51
|
Laureano A, Kim J, Martinez E, Kwan KY. Chromodomain helicase DNA binding protein 4 in cell fate decisions. Hear Res 2023; 436:108813. [PMID: 37329862 PMCID: PMC10463912 DOI: 10.1016/j.heares.2023.108813] [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: 01/06/2022] [Revised: 05/09/2023] [Accepted: 05/24/2023] [Indexed: 06/19/2023]
Abstract
Loss of spiral ganglion neurons (SGNs) in the cochlea causes hearing loss. Understanding the mechanisms of cell fate transition accelerates efforts that employ directed differentiation and lineage conversion to repopulate lost SGNs. Proposed strategies to regenerate SGNs rely on altering cell fate by activating transcriptional regulatory networks, but repressing networks for alternative cell lineages is also essential. Epigenomic changes during cell fate transitions suggest that CHD4 represses gene expression by altering the chromatin status. Despite limited direct investigations, human genetic studies implicate CHD4 function in the inner ear. The possibility of CHD4 in suppressing alternative cell fates to promote inner ear regeneration is discussed.
Collapse
Affiliation(s)
- Alejandra Laureano
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Jihyun Kim
- Department of Cell Biology & Neuroscience, Rutgers University, Nelson Labs D250 604 Allison Rd., Piscataway, NJ 08854, USA; Stem Cell Research Center and Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Edward Martinez
- Department of Cell Biology & Neuroscience, Rutgers University, Nelson Labs D250 604 Allison Rd., Piscataway, NJ 08854, USA; Stem Cell Research Center and Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Kelvin Y Kwan
- Department of Cell Biology & Neuroscience, Rutgers University, Nelson Labs D250 604 Allison Rd., Piscataway, NJ 08854, USA; Stem Cell Research Center and Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
52
|
Tureček R, Melichar A, Králíková M, Hrušková B. The role of GABA B receptors in the subcortical pathways of the mammalian auditory system. Front Endocrinol (Lausanne) 2023; 14:1195038. [PMID: 37635966 PMCID: PMC10456889 DOI: 10.3389/fendo.2023.1195038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
GABAB receptors are G-protein coupled receptors for the inhibitory neurotransmitter GABA. Functional GABAB receptors are formed as heteromers of GABAB1 and GABAB2 subunits, which further associate with various regulatory and signaling proteins to provide receptor complexes with distinct pharmacological and physiological properties. GABAB receptors are widely distributed in nervous tissue, where they are involved in a number of processes and in turn are subject to a number of regulatory mechanisms. In this review, we summarize current knowledge of the cellular distribution and function of the receptors in the inner ear and auditory pathway of the mammalian brainstem and midbrain. The findings suggest that in these regions, GABAB receptors are involved in processes essential for proper auditory function, such as cochlear amplifier modulation, regulation of spontaneous activity, binaural and temporal information processing, and predictive coding. Since impaired GABAergic inhibition has been found to be associated with various forms of hearing loss, GABAB dysfunction could also play a role in some pathologies of the auditory system.
Collapse
Affiliation(s)
- Rostislav Tureček
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czechia
| | - Adolf Melichar
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czechia
- Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Michaela Králíková
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czechia
| | - Bohdana Hrušková
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czechia
| |
Collapse
|
53
|
Siebald C, Vincent PFY, Bottom RT, Sun S, Reijntjes DOJ, Manca M, Glowatzki E, Müller U. Molecular signatures define subtypes of auditory afferents with distinct peripheral projection patterns and physiological properties. Proc Natl Acad Sci U S A 2023; 120:e2217033120. [PMID: 37487063 PMCID: PMC10400978 DOI: 10.1073/pnas.2217033120] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 06/06/2023] [Indexed: 07/26/2023] Open
Abstract
Type I spiral ganglion neurons (SGNs) are the auditory afferents that transmit sound information from cochlear inner hair cells (IHCs) to the brainstem. These afferents consist of physiological subtypes that differ in their spontaneous firing rate (SR), activation threshold, and dynamic range and have been described as low, medium, and high SR fibers. Lately, single-cell RNA sequencing experiments have revealed three molecularly defined type I SGN subtypes. The extent to which physiological type I SGN subtypes correspond to molecularly defined subtypes is unclear. To address this question, we have generated mouse lines expressing CreERT2 in SGN subtypes that allow for a physiological assessment of molecular subtypes. We show that Lypd1-CreERT2 expressing SGNs represent a well-defined group of neurons that preferentially innervate the IHC modiolar side and exhibit a narrow range of low SRs. In contrast, Calb2-CreERT2 expressing SGNs preferentially innervate the IHC pillar side and exhibit a wider range of SRs, thus suggesting that a strict stratification of all SGNs into three molecular subclasses is not obvious, at least not with the CreERT2 tools used here. Genetically marked neuronal subtypes refine their innervation specificity onto IHCs postnatally during the time when activity is required to refine their molecular phenotype. Type I SGNs thus consist of genetically defined subtypes with distinct physiological properties and innervation patterns. The molecular subtype-specific lines characterized here will provide important tools for investigating the role of the physiologically distinct type I SGNs in encoding sound signals.
Collapse
Affiliation(s)
- Caroline Siebald
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Philippe F. Y. Vincent
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Riley T. Bottom
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Shuohao Sun
- National Institute of Biological Science, Beijing102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing100084, China
| | - Daniel O. J. Reijntjes
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Marco Manca
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Elisabeth Glowatzki
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Ulrich Müller
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
| |
Collapse
|
54
|
Sun Y, Liu Z. Recent advances in molecular studies on cochlear development and regeneration. Curr Opin Neurobiol 2023; 81:102745. [PMID: 37356371 DOI: 10.1016/j.conb.2023.102745] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/06/2023] [Accepted: 05/25/2023] [Indexed: 06/27/2023]
Abstract
The auditory organ cochlea harbors two types of sound receptors, inner hair cells (IHCs) and outer hair cells (OHCs), which are innervated by spiral (auditory) ganglion neurons (SGNs). Recent transcriptomic, epigenetic, and genetic studies have started to reveal various aspects of cochlear development, including how prosensory progenitors are specified and diversified into IHCs or OHCs, as well as the heterogeneity among SGNs and how SGN subtypes are formed. Here, we primarily review advances in this line of research over the past five years and discuss a few key studies (from the past two years) to elucidate (1) how prosensory progenitors are specified; (2) the cis-regulatory control of Atoh1 expression and the synergistic interaction between Atoh1 and Pou4f3; and (3) the essential roles of Insm1 and Ikzf2 in OHC development and Tbx2 in IHC development. Moreover, we highlight the contribution of recent molecular studies on cochlear development toward the goal of regenerating IHCs and OHCs, which holds considerable potential for application in treating human deafness. Lastly, we briefly summarize the most recent progress on uncovering when and how SGN diversity is generated.
Collapse
Affiliation(s)
- Yuwei Sun
- 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.
| |
Collapse
|
55
|
Shi L, Hong G, Chen C, Li X, Zhang H, Chai R, Sun D. Growth of spiral ganglion neurons induced by graphene oxide/oxidized bacterial cellulose composite hydrogel. Carbohydr Polym 2023; 311:120749. [PMID: 37028876 DOI: 10.1016/j.carbpol.2023.120749] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/20/2023] [Accepted: 02/23/2023] [Indexed: 03/06/2023]
Abstract
The damage or degeneration of spiral ganglion neurons (SGNs) can impair the auditory signals transduction from hair cells to the central auditory system, and cause significant hearing loss. Herein, a new form of bioactive hydrogel incorporating topological graphene oxide (GO) and TEMPO-oxidized bacterial cellulose (GO/TOBC hydrogel) was developed to provide a favorable microenvironment for SGN neurite outgrowth. As the network structure of lamellar interspersed fiber cross-linked by GO/TOBC hydrogels well simulated the structure and morphology of ECM, with the controllable hydrophilic property and appropriate Young's modulus well met those requirements of SGNs microenvironment, the GO/TOBC hybrid matrix exhibited great potential to promote the growth of SGNs. The quantitative real-time PCR result confirmed that the GO/TOBC hydrogel can significantly accelerate the development of growth cones and filopodia, by increasing the mRNA expression levels of diap3, fscn2, and integrin β1. These results suggest that GO/TOBC hydrogel scaffolds have the potential to be used to construct biomimetic nerve grafts for repairing or replacing nerve defects.
Collapse
Affiliation(s)
- Lin Shi
- Institute of Chemicobiology and Functional Materials, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing 210094, Jiangsu Province, China
| | - Guodong Hong
- 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
| | - Chuntao Chen
- Institute of Chemicobiology and Functional Materials, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing 210094, Jiangsu Province, China.
| | - Xueqian Li
- Institute of Chemicobiology and Functional Materials, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing 210094, Jiangsu Province, China
| | - Heng Zhang
- Institute of Chemicobiology and Functional Materials, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing 210094, Jiangsu Province, 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; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.
| | - Dongping Sun
- Institute of Chemicobiology and Functional Materials, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing 210094, Jiangsu Province, China.
| |
Collapse
|
56
|
Matern MS, Durruthy-Durruthy R, Birol O, Darmanis S, Scheibinger M, Groves AK, Heller S. Transcriptional dynamics of delaminating neuroblasts in the mouse otic vesicle. Cell Rep 2023; 42:112545. [PMID: 37227818 PMCID: PMC10592509 DOI: 10.1016/j.celrep.2023.112545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 02/23/2023] [Accepted: 05/04/2023] [Indexed: 05/27/2023] Open
Abstract
An abundance of research has recently highlighted the susceptibility of cochleovestibular ganglion (CVG) neurons to noise damage and aging in the adult cochlea, resulting in hearing deficits. Furthering our understanding of the transcriptional cascades that contribute to CVG development may provide insight into how these cells can be regenerated to treat inner ear dysfunction. Here we perform a high-depth single-cell RNA sequencing analysis of the E10.5 otic vesicle and its surrounding tissues, including CVG precursor neuroblasts and emerging CVG neurons. Clustering and trajectory analysis of otic-lineage cells reveals otic markers and the changes in gene expression that occur from neuroblast delamination toward the development of the CVG. This dataset provides a valuable resource for further identifying the mechanisms associated with CVG development from neurosensory competent cells within the otic vesicle.
Collapse
Affiliation(s)
- Maggie S Matern
- Department of Otolaryngology Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Robert Durruthy-Durruthy
- Department of Otolaryngology Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Onur Birol
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Spyros Darmanis
- Departments of Bioengineering and Applied Physics and Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Mirko Scheibinger
- Department of Otolaryngology Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Andrew K Groves
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Stefan Heller
- Department of Otolaryngology Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
57
|
Spirou GA, Kersting M, Carr S, Razzaq B, Yamamoto Alves Pinto C, Dawson M, Ellisman MH, Manis PB. High-resolution volumetric imaging constrains compartmental models to explore synaptic integration and temporal processing by cochlear nucleus globular bushy cells. eLife 2023; 12:e83393. [PMID: 37288824 PMCID: PMC10435236 DOI: 10.7554/elife.83393] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 06/07/2023] [Indexed: 06/09/2023] Open
Abstract
Globular bushy cells (GBCs) of the cochlear nucleus play central roles in the temporal processing of sound. Despite investigation over many decades, fundamental questions remain about their dendrite structure, afferent innervation, and integration of synaptic inputs. Here, we use volume electron microscopy (EM) of the mouse cochlear nucleus to construct synaptic maps that precisely specify convergence ratios and synaptic weights for auditory nerve innervation and accurate surface areas of all postsynaptic compartments. Detailed biophysically based compartmental models can help develop hypotheses regarding how GBCs integrate inputs to yield their recorded responses to sound. We established a pipeline to export a precise reconstruction of auditory nerve axons and their endbulb terminals together with high-resolution dendrite, soma, and axon reconstructions into biophysically detailed compartmental models that could be activated by a standard cochlear transduction model. With these constraints, the models predict auditory nerve input profiles whereby all endbulbs onto a GBC are subthreshold (coincidence detection mode), or one or two inputs are suprathreshold (mixed mode). The models also predict the relative importance of dendrite geometry, soma size, and axon initial segment length in setting action potential threshold and generating heterogeneity in sound-evoked responses, and thereby propose mechanisms by which GBCs may homeostatically adjust their excitability. Volume EM also reveals new dendritic structures and dendrites that lack innervation. This framework defines a pathway from subcellular morphology to synaptic connectivity, and facilitates investigation into the roles of specific cellular features in sound encoding. We also clarify the need for new experimental measurements to provide missing cellular parameters, and predict responses to sound for further in vivo studies, thereby serving as a template for investigation of other neuron classes.
Collapse
Affiliation(s)
- George A Spirou
- Department of Medical Engineering, University of South FloridaTampaUnited States
| | - Matthew Kersting
- Department of Medical Engineering, University of South FloridaTampaUnited States
| | - Sean Carr
- Department of Medical Engineering, University of South FloridaTampaUnited States
| | - Bayan Razzaq
- Department of Otolaryngology, Head and Neck Surgery, West Virginia UniversityMorgantownUnited States
| | | | - Mariah Dawson
- Department of Otolaryngology, Head and Neck Surgery, West Virginia UniversityMorgantownUnited States
| | - Mark H Ellisman
- Department of Neurosciences, University of California, San DiegoSan DiegoUnited States
- National Center for Microscopy and Imaging Research,University of California, San DiegoSan DiegoUnited States
| | - Paul B Manis
- Department of Otolaryngology/Head and Neck Surgery, University of North Carolina at Chapel HillChapel HillUnited States
- Department of Cell Biology and Physiology, University of North CarolinaChapel HillUnited States
| |
Collapse
|
58
|
Liu H, Liu H, Wang L, Song L, Jiang G, Lu Q, Yang T, Peng H, Cai R, Zhao X, Zhao T, Wu H. Cochlear transcript diversity and its role in auditory functions implied by an otoferlin short isoform. Nat Commun 2023; 14:3085. [PMID: 37248244 DOI: 10.1038/s41467-023-38621-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 05/10/2023] [Indexed: 05/31/2023] Open
Abstract
Isoforms of a gene may contribute to diverse biological functions. In the cochlea, the repertoire of alternative isoforms remains unexplored. We integrated single-cell short-read and long-read RNA sequencing techniques and identified 236,012 transcripts, 126,612 of which were unannotated in the GENCODE database. Then we analyzed and verified the unannotated transcripts using RNA-seq, RT-PCR, Sanger sequencing, and MS-based proteomics approaches. To illustrate the importance of identifying spliced isoforms, we investigated otoferlin, a key protein involved in synaptic transmission in inner hair cells (IHCs). Upon deletion of the canonical otoferlin isoform, the identified short isoform is able to support normal hearing thresholds but with reduced sustained exocytosis of IHCs, and further revealed otoferlin functions in endocytic membrane retrieval that was not well-addressed previously. Furthermore, we found that otoferlin isoforms are associated with IHC functions and auditory phenotypes. This work expands our mechanistic understanding of auditory functions at the level of isoform resolution.
Collapse
Affiliation(s)
- Huihui Liu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200011, China
| | - Hongchao Liu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200011, China
| | - Longhao Wang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200011, China
| | - Lei Song
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200011, China
| | - Guixian Jiang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200011, China
| | - Qing Lu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200011, China
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Tao Yang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200011, China
| | - Hu Peng
- Department of Otolaryngology-Head and Neck Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Ruijie Cai
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200011, China
| | - Xingle Zhao
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200011, China
| | - Ting Zhao
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200011, China
| | - Hao Wu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200011, China.
| |
Collapse
|
59
|
Yu X, Wang Y. Peripheral Fragile X messenger ribonucleoprotein is required for the timely closure of a critical period for neuronal susceptibility in the ventral cochlear nucleus. Front Cell Neurosci 2023; 17:1186630. [PMID: 37305436 PMCID: PMC10248243 DOI: 10.3389/fncel.2023.1186630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/08/2023] [Indexed: 06/13/2023] Open
Abstract
Alterations in neuronal plasticity and critical periods are common across neurodevelopmental diseases, including Fragile X syndrome (FXS), the leading single-gene cause of autism. Characterized with sensory dysfunction, FXS is the result of gene silencing of Fragile X messenger ribonucleoprotein 1 (FMR1) and loss of its product, Fragile X messenger ribonucleoprotein (FMRP). The mechanisms underlying altered critical period and sensory dysfunction in FXS are obscure. Here, we performed genetic and surgical deprivation of peripheral auditory inputs in wildtype and Fmr1 knockout (KO) mice across ages and investigated the effects of global FMRP loss on deafferentation-induced neuronal changes in the ventral cochlear nucleus (VCN) and auditory brainstem responses. The degree of neuronal cell loss during the critical period was unchanged in Fmr1 KO mice. However, the closure of the critical period was delayed. Importantly, this delay was temporally coincidental with reduced hearing sensitivity, implying an association with sensory inputs. Functional analyses further identified early-onset and long-lasting alterations in signal transmission from the spiral ganglion to the VCN, suggesting a peripheral site of FMRP action. Finally, we generated conditional Fmr1 KO (cKO) mice with selective deletion of FMRP in spiral ganglion but not VCN neurons. cKO mice recapitulated the delay in the VCN critical period closure in Fmr1 KO mice, confirming an involvement of cochlear FMRP in shaping the temporal features of neuronal critical periods in the brain. Together, these results identify a novel peripheral mechanism of neurodevelopmental pathogenesis.
Collapse
Affiliation(s)
| | - Yuan Wang
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| |
Collapse
|
60
|
Li Q, Cui C, Liao R, Yin X, Wang D, Cheng Y, Huang B, Wang L, Yan M, Zhou J, Zhao J, Tang W, Wang Y, Wang X, Lv J, Li J, Li H, Shu Y. The pathogenesis of common Gjb2 mutations associated with human hereditary deafness in mice. Cell Mol Life Sci 2023; 80:148. [PMID: 37178259 PMCID: PMC10182940 DOI: 10.1007/s00018-023-04794-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/31/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023]
Abstract
Mutations in GJB2 (Gap junction protein beta 2) are the most common genetic cause of non-syndromic hereditary deafness in humans, especially the 35delG and 235delC mutations. Owing to the homozygous lethality of Gjb2 mutations in mice, there are currently no perfect mouse models carrying Gjb2 mutations derived from patients for mimicking human hereditary deafness and for unveiling the pathogenesis of the disease. Here, we successfully constructed heterozygous Gjb2+/35delG and Gjb2+/235delC mutant mice through advanced androgenic haploid embryonic stem cell (AG-haESC)-mediated semi-cloning technology, and these mice showed normal hearing at postnatal day (P) 28. A homozygous mutant mouse model, Gjb235delG/35delG, was then generated using enhanced tetraploid embryo complementation, demonstrating that GJB2 plays an indispensable role in mouse placenta development. These mice exhibited profound hearing loss similar to human patients at P14, i.e., soon after the onset of hearing. Mechanistic analyses showed that Gjb2 35delG disrupts the function and formation of intercellular gap junction channels of the cochlea rather than affecting the survival and function of hair cells. Collectively, our study provides ideal mouse models for understanding the pathogenic mechanism of DFNB1A-related hereditary deafness and opens up a new avenue for investigating the treatment of this disease.
Collapse
Affiliation(s)
- Qing Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China.
| | - Chong Cui
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Rongyu Liao
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Xidi Yin
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Daqi Wang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Yanbo Cheng
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Bowei Huang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Liqin Wang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Meng Yan
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Jinan Zhou
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Jingjing Zhao
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Wei Tang
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Yingyi Wang
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | | | - Jun Lv
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Jinsong Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Huawei Li
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China.
| | - Yilai Shu
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China.
| |
Collapse
|
61
|
Li S, He S, Lu Y, Jia S, Liu Z. Epistatic genetic interactions between Insm1 and Ikzf2 during cochlear outer hair cell development. Cell Rep 2023; 42:112504. [PMID: 37171961 DOI: 10.1016/j.celrep.2023.112504] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/17/2023] [Accepted: 04/27/2023] [Indexed: 05/14/2023] Open
Abstract
The cochlea harbors two types of sound receptors, outer hair cells (OHCs) and inner hair cells (IHCs). OHCs transdifferentiate into IHCs in Insm1 mutants, and OHCs in Ikzf2-deficient mice are dysfunctional and maintain partial IHC gene expression. Insm1 potentially acts as a positive but indirect regulator of Ikzf2, considering that Insm1 is expressed earlier than Ikzf2 and primarily functions as a transcriptional repressor. However, direct evidence of this possibility is lacking. Here, we report the following results: first, Insm1 overexpression in IHCs leads to ectopic Ikzf2 expression. Second, Ikzf2 expression is repressed in Insm1-deficient OHCs, and forced expression of Ikzf2 mitigates the OHC abnormality in Insm1 mutants. Last, dual ablation of Insm1 and Ikzf2 generates a similar OHC phenotype as does Insm1 ablation alone. Collectively, our findings reveal the transcriptional cascade from Insm1 to Ikzf2, which should facilitate future investigation of the molecular mechanisms underlying OHC development and regeneration.
Collapse
Affiliation(s)
- 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
| | - Ying Lu
- 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
| | - Shiqi Jia
- The First Affiliated Hospital, Jinan University, Guangzhou 510630, 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.
| |
Collapse
|
62
|
Capra D, DosSantos MF, Sanz CK, Acosta Filha LG, Nunes P, Heringer M, Ximenes-da-Silva A, Pessoa L, de Mattos Coelho-Aguiar J, da Fonseca ACC, Mendes CB, da Rocha LS, Devalle S, Niemeyer Soares Filho P, Moura-Neto V. Pathophysiology and mechanisms of hearing impairment related to neonatal infection diseases. Front Microbiol 2023; 14:1162554. [PMID: 37125179 PMCID: PMC10140533 DOI: 10.3389/fmicb.2023.1162554] [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: 02/09/2023] [Accepted: 03/24/2023] [Indexed: 05/02/2023] Open
Abstract
The inner ear, the organ of equilibrium and hearing, has an extraordinarily complex and intricate arrangement. It contains highly specialized structures meticulously tailored to permit auditory processing. However, hearing also relies on both peripheral and central pathways responsible for the neuronal transmission of auditory information from the cochlea to the corresponding cortical regions. Understanding the anatomy and physiology of all components forming the auditory system is key to better comprehending the pathophysiology of each disease that causes hearing impairment. In this narrative review, the authors focus on the pathophysiology as well as on cellular and molecular mechanisms that lead to hearing loss in different neonatal infectious diseases. To accomplish this objective, the morphology and function of the main structures responsible for auditory processing and the immune response leading to hearing loss were explored. Altogether, this information permits the proper understanding of each infectious disease discussed.
Collapse
Affiliation(s)
- Daniela Capra
- Laboratório de Morfogênese Celular (LMC), Instituto de Ciências Biomédicas (ICB), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Neurociência Translacional, Instituto Nacional de Neurociência Translacional (INNT-UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcos F. DosSantos
- Laboratório de Morfogênese Celular (LMC), Instituto de Ciências Biomédicas (ICB), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Neurociência Translacional, Instituto Nacional de Neurociência Translacional (INNT-UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Odontologia (PPGO), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Laboratório de Propriedades Mecânicas e Biologia Celular (PropBio), Departamento de Prótese e Materiais Dentários, Faculdade de Odontologia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Carolina K. Sanz
- Laboratório de Propriedades Mecânicas e Biologia Celular (PropBio), Departamento de Prótese e Materiais Dentários, Faculdade de Odontologia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Lionete Gall Acosta Filha
- Laboratório de Morfogênese Celular (LMC), Instituto de Ciências Biomédicas (ICB), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Neurociência Translacional, Instituto Nacional de Neurociência Translacional (INNT-UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Propriedades Mecânicas e Biologia Celular (PropBio), Departamento de Prótese e Materiais Dentários, Faculdade de Odontologia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Priscila Nunes
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | - Manoela Heringer
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | | | - Luciana Pessoa
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | - Juliana de Mattos Coelho-Aguiar
- Laboratório de Morfogênese Celular (LMC), Instituto de Ciências Biomédicas (ICB), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Neurociência Translacional, Instituto Nacional de Neurociência Translacional (INNT-UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Anatomia Patológica, Hospital Universitário Clementino Fraga Filho (HUCFF), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Anna Carolina Carvalho da Fonseca
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | | | | | - Sylvie Devalle
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | - Paulo Niemeyer Soares Filho
- Programa de Pós-Graduação em Neurociência Translacional, Instituto Nacional de Neurociência Translacional (INNT-UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vivaldo Moura-Neto
- Laboratório de Morfogênese Celular (LMC), Instituto de Ciências Biomédicas (ICB), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Neurociência Translacional, Instituto Nacional de Neurociência Translacional (INNT-UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Anatomia Patológica, Hospital Universitário Clementino Fraga Filho (HUCFF), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
63
|
Sun G, Zheng Y, Fu X, Zhang W, Ren J, Ma S, Sun S, He X, Wang Q, Ji Z, Cheng F, Yan K, Liu Z, Belmonte JCI, Qu J, Wang S, Chai R, Liu GH. Single-cell transcriptomic atlas of mouse cochlear aging. Protein Cell 2023; 14:180-201. [PMID: 36933008 PMCID: PMC10098046 DOI: 10.1093/procel/pwac058] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 09/16/2022] [Indexed: 11/13/2022] Open
Abstract
Progressive functional deterioration in the cochlea is associated with age-related hearing loss (ARHL). However, the cellular and molecular basis underlying cochlear aging remains largely unknown. Here, we established a dynamic single-cell transcriptomic landscape of mouse cochlear aging, in which we characterized aging-associated transcriptomic changes in 27 different cochlear cell types across five different time points. Overall, our analysis pinpoints loss of proteostasis and elevated apoptosis as the hallmark features of cochlear aging, highlights unexpected age-related transcriptional fluctuations in intermediate cells localized in the stria vascularis (SV) and demonstrates that upregulation of endoplasmic reticulum (ER) chaperon protein HSP90AA1 mitigates ER stress-induced damages associated with aging. Our work suggests that targeting unfolded protein response pathways may help alleviate aging-related SV atrophy and hence delay the progression of ARHL.
Collapse
Affiliation(s)
- Guoqiang Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yandong Zheng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaolong Fu
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, School of Life Sciences and Technology, Southeast University, Nanjing 211189, China
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China
- China National Center for Bioinformation, Beijing 100101, China
| | - Jie Ren
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China
- China National Center for Bioinformation, Beijing 100101, China
| | - Shuai Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- The Fifth People’s Hospital of Chongqing, Chongqing 400062, China
| | - Shuhui Sun
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Xiaojuan He
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Qiaoran Wang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- China National Center for Bioinformation, Beijing 100101, China
| | - Zhejun Ji
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Fang Cheng
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kaowen Yan
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Ziyi Liu
- Shandong Provincial Hospital and School of Laboratory Animal Science, Shandong First Medical University, Jinan 250000, China
| | | | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- The Fifth People’s Hospital of Chongqing, Chongqing 400062, China
| | - Renjie Chai
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, School of Life Sciences and Technology, Southeast University, Nanjing 211189, China
- Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| |
Collapse
|
64
|
Kaczmarek LK. Modulation of potassium conductances optimizes fidelity of auditory information. Proc Natl Acad Sci U S A 2023; 120:e2216440120. [PMID: 36930599 PMCID: PMC10041146 DOI: 10.1073/pnas.2216440120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 02/08/2023] [Indexed: 03/18/2023] Open
Abstract
Potassium channels in auditory neurons are rapidly modified by changes in the auditory environment. In response to elevated auditory stimulation, short-term mechanisms such as protein phosphorylation and longer-term mechanisms such as accelerated channel synthesis increase the amplitude of currents that promote high-frequency firing. It has been suggested that this allows neurons to fire at high rates in response to high sound levels. We have carried out simple simulations of the response to postsynaptic neurons to patterns of neurotransmitter release triggered by auditory stimuli. These demonstrate that the amplitudes of potassium currents required for optimal encoding of a low-amplitude auditory signal differ from those for louder sounds. Specifically, the cross-correlation of the output of a neuron with an auditory stimulus is improved by increasing potassium currents as sound amplitude increases. Temporal fidelity for low-frequency stimuli is improved by increasing potassium currents that activate at negative potentials, while that for high-frequency stimuli requires increases in currents that activate at positive membrane potentials. These effects are independent of the firing rate. Moreover, levels of potassium currents that maximize the fidelity of the output of an ensemble of neurons differ from those that maximize fidelity for a single neuron. This suggests that the modulatory mechanisms must coordinate channel activity in groups of neurons or an entire nucleus. The simulations provide an explanation for the modulation of the intrinsic excitability of auditory brainstem neurons by changes in environmental sound levels, and the results may extend to information processing in other neural systems.
Collapse
Affiliation(s)
- Leonard K. Kaczmarek
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT06520
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT06520
| |
Collapse
|
65
|
Wang M, Zhang C, Lin S, Xie R. Dendritic Degeneration and Altered Synaptic Innervation of a Central Auditory Neuron During Age-related Hearing Loss. Neuroscience 2023; 514:25-37. [PMID: 36738912 PMCID: PMC9992229 DOI: 10.1016/j.neuroscience.2023.01.037] [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: 10/21/2022] [Revised: 01/08/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Cellular morphology and synaptic configuration are key determinants of neuronal function and are often modified under pathological conditions. In the first nucleus of the central auditory system, the cochlear nucleus (CN), principal bushy neurons specialize in processing temporal information of sound critical for hearing. These neurons alter their physiological properties during aging that contribute to age-related hearing loss (ARHL). The structural basis of such changes remains unclear, especially age-related modifications in their dendritic morphology and the innervating auditory nerve (AN) synapses. Using young (2-5 months) and aged (28-33 months) CBA/CaJ mice of either sex, we filled individual bushy neurons with fluorescent dye in acute brain slices to characterize their dendritic morphology, followed by immunostaining against vesicular glutamate transporter 1 (VGluT1) and calretinin (CR) to identify innervating AN synapses. We found that dendritic morphology of aged bushy neurons had significantly reduced complexity, suggesting age-dependent dendritic degeneration, especially in neurons with predominantly non-CR-expressing synapses on the soma. These dendrites were innervated by AN bouton synapses, which were predominantly non-CR-expressing in young mice but had increased proportion of CR-expressing synapses in old mice. While somatic AN synapses degenerated substantially with age, as quantified by VGluT1-labeled puncta volume, no significant difference was observed in the total volume of dendritic synapses between young and old mice. Consequently, synaptic density on dendrites was significantly higher in old mice. The findings suggest that dendritic degeneration and altered synaptic innervation in bushy neurons during aging may underlie their changed physiological activity and contribute to the development of ARHL.
Collapse
Affiliation(s)
- Meijian Wang
- Department of Otolaryngology, The Ohio State University, Columbus, OH, USA
| | - Chuangeng Zhang
- Department of Otolaryngology, The Ohio State University, Columbus, OH, USA
| | - Shengyin Lin
- Department of Otolaryngology, The Ohio State University, Columbus, OH, USA
| | - Ruili Xie
- Department of Otolaryngology, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
66
|
Frank MM, Sitko AA, Suthakar K, Torres Cadenas L, Hunt M, Yuk MC, Weisz CJC, Goodrich LV. Experience-dependent flexibility in a molecularly diverse central-to-peripheral auditory feedback system. eLife 2023; 12:e83855. [PMID: 36876911 PMCID: PMC10147377 DOI: 10.7554/elife.83855] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 03/03/2023] [Indexed: 03/07/2023] Open
Abstract
Brainstem olivocochlear neurons (OCNs) modulate the earliest stages of auditory processing through feedback projections to the cochlea and have been shown to influence hearing and protect the ear from sound-induced damage. Here, we used single-nucleus sequencing, anatomical reconstructions, and electrophysiology to characterize murine OCNs during postnatal development, in mature animals, and after sound exposure. We identified markers for known medial (MOC) and lateral (LOC) OCN subtypes, and show that they express distinct cohorts of physiologically relevant genes that change over development. In addition, we discovered a neuropeptide-enriched LOC subtype that produces Neuropeptide Y along with other neurotransmitters. Throughout the cochlea, both LOC subtypes extend arborizations over wide frequency domains. Moreover, LOC neuropeptide expression is strongly upregulated days after acoustic trauma, potentially providing a sustained protective signal to the cochlea. OCNs are therefore poised to have diffuse, dynamic effects on early auditory processing over timescales ranging from milliseconds to days.
Collapse
Affiliation(s)
- Michelle M Frank
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Austen A Sitko
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Kirupa Suthakar
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication DisordersBethesdaUnited States
| | - Lester Torres Cadenas
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication DisordersBethesdaUnited States
| | - Mackenzie Hunt
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Mary Caroline Yuk
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Catherine JC Weisz
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication DisordersBethesdaUnited States
| | - Lisa V Goodrich
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
67
|
Shrestha BR, Wu L, Goodrich LV. Runx1 controls auditory sensory neuron diversity in mice. Dev Cell 2023; 58:306-319.e5. [PMID: 36800995 PMCID: PMC10202259 DOI: 10.1016/j.devcel.2023.01.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/28/2022] [Accepted: 01/26/2023] [Indexed: 02/18/2023]
Abstract
Sound stimulus is encoded in mice by three molecularly and physiologically diverse subtypes of sensory neurons, called Ia, Ib, and Ic spiral ganglion neurons (SGNs). Here, we show that the transcription factor Runx1 controls SGN subtype composition in the murine cochlea. Runx1 is enriched in Ib/Ic precursors by late embryogenesis. Upon the loss of Runx1 from embryonic SGNs, more SGNs take on Ia rather than Ib or Ic identities. This conversion was more complete for genes linked to neuronal function than to connectivity. Accordingly, synapses in the Ib/Ic location acquired Ia properties. Suprathreshold SGN responses to sound were enhanced in Runx1CKO mice, confirming the expansion of neurons with Ia-like functional properties. Runx1 deletion after birth also redirected Ib/Ic SGNs toward Ia identity, indicating that SGN identities are plastic postnatally. Altogether, these findings show that diverse neuronal identities essential for normal auditory stimulus coding arise hierarchically and remain malleable during postnatal development.
Collapse
Affiliation(s)
- Brikha R Shrestha
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA 02114, USA; Department of Otolaryngology, Harvard Medical School, Boston, MA 02114, USA.
| | - Lorna Wu
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Lisa V Goodrich
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
68
|
Chakraborty B, Mukhopadhyay S. Heterogeneous recurrent spiking neural network for spatio-temporal classification. Front Neurosci 2023; 17:994517. [PMID: 36793542 PMCID: PMC9922697 DOI: 10.3389/fnins.2023.994517] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 01/04/2023] [Indexed: 02/01/2023] Open
Abstract
Spiking Neural Networks are often touted as brain-inspired learning models for the third wave of Artificial Intelligence. Although recent SNNs trained with supervised backpropagation show classification accuracy comparable to deep networks, the performance of unsupervised learning-based SNNs remains much lower. This paper presents a heterogeneous recurrent spiking neural network (HRSNN) with unsupervised learning for spatio-temporal classification of video activity recognition tasks on RGB (KTH, UCF11, UCF101) and event-based datasets (DVS128 Gesture). We observed an accuracy of 94.32% for the KTH dataset, 79.58% and 77.53% for the UCF11 and UCF101 datasets, respectively, and an accuracy of 96.54% on the event-based DVS Gesture dataset using the novel unsupervised HRSNN model. The key novelty of the HRSNN is that the recurrent layer in HRSNN consists of heterogeneous neurons with varying firing/relaxation dynamics, and they are trained via heterogeneous spike-time-dependent-plasticity (STDP) with varying learning dynamics for each synapse. We show that this novel combination of heterogeneity in architecture and learning method outperforms current homogeneous spiking neural networks. We further show that HRSNN can achieve similar performance to state-of-the-art backpropagation trained supervised SNN, but with less computation (fewer neurons and sparse connection) and less training data.
Collapse
|
69
|
Mittring A, Moser T, Huet AT. Graded optogenetic activation of the auditory pathway for hearing restoration. Brain Stimul 2023; 16:466-483. [PMID: 36702442 PMCID: PMC10159867 DOI: 10.1016/j.brs.2023.01.1671] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
Optogenetic control of neural activity enables innovative approaches to improve functional restoration of diseased sensory and motor systems. For clinical translation to succeed, optogenetic stimulation needs to closely match the coding properties of the targeted neuronal population and employ optimally operating emitters. This requires the customization of channelrhodopsins, emitters and coding strategies. Here, we provide a framework to parametrize optogenetic neural control and apply it to the auditory pathway that requires high temporal fidelity of stimulation. We used a viral gene transfer of ultrafast targeting-optimized Chronos into spiral ganglion neurons (SGNs) of the cochlea of mice. We characterized the light-evoked response by in vivo recordings from individual SGNs and neurons of the anteroventral cochlear nucleus (AVCN) that detect coincident SGN inputs. Our recordings from single SGNs demonstrated that their spike probability can be graded by adjusting the duration of light pulses at constant intensity, which optimally serves efficient laser diode operation. We identified an effective pulse width of 1.6 ms to maximize encoding in SGNs at the maximal light intensity employed here (∼35 mW). Alternatively, SGNs were activated at lower energy thresholds using short light pulses (<1 ms). An upper boundary of optical stimulation rates was identified at 316 Hz, inducing a robust spike rate adaptation that required a few tens of milliseconds to recover. We developed a semi-stochastic stimulation paradigm to rapidly (within minutes) estimate the input/output function from light to SGN firing and approximate the time constant of neuronal integration in the AVCN. By that, our data pave the way to design the sound coding strategies of future optical cochlear implants.
Collapse
Affiliation(s)
- Artur Mittring
- Auditory Circuit Lab, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany; Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany; Auditory Neuroscience and Optogenetics Laboratory, German Primate Center, Göttingen, Germany; Auditory Neuroscience and Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
| | - Antoine Tarquin Huet
- Auditory Circuit Lab, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany; Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany; Auditory Neuroscience and Optogenetics Laboratory, German Primate Center, Göttingen, Germany.
| |
Collapse
|
70
|
Rutherford MA, Bhattacharyya A, Xiao M, Cai HM, Pal I, Rubio ME. GluA3 subunits are required for appropriate assembly of AMPAR GluA2 and GluA4 subunits on cochlear afferent synapses and for presynaptic ribbon modiolar-pillar morphology. eLife 2023; 12:e80950. [PMID: 36648432 PMCID: PMC9891727 DOI: 10.7554/elife.80950] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 01/16/2023] [Indexed: 01/18/2023] Open
Abstract
Cochlear sound encoding depends on α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPARs), but reliance on specific pore-forming subunits is unknown. With 5-week-old male C57BL/6J Gria3-knockout mice (i.e., subunit GluA3KO) we determined cochlear function, synapse ultrastructure, and AMPAR molecular anatomy at ribbon synapses between inner hair cells (IHCs) and spiral ganglion neurons. GluA3KO and wild-type (GluA3WT) mice reared in ambient sound pressure level (SPL) of 55-75 dB had similar auditory brainstem response (ABR) thresholds, wave-1 amplitudes, and latencies. Postsynaptic densities (PSDs), presynaptic ribbons, and synaptic vesicle sizes were all larger on the modiolar side of the IHCs from GluA3WT, but not GluA3KO, demonstrating GluA3 is required for modiolar-pillar synapse differentiation. Presynaptic ribbons juxtaposed with postsynaptic GluA2/4 subunits were similar in quantity, however, lone ribbons were more frequent in GluA3KO and GluA2-lacking synapses were observed only in GluA3KO. GluA2 and GluA4 immunofluorescence volumes were smaller on the pillar side than the modiolar side in GluA3KO, despite increased pillar-side PSD size. Overall, the fluorescent puncta volumes of GluA2 and GluA4 were smaller in GluA3KO than GluA3WT. However, GluA3KO contained less GluA2 and greater GluA4 immunofluorescence intensity relative to GluA3WT (threefold greater mean GluA4:GluA2 ratio). Thus, GluA3 is essential in development, as germline disruption of Gria3 caused anatomical synapse pathology before cochlear output became symptomatic by ABR. We propose the hearing loss in older male GluA3KO mice results from progressive synaptopathy evident in 5-week-old mice as decreased abundance of GluA2 subunits and an increase in GluA2-lacking, GluA4-monomeric Ca2+-permeable AMPARs.
Collapse
Affiliation(s)
- Mark A Rutherford
- Department of Otolaryngology, Washington University School of MedicineSt LouisUnited States
| | - Atri Bhattacharyya
- Department of Otolaryngology, Washington University School of MedicineSt LouisUnited States
| | - Maolei Xiao
- Department of Otolaryngology, Washington University School of MedicineSt LouisUnited States
| | - Hou-Ming Cai
- Department of Neurobiology, University of Pittsburgh School of MedicinePittsburghUnited States
| | - Indra Pal
- Department of Neurobiology, University of Pittsburgh School of MedicinePittsburghUnited States
| | - Maria Eulalia Rubio
- Department of Neurobiology, University of Pittsburgh School of MedicinePittsburghUnited States
- Department of Otolaryngology, University of Pittsburgh School of MedicinePittsburghUnited States
| |
Collapse
|
71
|
Kim KS, Koo HY, Bok J. Alternative splicing in shaping the molecular landscape of the cochlea. Front Cell Dev Biol 2023; 11:1143428. [PMID: 36936679 PMCID: PMC10018040 DOI: 10.3389/fcell.2023.1143428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/16/2023] [Indexed: 03/06/2023] Open
Abstract
The cochlea is a complex organ comprising diverse cell types with highly specialized morphology and function. Until now, the molecular underpinnings of its specializations have mostly been studied from a transcriptional perspective, but accumulating evidence points to post-transcriptional regulation as a major source of molecular diversity. Alternative splicing is one of the most prevalent and well-characterized post-transcriptional regulatory mechanisms. Many molecules important for hearing, such as cadherin 23 or harmonin, undergo alternative splicing to produce functionally distinct isoforms. Some isoforms are expressed specifically in the cochlea, while some show differential expression across the various cochlear cell types and anatomical regions. Clinical phenotypes that arise from mutations affecting specific splice variants testify to the functional relevance of these isoforms. All these clues point to an essential role for alternative splicing in shaping the unique molecular landscape of the cochlea. Although the regulatory mechanisms controlling alternative splicing in the cochlea are poorly characterized, there are animal models with defective splicing regulators that demonstrate the importance of RNA-binding proteins in maintaining cochlear function and cell survival. Recent technological breakthroughs offer exciting prospects for overcoming some of the long-standing hurdles that have complicated the analysis of alternative splicing in the cochlea. Efforts toward this end will help clarify how the remarkable diversity of the cochlear transcriptome is both established and maintained.
Collapse
Affiliation(s)
- Kwan Soo Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hei Yeun Koo
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jinwoong Bok
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
- *Correspondence: Jinwoong Bok,
| |
Collapse
|
72
|
Faure L, Techameena P, Hadjab S. Emergence of neuron types. Curr Opin Cell Biol 2022; 79:102133. [PMID: 36347131 DOI: 10.1016/j.ceb.2022.102133] [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: 07/14/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 01/31/2023]
Abstract
Neuron types are the building blocks of the nervous system, and therefore, of functional circuits. Understanding the origin of neuronal diversity has always been an essential question in neuroscience and developmental biology. While knowledge on the molecular control of their diversification has largely increased during the last decades, it is now possible to reveal the dynamic mechanisms and the actual stepwise molecular changes occurring at single-cell level with the advent of single-cell omics technologies and analysis with high temporal resolution. Here, we focus on recent advances in the field and in technical and analytical tools that enable detailed insights into the emergence of neuron types in the central and peripheral nervous systems.
Collapse
Affiliation(s)
- Louis Faure
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, 1090, Vienna, Austria
| | - Prach Techameena
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Saida Hadjab
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
73
|
Glover JC, Fritzsch B. Molecular mechanisms governing development of the hindbrain choroid plexus and auditory projection: A validation of the seminal observations of Wilhelm His. IBRO Neurosci Rep 2022; 13:306-313. [PMID: 36247525 PMCID: PMC9561746 DOI: 10.1016/j.ibneur.2022.09.011] [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: 04/29/2022] [Revised: 09/29/2022] [Accepted: 09/29/2022] [Indexed: 11/05/2022] Open
Abstract
Studies by His from 1868 to 1904 delineated the critical role of the dorsal roof plate in the development of the hindbrain choroid plexus, and of the rhombic lips in the development of hindbrain auditory centers. Modern molecular studies have confirmed these observations and placed them in a mechanistic context. Expression of the transcription factor Lmx1a/b is crucial to the development of the hindbrain choroid plexus, and also regulates the expression of Atoh1, a transcription factor that is essential for the formation of the cochlear hair cells and auditory nuclei. By contrast, development of the vestibular hair cells, vestibular ganglion and vestibular nuclei does not depend on Lmx1a/b. These findings demonstrate a common dependence on a specific gene for the hindbrain choroid plexus and the primary auditory projection from hair cells to sensory neurons to hindbrain nuclei. Thus, His' conclusions regarding the origins of specific hindbrain structures are borne out by molecular genetic experiments conducted more than a hundred years later.
Collapse
Affiliation(s)
- Joel C. Glover
- Department of Molecular Medicine, University of Oslo, Oslo, Norway
- Norwegian Center for Stem Cell Research, Oslo University Hospital, Oslo, Norway
- Sars International Centre for Marine Molecular Biology, University of Bergen, Bergen, Norway
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa, IA 52242, USA
| |
Collapse
|
74
|
Qiu X, Müller U. Sensing sound: Cellular specializations and molecular force sensors. Neuron 2022; 110:3667-3687. [PMID: 36223766 PMCID: PMC9671866 DOI: 10.1016/j.neuron.2022.09.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/03/2022] [Accepted: 09/14/2022] [Indexed: 11/08/2022]
Abstract
Organisms of all phyla express mechanosensitive ion channels with a wide range of physiological functions. In recent years, several classes of mechanically gated ion channels have been identified. Some of these ion channels are intrinsically mechanosensitive. Others depend on accessory proteins to regulate their response to mechanical force. The mechanotransduction machinery of cochlear hair cells provides a particularly striking example of a complex force-sensing machine. This molecular ensemble is embedded into a specialized cellular compartment that is crucial for its function. Notably, mechanotransduction channels of cochlear hair cells are not only critical for auditory perception. They also shape their cellular environment and regulate the development of auditory circuitry. Here, we summarize recent discoveries that have shed light on the composition of the mechanotransduction machinery of cochlear hair cells and how this machinery contributes to the development and function of the auditory system.
Collapse
Affiliation(s)
- Xufeng Qiu
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ulrich Müller
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
75
|
McChesney N, Barth JL, Rumschlag JA, Tan J, Harrington AJ, Noble KV, McClaskey CM, Elvis P, Vaena SG, Romeo MJ, Harris KC, Cowan CW, Lang H. Peripheral Auditory Nerve Impairment in a Mouse Model of Syndromic Autism. J Neurosci 2022; 42:8002-8018. [PMID: 36180228 PMCID: PMC9617620 DOI: 10.1523/jneurosci.0253-22.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 07/27/2022] [Accepted: 08/13/2022] [Indexed: 11/21/2022] Open
Abstract
Dysfunction of the peripheral auditory nerve (AN) contributes to dynamic changes throughout the central auditory system, resulting in abnormal auditory processing, including hypersensitivity. Altered sound sensitivity is frequently observed in autism spectrum disorder (ASD), suggesting that AN deficits and changes in auditory information processing may contribute to ASD-associated symptoms, including social communication deficits and hyperacusis. The MEF2C transcription factor is associated with risk for several neurodevelopmental disorders, and mutations or deletions of MEF2C produce a haploinsufficiency syndrome characterized by ASD, language, and cognitive deficits. A mouse model of this syndromic ASD (Mef2c-Het) recapitulates many of the MEF2C haploinsufficiency syndrome-linked behaviors, including communication deficits. We show here that Mef2c-Het mice of both sexes exhibit functional impairment of the peripheral AN and a modest reduction in hearing sensitivity. We find that MEF2C is expressed during development in multiple AN and cochlear cell types; and in Mef2c-Het mice, we observe multiple cellular and molecular alterations associated with the AN, including abnormal myelination, neuronal degeneration, neuronal mitochondria dysfunction, and increased macrophage activation and cochlear inflammation. These results reveal the importance of MEF2C function in inner ear development and function and the engagement of immune cells and other non-neuronal cells, which suggests that microglia/macrophages and other non-neuronal cells might contribute, directly or indirectly, to AN dysfunction and ASD-related phenotypes. Finally, our study establishes a comprehensive approach for characterizing AN function at the physiological, cellular, and molecular levels in mice, which can be applied to animal models with a wide range of human auditory processing impairments.SIGNIFICANCE STATEMENT This is the first report of peripheral auditory nerve (AN) impairment in a mouse model of human MEF2C haploinsufficiency syndrome that has well-characterized ASD-related behaviors, including communication deficits, hyperactivity, repetitive behavior, and social deficits. We identify multiple underlying cellular, subcellular, and molecular abnormalities that may contribute to peripheral AN impairment. Our findings also highlight the important roles of immune cells (e.g., cochlear macrophages) and other non-neuronal elements (e.g., glial cells and cells in the stria vascularis) in auditory impairment in ASD. The methodological significance of the study is the establishment of a comprehensive approach for evaluating peripheral AN function and impact of peripheral AN deficits with minimal hearing loss.
Collapse
Affiliation(s)
- Nathan McChesney
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Jeremy L Barth
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Jeffrey A Rumschlag
- Department of Otolaryngology & Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Junying Tan
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Adam J Harrington
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Kenyaria V Noble
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Carolyn M McClaskey
- Department of Otolaryngology & Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Phillip Elvis
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Silvia G Vaena
- Hollings Cancer Institute, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Martin J Romeo
- Hollings Cancer Institute, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Kelly C Harris
- Department of Otolaryngology & Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Christopher W Cowan
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Hainan Lang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
| |
Collapse
|
76
|
Wan H, Zhang Y, Hua Q. Cellular autophagy, the compelling roles in hearing function and dysfunction. Front Cell Neurosci 2022; 16:966202. [PMID: 36246522 PMCID: PMC9561951 DOI: 10.3389/fncel.2022.966202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/14/2022] [Indexed: 11/21/2022] Open
Abstract
Sensorineural hearing loss (SNHL) is currently a major health issue. As one of the most common neurodegenerative diseases, SNHL is associated with the degradation of hair cells (HCs), spiral ganglion neurons (SGNs), the stria vascularis, supporting cells and central auditory system cells. Autophagy is a highly integrated cellular system that eliminates impaired components and replenishes energy to benefit cellular homeostasis. Etiological links between autophagy alterations and neurodegenerative diseases, such as SNHL, have been established. The hearing pathway is complex and depends on the comprehensive functions of many types of tissues and cells in auditory system. In this review, we discuss the roles of autophagy in promoting and inhibiting hearing, paying particular attention to specific cells in the auditory system, as discerned through research. Hence, our review provides enlightening ideas for the role of autophagy in hearing development and impairment.
Collapse
Affiliation(s)
- Huanzhi Wan
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuanyuan Zhang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Yuanyuan Zhang,
| | - Qingquan Hua
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Qingquan Hua,
| |
Collapse
|
77
|
Filova I, Pysanenko K, Tavakoli M, Vochyanova S, Dvorakova M, Bohuslavova R, Smolik O, Fabriciova V, Hrabalova P, Benesova S, Valihrach L, Cerny J, Yamoah EN, Syka J, Fritzsch B, Pavlinkova G. ISL1 is necessary for auditory neuron development and contributes toward tonotopic organization. Proc Natl Acad Sci U S A 2022; 119:e2207433119. [PMID: 36074819 PMCID: PMC9478650 DOI: 10.1073/pnas.2207433119] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 08/04/2022] [Indexed: 11/18/2022] Open
Abstract
A cardinal feature of the auditory pathway is frequency selectivity, represented in a tonotopic map from the cochlea to the cortex. The molecular determinants of the auditory frequency map are unknown. Here, we discovered that the transcription factor ISL1 regulates the molecular and cellular features of auditory neurons, including the formation of the spiral ganglion and peripheral and central processes that shape the tonotopic representation of the auditory map. We selectively knocked out Isl1 in auditory neurons using Neurod1Cre strategies. In the absence of Isl1, spiral ganglion neurons migrate into the central cochlea and beyond, and the cochlear wiring is profoundly reduced and disrupted. The central axons of Isl1 mutants lose their topographic projections and segregation at the cochlear nucleus. Transcriptome analysis of spiral ganglion neurons shows that Isl1 regulates neurogenesis, axonogenesis, migration, neurotransmission-related machinery, and synaptic communication patterns. We show that peripheral disorganization in the cochlea affects the physiological properties of hearing in the midbrain and auditory behavior. Surprisingly, auditory processing features are preserved despite the significant hearing impairment, revealing central auditory pathway resilience and plasticity in Isl1 mutant mice. Mutant mice have a reduced acoustic startle reflex, altered prepulse inhibition, and characteristics of compensatory neural hyperactivity centrally. Our findings show that ISL1 is one of the obligatory factors required to sculpt auditory structural and functional tonotopic maps. Still, upon Isl1 deletion, the ensuing central plasticity of the auditory pathway does not suffice to overcome developmentally induced peripheral dysfunction of the cochlea.
Collapse
Affiliation(s)
- Iva Filova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Kateryna Pysanenko
- Department of Auditory Neuroscience, Institute of Experimental Medicine Czech Academy of Sciences, 14220 Prague, Czechia
| | - Mitra Tavakoli
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Simona Vochyanova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Martina Dvorakova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Romana Bohuslavova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Ondrej Smolik
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Valeria Fabriciova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Petra Hrabalova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Sarka Benesova
- Laboratory of Gene Expression, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Lukas Valihrach
- Laboratory of Gene Expression, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Jiri Cerny
- Laboratory of Light Microscopy, Institute of Molecular Genetics Czech Academy of Sciences, 14220 Prague, Czechia
| | - Ebenezer N. Yamoah
- Department of Physiology, School of Medicine, University of Nevada, Reno, NV 89557
| | - Josef Syka
- Department of Auditory Neuroscience, Institute of Experimental Medicine Czech Academy of Sciences, 14220 Prague, Czechia
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, IA 52242-1324
- Department of Otolaryngology, University of Iowa, Iowa City, IA 52242-1324
| | - Gabriela Pavlinkova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| |
Collapse
|
78
|
Sharma K, Kang KW, Seo YW, Glowatzki E, Yi E. Low-voltage Activating K + Channels in Cochlear Afferent Nerve Fiber Dendrites. Exp Neurobiol 2022; 31:243-259. [PMID: 36050224 PMCID: PMC9471414 DOI: 10.5607/en22013] [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: 04/01/2022] [Revised: 06/18/2022] [Accepted: 07/29/2022] [Indexed: 11/19/2022] Open
Abstract
Cochlear afferent nerve fibers (ANF) are the first neurons in the ascending auditory pathway. We investigated the low-voltage activating K+ channels expressed in ANF dendrites using isolated rat cochlear segments. Whole cell patch clamp recordings were made from the dendritic terminals of ANFs. Outward currents activating at membrane potentials as low as -64 mV were observed in all dendrites studied. These currents were inhibited by 4-aminopyridine (4-AP), a blocker known to preferentially inhibit low-voltage activating K+ currents (IKL) in CNS auditory neurons and spiral ganglion neurons. When the dendritic IKL was blocked by 4-AP, the EPSP decay time was significantly prolonged, suggesting that dendritic IKL speeds up the decay of EPSPs and likely modulates action potentials of ANFs. To reveal molecular subtype of dendritic IKL, α-dendrotoxin (α-DTX), a selective inhibitor for Kv1.1, Kv1.2, and Kv1.6 containing channels, was tested. α-DTX inhibited 23±9% of dendritic IKL. To identify the α-DTXsensitive and α-DTX-insensitive components of IKL, immunofluorescence labeling was performed. Strong Kv1.1- and Kv1.2-immunoreactivity was found at unmyelinated dendritic segments, nodes of Ranvier, and cell bodies of most ANFs. A small fraction of ANF dendrites showed Kv7.2- immunoreactivity. These data suggest that dendritic IKL is conducted through Kv1.1and Kv1.2 channels, with a minor contribution from Kv7.2 and other as yet unidentified channels.
Collapse
Affiliation(s)
- Kushal Sharma
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Muan 58554, Korea
| | - Kwon Woo Kang
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Muan 58554, Korea
| | - Young-Woo Seo
- KBSI Gwangju Center, Korea Basic Science Institute, Gwangju 61186, Korea
| | - Elisabeth Glowatzki
- Department of Otolaryngology-Head and Neck Surgery and Neuroscience, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Eunyoung Yi
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Muan 58554, Korea
| |
Collapse
|
79
|
Abstract
Current estimates suggest that nearly half a billion people worldwide are affected by hearing loss. Because of the major psychological, social, economic, and health ramifications, considerable efforts have been invested in identifying the genes and molecular pathways involved in hearing loss, whether genetic or environmental, to promote prevention, improve rehabilitation, and develop therapeutics. Genomic sequencing technologies have led to the discovery of genes associated with hearing loss. Studies of the transcriptome and epigenome of the inner ear have characterized key regulators and pathways involved in the development of the inner ear and have paved the way for their use in regenerative medicine. In parallel, the immense preclinical success of using viral vectors for gene delivery in animal models of hearing loss has motivated the industry to work on translating such approaches into the clinic. Here, we review the recent advances in the genomics of auditory function and dysfunction, from patient diagnostics to epigenetics and gene therapy.
Collapse
Affiliation(s)
- Shahar Taiber
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; ,
| | - Kathleen Gwilliam
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA; ,
| | - Ronna Hertzano
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA; ,
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Karen B Avraham
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; ,
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
80
|
Ramirez MA, Ninoyu Y, Miller C, Andrade LR, Edassery S, Bomba-Warczak E, Ortega B, Manor U, Rutherford MA, Friedman RA, Savas JN. Cochlear ribbon synapse maturation requires Nlgn1 and Nlgn3. iScience 2022; 25:104803. [PMID: 35992071 PMCID: PMC9386149 DOI: 10.1016/j.isci.2022.104803] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 05/10/2022] [Accepted: 07/15/2022] [Indexed: 01/13/2023] Open
Abstract
Hearing depends on precise synaptic transmission between cochlear inner hair cells and spiral ganglion neurons through afferent ribbon synapses. Neuroligins (Nlgns) facilitate synapse maturation in the brain, but they have gone unstudied in the cochlea. We report Nlgn3 and Nlgn1 knockout (KO) cochleae have fewer ribbon synapses and have impaired hearing. Nlgn3 KO is more vulnerable to noise trauma with limited activity at high frequencies one day after noise. Furthermore, Nlgn3 KO cochleae have a 5-fold reduction in synapse number compared to wild type after two weeks of recovery. Double KO cochlear phenotypes are more prominent than the KOs, for example, 5-fold smaller synapses, 25% reduction in synapse density, and 30% less synaptic output. These observations indicate Nlgn3 and Nlgn1 are essential to cochlear ribbon synapse maturation and function.
Collapse
Affiliation(s)
- Miguel A. Ramirez
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Yuzuru Ninoyu
- Division of Otolaryngology, Department of Surgery, University of California, San Diego, 9500 Gilman Drive, Mail Code 0666, La Jolla, CA 92093, USA
| | - Cayla Miller
- Waitt Advanced Biophotonics Core, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Leonardo R. Andrade
- Waitt Advanced Biophotonics Core, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Seby Edassery
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ewa Bomba-Warczak
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Briana Ortega
- Division of Otolaryngology, Department of Surgery, University of California, San Diego, 9500 Gilman Drive, Mail Code 0666, La Jolla, CA 92093, USA
| | - Uri Manor
- Waitt Advanced Biophotonics Core, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Mark A. Rutherford
- Department of Otolaryngology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Rick A. Friedman
- Division of Otolaryngology, Department of Surgery, University of California, San Diego, 9500 Gilman Drive, Mail Code 0666, La Jolla, CA 92093, USA
| | - Jeffrey N. Savas
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
81
|
Bali B, Gruber-Dujardin E, Kusch K, Rankovic V, Moser T. Analyzing efficacy, stability, and safety of AAV-mediated optogenetic hearing restoration in mice. Life Sci Alliance 2022; 5:e202101338. [PMID: 35512833 PMCID: PMC9258265 DOI: 10.26508/lsa.202101338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 11/24/2022] Open
Abstract
AAV-mediated optogenetic neural stimulation has become a clinical approach for restoring function in sensory disorders and feasibility for hearing restoration has been indicated in rodents. Nonetheless, long-term stability and safety of AAV-mediated channelrhodopsin (ChR) expression in spiral ganglion neurons (SGNs) remained to be addressed. Here, we used longitudinal studies on mice subjected to early postnatal administration of AAV2/6 carrying fast gating ChR f-Chrimson under the control of the human synapsin promoter unilaterally to the cochlea. f-Chrimson expression in SGNs in both ears and the brain was probed in animals aged 1 mo to 2 yr. f-Chrimson was observed in SGNs at all ages indicating longevity of ChR-expression. SGN numbers in the AAV-injected cochleae declined with age faster than in controls. Investigations were extended to the brain in which viral transduction was observed across the organ at varying degrees irrespective of age without observing viral spread-related pathologies. No viral DNA or virus-related histopathological findings in visceral organs were encountered. In summary, our study demonstrates life-long (24 mo in mice) expression of f-Chrimson in SGNs upon single AAV-dosing of the cochlea.
Collapse
Affiliation(s)
- Burak Bali
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Göttingen Graduate School for Neurosciences and Molecular Biosciences, University of Göttingen, Göttingen, Germany
- Restorative Cochlear Genomics Group, Auditory Neuroscience and Optogenetics Laboratory, German Primate Center, Göttingen, Germany
| | | | - Kathrin Kusch
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Functional Auditory Genomics, Auditory Neuroscience and Optogenetics Laboratory, German Primate Center, Göttingen, Germany
| | - Vladan Rankovic
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Restorative Cochlear Genomics Group, Auditory Neuroscience and Optogenetics Laboratory, German Primate Center, Göttingen, Germany
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience and Optogenetics Laboratory, German Primate Center, Göttingen, Germany
- Auditory Neuroscience and 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 Goettingen, Göttingen, Germany
| |
Collapse
|
82
|
Petitpré C, Faure L, Uhl P, Fontanet P, Filova I, Pavlinkova G, Adameyko I, Hadjab S, Lallemend F. Single-cell RNA-sequencing analysis of the developing mouse inner ear identifies molecular logic of auditory neuron diversification. Nat Commun 2022; 13:3878. [PMID: 35790771 PMCID: PMC9256748 DOI: 10.1038/s41467-022-31580-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 06/21/2022] [Indexed: 11/08/2022] Open
Abstract
Different types of spiral ganglion neurons (SGNs) are essential for auditory perception by transmitting complex auditory information from hair cells (HCs) to the brain. Here, we use deep, single cell transcriptomics to study the molecular mechanisms that govern their identity and organization in mice. We identify a core set of temporally patterned genes and gene regulatory networks that may contribute to the diversification of SGNs through sequential binary decisions and demonstrate a role for NEUROD1 in driving specification of a Ic-SGN phenotype. We also find that each trajectory of the decision tree is defined by initial co-expression of alternative subtype molecular controls followed by gradual shifts toward cell fate resolution. Finally, analysis of both developing SGN and HC types reveals cell-cell signaling potentially playing a role in the differentiation of SGNs. Our results indicate that SGN identities are drafted prior to birth and reveal molecular principles that shape their differentiation and will facilitate studies of their development, physiology, and dysfunction.
Collapse
Affiliation(s)
- Charles Petitpré
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Louis Faure
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, 1090, Vienna, Austria
| | - Phoebe Uhl
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Paula Fontanet
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Iva Filova
- Institute of Biotechnology CAS, 25250, Vestec, Czech Republic
| | | | - Igor Adameyko
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, 1090, Vienna, Austria
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Saida Hadjab
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Francois Lallemend
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
- Ming-Wai Lau Centre for Reparative Medicine, Stockholm Node, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
83
|
Lewis MA, Schulte BA, Dubno JR, Steel KP. Investigating the characteristics of genes and variants associated with self-reported hearing difficulty in older adults in the UK Biobank. BMC Biol 2022; 20:150. [PMID: 35761239 PMCID: PMC9238072 DOI: 10.1186/s12915-022-01349-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 06/10/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Age-related hearing loss is a common, heterogeneous disease with a strong genetic component. More than 100 loci have been reported to be involved in human hearing impairment to date, but most of the genes underlying human adult-onset hearing loss remain unknown. Most genetic studies have focussed on very rare variants (such as family studies and patient cohort screens) or very common variants (genome-wide association studies). However, the contribution of variants present in the human population at intermediate frequencies is hard to quantify using these methods, and as a result, the landscape of variation associated with adult-onset hearing loss remains largely unknown. RESULTS Here we present a study based on exome sequencing and self-reported hearing difficulty in the UK Biobank, a large-scale biomedical database. We have carried out variant load analyses using different minor allele frequency and impact filters, and compared the resulting gene lists to a manually curated list of nearly 700 genes known to be involved in hearing in humans and/or mice. An allele frequency cutoff of 0.1, combined with a high predicted variant impact, was found to be the most effective filter setting for our analysis. We also found that separating the participants by sex produced markedly different gene lists. The gene lists obtained were investigated using gene ontology annotation, functional prioritisation and expression analysis, and this identified good candidates for further study. CONCLUSIONS Our results suggest that relatively common as well as rare variants with a high predicted impact contribute to age-related hearing impairment and that the genetic contributions to adult hearing difficulty may differ between the sexes. Our manually curated list of deafness genes is a useful resource for candidate gene prioritisation in hearing loss.
Collapse
Affiliation(s)
- Morag A Lewis
- Wolfson Centre for Age-Related Diseases, King's College London, London, SE1 1UL, UK.
| | | | - Judy R Dubno
- The Medical University of South Carolina, Charleston, SC, USA
| | - Karen P Steel
- Wolfson Centre for Age-Related Diseases, King's College London, London, SE1 1UL, UK
| |
Collapse
|
84
|
The role of Aquaporins in Hearing Function and Dysfunction. Eur J Cell Biol 2022; 101:151252. [DOI: 10.1016/j.ejcb.2022.151252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/09/2022] [Accepted: 06/22/2022] [Indexed: 11/23/2022] Open
|
85
|
Garrido-Charles A, Huet A, Matera C, Thirumalai A, Hernando J, Llebaria A, Moser T, Gorostiza P. Fast Photoswitchable Molecular Prosthetics Control Neuronal Activity in the Cochlea. J Am Chem Soc 2022; 144:9229-9239. [PMID: 35584208 PMCID: PMC9164239 DOI: 10.1021/jacs.1c12314] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Indexed: 12/15/2022]
Abstract
Artificial control of neuronal activity enables the study of neural circuits and restoration of neural functions. Direct, rapid, and sustained photocontrol of intact neurons could overcome the limitations of established electrical stimulation such as poor selectivity. We have developed fast photoswitchable ligands of glutamate receptors (GluRs) to enable neuronal control in the auditory system. The new photoswitchable ligands induced photocurrents in untransfected neurons upon covalently tethering to endogenous GluRs and activating them reversibly with visible light pulses of a few milliseconds. As a proof of concept of these molecular prostheses, we applied them to the ultrafast synapses of auditory neurons of the cochlea that encode sound and provide auditory input to the brain. This drug-based method afforded the optical stimulation of auditory neurons of adult gerbils at hundreds of hertz without genetic manipulation that would be required for their optogenetic control. This indicates that the new photoswitchable ligands are also applicable to the spatiotemporal control of fast spiking interneurons in the brain.
Collapse
Affiliation(s)
- Aida Garrido-Charles
- Institute
for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science
and Technology, Carrer
de Baldiri Reixac 15-21, 08028 Barcelona, Spain
- Network
Biomedical Research Center in Bioengineering, Biomaterials, and Nanomedicine
(CIBER-BBN), 28029 Madrid, Spain
- Institute
for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany
- Auditory
Neuroscience and Optogenetics Group, German
Primate Center, 37077 Göttingen, Germany
- Cluster
of Excellence “Multiscale Bioimaging: from Molecular Machines
to Networks of Excitable Cells” (MBExC), University of Göttingen, 37075 Göttingen, Germany
| | - Antoine Huet
- Institute
for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany
- Auditory
Neuroscience and Optogenetics Group, German
Primate Center, 37077 Göttingen, Germany
- Cluster
of Excellence “Multiscale Bioimaging: from Molecular Machines
to Networks of Excitable Cells” (MBExC), University of Göttingen, 37075 Göttingen, Germany
- Auditory
Circuit Lab, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Carlo Matera
- Institute
for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science
and Technology, Carrer
de Baldiri Reixac 15-21, 08028 Barcelona, Spain
- Network
Biomedical Research Center in Bioengineering, Biomaterials, and Nanomedicine
(CIBER-BBN), 28029 Madrid, Spain
- Department
of Pharmaceutical Sciences, University of
Milan, Via Luigi Mangiagalli
25, 20133 Milan, Italy
| | - Anupriya Thirumalai
- Institute
for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany
- Auditory
Neuroscience and Optogenetics Group, German
Primate Center, 37077 Göttingen, Germany
- Auditory
Circuit Lab, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Jordi Hernando
- Departament
de Química, Universitat Autònoma
de Barcelona (UAB), Cerdanyola
del Vallès 08193, Spain
| | - Amadeu Llebaria
- Consejo
Superior de Investigaciones Científicas (IQAC-CSIC), Institute of Advanced Chemistry of Catalonia, 08034 Barcelona, Spain
| | - Tobias Moser
- Institute
for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany
- Auditory
Neuroscience and Optogenetics Group, German
Primate Center, 37077 Göttingen, Germany
- Cluster
of Excellence “Multiscale Bioimaging: from Molecular Machines
to Networks of Excitable Cells” (MBExC), University of Göttingen, 37075 Göttingen, Germany
| | - Pau Gorostiza
- Institute
for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science
and Technology, Carrer
de Baldiri Reixac 15-21, 08028 Barcelona, Spain
- Network
Biomedical Research Center in Bioengineering, Biomaterials, and Nanomedicine
(CIBER-BBN), 28029 Madrid, Spain
- Catalan Institution for Research and Advanced
Studies (ICREA), 08010 Barcelona, Spain
| |
Collapse
|
86
|
Crozier RA, Wismer ZQ, Parra-Munevar J, Plummer MR, Davis RL. Amplification of input differences by dynamic heterogeneity in the spiral ganglion. J Neurophysiol 2022; 127:1317-1333. [PMID: 35389760 PMCID: PMC9054264 DOI: 10.1152/jn.00544.2021] [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: 12/20/2021] [Revised: 03/30/2022] [Accepted: 03/30/2022] [Indexed: 11/22/2022] Open
Abstract
A defining feature of type I primary auditory afferents that compose ∼95% of the spiral ganglion is their intrinsic electrophysiological heterogeneity. This diversity is evident both between and within unitary, rapid, and slow adaptation (UA, RA, and SA) classes indicative of specializations designed to shape sensory receptor input. But to what end? Our initial impulse is to expect the opposite: that auditory afferents fire uniformly to represent acoustic stimuli with accuracy and high fidelity. Yet this is clearly not the case. One explanation for this neural signaling strategy is to coordinate a system in which differences between input stimuli are amplified. If this is correct, then stimulus disparity enhancements within the primary afferents should be transmitted seamlessly into auditory processing pathways that utilize population coding for difference detection. Using sound localization as an example, one would expect to observe separately regulated differences in intensity level compared with timing or spectral cues within a graded tonotopic distribution. This possibility was evaluated by examining the neuromodulatory effects of cAMP on immature neurons with high excitability and slow membrane kinetics. We found that electrophysiological correlates of intensity and timing were indeed independently regulated and tonotopically distributed, depending on intracellular cAMP signaling level. These observations, therefore, are indicative of a system in which differences between signaling elements of individual stimulus attributes are systematically amplified according to auditory processing constraints. Thus, dynamic heterogeneity mediated by cAMP in the spiral ganglion has the potential to enhance the representations of stimulus input disparities transmitted into higher level difference detection circuitry.NEW & NOTEWORTHY Can changes in intracellular second messenger signaling within primary auditory afferents shift our perception of sound? Results presented herein lead to this conclusion. We found that intracellular cAMP signaling level systematically altered the kinetics and excitability of primary auditory afferents, exemplifying how dynamic heterogeneity can enhance differences between electrophysiological correlates of timing and intensity.
Collapse
Affiliation(s)
| | - Zachary Q Wismer
- AtlantiCare Regional Medical Center, Department of Family Medicine, Atlantic City, New Jersey
| | - Jeffrey Parra-Munevar
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey
| | - Mark R Plummer
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey
| | - Robin L Davis
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey
| |
Collapse
|
87
|
Signatures of cochlear processing in neuronal coding of auditory information. Mol Cell Neurosci 2022; 120:103732. [PMID: 35489636 DOI: 10.1016/j.mcn.2022.103732] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 11/22/2022] Open
Abstract
The vertebrate ear is endowed with remarkable perceptual capabilities. The faintest sounds produce vibrations of magnitudes comparable to those generated by thermal noise and can nonetheless be detected through efficient amplification of small acoustic stimuli. Two mechanisms have been proposed to underlie such sound amplification in the mammalian cochlea: somatic electromotility and active hair-bundle motility. These biomechanical mechanisms may work in concert to tune auditory sensitivity. In addition to amplitude sensitivity, the hearing system shows exceptional frequency discrimination allowing mammals to distinguish complex sounds with great accuracy. For instance, although the wide hearing range of humans encompasses frequencies from 20 Hz to 20 kHz, our frequency resolution extends to one-thirtieth of the interval between successive keys on a piano. In this article, we review the different cochlear mechanisms underlying sound encoding in the auditory system, with a particular focus on the frequency decomposition of sounds. The relation between peak frequency of activation and location along the cochlea - known as tonotopy - arises from multiple gradients in biophysical properties of the sensory epithelium. Tonotopic mapping represents a major organizational principle both in the peripheral hearing system and in higher processing levels and permits the spectral decomposition of complex tones. The ribbon synapses connecting sensory hair cells to auditory afferents and the downstream spiral ganglion neurons are also tuned to process periodic stimuli according to their preferred frequency. Though sensory hair cells and neurons necessarily filter signals beyond a few kHz, many animals can hear well beyond this range. We finally describe how the cochlear structure shapes the neural code for further processing in order to send meaningful information to the brain. Both the phase-locked response of auditory nerve fibers and tonotopy are key to decode sound frequency information and place specific constraints on the downstream neuronal network.
Collapse
|
88
|
Romano DR, Hashino E, Nelson RF. Deafness-in-a-dish: modeling hereditary deafness with inner ear organoids. Hum Genet 2022; 141:347-362. [PMID: 34342719 PMCID: PMC9035009 DOI: 10.1007/s00439-021-02325-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/24/2021] [Indexed: 12/27/2022]
Abstract
Sensorineural hearing loss (SNHL) is a major cause of functional disability in both the developed and developing world. While hearing aids and cochlear implants provide significant benefit to many with SNHL, neither targets the cellular and molecular dysfunction that ultimately underlies SNHL. The successful development of more targeted approaches, such as growth factor, stem cell, and gene therapies, will require a yet deeper understanding of the underlying molecular mechanisms of human hearing and deafness. Unfortunately, the human inner ear cannot be biopsied without causing significant, irreversible damage to the hearing or balance organ. Thus, much of our current understanding of the cellular and molecular biology of human deafness, and of the human auditory system more broadly, has been inferred from observational and experimental studies in animal models, each of which has its own advantages and limitations. In 2013, researchers described a protocol for the generation of inner ear organoids from pluripotent stem cells (PSCs), which could serve as scalable, high-fidelity alternatives to animal models. Here, we discuss the advantages and limitations of conventional models of the human auditory system, describe the generation and characteristics of PSC-derived inner ear organoids, and discuss several strategies and recent attempts to model hereditary deafness in vitro. Finally, we suggest and discuss several focus areas for the further, intensive characterization of inner ear organoids and discuss the translational applications of these novel models of the human inner ear.
Collapse
Affiliation(s)
- Daniel R Romano
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, 980 W. Walnut Street, WH-C426C, Indianapolis, IN, 46202, USA
| | - Eri Hashino
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, 980 W. Walnut Street, WH-C426C, Indianapolis, IN, 46202, USA
| | - Rick F Nelson
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, 980 W. Walnut Street, WH-C426C, Indianapolis, IN, 46202, USA.
| |
Collapse
|
89
|
Michalski N, Petit C. Central auditory deficits associated with genetic forms of peripheral deafness. Hum Genet 2022; 141:335-345. [PMID: 34435241 PMCID: PMC9034985 DOI: 10.1007/s00439-021-02339-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 08/09/2021] [Indexed: 01/11/2023]
Abstract
Since the 1990s, the study of inherited hearing disorders, mostly those detected at birth, in the prelingual period or in young adults, has led to the identification of their causal genes. The genes responsible for more than 140 isolated (non-syndromic) and about 400 syndromic forms of deafness have already been discovered. Studies of mouse models of these monogenic forms of deafness have provided considerable insight into the molecular mechanisms of hearing, particularly those involved in the development and/or physiology of the auditory sensory organ, the cochlea. In parallel, studies of these models have also made it possible to decipher the pathophysiological mechanisms underlying hearing impairment. This has led a number of laboratories to investigate the potential of gene therapy for curing these forms of deafness. Proof-of-concept has now been obtained for the treatment of several forms of deafness in mouse models, paving the way for clinical trials of cochlear gene therapy in patients in the near future. Nevertheless, peripheral deafness may also be associated with central auditory dysfunctions and may extend well beyond the auditory system itself, as a consequence of alterations to the encoded sensory inputs or involvement of the causal deafness genes in the development and/or functioning of central auditory circuits. Investigating the diversity, causes and underlying mechanisms of these central dysfunctions, the ways in which they could impede the expected benefits of hearing restoration by peripheral gene therapy, and determining how these problems could be remedied is becoming a research field in its own right. Here, we provide an overview of the current knowledge about the central deficits associated with genetic forms of deafness.
Collapse
Affiliation(s)
- Nicolas Michalski
- Institut de l'Audition, Institut Pasteur, INSERM, 75012, Paris, France.
| | - Christine Petit
- Institut de l'Audition, Institut Pasteur, INSERM, 75012, Paris, France.
| |
Collapse
|
90
|
Liu W, Rask-Andersen H. Na/K-ATPase Gene Expression in the Human Cochlea: A Study Using mRNA in situ Hybridization and Super-Resolution Structured Illumination Microscopy. Front Mol Neurosci 2022; 15:857216. [PMID: 35431803 PMCID: PMC9009265 DOI: 10.3389/fnmol.2022.857216] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 02/23/2022] [Indexed: 12/03/2022] Open
Abstract
Background The pervasive Na/K-ATPase pump is highly expressed in the human cochlea and is involved in the generation of the endocochlear potential as well as auditory nerve signaling and relay. Its distribution, molecular organization and gene regulation are essential to establish to better understand inner ear function and disease. Here, we analyzed the expression and distribution of the ATP1A1, ATP1B1, and ATP1A3 gene transcripts encoding the Na/K-ATPase α1, α3, and β1 isoforms in different domains of the human cochlea using RNA in situ hybridization. Materials and Methods Archival paraformaldehyde-fixed sections derived from surgically obtained human cochleae were used to label single mRNA gene transcripts using the highly sensitive multiplex RNAscope® technique. Localization of gene transcripts was performed by super-resolution structured illumination microscopy (SR-SIM) using fluorescent-tagged probes. GJB6 encoding of the protein connexin30 served as an additional control. Results Single mRNA gene transcripts were seen as brightly stained puncta. Positive and negative controls verified the specificity of the labeling. ATP1A1 and ATP1B1 gene transcripts were demonstrated in the organ of Corti, including the hair and supporting cells. In the stria vascularis, these transcripts were solely expressed in the marginal cells. A large number of ATP1B1 gene transcripts were found in the spiral ganglion cell soma, outer sulcus, root cells, and type II fibrocytes. The ATP1B1 and ATP1A3 gene transcripts were rarely detected in axons. Discussion Surgically obtained inner ear tissue can be used to identify single mRNA gene transcripts using high-resolution fluorescence microscopy after prompt formaldehyde fixation and chelate decalcification. A large number of Na/K-ATPase gene transcripts were localized in selected areas of the cochlear wall epithelium, fibrocyte networks, and spiral ganglion, confirming the enzyme’s essential role for human cochlear function.
Collapse
|
91
|
Zhang C, Wang M, Lin S, Xie R. Calretinin-Expressing Synapses Show Improved Synaptic Efficacy with Reduced Asynchronous Release during High-Rate Activity. J Neurosci 2022; 42:2729-2742. [PMID: 35165172 PMCID: PMC8973423 DOI: 10.1523/jneurosci.1773-21.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/27/2022] [Accepted: 02/07/2022] [Indexed: 11/21/2022] Open
Abstract
Calretinin (CR) is a major calcium binding protein widely expressed in the CNS. However, its synaptic function remains largely elusive. At the auditory synapse of the endbulb of Held, CR is selectively expressed in different subtypes. Combining electrophysiology with immunohistochemistry, we investigated the synaptic transmission at the endbulb of Held synapses with and without endogenous CR expression in mature CBA/CAJ mice of either sex. Two synapse subtypes showed similar basal synaptic transmission, except a larger quantal size in CR-expressing synapses. During high-rate stimulus trains, CR-expressing synapses showed improved synaptic efficacy with significantly less depression and lower asynchronous release, suggesting more efficient exocytosis than non-CR-expressing synapses. Conversely, CR-expressing synapses had a smaller readily releasable pool size, which was countered by higher release probability and faster synaptic recovery to support sustained release during high-rate activity. EGTA-AM treatment did not change the synaptic transmission of CR-expressing synapses, but reduced synaptic depression and decreased asynchronous release at non-CR-expressing synapses, suggesting that CR helps to minimize calcium accumulation during high-rate activity. Both synapses express parvalbumin, another calcium-binding protein with slower kinetics and higher affinity than CR, but not calbindin. Furthermore, CR-expressing synapses only express the fast isoform of vesicular glutamate transporter 1 (VGluT1), while most non-CR-expressing synapses express both VGluT1 and the slower VGluT2, which may underlie their lagged synaptic recovery. The findings suggest that, paired with associated synaptic machinery, differential CR expression regulates synaptic efficacy among different subtypes of auditory nerve synapses to accomplish distinctive physiological functions in transmitting auditory information at high rates.SIGNIFICANCE STATEMENT CR is a major calcium-binding protein in the brain. It remains unclear how endogenous CR impacts synaptic transmission. We investigated the question at the large endbulb of Held synapses with selective CR expression and found that CR-expressing and non-CR-expressing synapses had similar release properties under basal synaptic transmission. During high-rate activity, however, CR-expressing synapses showed improved synaptic efficacy with less depression, lower asynchronous release, and faster recovery. Furthermore, CR-expressing synapses use exclusive VGluT1 to refill synaptic vesicles, while non-CR-expressing synapses use both VGluT1 and the slower isoform of VGluT2. Our findings suggest that CR may play significant roles in promoting synaptic efficacy during high-rate activity, and selective CR expression can differentially impact signal processing among different synapses.
Collapse
Affiliation(s)
- Chuangeng Zhang
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University, Columbus, Ohio 43210
| | - Meijian Wang
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University, Columbus, Ohio 43210
| | - Shengyin Lin
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University, Columbus, Ohio 43210
| | - Ruili Xie
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University, Columbus, Ohio 43210
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210
| |
Collapse
|
92
|
Perks KE, Sawtell NB. Neural readout of a latency code in the active electrosensory system. Cell Rep 2022; 38:110605. [PMID: 35354029 PMCID: PMC9045710 DOI: 10.1016/j.celrep.2022.110605] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/03/2022] [Accepted: 03/10/2022] [Indexed: 11/29/2022] Open
Abstract
The latency of spikes relative to a stimulus conveys sensory information across modalities. However, in most cases, it remains unclear whether and how such latency codes are utilized by postsynaptic neurons. In the active electrosensory system of mormyrid fish, a latency code for stimulus amplitude in electroreceptor afferent nerve fibers (EAs) is hypothesized to be read out by a central reference provided by motor corollary discharge (CD). Here, we demonstrate that CD enhances sensory responses in postsynaptic granular cells of the electrosensory lobe but is not required for reading out EA input. Instead, diverse latency and spike count tuning across the EA population give rise to graded information about stimulus amplitude that can be read out by standard integration of converging excitatory synaptic inputs. Inhibitory control over the temporal window of integration renders two granular cell subclasses differentially sensitive to information derived from relative spike latency versus spike count.
Collapse
Affiliation(s)
- Krista E Perks
- Department of Biology, Wesleyan University, Middletown, CT 06459, USA; Zuckerman Mind Brain Behavior Institute, Department of Neuroscience, Columbia University, New York, NY 10027, USA
| | - Nathaniel B Sawtell
- Zuckerman Mind Brain Behavior Institute, Department of Neuroscience, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
93
|
Single-cell transcriptomic landscapes of the otic neuronal lineage at multiple early embryonic ages. Cell Rep 2022; 38:110542. [PMID: 35320729 DOI: 10.1016/j.celrep.2022.110542] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/03/2021] [Accepted: 02/25/2022] [Indexed: 11/20/2022] Open
Abstract
Inner ear vestibular and spiral ganglion neurons (VGNs and SGNs) are known to play pivotal roles in balance control and sound detection. However, the molecular mechanisms underlying otic neurogenesis at early embryonic ages have remained unclear. Here, we use single-cell RNA sequencing to reveal the transcriptomes of mouse otic tissues at three embryonic ages, embryonic day 9.5 (E9.5), E11.5, and E13.5, covering proliferating and undifferentiated otic neuroblasts and differentiating VGNs and SGNs. We validate the high quality of our studies by using multiple assays, including genetic fate mapping analysis, and we uncover several genes upregulated in neuroblasts or differentiating VGNs and SGNs, such as Shox2, Myt1, Casz1, and Sall3. Notably, our findings suggest a general cascaded differentiation trajectory during early otic neurogenesis. The comprehensive understanding of early otic neurogenesis provided by our study holds critical implications for both basic and translational research.
Collapse
|
94
|
Sun Y, Zhang Y, Zhang D, Wang G, Song L, Liu Z. In vivo CRISPR-Cas9-mediated DNA chop identifies a cochlear outer hair cell-specific enhancer. FASEB J 2022; 36:e22233. [PMID: 35225354 DOI: 10.1096/fj.202100421rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 02/06/2022] [Accepted: 02/14/2022] [Indexed: 01/18/2023]
Abstract
Cochlear outer hair cells (OHCs) are essential for hearing. A short, OHC-specific enhancer is necessary but not yet available for gene therapeutic applications in OHC damage. Such damage is a major cause of deafness. Prestin is a motor protein exclusively expressed in OHCs. We hypothesized that the cis-regulatory DNA fragment deletion of Slc26a5 would affect its expression. We tested this hypothesis by conducting CRISPR/Cas9-mediated large DNA fragment deletion of mouse Slc26a5 intron regions. First, starting from a ~13 kbp fragment, step-by-step, we narrowed down the sequence to a 1.4 kbp segment. By deleting either a 13 kbp or 1.4 kbp fragment, we observed delayed Prestin expression. Second, we showed that 1.4 kbp was an OHC-specific enhancer because enhanced green fluorescent protein (EGFP) was highly and specifically expressed in OHCs in a transgenic mouse where EGFP was driven by the 1.4 kbp segment. More importantly, specific EGFP was also driven by its homologous 398 bp fragment in human Slc26a5. This suggests that the enhancer is likely to be evolutionarily conserved across different species.
Collapse
Affiliation(s)
- Yuwei Sun
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yu Zhang
- Department of Otolaryngology-Head and Neck Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - 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, China.,University of Chinese Academy of Sciences, Beijing, 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, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Lei Song
- Department of Otolaryngology-Head and Neck Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 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, China.,University of Chinese Academy of Sciences, Beijing, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| |
Collapse
|
95
|
Elliott KL, Fritzsch B, Yamoah EN, Zine A. Age-Related Hearing Loss: Sensory and Neural Etiology and Their Interdependence. Front Aging Neurosci 2022; 14:814528. [PMID: 35250542 PMCID: PMC8891613 DOI: 10.3389/fnagi.2022.814528] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 01/03/2022] [Indexed: 12/19/2022] Open
Abstract
Age-related hearing loss (ARHL) is a common, increasing problem for older adults, affecting about 1 billion people by 2050. We aim to correlate the different reductions of hearing from cochlear hair cells (HCs), spiral ganglion neurons (SGNs), cochlear nuclei (CN), and superior olivary complex (SOC) with the analysis of various reasons for each one on the sensory deficit profiles. Outer HCs show a progressive loss in a basal-to-apical gradient, and inner HCs show a loss in a apex-to-base progression that results in ARHL at high frequencies after 70 years of age. In early neonates, SGNs innervation of cochlear HCs is maintained. Loss of SGNs results in a considerable decrease (~50% or more) of cochlear nuclei in neonates, though the loss is milder in older mice and humans. The dorsal cochlear nuclei (fusiform neurons) project directly to the inferior colliculi while most anterior cochlear nuclei reach the SOC. Reducing the number of neurons in the medial nucleus of the trapezoid body (MNTB) affects the interactions with the lateral superior olive to fine-tune ipsi- and contralateral projections that may remain normal in mice, possibly humans. The inferior colliculi receive direct cochlear fibers and second-order fibers from the superior olivary complex. Loss of the second-order fibers leads to hearing loss in mice and humans. Although ARHL may arise from many complex causes, HC degeneration remains the more significant problem of hearing restoration that would replace the cochlear implant. The review presents recent findings of older humans and mice with hearing loss.
Collapse
Affiliation(s)
- Karen L. Elliott
- Department of Biology, University of Iowa, Iowa City, IA, United States
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, IA, United States
- *Correspondence: Bernd Fritzsch
| | - Ebenezer N. Yamoah
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, United States
| | - Azel Zine
- LBN, Laboratory of Bioengineering and Nanoscience, University of Montpellier, Montpellier, France
| |
Collapse
|
96
|
Panganiban CH, Barth JL, Tan J, Noble KV, McClaskey CM, Howard BA, Jafri SH, Dias JW, Harris KC, Lang H. Two distinct types of nodes of Ranvier support auditory nerve function in the mouse cochlea. Glia 2021; 70:768-791. [DOI: 10.1002/glia.24138] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 11/12/2021] [Accepted: 12/17/2021] [Indexed: 11/09/2022]
Affiliation(s)
- Clarisse H. Panganiban
- Department of Pathology and Laboratory Medicine Medical University of South Carolina Charleston South Carolina USA
- Wolfson Centre for Age‐Related Diseases King's College London London UK
| | - Jeremy L. Barth
- Department of Regenerative Medicine and Cell Biology Medical University of South Carolina Charleston South Carolina USA
| | - Junying Tan
- Department of Pathology and Laboratory Medicine Medical University of South Carolina Charleston South Carolina USA
| | - Kenyaria V. Noble
- Department of Pathology and Laboratory Medicine Medical University of South Carolina Charleston South Carolina USA
| | - Carolyn M. McClaskey
- Department of Otolaryngology & Head and Neck Surgery Medical University of South Carolina Charleston South Carolina USA
| | - Blake A. Howard
- Department of Pathology and Laboratory Medicine Medical University of South Carolina Charleston South Carolina USA
| | - Shabih H. Jafri
- Department of Pathology and Laboratory Medicine Medical University of South Carolina Charleston South Carolina USA
| | - James W. Dias
- Department of Otolaryngology & Head and Neck Surgery Medical University of South Carolina Charleston South Carolina USA
| | - Kelly C. Harris
- Department of Otolaryngology & Head and Neck Surgery Medical University of South Carolina Charleston South Carolina USA
| | - Hainan Lang
- Department of Pathology and Laboratory Medicine Medical University of South Carolina Charleston South Carolina USA
| |
Collapse
|
97
|
Suthakar K, Liberman MC. Auditory-nerve responses in mice with noise-induced cochlear synaptopathy. J Neurophysiol 2021; 126:2027-2038. [PMID: 34788179 DOI: 10.1152/jn.00342.2021] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Cochlear synaptopathy is the noise-induced or age-related loss of ribbon synapses between inner hair cells (IHCs) and auditory-nerve fibers (ANFs), first reported in CBA/CaJ mice. Recordings from single ANFs in anesthetized, noise-exposed guinea pigs suggested that neurons with low spontaneous rates (SRs) and high thresholds are more vulnerable than low-threshold, high-SR fibers. However, there is extensive postexposure regeneration of ANFs in guinea pigs but not in mice. Here, we exposed CBA/CaJ mice to octave-band noise and recorded sound-evoked and spontaneous activity from single ANFs at least 2 wk later. Confocal analysis of cochleae immunostained for pre- and postsynaptic markers confirmed the expected loss of 40%-50% of ANF synapses in the basal half of the cochlea; however, our data were not consistent with a selective loss of low-SR fibers. Rather they suggested a loss of both SR groups in synaptopathic regions. Single-fiber thresholds and frequency tuning recovered to pre-exposure levels; however, response to tone bursts showed increased peak and steady-state firing rates, as well as decreased jitter in first-spike latencies. This apparent gain-of-function increased the robustness of tone-burst responses in the presence of continuous masking noise. This study suggests that the nature of noise-induced synaptic damage varies between different species and that, in mouse, the noise-induced hyperexcitability seen in central auditory circuits is also observed at the level of the auditory nerve.NEW & NOTEWORTHY Noise-induced damage to synapses between inner hair cells and auditory-nerve fibers (ANFs) can occur without permanent hair cell damage, resulting in pathophysiology that "hides" behind normal thresholds. Prior single-fiber neurophysiology in guinea pig suggested that noise selectively targets high-threshold ANFs. Here, we show that the lingering pathophysiology differs in mouse, with both ANF groups affected and a paradoxical gain-of-function in surviving low-threshold fibers, including increased onset rate, decreased onset jitter, and reduced maskability.
Collapse
Affiliation(s)
- Kirupa Suthakar
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, Massachusetts.,Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts
| | - M Charles Liberman
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, Massachusetts.,Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
98
|
Liu J, Wang S, Lu Y, Wang H, Wang F, Qiu M, Xie Q, Han H, Hua Y. Aligned Organization of Synapses and Mitochondria in Auditory Hair Cells. Neurosci Bull 2021; 38:235-248. [PMID: 34837647 PMCID: PMC8975952 DOI: 10.1007/s12264-021-00801-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/25/2021] [Indexed: 10/19/2022] Open
Abstract
Recent studies have revealed great functional and structural heterogeneity in the ribbon-type synapses at the basolateral pole of the isopotential inner hair cell (IHC). This feature is believed to be critical for audition over a wide dynamic range, but whether the spatial gradient of ribbon morphology is fine-tuned in each IHC and how the mitochondrial network is organized to meet local energy demands of synaptic transmission remain unclear. By means of three-dimensional electron microscopy and artificial intelligence-based algorithms, we demonstrated the cell-wide structural quantification of ribbons and mitochondria in mature mid-cochlear IHCs of mice. We found that adjacent IHCs in staggered pairs differ substantially in cell body shape and ribbon morphology gradient as well as mitochondrial organization. Moreover, our analysis argues for a location-specific arrangement of correlated ribbon and mitochondrial function at the basolateral IHC pole.
Collapse
Affiliation(s)
- Jing Liu
- grid.9227.e0000000119573309National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190 China ,grid.410726.60000 0004 1797 8419School of Artificial Intelligence, School of Future Technology, University of Chinese Academy of Sciences, Beijing, 101408 China ,grid.507732.4CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai, 200031 China
| | - Shengxiong Wang
- grid.24516.340000000123704535Putuo People’s Hospital, Tongji University, Shanghai, 200060 China ,grid.16821.3c0000 0004 0368 8293Shanghai Institute of Precision Medicine, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125 China
| | - Yan Lu
- grid.16821.3c0000 0004 0368 8293Shanghai Institute of Precision Medicine, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125 China ,grid.412523.3Department of Otolaryngology–Head and Neck Surgery, Shanghai Ninth People’s Hospital, Shanghai, 200125 China ,grid.16821.3c0000 0004 0368 8293Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125 China ,grid.412987.10000 0004 0630 1330Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200125 China
| | - Haoyu Wang
- grid.16821.3c0000 0004 0368 8293Shanghai Institute of Precision Medicine, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125 China ,grid.412523.3Department of Otolaryngology–Head and Neck Surgery, Shanghai Ninth People’s Hospital, Shanghai, 200125 China ,grid.16821.3c0000 0004 0368 8293Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125 China ,grid.412987.10000 0004 0630 1330Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200125 China
| | - Fangfang Wang
- grid.16821.3c0000 0004 0368 8293Shanghai Institute of Precision Medicine, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125 China
| | - Miaoxin Qiu
- grid.24516.340000000123704535Putuo People’s Hospital, Tongji University, Shanghai, 200060 China
| | - Qiwei Xie
- grid.28703.3e0000 0000 9040 3743Research Base of Beijing Modern Manufacturing Development, Beijing University of Technology, Beijing, 100124 China
| | - Hua Han
- National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China. .,School of Artificial Intelligence, School of Future Technology, University of Chinese Academy of Sciences, Beijing, 101408, China. .,CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai, 200031, China.
| | - Yunfeng Hua
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China. .,Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai, 200125, China. .,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China. .,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200125, China.
| |
Collapse
|
99
|
Wang Q, Shen Y, Pan Y, Chen K, Ding R, Zou T, Zhang A, Guo D, Ji P, Fan C, Mei L, Hu H, Ye B, Xiang M. Tlr2/4 Double Knockout Attenuates the Degeneration of Primary Auditory Neurons: Potential Mechanisms From Transcriptomic Perspectives. Front Cell Dev Biol 2021; 9:750271. [PMID: 34760891 PMCID: PMC8573328 DOI: 10.3389/fcell.2021.750271] [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: 07/30/2021] [Accepted: 09/28/2021] [Indexed: 11/30/2022] Open
Abstract
The transcriptomic landscape of mice with primary auditory neurons degeneration (PAND) indicates key pathways in its pathogenesis, including complement cascades, immune responses, tumor necrosis factor (TNF) signaling pathway, and cytokine-cytokine receptor interaction. Toll-like receptors (TLRs) are important immune and inflammatory molecules that have been shown to disrupt the disease network of PAND. In a PAND model involving administration of kanamycin combined with furosemide to destroy cochlear hair cells, Tlr 2/4 double knockout (DKO) mice had auditory preservation advantages, which were mainly manifested at 4–16 kHz. DKO mice and wild type (WT) mice had completely damaged cochlear hair cells on the 30th day, but the density of spiral ganglion neurons (SGN) in the Rosenthal canal was significantly higher in the DKO group than in the WT group. The results of immunohistochemistry for p38 and p65 showed that the attenuation of SGN degeneration in DKO mice may not be mediated by canonical Tlr signaling pathways. The SGN transcriptome of DKO and WT mice indicated that there was an inverted gene set enrichment relationship between their different transcriptomes and the SGN degeneration transcriptome, which is consistent with the morphology results. Core module analysis suggested that DKO mice may modulate SGN degeneration by activating two clusters, and the involved molecules include EGF, STAT3, CALB2, LOX, SNAP25, CAV2, SDC4, MYL1, NCS1, PVALB, TPM4, and TMOD4.
Collapse
Affiliation(s)
- Quan Wang
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yilin Shen
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Pan
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kaili Chen
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Ding
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianyuan Zou
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Andi Zhang
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongye Guo
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peilin Ji
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cui Fan
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Mei
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haixia Hu
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Ye
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingliang Xiang
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
100
|
Ahmed M, Moon R, Prajapati RS, James E, Basson MA, Streit A. The chromatin remodelling factor Chd7 protects auditory neurons and sensory hair cells from stress-induced degeneration. Commun Biol 2021; 4:1260. [PMID: 34732824 PMCID: PMC8566505 DOI: 10.1038/s42003-021-02788-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 10/08/2021] [Indexed: 11/08/2022] Open
Abstract
Neurons and sensory cells are particularly vulnerable to oxidative stress due to their high oxygen demand during stimulus perception and transmission. The mechanisms that protect them from stress-induced death and degeneration remain elusive. Here we show that embryonic deletion of the chromodomain helicase DNA-binding protein 7 (CHD7) in auditory neurons or hair cells leads to sensorineural hearing loss due to postnatal degeneration of both cell types. Mechanistically, we demonstrate that CHD7 controls the expression of major stress pathway components. In its absence, hair cells are hypersensitive, dying rapidly after brief exposure to stress inducers, suggesting that sound at the onset of hearing triggers their degeneration. In humans, CHD7 haploinsufficiency causes CHARGE syndrome, a disorder affecting multiple organs including the ear. Our findings suggest that CHD7 mutations cause developmentally silent phenotypes that predispose cells to postnatal degeneration due to a failure of protective mechanisms.
Collapse
Affiliation(s)
- Mohi Ahmed
- Centre for Craniofacial and Regenerative Biology, Floor 27 Tower Wing, Guy's Hospital, King's College London, London, SE1 9RT, UK.
| | - Ruth Moon
- Centre for Craniofacial and Regenerative Biology, Floor 27 Tower Wing, Guy's Hospital, King's College London, London, SE1 9RT, UK
| | - Ravindra Singh Prajapati
- Centre for Craniofacial and Regenerative Biology, Floor 27 Tower Wing, Guy's Hospital, King's College London, London, SE1 9RT, UK
- Leukaemia and Stem Cell Biology Group, School of Cancer and Pharmaceutical Sciences, King's College London, London, SE5 9NU, UK
| | - Elysia James
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK
| | - M Albert Basson
- Centre for Craniofacial and Regenerative Biology, Floor 27 Tower Wing, Guy's Hospital, King's College London, London, SE1 9RT, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, SE1 1UL, UK
| | - Andrea Streit
- Centre for Craniofacial and Regenerative Biology, Floor 27 Tower Wing, Guy's Hospital, King's College London, London, SE1 9RT, UK.
| |
Collapse
|