1
|
Gagliardini M, Mechaussier S, Campos Pina C, Morais M, Postal O, Jean P, Dupont T, Singh-Estivalet A, Udugampolage S, Scandola C, Verpy E, Libé-Philippot B, Inbar TC, Schwenkgrub J, Spinola CMB, Etournay R, El-Amraoui A, Bathellier B, Mallet A, Delmaghani S, Giraudet F, Petit C, Gourévitch B, Avan P, Michalski N. Deciphering Auditory Hyperexcitability in Otogl Mutant Mice Unravels an Auditory Neuropathy Mechanism. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410776. [PMID: 39965080 DOI: 10.1002/advs.202410776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 01/09/2025] [Indexed: 02/20/2025]
Abstract
Auditory neuropathies affect the spiral ganglion neurons of the auditory nerve or their synapses with the sensory hair cells, distorting the sound information transmitted from the ear to the brain. Deciphering the underlying pathophysiological mechanisms remains challenging owing to the diversity of spiral ganglion neuron subtypes and associated central auditory circuits. An auditory neuropathy mechanism is unraveled by investigating the origin of auditory hyperexcitability in a mouse model for hereditary congenital deafness. Otogl encodes the large Otogelin-like protein, which is related to secreted epithelial mucins and is implicated in the mechanical stimulation of cochlear outer hair cells. Heterozygous Otogl+/- mutant mice display auditory hyperexcitability, highlighted by their susceptibility to audiogenic seizures induced by loud sounds. It is shown that Otogl is transiently expressed in a subpopulation of spiral ganglion neurons during cochlear development. Despite their apparently normal hearing, Otogl+/- mice display poor activation of the spiral ganglion neurons processing loud sounds and an elevation of the activation threshold of the middle the ear muscle reflex that attenuates loud sounds. The findings reveal how a neuropathy affecting spiral ganglion neurons specialized in loud sound processing and associated with the middle the ear muscle reflex can manifest itself as auditory hyperexcitability.
Collapse
Affiliation(s)
- Mathilde Gagliardini
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Plasticity of Central Auditory Circuits, Paris, F-75012, France
- Sorbonne Université, Collège Doctoral, Paris, F-75005, France
| | - Sabrina Mechaussier
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Plasticity of Central Auditory Circuits, Paris, F-75012, France
| | - Carolina Campos Pina
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Plasticity of Central Auditory Circuits, Paris, F-75012, France
- Sorbonne Université, Collège Doctoral, Paris, F-75005, France
| | - Monica Morais
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Plasticity of Central Auditory Circuits, Paris, F-75012, France
| | - Olivier Postal
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Plasticity of Central Auditory Circuits, Paris, F-75012, France
- Sorbonne Université, Collège Doctoral, Paris, F-75005, France
| | - Philippe Jean
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Plasticity of Central Auditory Circuits, Paris, F-75012, France
| | - Typhaine Dupont
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Plasticity of Central Auditory Circuits, Paris, F-75012, France
| | - Amrit Singh-Estivalet
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Auditory Therapies Innovation Laboratory, Paris, F-75012, France
| | - Shéhanie Udugampolage
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Plasticity of Central Auditory Circuits, Paris, F-75012, France
| | - Cyril Scandola
- Institut Pasteur, Université Paris Cité, Ultrastructural Bioimaging Unit, Paris, F-75015, France
| | - Elisabeth Verpy
- Institut Pasteur, IUF, Université Paris Cité, Human Genetics and Cognitive Functions, Paris, F-75015, France
| | - Baptiste Libé-Philippot
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Plasticity of Central Auditory Circuits, Paris, F-75012, France
| | - Talya C Inbar
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Plasticity of Central Auditory Circuits, Paris, F-75012, France
| | - Joanna Schwenkgrub
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Auditory System Dynamics and Multisensory Processing, Paris, F-75012, France
| | - Carla Maria Barbosa Spinola
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Cochlear Development and Therapeutic Perspectives, Paris, F-75012, France
| | - Raphaël Etournay
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Cochlear Development and Therapeutic Perspectives, Paris, F-75012, France
| | - Aziz El-Amraoui
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Progressive Sensory Disorders, Pathophysiology and Therapy, Paris, F-75012, France
| | - Brice Bathellier
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Auditory System Dynamics and Multisensory Processing, Paris, F-75012, France
| | - Adeline Mallet
- Institut Pasteur, Université Paris Cité, Ultrastructural Bioimaging Unit, Paris, F-75015, France
| | - Sedigheh Delmaghani
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Progressive Sensory Disorders, Pathophysiology and Therapy, Paris, F-75012, France
| | - Fabrice Giraudet
- Laboratoire de Biophysique Neurosensorielle, INSERM 1107, Université Clermont Auvergne, Clermont-Ferrand, F-63000, France
- Service de Génétique Médicale, CHU de Clermont-Ferrand, Clermont-Ferrand, F-63000, France
| | - Christine Petit
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Auditory Therapies Innovation Laboratory, Paris, F-75012, France
- Collège de France, Paris, F-75005, France
| | - Boris Gourévitch
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Plasticity of Central Auditory Circuits, Paris, F-75012, France
| | - Paul Avan
- Laboratoire de Biophysique Neurosensorielle, INSERM 1107, Université Clermont Auvergne, Clermont-Ferrand, F-63000, France
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Centre de Recherche et d'Innovation en Audiologie Humaine, Paris, F-75015, France
| | - Nicolas Michalski
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Plasticity of Central Auditory Circuits, Paris, F-75012, France
| |
Collapse
|
2
|
Kreeger LJ, Honnuraiah S, Maeker S, Shea S, Fishell G, Goodrich L. An anatomical and physiological basis for flexible coincidence detection in the auditory system. eLife 2025; 13:RP100492. [PMID: 40232945 PMCID: PMC11999698 DOI: 10.7554/elife.100492] [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: 04/17/2025] Open
Abstract
Animals navigate the auditory world by recognizing complex sounds, from the rustle of a predator to the call of a potential mate. This ability depends in part on the octopus cells of the auditory brainstem, which respond to multiple frequencies that change over time, as occurs in natural stimuli. Unlike the average neuron, which integrates inputs over time on the order of tens of milliseconds, octopus cells must detect momentary coincidence of excitatory inputs from the cochlea during an ongoing sound on both the millisecond and submillisecond time scale. Here, we show that octopus cells receive inhibitory inputs on their dendrites that enhance opportunities for coincidence detection in the cell body, thereby allowing for responses both to rapid onsets at the beginning of a sound and to frequency modulations during the sound. This mechanism is crucial for the fundamental process of integrating the synchronized frequencies of natural auditory signals over time.
Collapse
Affiliation(s)
- Lauren J Kreeger
- Harvard Medical School, Department of NeurobiologyBostonUnited States
| | - Suraj Honnuraiah
- Harvard Medical School, Department of NeurobiologyBostonUnited States
- Stanley Center for Psychiatric Research, Broad Institute of MIT and HarvardCambridgeUnited States
| | - Sydney Maeker
- Harvard Medical School, Department of NeurobiologyBostonUnited States
| | - Siobhan Shea
- Harvard Medical School, Department of NeurobiologyBostonUnited States
| | - Gordon Fishell
- Harvard Medical School, Department of NeurobiologyBostonUnited States
- Stanley Center for Psychiatric Research, Broad Institute of MIT and HarvardCambridgeUnited States
| | - Lisa Goodrich
- Harvard Medical School, Department of NeurobiologyBostonUnited States
| |
Collapse
|
3
|
de Haan S, He J, Corbat AA, Belicova L, Ratz M, Vinsland E, Frisén J, Kelley MW, Andersson ER. Ectoderm barcoding reveals neural and cochlear compartmentalization. Science 2025; 388:60-68. [PMID: 40179197 DOI: 10.1126/science.adq9248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 02/12/2025] [Indexed: 04/05/2025]
Abstract
Placodes and the neural crest are defining features of vertebrates. In this study, we investigate their lineages in mice using in utero approaches. We demonstrated that nanoinjection at embryonic day 7.5 targeted the ectoderm, including the future nervous system, placodes, and neural crest, allowing highly efficient manipulation of the future nervous system and inner ear. By using heritable DNA barcodes and high-throughput next-generation single-cell lineage tracing, we elucidated convergent differentiation pathways and identified distinct nervous system-, neural crest-, and otic placode-derived lineages. Clonal analyses identified early neural and cochlear compartmentalization, linking differentiated cell types to their progenitors or cellular siblings. This provides foundational insights for neuroscience and developmental biology.
Collapse
Affiliation(s)
- Sandra de Haan
- Department of Cell and Molecular Biology, Karolinska Institutet, Solna, Sweden
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Jingyan He
- Department of Cell and Molecular Biology, Karolinska Institutet, Solna, Sweden
| | - Agustin A Corbat
- Department of Cell and Molecular Biology, Karolinska Institutet, Solna, Sweden
| | - Lenka Belicova
- Department of Cell and Molecular Biology, Karolinska Institutet, Solna, Sweden
| | - Michael Ratz
- Department of Cell and Molecular Biology, Karolinska Institutet, Solna, Sweden
| | - Elin Vinsland
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
| | - Jonas Frisén
- Department of Cell and Molecular Biology, Karolinska Institutet, Solna, Sweden
| | - Matthew W Kelley
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Emma R Andersson
- Department of Cell and Molecular Biology, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
4
|
Huang Y, Chen Z, Chen J, Liu J, Qiu C, Liu Q, Zhang L, Zhu G, Ma X, Sun S, Shi YS, Wan G. Direct reprogramming of fibroblasts into spiral ganglion neurons by defined transcription factors. Cell Prolif 2025; 58:e13775. [PMID: 39551613 PMCID: PMC11969255 DOI: 10.1111/cpr.13775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/19/2024] Open
Abstract
Degeneration of the cochlear spiral ganglion neurons (SGNs) is one of the major causes of sensorineural hearing loss and significantly impacts the outcomes of cochlear implantation. Functional regeneration of SGNs holds great promise for treating sensorineural hearing loss. In this study, we systematically screened 33 transcriptional regulators implicated in neuronal and SGN fate. Using gene expression array and principal component analyses, we identified a sequential combination of Ascl1, Pou4f1 and Myt1l (APM) in promoting functional reprogramming of SGNs. The neurons induced by APM expressed mature neuronal and SGN lineage-specific markers, displayed mature SGN-like electrophysiological characteristics and exhibited single-cell transcriptomes resembling the endogenous SGNs. Thus, transcription factors APM may serve as novel candidates for direct reprogramming of SGNs and hearing recovery due to SGN damages.
Collapse
Affiliation(s)
- Yuhang Huang
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
| | - Zhen Chen
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
| | - Jiang Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
- Department of Neurology, The Affiliated Drum Tower Hospital of Medical School and Institute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjingChina
| | - Jingyue Liu
- National Institute of Biological SciencesBeijingChina
- Tsinghua Institute of Multidisciplinary Biomedical ResearchTsinghua UniversityBeijingChina
| | - Cui Qiu
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
| | - Qing Liu
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Linqing Zhang
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
| | - Guang‐Jie Zhu
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Xiaofeng Ma
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Shuohao Sun
- National Institute of Biological SciencesBeijingChina
- Tsinghua Institute of Multidisciplinary Biomedical ResearchTsinghua UniversityBeijingChina
| | - Yun Stone Shi
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
- Guangdong Institute of Intelligence Science and TechnologyZhuhaiChina
| | - Guoqiang Wan
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| |
Collapse
|
5
|
Pearson LJ, Pinyon JL, Cederholm JME, von Jonquieres G, Bartlett F, Vázquez-Campos X, Delerue F, Ittner LM, Housley GD. Developmental differentiation of mouse inner ear neuron subpopulations resolved with a peripherin-promoter reporter within the Grm8 locus. Sci Rep 2025; 15:10281. [PMID: 40133378 PMCID: PMC11937576 DOI: 10.1038/s41598-025-94011-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 03/11/2025] [Indexed: 03/27/2025] Open
Abstract
Molecular profiling of inner ear neurons has broadened the classification of the primary afferents that support neural coding for hearing and balance. To extend spatiotemporal characterization of auditory and vestibular neuron diversity, we established a transgenic reporter mouse model (Prphp-mCherry), where elements of the peripherin promoter (Prphp) drive expression of the mCherry fluorescent reporter. Type III intermediate filament protein peripherin expression is a marker for type II spiral ganglion neurons (SGN) that innervate the cochlear outer hair cells, and the small diameter 'bouton' vestibular ganglion neurons (VGN) innervating the type II vestibular hair cells. Using Nanopore genome sequencing, the integration site of the transgene construct was identified within the class III metabotropic glutamate receptor 8 gene (Grm8, chromosome 6). Use of CUBIC / PEGASOS clearing of early postnatal to adult inner ear tissues enabled in situ 3D spatial localization of a dispersed population of cochlear mCherry + ve SGN, with highest expression and density in the hook (high frequency encoding) basal region. Of these mCherry + ve SGN, type II SGN (peripherin-immunopositive) were all co-labeled in the basal region, but the majority of the overall mCherry-delineated SGN auditory subpopulation were type I SGN innervating inner hair cells. In the VGN, mCherry + ve neurons represented ~ 15% of the adult population, dispersed as a small diameter subpopulation throughout both the inferior and superior VGN regions. These findings resolve heterogeneous type I and type II cochlear SGN subpopulations, particularly in the structurally complex hook region, and further differentiate vestibular primary afferents across postnatal development.
Collapse
Affiliation(s)
- Lily J Pearson
- Translational Neuroscience Facility, Department of Physiology, School of Biomedical Sciences, UNSW, Sydney, NSW, Australia
| | - Jeremy L Pinyon
- Translational Neuroscience Facility, Department of Physiology, School of Biomedical Sciences, UNSW, Sydney, NSW, Australia
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Camperdown, NSW, Australia
| | - Jennie M E Cederholm
- Translational Neuroscience Facility, Department of Physiology, School of Biomedical Sciences, UNSW, Sydney, NSW, Australia
| | - Georg von Jonquieres
- Translational Neuroscience Facility, Department of Physiology, School of Biomedical Sciences, UNSW, Sydney, NSW, Australia
| | - Florence Bartlett
- KGLMF Mark Wainwright Analytical Centre, UNSW, Sydney, NSW, Australia
| | | | - Fabien Delerue
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lars M Ittner
- Faculty of Medicine, Health and Human Sciences, Department of Biomedical Sciences, Dementia Research Centre, Macquarie University, Sydney, NSW, Australia
| | - Gary D Housley
- Translational Neuroscience Facility, Department of Physiology, School of Biomedical Sciences, UNSW, Sydney, NSW, Australia.
| |
Collapse
|
6
|
Thirumalai A, Henseler J, Enayati M, Kusch K, Hessler R, Moser T, Huet AT. Improved optogenetic modification of spiral ganglion neurons for future optical cochlear implants. Theranostics 2025; 15:4270-4286. [PMID: 40225583 PMCID: PMC11984395 DOI: 10.7150/thno.104474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 02/19/2025] [Indexed: 04/15/2025] Open
Abstract
Optogenetic stimulation has become a promising approach for restoring lost body function. For example, partial restoration of vision has been achieved in a blind patient and preclinical proof-of-concept has been demonstrated for optogenetic hearing restoration. In preparation for clinical translation of hearing restoration, efficient and safe optogenetic modification of spiral ganglion neurons (SGNs) in the mature cochlea remained to be developed. Methods: Here, we established microcatheter-based administration of adeno-associated virus (AAV) into scala tympani of the cochlea of Mongolian gerbils and compared it to the previously developed direct AAV-injection into the spiral ganglion. We probed the potential of AAV-PHP.S to express channelrhodopsins (ChRs) under the control of the human synapsin promotor in mature SGNs of hearing and deafened gerbils. Results: Using the microcatheter approach, but not with the AAV-modiolus injection, we achieved reliable ChR expression in SGN enabling optogenetic stimulation of the auditory pathway in 80% of the treated animals. Yet, the efficiency of SGN transduction was modest with only ~30% ChR-expressing SGNs. Moreover, we encountered off-target expression in hair cells in hearing gerbils in both approaches. We did not detect ChR expression in the central nervous system using microcatheter administration. Comparing optogenetic auditory brainstem responses of gerbils with and without hair cell transduction confirmed that SGNs were the primary site of optogenetic stimulation of the pathway. Conclusions: The AAV.PHP-S microcatheter administration via the round window with pressure relief at the round window is a reliable approach to optogenetically modify the SGNs in order to restore hearing with future optical cochlear implants.
Collapse
Affiliation(s)
- Anupriya Thirumalai
- Institute for Auditory Neuroscience, University Medical Center Göttingen, 37075 Göttingen, Germany
- InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany
- Auditory Circuit Lab, University Medical Center Göttingen, 37075 Göttingen, Germany
- Göttingen Graduate School for Neurosciences and Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Jana Henseler
- Institute for Auditory Neuroscience, University Medical Center Göttingen, 37075 Göttingen, Germany
- InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany
- Auditory Circuit Lab, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Marzieh Enayati
- Institute for Auditory Neuroscience, University Medical Center Göttingen, 37075 Göttingen, Germany
- InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany
- Auditory Circuit Lab, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Kathrin Kusch
- Institute for Auditory Neuroscience, University Medical Center Göttingen, 37075 Göttingen, Germany
- InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany
- Auditory Neuroscience and Optogenetics Laboratory, German Primate Center, Göttingen, Germany
- Functional Auditory Genomics group, University Medical Center Göttingen, 37075 Göttingen, Germany
- Else Kroener Fresenius Center for Optogenetic Therapies, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Roland Hessler
- 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, University Medical Center Göttingen, 37075 Göttingen, Germany
- InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany
- Auditory Neuroscience and Optogenetics Laboratory, German Primate Center, Göttingen, Germany
- Else Kroener Fresenius Center for Optogenetic Therapies, University Medical Center Göttingen, 37075 Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
- Auditory Neuroscience and Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Antoine Tarquin Huet
- Institute for Auditory Neuroscience, University Medical Center Göttingen, 37075 Göttingen, Germany
- InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany
- Auditory Circuit Lab, University Medical Center Göttingen, 37075 Göttingen, Germany
- Auditory Neuroscience and Optogenetics Laboratory, German Primate Center, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
- Auditory Neuroscience and Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Current address: Institute for Neurosciences Montpellier, Institut National de la Santé et de la Recherche Médical, University of Montpellier, Montpellier, France
| |
Collapse
|
7
|
Tabassum A, Deng M, Hu Z. Role of Neurotrophins in the Generation of Spiral Ganglion Neuron-Like Cells from Embryonic Stem Cells. Stem Cells Dev 2025; 34:127-135. [PMID: 39946227 DOI: 10.1089/scd.2024.0164] [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] [Indexed: 03/15/2025] Open
Abstract
Spiral ganglion neurons (SGNs) are crucial for transferring auditory signals from cochlear sensory hair cells to the brainstem. However, SGNs are usually damaged in sensorineural hearing loss. Embryonic stem cells (ESCs) have been used to regenerate SGNs, but it is obscure whether ESC-derived neurons can fully resemble SGN subtype features. This study aimed to understand the effect of neurotrophins on the generation of SGN-like cells from ESCs and their subsequent subtype specification. This study utilized a stepwise neuronal generation approach to direct DsRed ESCs toward neural progenitors and eventually SGN-like cells. The derived SGN-like cells expressed multiple neuronal markers, including Tuj1, Map2, and NeuN, indicating maturity. Neurotrophins, including brain-derived neurotrophic factor, neutrotrophin-3, and nerve growth factor, seemed to regulate the generation of mature neurons from ESCs. In addition, derived neuron-like cells expressed the otic protein marker Gata3 and glutamatergic marker VGluT1, suggesting that they are SGN-like the glutamatergic cells. Significantly more SGN subtype marker-positive cells, including Pou4f1, calbindin, and calretinin-positive cells, were observed in the neurotrophin treatment groups. Overall, this study indicates the potential of SGN subtype generation from ESCs, which could be significant for cochlear implant therapy or stem cell-based replacement studies.
Collapse
Affiliation(s)
- Anika Tabassum
- Department of Otolaryngology-HNS, Wayne State University School of Medicine, Detroit, MI, United States
- John D Dingell VA Medical Center, Detroit, MI, United States
| | - Meng Deng
- Department of Otolaryngology-HNS, Wayne State University School of Medicine, Detroit, MI, United States
- John D Dingell VA Medical Center, Detroit, MI, United States
| | - Zhengqing Hu
- Department of Otolaryngology-HNS, Wayne State University School of Medicine, Detroit, MI, United States
- John D Dingell VA Medical Center, Detroit, MI, United States
| |
Collapse
|
8
|
Wong NF, Brongo SE, Forero EA, Sun S, Cook CJ, Lauer AM, Müller U, Xu-Friedman MA. Convergence of Type 1 Spiral Ganglion Neuron Subtypes onto Principal Neurons of the Anteroventral Cochlear Nucleus. J Neurosci 2025; 45:e1507242024. [PMID: 39663118 PMCID: PMC11800758 DOI: 10.1523/jneurosci.1507-24.2024] [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: 08/09/2024] [Revised: 11/07/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024] Open
Abstract
The mammalian auditory system encodes sounds with subtypes of spiral ganglion neurons (SGNs) that differ in sound level sensitivity, permitting discrimination across a wide range of levels. Recent work suggests the physiologically defined SGN subtypes correspond to at least three molecular subtypes. It is not known how information from the different subtypes converges within the cochlear nucleus. We examined this issue using transgenic mice of both sexes that express Cre recombinase in SGNs that are positive for markers of two subtypes: CALB2 (calretinin) in type 1a SGNs and LYPD1 in type 1c SGNs, which correspond to high- and low-sensitivity subtypes, respectively. We crossed these with mice expressing floxed channelrhodopsin, which allowed specific activation of axons from type 1a or 1c SGNs using optogenetics. We made voltage-clamp recordings from bushy cells in the anteroventral cochlear nucleus (AVCN) and found that the synapses formed by CALB2- and LYPD1-positive SGNs had similar EPSC amplitudes and short-term plasticity. Immunohistochemistry revealed that individual bushy cells receive a mix of 1a, 1b, and 1c synapses with VGluT1-positive puncta of similar sizes. We used optogenetic stimulation during in vivo recordings to classify chopper and primary-like units as receiving versus nonreceiving 1a- or 1c-type inputs. These groups showed no significant difference in threshold or spontaneous rate, suggesting the subtypes do not segregate into distinct processing streams in the AVCN. Our results indicate that principal cells in the AVCN integrate information from all SGN subtypes with extensive convergence, which could optimize sound encoding across a large dynamic range.
Collapse
Affiliation(s)
- Nicole F Wong
- Department of Biological Sciences, University at Buffalo, State University of New York, Buffalo, New York 14260
| | - Sydney E Brongo
- Department of Biological Sciences, University at Buffalo, State University of New York, Buffalo, New York 14260
| | - Evan A Forero
- Department of Biological Sciences, University at Buffalo, State University of New York, Buffalo, New York 14260
| | - Shuohao Sun
- The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Connor J Cook
- Department of Biological Sciences, University at Buffalo, State University of New York, Buffalo, New York 14260
| | - Amanda M Lauer
- The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
- Department of Otolaryngology-HNS, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
- Department of Functional Anatomy and Evolution, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Ulrich Müller
- The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Matthew A Xu-Friedman
- Department of Biological Sciences, University at Buffalo, State University of New York, Buffalo, New York 14260
| |
Collapse
|
9
|
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 2025; 456:109172. [PMID: 39708764 PMCID: PMC11772111 DOI: 10.1016/j.heares.2024.109172] [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: 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
|
10
|
Bao M, Wang X, Li X, Sun R, Wang Z, Jiang T, Wang H, Feng J. Single-Cell Landscape of the Cochlea Revealed Cell-Type-Specific Diversification in Hipposideros armiger Based on PacBio Long-Read Sequencing. Biomolecules 2025; 15:211. [PMID: 40001514 PMCID: PMC11853400 DOI: 10.3390/biom15020211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
Echolocation represents one of the most rapid adaptive sensorimotor modulation behaviors observed in mammals, establishing bats as one of the most evolutionarily successful mammals. Bats rely on high-frequency hearing for survival, but our understanding of its cellular molecular basis is scattered and segmented. Herein, we constructed the first single-cell transcriptomic landscape of the cochlea in Hipposideros armiger, a CF-FM bat, using a PacBio-optimized genome and compared it with the results obtained from unoptimized original genomes. Sixteen distinct cell types were distributed across five spatial regions of the cochlea. Notably, through hematoxylin and eosin staining and fluorescence in situ hybridization, we identified new types of spiral ganglion neuron (SGN) cells in the cochlea of H. armiger. These SGN cells are likely critical for auditory perception and may have driven the adaptive evolution of high-frequency hearing in this species. Furthermore, we uncovered the differentiation relationships of among specific cell types, such as the transition from supporting cells to hair cells. Using the cochlear cell atlas as a reference, cell types susceptible to deafness-associated genes (in the human) were also identified. In summary, this study provides novel insights into the cellular and molecular mechanisms underlying the adaptive high-frequency hearing in bats and highlights potential candidate cell types and genes for therapeutic interventions in hearing loss.
Collapse
Affiliation(s)
- Mingyue Bao
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
| | - Xue Wang
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
| | - Xintong Li
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
| | - Ruyi Sun
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
| | - Zhiqiang Wang
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun 130117, China
| | - Tinglei Jiang
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun 130117, China
| | - Hui Wang
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
| | - Jiang Feng
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun 130117, China
| |
Collapse
|
11
|
Sitko AA, Frank MM, Romero GE, Hunt M, Goodrich LV. Lateral olivocochlear neurons modulate cochlear responses to noise exposure. Proc Natl Acad Sci U S A 2025; 122:e2404558122. [PMID: 39854232 PMCID: PMC11789013 DOI: 10.1073/pnas.2404558122] [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: 03/04/2024] [Accepted: 12/04/2024] [Indexed: 01/26/2025] Open
Abstract
The sense of hearing originates in the cochlea, which detects sounds across dynamic sensory environments. Like other peripheral organs, the cochlea is subjected to environmental insults, including loud, damage-inducing sounds. In response to internal and external stimuli, the central nervous system directly modulates cochlear function through olivocochlear neurons (OCNs), which are located in the brainstem and innervate the cochlear sensory epithelium. One population of OCNs, the lateral olivocochlear (LOC) neurons, target spiral ganglion neurons (SGNs), the primary sensory neurons of the ear. LOCs alter their transmitter expression for days to weeks in response to noise exposure (NE), suggesting that they could tune SGN excitability over long time periods in response to auditory experience. To examine how LOCs affect auditory function after NE, we characterized OCN transcriptional profiles and found transient LOC-specific gene expression changes after NE, including upregulation of multiple neuropeptide-encoding genes. Next, by generating intersectional mouse lines that selectively target LOCs, we chemogenetically ablated LOCs and assayed auditory responses at baseline and after NE. Compared to controls, mice with reduced LOC innervation showed greater NE-induced functional deficits 1 d later and had worse auditory function after a 2-wk recovery period. The number of remaining presynaptic puncta at the SGN synapse with inner hair cells did not differ between control and LOC-ablated animals, suggesting that the primary role of LOCs after NE is likely not to protect but instead to compensate, ensuring that SGN function is enhanced during periods of need.
Collapse
Affiliation(s)
- Austen A. Sitko
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | | | | | - Mackenzie Hunt
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | - Lisa V. Goodrich
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| |
Collapse
|
12
|
Jing J, Hu M, Ngodup T, Ma Q, Lau SNN, Ljungberg MC, McGinley MJ, Trussell LO, Jiang X. Molecular logic for cellular specializations that initiate the auditory parallel processing pathways. Nat Commun 2025; 16:489. [PMID: 39788966 PMCID: PMC11717940 DOI: 10.1038/s41467-024-55257-z] [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: 04/18/2024] [Accepted: 12/04/2024] [Indexed: 01/12/2025] Open
Abstract
The cochlear nuclear complex (CN), the starting point for all central auditory processing, encompasses a suite of neuronal cell types highly specialized for neural coding of acoustic signals. However, the molecular logic governing these specializations remains unknown. By combining single-nucleus RNA sequencing and Patch-seq analysis, we reveal a set of transcriptionally distinct cell populations encompassing all previously observed types and discover multiple hitherto unknown subtypes with anatomical and physiological identity. The resulting comprehensive cell-type taxonomy reconciles anatomical position, morphological, physiological, and molecular criteria, enabling the determination of the molecular basis of the specialized cellular phenotypes in the CN. In particular, CN cell-type identity is encoded in a transcriptional architecture that orchestrates functionally congruent expression across a small set of gene families to customize projection patterns, input-output synaptic communication, and biophysical features required for encoding distinct aspects of acoustic signals. This high-resolution account of cellular heterogeneity from the molecular to the circuit level reveals the molecular logic driving cellular specializations, thus enabling the genetic dissection of auditory processing and hearing disorders with a high specificity.
Collapse
Affiliation(s)
- Junzhan Jing
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Ming Hu
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Tenzin Ngodup
- Oregon Hearing Research Center and Vollum Institute, Oregon Health and Science University, Portland, OR, USA
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology-HNS, University of Washington, Seattle, WA, USA
| | - Qianqian Ma
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Shu-Ning Natalie Lau
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - M Cecilia Ljungberg
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Matthew J McGinley
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
| | - Laurence O Trussell
- Oregon Hearing Research Center and Vollum Institute, Oregon Health and Science University, Portland, OR, USA.
| | - Xiaolong Jiang
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
13
|
Bottom RT, Xu Y, Siebald C, Jung J, Müller U. Defects in hair cells disrupt the development of auditory peripheral circuitry. Nat Commun 2024; 15:10899. [PMID: 39738090 PMCID: PMC11686150 DOI: 10.1038/s41467-024-55275-x] [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: 02/22/2024] [Accepted: 12/05/2024] [Indexed: 01/01/2025] Open
Abstract
Deafness is the most common form of sensory impairment in humans and frequently caused by defects in hair cells of the inner ear. Here we demonstrate that in male mice which model recessive non-syndromic deafness (DFNB6), inactivation of Tmie in hair cells disrupts gene expression in the neurons that innervate them. This includes genes regulating axonal pathfinding and synaptogenesis, two processes that are disrupted in the inner ear of the mutant mice. Similar defects are observed in mouse models for deafness caused by mutations in other genes with primary functions in hair cells. Gene therapy targeting hair cells restores hearing and inner ear circuitry in DFNB6 model mice. We conclude that hair cell function is crucial for the establishment of peripheral auditory circuitry. Treatment modalities for deafness thus need to consider restoration of the function of both hair cells and neurons, even when the primary defect occurs in hair cells.
Collapse
Affiliation(s)
- Riley T Bottom
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Yijun Xu
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Caroline Siebald
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Jinsei Jung
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Ulrich Müller
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
14
|
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
|
15
|
Chen B, Sun Y, Sun H, Cong N, Ma R, Qian X, Lyu J, Fu X, Chi F, Li H, Liu Y, Ren D, Bu W. Ultrasound-Triggered NO Release to Promote Axonal Regeneration for Noise-Induced Hearing Loss Therapy. ACS NANO 2024; 18:33232-33244. [PMID: 39561026 DOI: 10.1021/acsnano.4c12676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
Intense noise poses a threat to spiral ganglion neurons (SGNs) in the inner ear, often resulting in limited axonal regeneration during noise injury and leading to noise-induced hearing loss (NIHL). Here, we propose an ultrasound-triggered nitric oxide (NO) release to enhance the sprouting and regeneration of injured axons in SGNs. We developed hollow silicon nanoparticles to load nitrosylated N-acetylcysteine, producing HMSN-SNO, which effectively protects NO from external interferences. Utilizing low-intensity ultrasound stimulation with bone penetration, we achieve the controlled release of NO from HMSN-SNO within the cochlea. In mice with NIHL, a rapid and extensive loss of synaptic connections between hair cells and SGNs is observed within 24 h after exposure to excessive noise. However, this loss could be reversed with the combined treatment, resulting in a hearing functional recovery from 83.57 to 65.00 dB SPL. This positive outcome is attributed to the multifunctional effects of HMSN-SNO, wherein they scavenge reactive oxygen species (ROS) to reverse the pathological microenvironment and simultaneously upregulate the CREB/BDNF/EGR1 signaling pathway, thereby enhancing neuroplasticity and promoting the regeneration of neuronal axons. These findings underscore the potential of nanomedicine for neuroplasticity modulation, which holds promise for advancing both basic research and the further treatment of neurological diseases.
Collapse
Affiliation(s)
- Binjun Chen
- ENT institute and Department of Otorhinolaryngology, NHC Key Laboratory of Hearing Medicine. Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai 200031, China
| | - Yanhong Sun
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Academy for Engineering and Technology, Fudan University, 220 Handan Road, Shanghai 200438, China
| | - Haojie Sun
- ENT institute and Department of Otorhinolaryngology, NHC Key Laboratory of Hearing Medicine. Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai 200031, China
| | - Ning Cong
- ENT institute and Department of Otorhinolaryngology, NHC Key Laboratory of Hearing Medicine. Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai 200031, China
| | - Rui Ma
- ENT institute and Department of Otorhinolaryngology, NHC Key Laboratory of Hearing Medicine. Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai 200031, China
| | - Xiaoqing Qian
- ENT institute and Department of Otorhinolaryngology, NHC Key Laboratory of Hearing Medicine. Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai 200031, China
| | - Jihan Lyu
- ENT institute and Department of Otorhinolaryngology, NHC Key Laboratory of Hearing Medicine. Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai 200031, China
| | - Xiao Fu
- ENT institute and Department of Otorhinolaryngology, NHC Key Laboratory of Hearing Medicine. Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai 200031, China
| | - Fanglu Chi
- ENT institute and Department of Otorhinolaryngology, NHC Key Laboratory of Hearing Medicine. Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai 200031, China
| | - Hongzhe Li
- Research Service, VA Loma Linda Healthcare System, 11201 Benton Street, Loma Linda, CA 92357, USA
| | - Yanyan Liu
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Academy for Engineering and Technology, Fudan University, 220 Handan Road, Shanghai 200438, China
| | - Dongdong Ren
- ENT institute and Department of Otorhinolaryngology, NHC Key Laboratory of Hearing Medicine. Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai 200031, China
| | - Wenbo Bu
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Academy for Engineering and Technology, Fudan University, 220 Handan Road, Shanghai 200438, China
| |
Collapse
|
16
|
Regalado Núñez K, Bronson D, Chang R, Kalluri R. Vestibular afferent neurons develop normally in the absence of quantal/glutamatergic input. Front Neurol 2024; 15:1441964. [PMID: 39655160 PMCID: PMC11625666 DOI: 10.3389/fneur.2024.1441964] [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: 05/31/2024] [Accepted: 10/29/2024] [Indexed: 12/12/2024] Open
Abstract
Introduction The vestibular nerve is comprised of neuron sub-groups with diverse functions related to their intrinsic biophysical properties. This diversity is partly due to differences in the types and numbers of low-voltage-gated potassium channels found in the neurons' membranes. Expression for some low-voltage gated ion channels like KCNQ4 is upregulated during early post-natal development; suggesting that ion channel composition and neuronal diversity may be shaped by hair cell activity. This idea is consistent with recent work showing that glutamatergic input from hair cells is necessary for the normal diversification auditory neurons. Methods To test if biophysical diversity is similarly dependent on glutamatergic input in vestibular neurons, we examined vestibular function and the maturation of the vestibular epithelium and ganglion neurons by immunohistochemistry and patch-clamp electrophysiology in Vglut3-ko mice whose hair cell synapses lack glutamate. Results The knockout mice showed no obvious balance deficits and crossed challenging balance beams with little difficulty. Immunolabeling of the Vglut3-ko vestibular epithelia showed normal development as indicated by an identifiable striolar zone with calyceal terminals labeled by molecular marker calretinin, and normal expression of KCNQ4 by the end of the second post-natal week. We found similar numbers of Type I and Type II hair cells in the knockout and wild-type animals, regardless of epithelial zone. Thus, the presumably quiescent Type II hair cells are not cleared from the epithelium. Patch-clamp recordings showed that biophysical diversity of vestibular ganglion neurons in the Vglut3-ko mice is comparable to that found in wild-type controls, with a similar range firing patterns at both immature and juvenile ages. However, our results suggest a subtle biophysical alteration to the largest ganglion cells (putative somata of central zone afferents); those in the knockout had smaller net conductance and were more excitable than those in the wild type. Discussion Thus, unlike in the auditory nerve, glutamatergic signaling is unnecessary for producing biophysical diversity in vestibular ganglion neurons. And yet, because the input signals from vestibular hair cells are complex and not solely reliant on quantal release of glutamate, whether diversity of vestibular ganglion neurons is simply hardwired or regulated by a more complex set of input signals remains to be determined.
Collapse
Affiliation(s)
- Katherine Regalado Núñez
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, United States
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Daniel Bronson
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Zilkha Neurogenetic Institute, Department of Otolaryngology, University of Southern California, Los Angeles, CA, United States
| | - Ryan Chang
- Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, United States
| | - Radha Kalluri
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Zilkha Neurogenetic Institute, Department of Otolaryngology, University of Southern California, Los Angeles, CA, United States
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
17
|
Kreeger LJ, Honnuraiah S, Maeker S, Shea S, Fishell G, Goodrich LV. An Anatomical and Physiological Basis for Flexible Coincidence Detection in the Auditory System. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.29.582808. [PMID: 38464181 PMCID: PMC10925315 DOI: 10.1101/2024.02.29.582808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Animals navigate the auditory world by recognizing complex sounds, from the rustle of a predator to the call of a potential mate. This ability depends in part on the octopus cells of the auditory brainstem, which respond to multiple frequencies that change over time, as occurs in natural stimuli. Unlike the average neuron, which integrates inputs over time on the order of tens of milliseconds, octopus cells must detect momentary coincidence of excitatory inputs from the cochlea during an ongoing sound on both the millisecond and submillisecond time scale. Here, we show that octopus cells receive inhibitory inputs on their dendrites that enhance opportunities for coincidence detection in the cell body, thereby allowing for responses both to rapid onsets at the beginning of a sound and to frequency modulations during the sound. This mechanism is crucial for the fundamental process of integrating the synchronized frequencies of natural auditory signals over time.
Collapse
Affiliation(s)
- Lauren J Kreeger
- Harvard Medical School, Department of Neurobiology, Boston, MA 02115, USA
| | - Suraj Honnuraiah
- Harvard Medical School, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sydney Maeker
- Harvard Medical School, Department of Neurobiology, Boston, MA 02115, USA
| | - Siobhan Shea
- Harvard Medical School, Department of Neurobiology, Boston, MA 02115, USA
| | - Gord Fishell
- Harvard Medical School, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Lisa V Goodrich
- Harvard Medical School, Department of Neurobiology, Boston, MA 02115, USA
| |
Collapse
|
18
|
Ma X, Chen X, Che Y, Zhu S, Wang X, Gao S, Wu J, Kong F, Cheng C, Wu Y, Guo J, Qi J, Chai R. The single-cell transcriptomic landscape of the topological differences in mammalian auditory receptors. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2398-2410. [PMID: 39083201 DOI: 10.1007/s11427-024-2672-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/01/2024] [Indexed: 10/22/2024]
Abstract
Mammalian hair cells (HCs) are arranged spirally along the cochlear axis and correspond to different frequency ranges. Serving as primary sound detectors, HCs spatially segregate component frequencies into a topographical map. HCs display significant diversity in anatomical and physiological characteristics, yet little is known about the organization of the cochleotopic map of HCs or the molecules involved in this process. Using single-cell RNA sequencing, we determined the distinct molecular profiles of inner hair cells and outer hair cells, and we identified numerous position-dependent genes that were expressed as gradients. Newly identified genes such as Ptn, Rxra, and Nfe2l2 were found to be associated with tonotopy. We employed the SCENIC algorithm to predict the transcription factors that potentially shape these tonotopic gradients. Furthermore, we confirmed that Nfe2l2, a tonotopy-related transcription factor, is critical in mice for sensing low-to-medium sound frequencies in vivo. the analysis of cell-cell communication revealed potential receptor-ligand networks linking inner hair cells to spiral ganglion neurons, including pathways such as BDNF-Ntrk and PTN-Scd4, which likely play essential roles in tonotopic maintenance. Overall, these findings suggest that molecular gradients serve as the organizing principle for maintaining the selection of sound frequencies by HCs.
Collapse
Affiliation(s)
- Xiangyu Ma
- 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, 210096, China
| | - Xin Chen
- 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, 210096, China
| | - Yuwei Che
- 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, 210096, China
| | - Siyao Zhu
- School of Engineering, Vanderbilt University, Nashville, 37240, USA
| | - Xinlin Wang
- 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, 210096, China
| | - Shan Gao
- 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, 210096, China
| | - Jiheng Wu
- 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, 210096, China
| | - Fanliang Kong
- 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, 210096, China
| | - Cheng Cheng
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing, 210096, China
- Research Institute of Otorhinolaryngology, Nanjing, 210096, China
| | - Yunhao Wu
- 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, 210096, China
| | - Jiamin Guo
- 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, 210096, China
| | - Jieyu Qi
- Department of Neurology, Aerospace Center Hospital, School of Life Sciences, Beijing Institute of Technology, Beijing, 100081, China.
- Advanced Technology Research Institute, Beijing Institute of Technology, Beijing, 100081, 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, 210096, China.
- Department of Neurology, Aerospace Center Hospital, School of Life Sciences, Beijing Institute of Technology, Beijing, 100081, China.
- Advanced Technology Research Institute, Beijing Institute of Technology, Beijing, 100081, China.
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Southeast University Shenzhen Research Institute, Shenzhen, 518063, China.
| |
Collapse
|
19
|
Hosoya M, Ueno M, Shimanuki MN, Nishiyama T, Oishi N, Ozawa H. A primate model animal revealed the inter-species differences and similarities in the subtype specifications of the spiral ganglion neurons. Sci Rep 2024; 14:25166. [PMID: 39448766 PMCID: PMC11502759 DOI: 10.1038/s41598-024-76892-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024] Open
Abstract
Type I spiral ganglion neurons are peripheral neurons essential for hearing perception. While they can be subdivided in mice based on characteristic gene expression patterns, detailed examinations of these subtypes in primates and humans are lacking. In this study, we investigated the developmental subtypes of spiral ganglion neurons in the common marmoset (Callithrix jacchus). We confirmed that Type I spiral ganglion can be divided based on the characteristic gene expression patterns of several marker genes. However, some combinations of these genes differ from those in rodents, suggesting common marmoset's suitability for advancing our understanding of human cochlear development. Additionally, identifying the essential time points for subtype specifications and subsequent maturation will aid in studying the primate-specific developmental biology of the inner ear. This could lead to new treatment strategies for hearing loss in humans and be valuable for studying age-related hearing loss, as well as designing regenerative therapies.
Collapse
Affiliation(s)
- Makoto Hosoya
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Masafumi Ueno
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Marie N Shimanuki
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Takanori Nishiyama
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Naoki Oishi
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hiroyuki Ozawa
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| |
Collapse
|
20
|
Cederroth CR, Dyhrfjeld-Johnsen J, Canlon B. Pharmacological Approaches to Hearing Loss. Pharmacol Rev 2024; 76:1063-1088. [PMID: 39164117 PMCID: PMC11549935 DOI: 10.1124/pharmrev.124.001195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/08/2024] [Accepted: 07/16/2024] [Indexed: 08/22/2024] Open
Abstract
Hearing disorders pose significant challenges to individuals experiencing them and their overall quality of life, emphasizing the critical need for advanced pharmacological approaches to address these conditions. Current treatment options often focus on amplification devices, cochlear implants, or other rehabilitative therapies, leaving a substantial gap regarding effective pharmacological interventions. Advancements in our understanding of the molecular and cellular mechanisms involved in hearing disorders induced by noise, aging, and ototoxicity have opened new avenues for drug development, some of which have led to numerous clinical trials, with promising results. The development of optimal drug delivery solutions in animals and humans can also enhance the targeted delivery of medications to the ear. Moreover, large genome studies contributing to a genetic understanding of hearing loss in humans combined with advanced molecular technologies in animal studies have shown a great potential to increase our understanding of the etiologies of hearing loss. The auditory system exhibits circadian rhythms and temporal variations in its physiology, its vulnerability to auditory insults, and its responsiveness to drug treatments. The cochlear clock rhythms are under the control of the glucocorticoid system, and preclinical evidence suggests that the risk/benefit profile of hearing disorder treatments using chronopharmacological approaches would be beneficial. If translatable to the bedside, such approaches may improve the outcome of clinical trials. Ongoing research into the molecular and genetic basis of auditory disorders, coupled with advancements in drug formulation and delivery as well as optimized timing of drug administration, holds great promise of more effective treatments. SIGNIFICANCE STATEMENT: Hearing disorders pose significant challenges to individuals and their overall quality of life, emphasizing the critical need for advanced pharmacological approaches to address these conditions. Ongoing research into the molecular and genetic basis of auditory disorders, coupled with advancements in drug delivery procedures and optimized timing of drug administration, holds the promise of more effective treatments.
Collapse
Affiliation(s)
- Christopher R Cederroth
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| | - Jonas Dyhrfjeld-Johnsen
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| | - Barbara Canlon
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| |
Collapse
|
21
|
Jukic A, Lei Z, Cebul ER, Pinter K, Tadesse Y, Jarysta A, David S, Mosqueda N, Tarchini B, Kindt K. Presynaptic Nrxn3 is essential for ribbon-synapse maturation in hair cells. Development 2024; 151:dev202723. [PMID: 39254120 PMCID: PMC11488651 DOI: 10.1242/dev.202723] [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: 01/25/2024] [Accepted: 08/28/2024] [Indexed: 09/11/2024]
Abstract
Hair cells of the inner ear and lateral-line system rely on specialized ribbon synapses to transmit sensory information to the central nervous system. The molecules required to assemble these synapses are not fully understood. We show that Nrxn3, a presynaptic adhesion molecule, is crucial for ribbon-synapse maturation in hair cells. In both mouse and zebrafish models, the loss of Nrxn3 results in significantly fewer intact ribbon synapses. We show in zebrafish that, initially, Nrxn3 loss does not alter pre- and postsynapse numbers but, later, synapses fail to pair, leading to postsynapse loss. We also demonstrate that Nrxn3 subtly influences synapse selectivity in zebrafish lateral-line hair cells that detect anterior flow. Loss of Nrxn3 leads to a 60% loss of synapses in zebrafish, which dramatically reduces pre- and postsynaptic responses. Despite fewer synapses, auditory responses in zebrafish and mice are unaffected. This work demonstrates that Nrxn3 is a crucial and conserved molecule required for the maturation of ribbon synapses. Understanding how ribbon synapses mature is essential to generating new therapies to treat synaptopathies linked to auditory or vestibular dysfunction.
Collapse
Affiliation(s)
- Alma Jukic
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Zhengchang Lei
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Elizabeth R. Cebul
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Katherine Pinter
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Yommi Tadesse
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | | | - Sandeep David
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Natalie Mosqueda
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Basile Tarchini
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Katie Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| |
Collapse
|
22
|
Blum K, Schepsky P, Derleder P, Schätzle P, Nasri F, Fischer P, Engel J, Kurt S. Noise-induced cochlear synaptopathy in C57BL/6 N mice as a function of trauma strength: ribbons are more vulnerable than postsynapses. Front Cell Neurosci 2024; 18:1465216. [PMID: 39411002 PMCID: PMC11473312 DOI: 10.3389/fncel.2024.1465216] [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: 07/15/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
Noise-induced cochlear synaptopathy is characterized by irreversible loss of synapses between inner hair cells (IHCs) and spiral ganglion neurons (SGNs) despite normal hearing thresholds. We analyzed hearing performance and cochlear structure in C57BL/6 N mice exposed to 100, 106, or 112 dB SPL broadband noise (8-16 kHz) for 2 h. Auditory brainstem responses (ABRs) were assessed before, directly after, and up to 28 days post-trauma. Finally, the number, size, and pairing of IHC presynaptic (CtBP2-positive) ribbons and postsynaptic AMPA receptor scaffold (Homer1-positive) clusters were analyzed along the cochlea. Four weeks after the 100 dB SPL trauma, a permanent threshold shift (PTS) was observed at 45 kHz, which after the higher traumata extended toward middle to low frequencies. Loss in ABR wave I amplitudes scaled with trauma strength indicating loss of functional IHC synaptic connections. Latencies of wave I mostly increased with trauma strength. No trauma-related OHC loss was found. The number of synaptic pairs was reduced in the midbasal and basal cochlear region in all trauma conditions, with ribbon loss amounting up to 46% of control. Ribbons surviving the trauma were paired, whereas 4-6 unpaired postsynapses/IHC were found in the medial, midbasal, and basal regions irrespective of trauma strength, contrasting findings in CBA/CaJ mice. Our data confirm the susceptibility of ribbon synapses and ABR wave I amplitudes to a noise trauma of 100 dB SPL or larger. Notably, peripheral dendrites bearing IHC postsynapses were less vulnerable than presynaptic ribbons in C57BL/6 N mice.
Collapse
Affiliation(s)
- Kerstin Blum
- Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Department of Biophysics, Saarland University, Homburg, Germany
- Center for Gender-specific Biology and Medicine (CGBM), School of Medicine, Saarland University, Homburg, Germany
| | - Pauline Schepsky
- Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Department of Biophysics, Saarland University, Homburg, Germany
| | - Philip Derleder
- Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Department of Biophysics, Saarland University, Homburg, Germany
| | - Philipp Schätzle
- Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Department of Biophysics, Saarland University, Homburg, Germany
| | - Fahmi Nasri
- Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Department of Biophysics, Saarland University, Homburg, Germany
| | - Philipp Fischer
- Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Department of Biophysics, Saarland University, Homburg, Germany
| | - Jutta Engel
- Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Department of Biophysics, Saarland University, Homburg, Germany
- Center for Gender-specific Biology and Medicine (CGBM), School of Medicine, Saarland University, Homburg, Germany
| | - Simone Kurt
- Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Department of Biophysics, Saarland University, Homburg, Germany
- Center for Gender-specific Biology and Medicine (CGBM), School of Medicine, Saarland University, Homburg, Germany
| |
Collapse
|
23
|
Conrad LJ, Grandi FC, Carlton AJ, Jeng JY, de Tomasi L, Zarecki P, Marcotti W, Johnson SL, Mustapha M. The upregulation of K + and HCN channels in developing spiral ganglion neurons is mediated by cochlear inner hair cells. J Physiol 2024; 602:5329-5351. [PMID: 39324853 DOI: 10.1113/jp286134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 09/02/2024] [Indexed: 09/27/2024] Open
Abstract
Spiral ganglion neurons (SGNs) are primary sensory afferent neurons that relay acoustic information from the cochlear inner hair cells (IHCs) to the brainstem. The response properties of different SGNs diverge to represent a wide range of sound intensities in an action-potential code. This biophysical heterogeneity is established during pre-hearing stages of development, a time when IHCs fire spontaneous Ca2+ action potentials that drive glutamate release from their ribbon synapses onto the SGN terminals. The role of spontaneous IHC activity in the refinement of SGN characteristics is still largely unknown. Using pre-hearing otoferlin knockout mice (Otof-/-), in which Ca2+-dependent exocytosis in IHCs is abolished, we found that developing SGNs fail to upregulate low-voltage-activated K+-channels and hyperpolarisation-activated cyclic-nucleotide-gated channels. This delayed maturation resulted in hyperexcitable SGNs with immature firing characteristics. We have also shown that SGNs that synapse with the pillar side of the IHCs selectively express a resurgent K+ current, highlighting a novel biophysical marker for these neurons. RNA-sequencing showed that several K+ channels are downregulated in Otof-/- mice, further supporting the electrophysiological recordings. Our data demonstrate that spontaneous Ca2+-dependent activity in pre-hearing IHCs regulates some of the key biophysical and molecular features of the developing SGNs. KEY POINTS: Ca2+-dependent exocytosis in inner hair cells (IHCs) is otoferlin-dependent as early as postnatal day 1. A lack of otoferlin in IHCs affects potassium channel expression in SGNs. The absence of otoferlin is associated with SGN hyperexcitability. We propose that type I spiral ganglion neuron functional maturation depends on IHC exocytosis.
Collapse
Affiliation(s)
- Linus J Conrad
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Fiorella C Grandi
- INSERM, Institute de Myologie, Centre de Recherche en Myologie F-75013, Sorbonne Université, Paris, France
| | - Adam J Carlton
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Jing-Yi Jeng
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Lara de Tomasi
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Patryk Zarecki
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Walter Marcotti
- School of Biosciences, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Stuart L Johnson
- School of Biosciences, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Mirna Mustapha
- School of Biosciences, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| |
Collapse
|
24
|
Chen J, Lewis MA, Wai A, Yin L, Dawson SJ, Ingham NJ, Steel KP. A new mutation of Sgms1 causes gradual hearing loss associated with a reduced endocochlear potential. Hear Res 2024; 451:109091. [PMID: 39067415 DOI: 10.1016/j.heares.2024.109091] [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: 04/25/2024] [Revised: 07/04/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Sgms1 encodes sphingomyelin synthase 1, an enzyme in the sphingosine-1-phosphate signalling pathway, and was previously reported to underlie hearing impairment in the mouse. A new mouse allele, Sgms1tm1a, unexpectedly showed normal Auditory Brainstem Response thresholds. We found that the Sgms1tm1a mutation led to incomplete knockdown of transcript to 20 % of normal values, which was enough to support normal hearing. The Sgms1tm1b allele was generated by knocking out exon 7, leading to a complete lack of detectable transcript in the inner ear. Sgms1tm1b homozygotes showed largely normal auditory brainstem response thresholds at first, followed by progressive loss of sensitivity until they showed severe impairment at 6 months old. The endocochlear potential was consistently reduced in Sgms1tm1b mutants at 3, 4 and 8 weeks old, to around 80 mV compared with around 120 mV in control littermates. The stria vascularis showed a characteristic irregularity of marginal cell surfaces and patchy loss of Kcnq1 expression at their apical membrane, and expression analysis of the lateral wall suggested that marginal cells were the most likely initial site of dysfunction in the mutants. Finally, significant association of auditory thresholds with DNA markers within and close to the human SGMS1 gene were found in the 1958 Birth Cohort, suggesting that SGMS1 variants may play a role in the range of hearing abilities in the human population.
Collapse
Affiliation(s)
- Jing Chen
- Wolfson Sensory, Pain and Regeneration Centre, King's College London, London SE1 1UL, United Kingdom
| | - Morag A Lewis
- Wolfson Sensory, Pain and Regeneration Centre, King's College London, London SE1 1UL, United Kingdom
| | - Alisa Wai
- Wolfson Sensory, Pain and Regeneration Centre, King's College London, London SE1 1UL, United Kingdom
| | - Lucia Yin
- Wolfson Sensory, Pain and Regeneration Centre, King's College London, London SE1 1UL, United Kingdom
| | - Sally J Dawson
- UCL Ear Institute, University College London, London WC1X 8EE, United Kingdom
| | - Neil J Ingham
- Wolfson Sensory, Pain and Regeneration Centre, King's College London, London SE1 1UL, United Kingdom
| | - Karen P Steel
- Wolfson Sensory, Pain and Regeneration Centre, King's College London, London SE1 1UL, United Kingdom.
| |
Collapse
|
25
|
Messat Y, Martin-Fernandez M, Assou S, Chung K, Guérin F, Gergely C, Cuisinier F, Zine A. Differentiation of Spiral Ganglion Neurons from Human Dental Pulp Stem Cells: A Further Step towards Autologous Auditory Nerve Recovery. Int J Mol Sci 2024; 25:9115. [PMID: 39201803 PMCID: PMC11354632 DOI: 10.3390/ijms25169115] [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: 06/14/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 09/03/2024] Open
Abstract
The degeneration of spiral ganglion neurons (SGNs), which convey auditory signals from hair cells to the brain, can be a primary cause of sensorineural hearing loss (SNHL) or can occur secondary to hair cell loss. Emerging therapies for SNHL include the replacement of damaged SGNs using stem cell-derived otic neuronal progenitors (ONPs). However, the availability of renewable, accessible, and patient-matched sources of human stem cells is a prerequisite for successful replacement of the auditory nerve. In this study, we derived ONP and SGN-like cells by a reliable and reproducible stepwise guidance differentiation procedure of self-renewing human dental pulp stem cells (hDPSCs). This in vitro differentiation protocol relies on the modulation of BMP and TGFβ pathways using a free-floating 3D neurosphere method, followed by differentiation on a Geltrex-coated surface using two culture paradigms to modulate the major factors and pathways involved in early otic neurogenesis. Gene and protein expression analyses revealed efficient induction of a comprehensive panel of known ONP and SGN-like cell markers during the time course of hDPSCs differentiation. Atomic force microscopy revealed that hDPSC-derived SGN-like cells exhibit similar nanomechanical properties as their in vivo SGN counterparts. Furthermore, spiral ganglion neurons from newborn rats come in close contact with hDPSC-derived ONPs 5 days after co-culturing. Our data demonstrate the capability of hDPSCs to generate SGN-like neurons with specific lineage marker expression, bipolar morphology, and the nanomechanical characteristics of SGNs, suggesting that the neurons could be used for next-generation cochlear implants and/or inner ear cell-based strategies for SNHL.
Collapse
Affiliation(s)
- Yassine Messat
- LBN, Laboratory of Bioengineering and Nanoscience, University of Montpellier, 34193 Montpellier, France
| | - Marta Martin-Fernandez
- L2C, Laboratoire Charles Coulomb, University of Montpellier, CNRS, 34095 Montpellier, France
| | - Said Assou
- IRMB, Institute for Regenerative Medicine & Biotherapy, University of Montpellier, INSERM, CHU Montpellier, 34295 Montpellier, France;
| | - Keshi Chung
- LBN, Laboratory of Bioengineering and Nanoscience, University of Montpellier, 34193 Montpellier, France
| | - Frederic Guérin
- Faculté de Médecine, University of Montpellier, 34090 Montpellier, France
| | - Csilla Gergely
- L2C, Laboratoire Charles Coulomb, University of Montpellier, CNRS, 34095 Montpellier, France
| | - Frederic Cuisinier
- LBN, Laboratory of Bioengineering and Nanoscience, University of Montpellier, 34193 Montpellier, France
| | - Azel Zine
- LBN, Laboratory of Bioengineering and Nanoscience, University of Montpellier, 34193 Montpellier, France
| |
Collapse
|
26
|
Pyott SJ, Pavlinkova G, Yamoah EN, Fritzsch B. Harmony in the Molecular Orchestra of Hearing: Developmental Mechanisms from the Ear to the Brain. Annu Rev Neurosci 2024; 47:1-20. [PMID: 38360566 PMCID: PMC11787624 DOI: 10.1146/annurev-neuro-081423-093942] [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: 02/17/2024]
Abstract
Auditory processing in mammals begins in the peripheral inner ear and extends to the auditory cortex. Sound is transduced from mechanical stimuli into electrochemical signals of hair cells, which relay auditory information via the primary auditory neurons to cochlear nuclei. Information is subsequently processed in the superior olivary complex, lateral lemniscus, and inferior colliculus and projects to the auditory cortex via the medial geniculate body in the thalamus. Recent advances have provided valuable insights into the development and functioning of auditory structures, complementing our understanding of the physiological mechanisms underlying auditory processing. This comprehensive review explores the genetic mechanisms required for auditory system development from the peripheral cochlea to the auditory cortex. We highlight transcription factors and other genes with key recurring and interacting roles in guiding auditory system development and organization. Understanding these gene regulatory networks holds promise for developing novel therapeutic strategies for hearing disorders, benefiting millions globally.
Collapse
Affiliation(s)
- Sonja J Pyott
- Department of Otorhinolaryngology and Head and Neck Surgery, University Medical Center Groningen, Graduate School of Medical Sciences, and Research School of Behavioral and Cognitive Neurosciences, University of Groningen, Groningen, The Netherlands
| | - Gabriela Pavlinkova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology, Czech Academy of Sciences, Vestec, Czechia
| | - Ebenezer N Yamoah
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, Nevada, USA
| | - Bernd Fritzsch
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA;
| |
Collapse
|
27
|
Lu Y, Jiang Y, Wang F, Wu H, Hua Y. Electron Microscopic Mapping of Mitochondrial Morphology in the Cochlear Nerve Fibers. J Assoc Res Otolaryngol 2024; 25:341-354. [PMID: 38937328 PMCID: PMC11349726 DOI: 10.1007/s10162-024-00957-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 06/12/2024] [Indexed: 06/29/2024] Open
Abstract
To enable nervous system function, neurons are powered in a use-dependent manner by mitochondria undergoing morphological-functional adaptation. In a well-studied model system-the mammalian cochlea, auditory nerve fibers (ANFs) display distinct electrophysiological properties, which is essential for collectively sampling acoustic information of a large dynamic range. How exactly the associated mitochondrial networks are deployed in functionally differentiated ANFs remains scarcely interrogated. Here, we leverage volume electron microscopy and machine-learning-assisted image analysis to phenotype mitochondrial morphology and distribution along ANFs of full-length in the mouse cochlea inner spiral bundle. This reveals greater variance in mitochondrial size with increased ANF habenula to terminal path length. Particularly, we analyzed the ANF terminal-residing mitochondria, which are critical for local calcium uptake during sustained afferent activities. Our results suggest that terminal-specific enrichment of mitochondria, in addition to terminal size and overall mitochondrial abundance of the ANF, correlates with heterogenous mitochondrial contents of the terminal.
Collapse
Affiliation(s)
- Yan Lu
- 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
| | - Yi Jiang
- 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
| | - 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
| | - 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.
| |
Collapse
|
28
|
Vincent PFY, Young ED, Edge ASB, Glowatzki E. Auditory hair cells and spiral ganglion neurons regenerate synapses with refined release properties in vitro. Proc Natl Acad Sci U S A 2024; 121:e2315599121. [PMID: 39058581 PMCID: PMC11294990 DOI: 10.1073/pnas.2315599121] [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: 09/10/2023] [Accepted: 06/12/2024] [Indexed: 07/28/2024] Open
Abstract
Ribbon synapses between inner hair cells (IHCs) and type I spiral ganglion neurons (SGNs) in the inner ear are damaged by noise trauma and with aging, causing "synaptopathy" and hearing loss. Cocultures of neonatal denervated organs of Corti and newly introduced SGNs have been developed to find strategies for improving IHC synapse regeneration, but evidence of the physiological normality of regenerated synapses is missing. This study utilizes IHC optogenetic stimulation and SGN recordings, showing that, when P3-5 denervated organs of Corti are cocultured with SGNs, newly formed IHC/SGN synapses are indeed functional, exhibiting glutamatergic excitatory postsynaptic currents. When using older organs of Corti at P10-11, synaptic activity probed by deconvolution showed more mature release properties, closer to the specialized mode of IHC synaptic transmission crucial for coding the sound signal. This functional assessment of newly formed IHC synapses developed here, provides a powerful tool for testing approaches to improve synapse regeneration.
Collapse
Affiliation(s)
- Philippe F. Y. Vincent
- The Center for Hearing and Balance, The Johns Hopkins School of Medicine, Baltimore, MD21205
- Department of Otolaryngology Head and Neck Surgery, The Johns Hopkins School of Medicine, Baltimore, MD21205
| | - Eric D. Young
- The Center for Hearing and Balance, The Johns Hopkins School of Medicine, Baltimore, MD21205
- Department of Otolaryngology Head and Neck Surgery, The Johns Hopkins School of Medicine, Baltimore, MD21205
- Department of Neuroscience, The Johns Hopkins School of Medicine, Baltimore, MD21205
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD21205
| | - Albert S. B. Edge
- Department of Otolaryngology, Harvard Medical School, Boston, MA02115
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, MA02114
- Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, MA02115
- Harvard Stem Cell Institute, Cambridge, MA02139
| | - Elisabeth Glowatzki
- The Center for Hearing and Balance, The Johns Hopkins School of Medicine, Baltimore, MD21205
- Department of Otolaryngology Head and Neck Surgery, The Johns Hopkins School of Medicine, Baltimore, MD21205
- Department of Neuroscience, The Johns Hopkins School of Medicine, Baltimore, MD21205
| |
Collapse
|
29
|
Kersbergen CJ, Bergles DE. Priming central sound processing circuits through induction of spontaneous activity in the cochlea before hearing onset. Trends Neurosci 2024; 47:522-537. [PMID: 38782701 PMCID: PMC11236524 DOI: 10.1016/j.tins.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/02/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024]
Abstract
Sensory systems experience a period of intrinsically generated neural activity before maturation is complete and sensory transduction occurs. Here we review evidence describing the mechanisms and functions of this 'spontaneous' activity in the auditory system. Both ex vivo and in vivo studies indicate that this correlated activity is initiated by non-sensory supporting cells within the developing cochlea, which induce depolarization and burst firing of groups of nearby hair cells in the sensory epithelium, activity that is conveyed to auditory neurons that will later process similar sound features. This stereotyped neural burst firing promotes cellular maturation, synaptic refinement, acoustic sensitivity, and establishment of sound-responsive domains in the brain. While sensitive to perturbation, the developing auditory system exhibits remarkable homeostatic mechanisms to preserve periodic burst firing in deaf mice. Preservation of this early spontaneous activity in the context of deafness may enhance the efficacy of later interventions to restore hearing.
Collapse
Affiliation(s)
- Calvin J Kersbergen
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Dwight E Bergles
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Otolaryngology - Head and Neck Surgery, Johns Hopkins University, Baltimore, MD, USA; Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
30
|
Ma X, Guo J, Tian M, Fu Y, Jiang P, Zhang Y, Chai R. Advance and Application of Single-cell Transcriptomics in Auditory Research. Neurosci Bull 2024; 40:963-980. [PMID: 38015350 PMCID: PMC11250760 DOI: 10.1007/s12264-023-01149-z] [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: 06/27/2023] [Accepted: 08/03/2023] [Indexed: 11/29/2023] Open
Abstract
Hearing loss and deafness, as a worldwide disability disease, have been troubling human beings. However, the auditory organ of the inner ear is highly heterogeneous and has a very limited number of cells, which are largely uncharacterized in depth. Recently, with the development and utilization of single-cell RNA sequencing (scRNA-seq), researchers have been able to unveil the complex and sophisticated biological mechanisms of various types of cells in the auditory organ at the single-cell level and address the challenges of cellular heterogeneity that are not resolved through by conventional bulk RNA sequencing (bulk RNA-seq). Herein, we reviewed the application of scRNA-seq technology in auditory research, with the aim of providing a reference for the development of auditory organs, the pathogenesis of hearing loss, and regenerative therapy. Prospects about spatial transcriptomic scRNA-seq, single-cell based genome, and Live-seq technology will also be discussed.
Collapse
Affiliation(s)
- Xiangyu Ma
- State Key Laboratory of Digital Medical Engineering, 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, 210096, China
| | - Jiamin Guo
- State Key Laboratory of Digital Medical Engineering, 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, 210096, China
| | - Mengyao Tian
- State Key Laboratory of Digital Medical Engineering, 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, 210096, China
| | - Yaoyang Fu
- Department of Psychiatry, Affiliated Hangzhou First People's Hospital, Zhejiang University school of Medicine, Hangzhou, 310030, China
| | - Pei Jiang
- State Key Laboratory of Digital Medical Engineering, 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, 210096, China
| | - Yuan Zhang
- Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing, China
- Research Institute of Otolaryngology, Nanjing, 210008, China
| | - Renjie Chai
- State Key Laboratory of Digital Medical Engineering, 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, 210096, China.
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing, 101408, China.
- Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
31
|
Núñez KR, Bronson D, Chang R, Kalluri R. Vestibular afferent neurons develop normally in the absence of quantal/glutamatergic input. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.597464. [PMID: 38915604 PMCID: PMC11195208 DOI: 10.1101/2024.06.12.597464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
The vestibular nerve is comprised of neuron sub-groups with diverse functions related to their intrinsic biophysical properties. This diversity is partly due to differences in the types and numbers of low-voltage-gated potassium channels found in the neurons' membranes. Expression for some low-voltage gated ion channels like KCNQ4 is upregulated during early post-natal development; suggesting that ion channel composition and neuronal diversity may be shaped by hair cell activity. This idea is consistent with recent work showing that glutamatergic input from hair cells is necessary for the normal diversification auditory neurons. To test if biophysical diversity is similarly dependent on glutamatergic input in vestibular neurons, we examined the maturation of the vestibular epithelium and ganglion neurons in Vglut3-ko mice whose hair cell synapses lack glutamate. Despite lacking glutamatergic input, the knockout mice showed no notable balance deficits and crossed challenging balance beams with little difficulty. Immunolabeling of the Vglut3-ko vestibular epithelia showed normal development as indicated by an identifiable striolar zone with calyceal terminals labeled by molecular marker calretinin, and normal expression of KCNQ4 by the end of the second post-natal week. We found similar numbers of Type I and Type II hair cells in the knockout and wildtype animals, regardless of epithelial zone. Thus, the presumably quiescent Type II hair cells are not cleared from the epithelium. Patch-clamp recordings showed that biophysical diversity of vestibular ganglion neurons in the Vglut3-ko mice is comparable to that found in wildtype controls, with a similar range firing patterns at both immature and juvenile ages. However, our results suggest a subtle biophysical alteration to the largest ganglion cells (putative somata of central zone afferents); those in the knockout had smaller net conductance and were more excitable than those in the wild type. Thus, unlike in the auditory nerve, glutamatergic signaling is unnecessary for producing biophysical diversity in vestibular ganglion neurons. And yet, because the input signals from vestibular hair cells are complex and not solely reliant on quantal release of glutamate, whether diversity of vestibular ganglion neurons is simply hardwired or regulated by a more complex set of input signals remains to be determined.
Collapse
|
32
|
Maraslioglu-Sperber A, Pizzi E, Fisch JO, Kattler K, Ritter T, Friauf E. Molecular and functional profiling of cell diversity and identity in the lateral superior olive, an auditory brainstem center with ascending and descending projections. Front Cell Neurosci 2024; 18:1354520. [PMID: 38846638 PMCID: PMC11153811 DOI: 10.3389/fncel.2024.1354520] [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: 12/12/2023] [Accepted: 03/15/2024] [Indexed: 06/09/2024] Open
Abstract
The lateral superior olive (LSO), a prominent integration center in the auditory brainstem, contains a remarkably heterogeneous population of neurons. Ascending neurons, predominantly principal neurons (pLSOs), process interaural level differences for sound localization. Descending neurons (lateral olivocochlear neurons, LOCs) provide feedback into the cochlea and are thought to protect against acoustic overload. The molecular determinants of the neuronal diversity in the LSO are largely unknown. Here, we used patch-seq analysis in mice at postnatal days P10-12 to classify developing LSO neurons according to their functional and molecular profiles. Across the entire sample (n = 86 neurons), genes involved in ATP synthesis were particularly highly expressed, confirming the energy expenditure of auditory neurons. Two clusters were identified, pLSOs and LOCs. They were distinguished by 353 differentially expressed genes (DEGs), most of which were novel for the LSO. Electrophysiological analysis confirmed the transcriptomic clustering. We focused on genes affecting neuronal input-output properties and validated some of them by immunohistochemistry, electrophysiology, and pharmacology. These genes encode proteins such as osteopontin, Kv11.3, and Kvβ3 (pLSO-specific), calcitonin-gene-related peptide (LOC-specific), or Kv7.2 and Kv7.3 (no DEGs). We identified 12 "Super DEGs" and 12 genes showing "Cluster similarity." Collectively, we provide fundamental and comprehensive insights into the molecular composition of individual ascending and descending neurons in the juvenile auditory brainstem and how this may relate to their specific functions, including developmental aspects.
Collapse
Affiliation(s)
- Ayse Maraslioglu-Sperber
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Erika Pizzi
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Jonas O. Fisch
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Kathrin Kattler
- Genetics/Epigenetics Group, Department of Biological Sciences, Saarland University, Saarbrücken, Germany
| | - Tamara Ritter
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Eckhard Friauf
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| |
Collapse
|
33
|
Carlton AJ, Jeng JY, Grandi FC, De Faveri F, Amariutei AE, De Tomasi L, O'Connor A, Johnson SL, Furness DN, Brown SDM, Ceriani F, Bowl MR, Mustapha M, Marcotti W. BAI1 localizes AMPA receptors at the cochlear afferent post-synaptic density and is essential for hearing. Cell Rep 2024; 43:114025. [PMID: 38564333 DOI: 10.1016/j.celrep.2024.114025] [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: 09/14/2023] [Revised: 01/25/2024] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
Type I spiral ganglion neurons (SGNs) convey sound information to the central auditory pathway by forming synapses with inner hair cells (IHCs) in the mammalian cochlea. The molecular mechanisms regulating the formation of the post-synaptic density (PSD) in the SGN afferent terminals are still unclear. Here, we demonstrate that brain-specific angiogenesis inhibitor 1 (BAI1) is required for the clustering of AMPA receptors GluR2-4 (glutamate receptors 2-4) at the PSD. Adult Bai1-deficient mice have functional IHCs but fail to transmit information to the SGNs, leading to highly raised hearing thresholds. Despite the almost complete absence of AMPA receptor subunits, the SGN fibers innervating the IHCs do not degenerate. Furthermore, we show that AMPA receptors are still expressed in the cochlea of Bai1-deficient mice, highlighting a role for BAI1 in trafficking or anchoring GluR2-4 to the PSDs. These findings identify molecular and functional mechanisms required for sound encoding at cochlear ribbon synapses.
Collapse
Affiliation(s)
- Adam J Carlton
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | - Jing-Yi Jeng
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | - Fiorella C Grandi
- Sorbonne Université, INSERM, Institute de Myologie, Centre de Recherche en Myologie, 75013 Paris, France
| | | | - Ana E Amariutei
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | - Lara De Tomasi
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | - Andrew O'Connor
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | - Stuart L Johnson
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK; Neuroscience Institute, University of Sheffield, Sheffield S10 2TN, UK
| | - David N Furness
- School of Life Sciences, Keele University, Keele ST5 5BG, UK
| | - Steve D M Brown
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK
| | - Federico Ceriani
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | - Michael R Bowl
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK
| | - Mirna Mustapha
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK; Neuroscience Institute, University of Sheffield, Sheffield S10 2TN, UK
| | - Walter Marcotti
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK; Neuroscience Institute, University of Sheffield, Sheffield S10 2TN, UK.
| |
Collapse
|
34
|
Wang G, Gu Y, Liu Z. Deciphering the genetic interactions between Pou4f3, Gfi1, and Rbm24 in maintaining mouse cochlear hair cell survival. eLife 2024; 12:RP90025. [PMID: 38483314 PMCID: PMC10939501 DOI: 10.7554/elife.90025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024] Open
Abstract
Mammals harbor a limited number of sound-receptor hair cells (HCs) that cannot be regenerated after damage. Thus, investigating the underlying molecular mechanisms that maintain HC survival is crucial for preventing hearing impairment. Intriguingly, Pou4f3-/- or Gfi1-/- HCs form initially but then rapidly degenerate, whereas Rbm24-/- HCs degenerate considerably later. However, the transcriptional cascades involving Pou4f3, Gfi1, and Rbm24 remain undescribed. Here, we demonstrate that Rbm24 expression is completely repressed in Pou4f3-/- HCs but unaltered in Gfi1-/- HCs, and further that the expression of both POU4F3 and GFI1 is intact in Rbm24-/- HCs. Moreover, by using in vivo mouse transgenic reporter assays, we identify three Rbm24 enhancers to which POU4F3 binds. Lastly, through in vivo genetic testing of whether Rbm24 restoration alleviates the degeneration of Pou4f3-/- HCs, we show that ectopic Rbm24 alone cannot prevent Pou4f3-/- HCs from degenerating. Collectively, our findings provide new molecular and genetic insights into how HC survival is regulated.
Collapse
Affiliation(s)
- Guangqin Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yunpeng Gu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Zhiyong Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
- Shanghai Center for Brain Science and Brain-Inspired Intelligence TechnologyShanghaiChina
| |
Collapse
|
35
|
Yu Q, Liu S, Guo R, Chen K, Li Y, Jiang D, Gong S, Yin L, Liu K. Complete Restoration of Hearing Loss and Cochlear Synaptopathy via Minimally Invasive, Single-Dose, and Controllable Middle Ear Delivery of Brain-Derived Neurotrophic Factor-Poly(dl-lactic acid- co-glycolic acid)-Loaded Hydrogel. ACS NANO 2024; 18:6298-6313. [PMID: 38345574 DOI: 10.1021/acsnano.3c11049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Noise-induced hearing loss (NIHL) often accompanies cochlear synaptopathy, which can be potentially reversed to restore hearing. However, there has been little success in achieving complete recovery of sensorineural deafness using nearly noninvasive middle ear drug delivery before. Here, we present a study demonstrating the efficacy of a middle ear delivery system employing brain-derived neurotrophic factor (BDNF)-poly-(dl-lactic acid-co-glycolic acid) (PLGA)-loaded hydrogel in reversing synaptopathy and restoring hearing function in a mouse model with NIHL. The mouse model achieved using the single noise exposure (NE, 115 dBL, 4 h) exhibited an average 20 dBL elevation of hearing thresholds with intact cochlear hair cells but a loss of ribbon synapses as the primary cause of hearing impairment. We developed a BDNF-PLGA-loaded thermosensitive hydrogel, which was administered via a single controllable injection into the tympanic cavity of noise-exposed mice, allowing its presence in the middle ear for a duration of 2 weeks. This intervention resulted in complete restoration of NIHL at frequencies of click, 4, 8, 16, and 32 kHz. Moreover, the cochlear ribbon synapses exhibited significant recovery, whereas other cochlear components (hair cells and auditory nerves) remained unchanged. Additionally, the cochlea of NE treated mice revealed activation of tropomyosin receptor kinase B (TRKB) signaling upon exposure to BDNF. These findings demonstrate a controllable and minimally invasive therapeutic approach that utilizes a BDNF-PLGA-loaded hydrogel to restore NIHL by specifically repairing cochlear synaptopathy. This tailored middle ear delivery system holds great promise for achieving ideal clinical outcomes in the treatment of NIHL and cochlear synaptopathy.
Collapse
Affiliation(s)
- Qianru Yu
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Shengnan Liu
- School of Materials Science and Engineering,Tsinghua University, Beijing 100084, China
| | - Rui Guo
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Kuntao Chen
- School of Materials Science and Engineering,Tsinghua University, Beijing 100084, China
| | - Yang Li
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Dan Jiang
- Hearing Implant Centre, Guy's and St. Thomas NHS Foundation Trust, London SE1 7EH, United Kingdom
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, United Kingdom
| | - Shusheng Gong
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
- Clinical Center for Hearing Loss, Capital Medical University, Beijing 100050, China
| | - Lan Yin
- School of Materials Science and Engineering,Tsinghua University, Beijing 100084, China
| | - Ke Liu
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
- Clinical Center for Hearing Loss, Capital Medical University, Beijing 100050, China
| |
Collapse
|
36
|
Lu Y, Liu J, Li B, Wang H, Wang F, Wang S, Wu H, Han H, Hua Y. Spatial patterns of noise-induced inner hair cell ribbon loss in the mouse mid-cochlea. iScience 2024; 27:108825. [PMID: 38313060 PMCID: PMC10835352 DOI: 10.1016/j.isci.2024.108825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/16/2023] [Accepted: 01/03/2024] [Indexed: 02/06/2024] Open
Abstract
In the mammalian cochlea, moderate acoustic overexposure leads to loss of ribbon-type synapse between the inner hair cell (IHC) and its postsynaptic spiral ganglion neuron (SGN), causing a reduced dynamic range of hearing but not a permanent threshold elevation. A prevailing view is that such ribbon loss (known as synaptopathy) selectively impacts the low-spontaneous-rate and high-threshold SGN fibers contacting predominantly the modiolar IHC face. However, the spatial pattern of synaptopathy remains scarcely characterized in the most sensitive mid-cochlear region, where two morphological subtypes of IHC with distinct ribbon size gradients coexist. Here, we used volume electron microscopy to investigate noise exposure-related changes in the mouse IHCs with and without ribbon loss. Our quantifications reveal that IHC subtypes differ in the worst-hit area of synaptopathy. Moreover, we show relative enrichment of mitochondria in the surviving SGN terminals, providing key experimental evidence for the long-proposed role of SGN-terminal mitochondria in synaptic vulnerability.
Collapse
Affiliation(s)
- Yan Lu
- 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
- Shanghai Institute of Precision Medicine, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Jing Liu
- Laboratory of Brain Atlas and Brain-inspired Intelligence, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Bei Li
- 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
| | - Haoyu Wang
- Shanghai Institute of Precision Medicine, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Fangfang Wang
- Shanghai Institute of Precision Medicine, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Shengxiong Wang
- Shanghai Institute of Precision Medicine, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Hao Wu
- 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
- Shanghai Institute of Precision Medicine, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Hua Han
- Laboratory of Brain Atlas and Brain-inspired Intelligence, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing 101408, China
- State Key Laboratory of Multimodal Artificial Intelligence Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Yunfeng Hua
- 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
- Shanghai Institute of Precision Medicine, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| |
Collapse
|
37
|
Jukic A, Lei Z, Cebul ER, Pinter K, Mosqueda N, David S, Tarchini B, Kindt K. Presynaptic Nrxn3 is essential for ribbon-synapse assembly in hair cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.14.580267. [PMID: 38410471 PMCID: PMC10896334 DOI: 10.1101/2024.02.14.580267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Hair cells of the inner ear rely on specialized ribbon synapses to transmit sensory information to the central nervous system. The molecules required to assemble these synapses are not fully understood. We show that Nrxn3, a presynaptic adhesion molecule, is critical for ribbon-synapse assembly in hair cells. In both mouse and zebrafish models, loss of Nrxn3 results in significantly fewer intact ribbon synapses. In zebrafish we demonstrate that a 60% loss of synapses in nrxn3 mutants dramatically reduces both presynaptic responses in hair cells and postsynaptic responses in afferent neurons. Despite a reduction in synapse function in this model, we find no deficits in the acoustic startle response, a behavior reliant on these synapses. Overall, this work demonstrates that Nrxn3 is a critical and conserved molecule required to assemble ribbon synapses. Understanding how ribbon synapses assemble is a key step towards generating novel therapies to treat forms of age-related and noise-induced hearing loss that occur due to loss of ribbon synapses.
Collapse
Affiliation(s)
- Alma Jukic
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, 20892, USA
| | - Zhengchang Lei
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, 20892, USA
| | - Elizabeth R Cebul
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, 20892, USA
| | - Katherine Pinter
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, 20892, USA
| | - Natalie Mosqueda
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, 20892, USA
| | - Sandeep David
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, 20892, USA
| | | | - Katie Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, 20892, USA
| |
Collapse
|
38
|
Kim J, Martinez E, Qiu J, Zhouli Ni J, Kwan KY. Chromatin remodeling protein CHD4 regulates axon guidance of spiral ganglion neurons in developing cochlea. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578202. [PMID: 38352369 PMCID: PMC10862897 DOI: 10.1101/2024.01.31.578202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
The chromodomain helicase binding protein 4 (CHD4) is an ATP-dependent chromatin remodeler. De-novo pathogenic variants of CHD4 cause Sifrim-Hitz-Weiss syndrome (SIHIWES). Patients with SIHIWES show delayed development, intellectual disability, facial dysmorphism, and hearing loss. Many cochlear cell types, including spiral ganglion neurons (SGNs), express CHD4. SGNs are the primary afferent neurons that convey sound information from the cochlea, but the function of CHD4 in SGNs is unknown. We employed the Neurog1(Ngn1) CreERT2 Chd4 conditional knockout animals to delete Chd4 in SGNs. SGNs are classified as type I and type II neurons. SGNs lacking CHD4 showed abnormal fasciculation of type I neurons along with improper pathfinding of type II fibers. CHD4 binding to chromatin from immortalized multipotent otic progenitor-derived neurons was used to identify candidate target genes in SGNs. Gene ontology analysis of CHD4 target genes revealed cellular processes involved in axon guidance, axonal fasciculation, and ephrin receptor signaling pathway. We validated increased Epha4 transcripts in SGNs from Chd4 conditional knockout cochleae. The results suggest that CHD4 attenuates the transcription of axon guidance genes to form the stereotypic pattern of SGN peripheral projections. The results implicate epigenetic changes in circuit wiring by modulating axon guidance molecule expression and provide insights into neurodevelopmental diseases.
Collapse
Affiliation(s)
- Jihyun Kim
- Keck Center for Collaborative Neuroscience and Stem Cell Research Center, Rutgers University, Piscataway, NJ 08854, USA
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Edward Martinez
- Keck Center for Collaborative Neuroscience and Stem Cell Research Center, Rutgers University, Piscataway, NJ 08854, USA
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Jingyun Qiu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Julie Zhouli Ni
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| | - Kelvin Y. Kwan
- Keck Center for Collaborative Neuroscience and Stem Cell Research Center, Rutgers University, Piscataway, NJ 08854, USA
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
39
|
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
|
40
|
Xie R, Wang M, Zhang C. Mechanisms of age-related hearing loss at the auditory nerve central synapses and postsynaptic neurons in the cochlear nucleus. Hear Res 2024; 442:108935. [PMID: 38113793 PMCID: PMC10842789 DOI: 10.1016/j.heares.2023.108935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 12/21/2023]
Abstract
Sound information is transduced from mechanical vibration to electrical signals in the cochlea, conveyed to and further processed in the brain to form auditory perception. During the process, spiral ganglion neurons (SGNs) are the key cells that connect the peripheral and central auditory systems by receiving information from hair cells in the cochlea and transmitting it to neurons of the cochlear nucleus (CN). Decades of research in the cochlea greatly improved our understanding of SGN function under normal and pathological conditions, especially about the roles of different subtypes of SGNs and their peripheral synapses. However, it remains less clear how SGN central terminals or auditory nerve (AN) synapses connect to CN neurons, and ultimately how peripheral pathology links to structural alterations and functional deficits in the central auditory nervous system. This review discusses recent progress about the morphological and physiological properties of different subtypes of AN synapses and associated postsynaptic CN neurons, their changes during aging, and the potential mechanisms underlying age-related hearing loss.
Collapse
Affiliation(s)
- Ruili Xie
- Department of Otolaryngology, The Ohio State University, 420 W 12th Ave, Columbus OH 43210, USA; Department of Neuroscience, The Ohio State University, 420W 12th Ave, Columbus, OH 43210, USA.
| | - Meijian Wang
- Department of Otolaryngology, The Ohio State University, 420 W 12th Ave, Columbus OH 43210, USA
| | - Chuangeng Zhang
- Department of Otolaryngology, The Ohio State University, 420 W 12th Ave, Columbus OH 43210, USA
| |
Collapse
|
41
|
Chen Y, Mu W, Wu Y, Xu J, Li X, Hu H, Wang S, Wang D, Hui B, Wang L, Dong Y, Chen W. Optogenetically modified human embryonic stem cell-derived otic neurons establish functional synaptic connection with cochlear nuclei. J Tissue Eng 2024; 15:20417314241265198. [PMID: 39092452 PMCID: PMC11292720 DOI: 10.1177/20417314241265198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 06/14/2024] [Indexed: 08/04/2024] Open
Abstract
Spiral ganglia neurons (SGNs) impairment can cause deafness. One important therapeutic approach involves utilizing stem cells to restore impaired auditory circuitry. Nevertheless, the inadequate implementation of research methodologies poses a challenge in accurately assessing the functionality of derived cells within the circuit. Here, we describe a novel method for converting human embryonic stem cells (hESCs) into otic neurons (ONs) and assess their functional connectivity using an optogenetic approach with cells or an organotypic slice of rat cochlear nucleus (CN) in coculture. Embryonic stem cell-derived otic neurons (eONs) exhibited SGN marker expression and generated functional synaptic connection when cocultured with cochlear nucleus neurons (CNNs). Synapsin 1 and VGLUT expression are found in the cochlear nucleus of brain slices, where eONs projected processes during the coculture of eONs and CN brain slices. Action potential spikes and INa+/IK+ of CNNs increased in tandem with light stimulations to eONs. These findings provide further evidence that eONs may be a candidate source to treat SGN-deafness.
Collapse
Affiliation(s)
- Yanni Chen
- Institute of Translational Medicine, and Children’s Hospital Affiliated and Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- College of Public Health, Shanghai University of Medicine & Health Sciences, Shanghai China
- Institute of Wound Prevention and Treatment, Shanghai University of Medicine & Health Sciences, Shanghai, China
- Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wenbo Mu
- Institute of Translational Medicine, and Children’s Hospital Affiliated and Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- College of Public Health, Shanghai University of Medicine & Health Sciences, Shanghai China
- Institute of Wound Prevention and Treatment, Shanghai University of Medicine & Health Sciences, Shanghai, China
- Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yongkang Wu
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, School of Physical Education & Health Care, East China Normal University, Shanghai, China
| | - Jiake Xu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Xiaofang Li
- Department of Neurology of the First Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Hu
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Siqi Wang
- College of Public Health, Shanghai University of Medicine & Health Sciences, Shanghai China
| | - Dali Wang
- Center for Clinical and Translational Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Bin Hui
- College of Pharmacy, Shanghai University of Medical & Health Sciences, Shanghai, China
| | - Lang Wang
- Department of Neurology of the First Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Dong
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, School of Physical Education & Health Care, East China Normal University, Shanghai, China
| | - Wei Chen
- Institute of Translational Medicine, and Children’s Hospital Affiliated and Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- College of Public Health, Shanghai University of Medicine & Health Sciences, Shanghai China
- Institute of Wound Prevention and Treatment, Shanghai University of Medicine & Health Sciences, Shanghai, China
- Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cellular Function and Pharmacology of Jilin Province, Yanbian University, Yanji, China
| |
Collapse
|
42
|
Vincent PF, Young ED, Edge AS, Glowatzki E. Auditory Hair Cells and Spiral Ganglion Neurons Regenerate Synapses with Refined Release Properties In Vitro. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.05.561095. [PMID: 38076928 PMCID: PMC10705289 DOI: 10.1101/2023.10.05.561095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Ribbon synapses between inner hair cells (IHCs) and type I spiral ganglion neurons (SGNs) in the inner ear are damaged by noise trauma and with aging, causing 'synaptopathy 'and hearing loss. Co-cultures of neonatal denervated organs of Corti and newly introduced SGNs have been developed to find strategies for improving IHC synapse regeneration, but evidence of the physiological normality of regenerated synapses is missing. This study utilizes IHC optogenetic stimulation and SGN recordings, showing that newly formed IHC synapses are indeed functional, exhibiting glutamatergic excitatory postsynaptic currents. When older organs of Corti were plated, synaptic activity probed by deconvolution, showed more mature release properties, closer to the highly specialized mode of IHC synaptic transmission that is crucial for coding the sound signal. This newly developed functional assessment of regenerated IHC synapses provides a powerful tool for testing approaches to improve synapse regeneration.
Collapse
Affiliation(s)
- Philippe F.Y. Vincent
- The Center for Hearing and Balance, The Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Otolaryngology Head and Neck Surgery, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Eric D. Young
- The Center for Hearing and Balance, The Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Otolaryngology Head and Neck Surgery, The Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Neuroscience, The Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Albert S.B. Edge
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts, USA
- Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Elisabeth Glowatzki
- The Center for Hearing and Balance, The Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Otolaryngology Head and Neck Surgery, The Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Neuroscience, The Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
43
|
Pan Y, Li S, He S, Wang G, Li C, Liu Z, Xiang M. Fgf8 P2A-3×GFP/+: A New Genetic Mouse Model for Specifically Labeling and Sorting Cochlear Inner Hair Cells. Neurosci Bull 2023; 39:1762-1774. [PMID: 37233921 PMCID: PMC10661496 DOI: 10.1007/s12264-023-01069-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/08/2023] [Indexed: 05/27/2023] Open
Abstract
The cochlear auditory epithelium contains two types of sound receptors, inner hair cells (IHCs) and outer hair cells (OHCs). Mouse models for labelling juvenile and adult IHCs or OHCs exist; however, labelling for embryonic and perinatal IHCs or OHCs are lacking. Here, we generated a new knock-in Fgf8P2A-3×GFP/+ (Fgf8GFP/+) strain, in which the expression of a series of three GFP fragments is controlled by endogenous Fgf8 cis-regulatory elements. After confirming that GFP expression accurately reflects the expression of Fgf8, we successfully obtained both embryonic and neonatal IHCs with high purity, highlighting the power of Fgf8GFP/+. Furthermore, our fate-mapping analysis revealed, unexpectedly, that IHCs are also derived from inner ear progenitors expressing Insm1, which is currently regarded as an OHC marker. Thus, besides serving as a highly favorable tool for sorting early IHCs, Fgf8GFP/+ will facilitate the isolation of pure early OHCs by excluding IHCs from the entire hair cell pool.
Collapse
Affiliation(s)
- Yi Pan
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shuting Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shunji He
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Guangqin Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chao Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Zhiyong Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 201210, China.
| | - Mingliang Xiang
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
44
|
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: 3.5] [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
|
45
|
Boussaty EC, Tedeschi N, Novotny M, Ninoyu Y, Du E, Draf C, Zhang Y, Manor U, Scheuermann RH, Friedman R. Cochlear transcriptome analysis of an outbred mouse population (CFW). Front Cell Neurosci 2023; 17:1256619. [PMID: 38094513 PMCID: PMC10716316 DOI: 10.3389/fncel.2023.1256619] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/11/2023] [Indexed: 12/20/2023] Open
Abstract
Age-related hearing loss (ARHL) is the most common cause of hearing loss and one of the most prevalent conditions affecting the elderly worldwide. Despite evidence from our lab and others about its polygenic nature, little is known about the specific genes, cell types, and pathways involved in ARHL, impeding the development of therapeutic interventions. In this manuscript, we describe, for the first time, the complete cell-type specific transcriptome of the aging mouse cochlea using snRNA-seq in an outbred mouse model in relation to auditory threshold variation. Cochlear cell types were identified using unsupervised clustering and annotated via a three-tiered approach-first by linking to expression of known marker genes, then using the NSForest algorithm to select minimum cluster-specific marker genes and reduce dimensional feature space for statistical comparison of our clusters with existing publicly-available data sets on the gEAR website, and finally, by validating and refining the annotations using Multiplexed Error Robust Fluorescence In Situ Hybridization (MERFISH) and the cluster-specific marker genes as probes. We report on 60 unique cell-types expanding the number of defined cochlear cell types by more than two times. Importantly, we show significant specific cell type increases and decreases associated with loss of hearing acuity implicating specific subsets of hair cell subtypes, ganglion cell subtypes, and cell subtypes within the stria vascularis in this model of ARHL. These results provide a view into the cellular and molecular mechanisms responsible for age-related hearing loss and pathways for therapeutic targeting.
Collapse
Affiliation(s)
- Ely Cheikh Boussaty
- Department of Otolaryngology, University of California, San Diego, La Jolla, CA, United States
| | - Neil Tedeschi
- J. Craig Venter Institute, La Jolla, CA, United States
| | - Mark Novotny
- J. Craig Venter Institute, La Jolla, CA, United States
| | - Yuzuru Ninoyu
- Department of Otolaryngology, University of California, San Diego, La Jolla, CA, United States
| | - Eric Du
- Department of Otolaryngology, University of California, San Diego, La Jolla, CA, United States
| | - Clara Draf
- Department of Otolaryngology, University of California, San Diego, La Jolla, CA, United States
| | - Yun Zhang
- J. Craig Venter Institute, La Jolla, CA, United States
| | - Uri Manor
- Department of Cell and Developmental Biology, University of California San Diego, Salk Institute for Biological Studies, Waitt Advanced Biophotonics Center, La Jolla, CA, United States
| | - Richard H. Scheuermann
- J. Craig Venter Institute, La Jolla, CA, United States
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States
| | - Rick Friedman
- Department of Otolaryngology, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
46
|
Hu Y, Fang L, Zhang H, Zheng S, Liao M, Cui Q, Wei H, Wu D, Cheng H, Qi Y, Wang H, Xin T, Wang T, Chai R. Emerging biotechnologies and biomedical engineering technologies for hearing reconstruction. SMART MEDICINE 2023; 2:e20230021. [PMID: 39188297 PMCID: PMC11235852 DOI: 10.1002/smmd.20230021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/09/2023] [Indexed: 08/28/2024]
Abstract
Hearing impairment is a global health problem that affects social communications and the economy. The damage and loss of cochlear hair cells and spiral ganglion neurons (SGNs) as well as the degeneration of neurites of SGNs are the core causes of sensorineural hearing loss. Biotechnologies and biomedical engineering technologies provide new hope for the treatment of auditory diseases, which utilizes biological strategies or tissue engineering methods to achieve drug delivery and the regeneration of cells, tissues, and even organs. Here, the advancements in the applications of biotechnologies (including gene therapy and cochlear organoids) and biomedical engineering technologies (including drug delivery, electrode coating, electrical stimulation and bionic scaffolds) in the field of hearing reconstruction are presented. Moreover, we summarize the challenges and provide a perspective on this field.
Collapse
Affiliation(s)
- Yangnan Hu
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologyAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
- Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Le Fang
- Department of NeurologyThe China‐Japan Union Hospital of Jilin UniversityChangchunJilinChina
| | - Hui Zhang
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologyAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
| | - Shasha Zheng
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologyAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
| | - Menghui Liao
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologyAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
| | - Qingyue Cui
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologyAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
| | - Hao Wei
- Department of Otolaryngology Head and Neck SurgeryAffiliated Drum Tower Hospital of Nanjing University Medical SchoolJiangsu Provincial Key Medical DisciplineNanjingChina
| | - Danqi Wu
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologyAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
| | - Hong Cheng
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologyAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
| | - Yanru Qi
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologyAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
| | - Huan Wang
- The Eighth Affiliated HospitalSun Yat‐Sen UniversityShenzhenChina
| | - Tao Xin
- Department of NeurosurgeryThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
- Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Tian Wang
- Department of Otolaryngology‐Head and Neck SurgeryStanford University School of MedicineStanfordCaliforniaUSA
- Department of Otolaryngology‐Head and Neck SurgeryThe Second Xiangya HospitalCentral South UniversityChangshaHunan ProvinceChina
| | - Renjie Chai
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologyAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
- Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
- Department of Otolaryngology Head and Neck SurgerySichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
- Beijing Key Laboratory of Neural Regeneration and RepairCapital Medical UniversityBeijingChina
| |
Collapse
|
47
|
Wang M, Lin S, Xie R. Apical-basal distribution of different subtypes of spiral ganglion neurons in the cochlea and the changes during aging. PLoS One 2023; 18:e0292676. [PMID: 37883357 PMCID: PMC10602254 DOI: 10.1371/journal.pone.0292676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023] Open
Abstract
Sound information is transmitted from the cochlea to the brain mainly by type I spiral ganglion neurons (SGNs), which consist of different subtypes with distinct physiological properties and selective expression of molecular markers. It remains unclear how these SGN subtypes distribute along the tonotopic axis, and whether the distribution pattern changes during aging that might underlie age-related hearing loss (ARHL). We investigated these questions using immunohistochemistry in three age groups of CBA/CaJ mice of either sex, including 2-5 months (young), 17-19 months (middle-age), and 28-32 months (old). Mouse cochleae were cryo-sectioned and triple-stained using antibodies against Tuj1, calretinin (CR) and calbindin (CB), which are reportedly expressed in all type I, subtype Ia, and subtype Ib SGNs, respectively. Labeled SGNs were classified into four groups based on the expression pattern of stained markers, including CR+ (subtype Ia), CB+ (subtype Ib), CR+CB+ (dual-labeled Ia/Ib), and CR-CB- (subtype Ic) neurons. The distribution of these SGN groups was analyzed in the apex, middle, and base regions of the cochleae. It showed that the prevalence of subtype Ia, Ib and dual-labeled Ia/Ib SGNs are high in the apex and low in the base. In contrast, the distribution pattern is reversed in Ic SGNs. Such frequency-dependent distribution is largely maintained during aging except for a preferential reduction of Ic SGNs, especially in the base. These findings corroborate the prior study based on RNAscope that SGN subtypes show differential vulnerability during aging. It suggests that sound processing of different frequencies involves distinct combinations of SGN subtypes, and the age-dependent loss of Ic SGNs in the base may especially impact high-frequency hearing during ARHL.
Collapse
Affiliation(s)
- Meijian Wang
- Department of Otolaryngology, The Ohio State University, Columbus, OH, United States of Ameirca
| | - Shengyin Lin
- Department of Otolaryngology, The Ohio State University, Columbus, OH, United States of Ameirca
| | - Ruili Xie
- Department of Otolaryngology, The Ohio State University, Columbus, OH, United States of Ameirca
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States of Ameirca
| |
Collapse
|
48
|
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: 1.0] [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
|
49
|
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
|
50
|
Zhang D, Ren M, Bi Z, Gu Y, Li S, Wang G, Li X, Liu Z. Lypd1-DTR/+: A New Mouse Model for Specifically Damaging the Type Ic Spiral Ganglion Neurons of the Cochlea. Neurosci Bull 2023; 39:1459-1462. [PMID: 37222854 PMCID: PMC10465461 DOI: 10.1007/s12264-023-01071-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 05/07/2023] [Indexed: 05/25/2023] Open
Affiliation(s)
- Di Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Minhui Ren
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - ZhengHong Bi
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yunpeng Gu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shuting Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guangqin Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiang Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Zhiyong Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 201210, China.
| |
Collapse
|