionic conductances in the basolateral membrane. Unique features enabling their specific function include prestin-based electromotility of OHCs and indefatigable transmitter release at the highest known rates by ribbon-type IHC active zones. Despite their compact morphology, the molecular machineries that either generate electrical signals or are driven by these signals are essentially all segregated into local subcellular structures. This review provides a brief account on recent insights into the molecular physiology of cochlear hair cells with a specific focus on organization into membrane domains.
Collapse
Affiliation(s)
- Thomas Effertz
- InnerEarLab, Department of Otorhinolaryngology, University Medical Center Göttingen, 37099 Göttingen, Germany
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37099 Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany
- Synaptic Nanophysiology Group, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
- Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, 37075 Göttingen, Germany
| | - Dominik Oliver
- Institute for Physiology and Pathophysiology, Philipps University, Deutschhausstraße 2, 35037 Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Universities of Marburg and Giessen, Germany
- DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodelling, GRK 2213, Philipps University, Marburg, Germany
| |
Collapse
|
44
|
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
|
45
|
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
|
46
|
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
|
47
|
Petitpré C, Bourien J, Wu H, Diuba A, Puel JL, Lallemend F. Genetic and functional diversity of primary auditory afferents. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
48
|
Voorn RA, Vogl C. Molecular Assembly and Structural Plasticity of Sensory Ribbon Synapses-A Presynaptic Perspective. Int J Mol Sci 2020; 21:E8758. [PMID: 33228215 PMCID: PMC7699581 DOI: 10.3390/ijms21228758] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/13/2022] Open
Abstract
In the mammalian cochlea, specialized ribbon-type synapses between sensory inner hair cells (IHCs) and postsynaptic spiral ganglion neurons ensure the temporal precision and indefatigability of synaptic sound encoding. These high-through-put synapses are presynaptically characterized by an electron-dense projection-the synaptic ribbon-which provides structural scaffolding and tethers a large pool of synaptic vesicles. While advances have been made in recent years in deciphering the molecular anatomy and function of these specialized active zones, the developmental assembly of this presynaptic interaction hub remains largely elusive. In this review, we discuss the dynamic nature of IHC (pre-) synaptogenesis and highlight molecular key players as well as the transport pathways underlying this process. Since developmental assembly appears to be a highly dynamic process, we further ask if this structural plasticity might be maintained into adulthood, how this may influence the functional properties of a given IHC synapse and how such plasticity could be regulated on the molecular level. To do so, we take a closer look at other ribbon-bearing systems, such as retinal photoreceptors and pinealocytes and aim to infer conserved mechanisms that may mediate these phenomena.
Collapse
MESH Headings
- Alcohol Oxidoreductases/genetics
- Alcohol Oxidoreductases/metabolism
- Animals
- Co-Repressor Proteins/genetics
- Co-Repressor Proteins/metabolism
- Cytoskeletal Proteins/genetics
- Cytoskeletal Proteins/metabolism
- Cytoskeleton/metabolism
- Cytoskeleton/ultrastructure
- Gene Expression Regulation, Developmental
- Hair Cells, Auditory, Inner/metabolism
- Hair Cells, Auditory, Inner/ultrastructure
- Hair Cells, Auditory, Outer/metabolism
- Hair Cells, Auditory, Outer/ultrastructure
- Hair Cells, Vestibular/metabolism
- Hair Cells, Vestibular/ultrastructure
- Mechanotransduction, Cellular
- Mice
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Neuronal Plasticity/genetics
- Neuropeptides/genetics
- Neuropeptides/metabolism
- Rats
- Synapses/metabolism
- Synapses/ultrastructure
- Synaptic Transmission/genetics
- Synaptic Vesicles/metabolism
- Synaptic Vesicles/ultrastructure
Collapse
Affiliation(s)
- Roos Anouk Voorn
- Presynaptogenesis and Intracellular Transport in Hair Cells Junior Research Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Goettingen, 37075 Goettingen, Germany;
- Göttingen Graduate Center for Neurosciences, Biophysics and Molecular Biosciences, 37075 Goettingen, Germany
- Collaborative Research Center 889 “Cellular Mechanisms of Sensory Processing”, 37075 Goettingen, Germany
| | - Christian Vogl
- Presynaptogenesis and Intracellular Transport in Hair Cells Junior Research Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Goettingen, 37075 Goettingen, Germany;
- Collaborative Research Center 889 “Cellular Mechanisms of Sensory Processing”, 37075 Goettingen, Germany
| |
Collapse
|
49
|
Dolphin AC. Functions of Presynaptic Voltage-gated Calcium Channels. FUNCTION (OXFORD, ENGLAND) 2020; 2:zqaa027. [PMID: 33313507 PMCID: PMC7709543 DOI: 10.1093/function/zqaa027] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/16/2020] [Accepted: 10/20/2020] [Indexed: 01/06/2023]
Abstract
Voltage-gated calcium channels are the principal conduits for depolarization-mediated Ca2+ entry into excitable cells. In this review, the biophysical properties of the relevant members of this family of channels, those that are present in presynaptic terminals, will be discussed in relation to their function in mediating neurotransmitter release. Voltage-gated calcium channels have properties that ensure they are specialized for particular roles, for example, differences in their activation voltage threshold, their various kinetic properties, and their voltage-dependence of inactivation. All these attributes play into the ability of the various voltage-gated calcium channels to participate in different patterns of presynaptic vesicular release. These include synaptic transmission resulting from single action potentials, and longer-term changes mediated by bursts or trains of action potentials, as well as release resulting from graded changes in membrane potential in specialized sensory synapses.
Collapse
Affiliation(s)
- Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, WC1E 6BT, UK,Address correspondence to A.C.D. (e-mail: )
| |
Collapse
|
50
|
Jeng JY, Ceriani F, Olt J, Brown SDM, Holley MC, Bowl MR, Johnson SL, Marcotti W. Pathophysiological changes in inner hair cell ribbon synapses in the ageing mammalian cochlea. J Physiol 2020; 598:4339-4355. [PMID: 32710572 DOI: 10.1113/jp280018] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/24/2020] [Indexed: 12/16/2022] Open
Abstract
KEY POINTS Age-related hearing loss (ARHL) is associated with the loss of inner hair cell (IHC) ribbon synapses, lower hearing sensitivity and decreased ability to understand speech, especially in a noisy environment. Little is known about the age-related physiological and morphological changes that occur at ribbon synapses. We show that the differing degrees of ARHL in four selected mouse stains is correlated with the loss of ribbon synapses, being most severe for the strains C57BL/6NTac and C57BL/6J, less so for C57BL/6NTacCdh23+ -Repaired and lowest for C3H/HeJ. Despite the loss of ribbon synapses with age, the volume of the remaining ribbons increased and the size and kinetics of Ca2+ -dependent exocytosis in IHCs was unaffected, indicating the presence of a previously unknown degree of functional compensation at ribbon synapses. Although the age-related morphological changes at IHC ribbon synapses contribute to the different progression of ARHL, without the observed functional compensation hearing loss could be greater. ABSTRACT Mammalian cochlear inner hair cells (IHCs) are specialized sensory receptors able to provide dynamic coding of sound signals. This ability is largely conferred by their ribbon synapses, which tether a large number of vesicles at the IHC's presynaptic active zones, allowing high rates of sustained synaptic transmission onto the afferent fibres. How the physiological and morphological properties of ribbon synapses change with age remains largely unknown. Here, we have investigated the biophysical and morphological properties of IHC ribbon synapses in the ageing cochlea (9-12 kHz region) of four mouse strains commonly used in hearing research: early-onset progressive hearing loss (C57BL/6J and C57BL/6NTac) and 'good hearing' strains (C57BL/6NTacCdh23+ and C3H/HeJ). We found that with age, both modiolar and pillar sides of the IHC exhibited a loss of ribbons, but there was an increased volume of those that remained. These morphological changes, which only occurred after 6 months of age, were correlated with the level of hearing loss in the different mouse strains, being most severe for C57BL/6NTac and C57BL/6J, less so for C57BL/6NTacCdh23+ and absent for C3H/HeJ strains. Despite the age-related reduction in ribbon number in three of the four strains, the size and kinetics of Ca2+ -dependent exocytosis, as well as the replenishment of synaptic vesicles, in IHCs was not affected. The degree of vesicle release at the fewer, but larger, individual remaining ribbon synapses colocalized with the post-synaptic afferent terminals is likely to increase, indicating the presence of a previously unknown degree of functional compensation in the ageing mouse cochlea.
Collapse
Affiliation(s)
- Jing-Yi Jeng
- Department of Biomedical Science, University of Sheffield, Sheffield, S10 2TN, UK.,Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Federico Ceriani
- Department of Biomedical Science, University of Sheffield, Sheffield, S10 2TN, UK.,Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Jennifer Olt
- Department of Biomedical Science, University of Sheffield, Sheffield, S10 2TN, UK
| | - Steve D M Brown
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, UK
| | - Matthew C Holley
- Department of Biomedical Science, University of Sheffield, Sheffield, S10 2TN, UK
| | - Michael R Bowl
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, UK
| | - Stuart L Johnson
- Department of Biomedical Science, University of Sheffield, Sheffield, S10 2TN, UK.,Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Walter Marcotti
- Department of Biomedical Science, University of Sheffield, Sheffield, S10 2TN, UK.,Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| |
Collapse
|