1
|
Wang T, Yang T, Kedaigle A, Pregernig G, McCarthy R, Holmes B, Wu X, Becker L, Pan N, So K, Chen L, He J, Mahmoudi A, Negi S, Kowalczyk M, Gibson T, Druckenbrod N, Cheng AG, Burns J. Precise genetic control of ATOH1 enhances maturation of regenerated hair cells in the mature mouse utricle. Nat Commun 2024; 15:9166. [PMID: 39448563 PMCID: PMC11502789 DOI: 10.1038/s41467-024-53153-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Abstract
Vestibular hair cells are mechanoreceptors critical for detecting head position and motion. In mammals, hair cell loss causes vestibular dysfunction as spontaneous regeneration is nearly absent. Constitutive expression of exogenous ATOH1, a hair cell transcription factor, increases hair cell regeneration, however, these cells fail to fully mature. Here, we profiled mouse utricles at 14 time points, and defined transcriptomes of developing and mature vestibular hair cells. To mimic native hair cells which downregulate endogenous ATOH1 as they mature, we engineered viral vectors carrying the supporting cell promoters GFAP and RLBP1. In utricles damaged ex vivo, both CMV-ATOH1 and GFAP-ATOH1 increased regeneration more effectively than RLBP1-ATOH1, while GFAP-ATOH1 and RLBP1-ATOH1 induced hair cells with more mature transcriptomes. In utricles damaged in vivo, GFAP-ATOH1 induced regeneration of hair cells expressing genes indicative of maturing type II hair cells, and more hair cells with bundles and synapses than untreated organs. Together our results demonstrate the efficacy of spatiotemporal control of ATOH1 overexpression in inner ear hair cell regeneration.
Collapse
Affiliation(s)
- Tian Wang
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, 94305, USA
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Tian Yang
- Decibel Therapeutics, Boston, MA, 02215, USA
| | | | - Gabriela Pregernig
- Decibel Therapeutics, Boston, MA, 02215, USA
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Ryan McCarthy
- Decibel Therapeutics, Boston, MA, 02215, USA
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Ben Holmes
- Decibel Therapeutics, Boston, MA, 02215, USA
| | - Xudong Wu
- Decibel Therapeutics, Boston, MA, 02215, USA
| | - Lars Becker
- Decibel Therapeutics, Boston, MA, 02215, USA
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Ning Pan
- Decibel Therapeutics, Boston, MA, 02215, USA
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Kathy So
- Decibel Therapeutics, Boston, MA, 02215, USA
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Leon Chen
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, 94305, USA
| | - Jun He
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, 94305, USA
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Ahmad Mahmoudi
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, 94305, USA
| | - Soumya Negi
- Decibel Therapeutics, Boston, MA, 02215, USA
| | | | | | | | - Alan G Cheng
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, 94305, USA.
| | | |
Collapse
|
2
|
David S, Pinter K, Nguyen KK, Lee DS, Lei Z, Sokolova Y, Sheets L, Kindt KS. Kif1a and intact microtubules maintain synaptic-vesicle populations at ribbon synapses in zebrafish hair cells. J Physiol 2024. [PMID: 39373584 DOI: 10.1113/jp286263] [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: 05/20/2024] [Accepted: 09/05/2024] [Indexed: 10/08/2024] Open
Abstract
Sensory hair cells of the inner ear utilize specialized ribbon synapses to transmit sensory stimuli to the central nervous system. This transmission necessitates rapid and sustained neurotransmitter release, which depends on a large pool of synaptic vesicles at the hair-cell presynapse. While previous work in neurons has shown that kinesin motor proteins traffic synaptic material along microtubules to the presynapse, the mechanisms of this process in hair cells remain unclear. Our study demonstrates that the kinesin motor protein Kif1a, along with an intact microtubule network, is essential for enriching synaptic vesicles at the presynapse in hair cells. Through genetic and pharmacological approaches, we disrupt Kif1a function and impair microtubule networks in hair cells of the zebrafish lateral-line system. These manipulations led to a significant reduction in synaptic-vesicle populations at the presynapse in hair cells. Using electron microscopy, in vivo calcium imaging, and electrophysiology, we show that a diminished supply of synaptic vesicles adversely affects ribbon-synapse function. Kif1aa mutants exhibit dramatic reductions in spontaneous vesicle release and evoked postsynaptic calcium responses. Furthermore, kif1aa mutants exhibit impaired rheotaxis, a behaviour reliant on the ability of hair cells in the lateral line to respond to sustained flow stimuli. Overall, our results demonstrate that Kif1a-mediated microtubule transport is critical to enrich synaptic vesicles at the active zone, a process that is vital for proper ribbon-synapse function in hair cells. KEY POINTS: Kif1a mRNAs are present in zebrafish hair cells. Loss of Kif1a disrupts the enrichment of synaptic vesicles at ribbon synapses. Disruption of microtubules depletes synaptic vesicles at ribbon synapses. Kif1aa mutants have impaired ribbon-synapse and sensory-system function.
Collapse
Affiliation(s)
- Sandeep David
- Section on Sensory Cell Development and Function, National Institute on Deafness and other Communication Disorders, Bethesda, Maryland, USA
- National Institutes of Health-Brown University Graduate Partnership Program, Bethesda, Maryland, USA
| | - Katherine Pinter
- Section on Sensory Cell Development and Function, National Institute on Deafness and other Communication Disorders, Bethesda, Maryland, USA
| | - Keziah-Khue Nguyen
- Department of Otolaryngology, Head and Neck Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - David S Lee
- Department of Otolaryngology, Head and Neck Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Zhengchang Lei
- Section on Sensory Cell Development and Function, National Institute on Deafness and other Communication Disorders, Bethesda, Maryland, USA
| | - Yuliya Sokolova
- Advanced Imaging Core, National Institute on Deafness and other Communication Disorders, Bethesda, Maryland, USA
| | - Lavinia Sheets
- Department of Otolaryngology, Head and Neck Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Katie S Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and other Communication Disorders, Bethesda, Maryland, USA
| |
Collapse
|
3
|
Shi W, Zhao Q, Gao H, Yang Y, Tan Z, Li N, Wang H, Ji Y, Zhou Y. Exploring the bioactive ingredients of three traditional Chinese medicine formulas against age-related hearing loss through network pharmacology and experimental validation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03464-2. [PMID: 39356317 DOI: 10.1007/s00210-024-03464-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/16/2024] [Indexed: 10/03/2024]
Abstract
Traditional Chinese medicine (TCM) formulas, including the Er-Long-Zuo-Ci pill, Tong-Qiao-Er-Long pill, and Er-Long pill, have long been utilized in China for managing age-related hearing loss (ARHL). However, the specific bioactive compounds, pharmacological targets, and underlying mechanisms remain elusive. This study aims to find the shared bioactive ingredients among these three formulas, uncover the molecular pathways they regulate, and identify potential therapeutic targets for ARHL. Furthermore, it seeks to validate the efficacy of these major components through both in vivo and in vitro experiments. Common bioactive ingredients were extracted from the TCMSP database, and their putative target proteins were predicted using the Swiss Target Prediction database. ARHL-related target proteins were collected from GeneCards and OMIM databases. Our approach involved constructing drug-target networks and drug-disease-specific protein-protein interaction networks and conducting clustering, topological property analyses, and functional annotation through GO and KEGG enrichment analysis. Molecular docking analysis was utilized to delineate interaction mechanisms between major bioactive ingredients and key target proteins. Finally, in vivo and in vitro experiments involving ABR recording, immunofluorescent staining, HE staining, and quantitative PCR were conducted to validate the treatment effects of flavonoids on the declining auditory function in DBA/2 J mice. We identified 11 common chemical compounds across the three formulas and their associated 276 putative targets. Additionally, 3350 ARHL-related targets were compiled. As an intersection of the putative targets of the common compounds and ARHL-related proteins, 145 shared targets were determined. Functional enrichment analysis indicated that these compounds may modulate various biological processes, including cell proliferation, apoptosis, inflammatory response, and synaptic connections. Notably, potential targets such as TNFα, MAPK1, SRC, AKT, EGFR, ESR1, and AR were implicated. Flavonoids emerged as major bioactive components against ARHL based on target numbers, with molecular docking demonstrating diverse interaction models between these flavonoids and protein targets. Furthermore, baicalin could mitigate the age-related cochlear damage and hearing loss of DBA/2 J mice through its multi-target and multi-pathway mechanism, involving anti-inflammation, modulation of sex hormone-related pathways, and activation of potassium channels. This study offers an integrated network pharmacology approach, validated by in vivo and in vitro experiments, shedding light on the potential mechanisms, major active components, and therapeutic targets of TCM formulas for treating ARHL.
Collapse
Affiliation(s)
- Wenying Shi
- School of Basic Medical Sciences, Hebei University, Baoding, 071030, China
| | - Qi Zhao
- School of Basic Medical Sciences, Hebei University, Baoding, 071030, China
| | - Hongwei Gao
- School of Basic Medical Sciences, Hebei University, Baoding, 071030, China
| | - Yaxin Yang
- School of Basic Medical Sciences, Hebei University, Baoding, 071030, China
| | - Zhiyong Tan
- School of Basic Medical Sciences, Hebei University, Baoding, 071030, China
| | - Na Li
- School of Basic Medical Sciences, Hebei University, Baoding, 071030, China
| | - Hongjie Wang
- School of Basic Medical Sciences, Hebei University, Baoding, 071030, China
| | - Yonghua Ji
- School of Basic Medical Sciences, Hebei University, Baoding, 071030, China
| | - You Zhou
- School of Basic Medical Sciences, Hebei University, Baoding, 071030, China.
| |
Collapse
|
4
|
Ye M, Marzullo B, Adler HJ, Hu BH. Expression profiling of cochlear genes uncovers sex-based cellular function in mouse cochleae. Hear Res 2024; 448:109030. [PMID: 38776705 DOI: 10.1016/j.heares.2024.109030] [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: 01/06/2024] [Revised: 04/13/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024]
Abstract
Sex is a pivotal biological factor that significantly impacts tissue homeostasis and disease susceptibility. In the auditory system, sex differences have been observed in cochlear physiology and responses to pathological conditions. However, the underlying molecular mechanisms responsible for these differences remain elusive. The current research explores the differences in gene expression profiles in the cochlea between male and female mice, aiming to understand the functional implication of sex-biased gene expression in each sex. Using RNA-sequencing analysis on cochlear tissues obtained from male and female mice, we identified a significant number of genes exhibiting sex-biased expression differences. While some of these differentially expressed genes are located on sex chromosomes, most are found on autosomal chromosomes. Further bioinformatic analysis revealed that these genes are involved in several key cellular functions. In males, these genes are notably linked to oxidative phosphorylation and RNA synthesis and processing, suggesting their involvement in mitochondrial energy production and regulatory control of gene expression. In contrast, sex-biased genes are associated with mechano-transduction and synaptic transmission within female cochleae. Collectively, our study provides valuable insights into the molecular differences between the sexes and emphasizes the need for future research to uncover their functional implications and relevance to auditory health and disease development.
Collapse
Affiliation(s)
- Mengxiao Ye
- The Department of Communicative Disorders and Sciences, University at Buffalo, 137 Cary Hall, 3435 Main Street, Buffalo, NY 14214, USA
| | - Brandon Marzullo
- UB Genomics and Bioinformatics Core NYS Center of Excellence in Bioinformatics & Life Sciences, University at Buffalo, 701 Ellicott Street, Buffalo, NY 14222, USA
| | - Henry J Adler
- The Department of Communicative Disorders and Sciences, University at Buffalo, 137 Cary Hall, 3435 Main Street, Buffalo, NY 14214, USA
| | - Bo Hua Hu
- The Department of Communicative Disorders and Sciences, University at Buffalo, 137 Cary Hall, 3435 Main Street, Buffalo, NY 14214, USA.
| |
Collapse
|
5
|
Shahab M, Rosati R, Stemmer PM, Dombkowski A, Jamesdaniel S. Quantitative profiling of cochlear synaptosomal proteins in cisplatin-induced synaptic dysfunction. Hear Res 2024; 447:109022. [PMID: 38705005 PMCID: PMC11116033 DOI: 10.1016/j.heares.2024.109022] [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: 01/09/2024] [Revised: 04/19/2024] [Accepted: 04/26/2024] [Indexed: 05/07/2024]
Abstract
The disruption of ribbon synapses in the cochlea impairs the transmission of auditory signals from the cochlear sensory receptor cells to the auditory cortex. Although cisplatin-induced loss of ribbon synapses is well-documented, and studies have reported nitration of cochlear proteins after cisplatin treatment, yet the underlying mechanism of cochlear synaptopathy is not fully understood. This study tests the hypothesis that cisplatin treatment alters the abundance of cochlear synaptosomal proteins, and selective targeting of nitrative stress prevents the associated synaptic dysfunction. Auditory brainstem responses of mice treated with cisplatin showed a reduction in amplitude and an increase in latency of wave I, indicating cisplatin-induced synaptic dysfunction. The mass spectrometry analysis of cochlear synaptosomal proteins identified 102 proteins that decreased in abundance and 249 that increased in abundance after cisplatin treatment. Pathway analysis suggested that the dysregulated proteins were involved in calcium binding, calcium ion regulation, synapses, and endocytosis pathways. Inhibition of nitrative stress by co-treatment with MnTBAP, a peroxynitrite scavenger, attenuated cisplatin-induced changes in the abundance of 27 proteins. Furthermore, MnTBAP co-treatment prevented the cisplatin-induced decrease in the amplitude and increase in the latency of wave I. Together, these findings suggest a potential role of oxidative/nitrative stress in cisplatin-induced cochlear synaptic dysfunction.
Collapse
Affiliation(s)
- Monazza Shahab
- Department of Pharmacology, Wayne State University, Detroit, MI, USA; Institute of Environment Health Science, Wayne State University, Detroit, MI, USA
| | - Rita Rosati
- Institute of Environment Health Science, Wayne State University, Detroit, MI, USA
| | - Paul M Stemmer
- Institute of Environment Health Science, Wayne State University, Detroit, MI, USA
| | - Alan Dombkowski
- Department of Pediatrics, Wayne State University, Detroit, MI, USA
| | - Samson Jamesdaniel
- Department of Pharmacology, Wayne State University, Detroit, MI, USA; Institute of Environment Health Science, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
6
|
David S, Pinter K, Nguyen KK, Lee DS, Lei Z, Sokolova Y, Sheets L, Kindt KS. Kif1a and intact microtubules maintain synaptic-vesicle populations at ribbon synapses in zebrafish hair cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.20.595037. [PMID: 38903095 PMCID: PMC11188139 DOI: 10.1101/2024.05.20.595037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Sensory hair cells of the inner ear utilize specialized ribbon synapses to transmit sensory stimuli to the central nervous system. This sensory transmission necessitates rapid and sustained neurotransmitter release, which relies on a large pool of synaptic vesicles at the hair-cell presynapse. Work in neurons has shown that kinesin motor proteins traffic synaptic material along microtubules to the presynapse, but how new synaptic material reaches the presynapse in hair cells is not known. We show that the kinesin motor protein Kif1a and an intact microtubule network are necessary to enrich synaptic vesicles at the presynapse in hair cells. We use genetics and pharmacology to disrupt Kif1a function and impair microtubule networks in hair cells of the zebrafish lateral-line system. We find that these manipulations decrease synaptic-vesicle populations at the presynapse in hair cells. Using electron microscopy, along with in vivo calcium imaging and electrophysiology, we show that a diminished supply of synaptic vesicles adversely affects ribbon-synapse function. Kif1a mutants exhibit dramatic reductions in spontaneous vesicle release and evoked postsynaptic calcium responses. Additionally, we find that kif1a mutants exhibit impaired rheotaxis, a behavior reliant on the ability of hair cells in the lateral line to respond to sustained flow stimuli. Overall, our results demonstrate that Kif1a-based microtubule transport is critical to enrich synaptic vesicles at the active zone in hair cells, a process that is vital for proper ribbon-synapse function.
Collapse
Affiliation(s)
- Sandeep David
- Section on Sensory Cell Development and Function, National Institute on Deafness and other Communication Disorders, Bethesda, MD, USA
- National Institutes of Health-Brown University Graduate Partnership Program, Bethesda, MD, USA
| | - Katherine Pinter
- Section on Sensory Cell Development and Function, National Institute on Deafness and other Communication Disorders, Bethesda, MD, USA
| | - Keziah-Khue Nguyen
- Department of Otolaryngology - Head and Neck Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - David S Lee
- Department of Otolaryngology - Head and Neck Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Zhengchang Lei
- Section on Sensory Cell Development and Function, National Institute on Deafness and other Communication Disorders, Bethesda, MD, USA
| | - Yuliya Sokolova
- Advanced Imaging Core, National Institute on Deafness and other Communication Disorders, Bethesda, MD, USA
| | - Lavinia Sheets
- Department of Otolaryngology - Head and Neck Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Katie S Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and other Communication Disorders, Bethesda, MD, USA
| |
Collapse
|
7
|
Hong J, Dai P, Liang H, Sun G, Qi W, Bi Y. Extrasynaptic distribution of NMDA receptors in cochlear inner hair cell afferent signaling complex. J Chem Neuroanat 2024; 137:102417. [PMID: 38570170 DOI: 10.1016/j.jchemneu.2024.102417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/30/2024] [Accepted: 03/30/2024] [Indexed: 04/05/2024]
Abstract
OBJECTIVE The distribution and role of NMDA receptors is unclear in the afferent signaling complex of the cochlea. The present study aimed to examine the distribution of NMDA receptors in cochlear afferent signaling complex of the adult mouse, and their relationship with ribbon synapses of inner hair cells (IHCs) and GABAergic efferent terminals of the lateral olivocochlear (LOC). METHODS Immunofluorescence staining in combination with confocal microscopy was used to investigate the distribution of glutamatergic NMDA and AMPA receptors in afferent terminals of SGNs, and their relationship with ribbon synapses of IHCs and GABAergic efferent terminals of LOC. RESULTS Terminals with AMPA receptors along with Ribbons of IHC formed afferent synapses in the basal pole of IHCs, and those with NMDA receptors were mainly distributed longitudinally in the IHCs nuclei region. Significant difference was found in the distribution of NMDA and AMPA receptors in IHC afferent signaling complex (P<0.05). Some GABAergic terminals colocalized with NMDA receptors at the IHC nucleus region (P>0.05). CONCLUSION There is significant difference in the distribution of NMDA and AMPA receptors in cochlear afferent signaling complex. NMDA receptors are present in the extra-synaptic region of ribbon synapses of IHCs, and they are related to GABA efferent terminals of the afferent signaling complex.
Collapse
Affiliation(s)
- Juan Hong
- Department of Otorhinolaryngology, Huashan Hospital of Fudan University, Shanghai 200040, China.
| | - Peidong Dai
- ENT Institute, Eye & ENT Hospital of Fudan University; NHC Hearing Medicine Key Laboratory (Fudan University), Shanghai 200031, China
| | - Huazheng Liang
- Monash Suzhou Research Institute, Suzhou Industrial Park, Jiangsu, China
| | - Guangbin Sun
- Department of Otorhinolaryngology, Huashan Hospital of Fudan University, Shanghai 200040, China
| | - Weidong Qi
- Department of Otorhinolaryngology, Huashan Hospital of Fudan University, Shanghai 200040, China
| | - Yong Bi
- Department of Neurology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China.
| |
Collapse
|
8
|
Shim M, Yi J, Pak JH, Chung JW. Zinc deficiency triggers hearing loss by reducing ribbon synapses of inner hair cells in CBA/N mice. Biochem Biophys Res Commun 2024; 693:149396. [PMID: 38118309 DOI: 10.1016/j.bbrc.2023.149396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 12/15/2023] [Indexed: 12/22/2023]
Abstract
Zinc plays a vital role in our metabolism, encompassing antioxidant regulation, immune response, and auditory function. Several studies have reported that zinc levels correlate with hearing loss. We have previously demonstrated that the auditory brainstem response (ABR) threshold increased in mice fed a zinc-deficient diet. However, the effects of zinc deficiency on hearing were not fully elucidated. The present study investigated whether zinc deficiency affects hearing in association with neuronal components or cochlear structures. CBA/N mice were fed a normal or zinc-deficient diet for 8 weeks and assessed for ABR and distortion product otoacoustic emissions (DPOAE). The cochlear sections were stained with hematoxylin and eosin solution. Also, we observed the expression of synaptic ribbons, neurofilaments, and alpha-synuclein (α-Syn). The 8-week zinc-deficient diet mice had an elevated ABR threshold but no changed DPOAE threshold or cochlear structures. A reduced number of synaptic ribbons of inner hair cells (IHCs) and impaired efferent nerve fibers were observed in the zinc-deficient diet mice. The number of outer hair cells (OHCs) and expression of α-Syn remained unchanged. Our results suggest that zinc-mediated hearing loss is associated with the loss of neuronal components of IHCs.
Collapse
Affiliation(s)
- Myungjoo Shim
- Department of Otorhinolaryngology, University of Ulsan Graduate School, Seoul, South Korea
| | - Junyeong Yi
- Department of Otorhinolaryngology-Head and Neck Surgery, AMIST, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea; Department of Otorhinolaryngology-Head and Neck Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jhang Ho Pak
- Department of Biochemistry, University of Ulsan College of Medicine and Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| | - Jong Woo Chung
- Department of Otorhinolaryngology-Head and Neck Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.
| |
Collapse
|
9
|
Ford CL, Riggs WJ, Quigley T, Keifer OP, Whitton JP, Valayannopoulos V. The natural history, clinical outcomes, and genotype-phenotype relationship of otoferlin-related hearing loss: a systematic, quantitative literature review. Hum Genet 2023; 142:1429-1449. [PMID: 37679651 PMCID: PMC10511631 DOI: 10.1007/s00439-023-02595-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 08/09/2023] [Indexed: 09/09/2023]
Abstract
Congenital hearing loss affects one in 500 newborns. Sequence variations in OTOF, which encodes the calcium-binding protein otoferlin, are responsible for 1-8% of congenital, nonsyndromic hearing loss and are the leading cause of auditory neuropathy spectrum disorders. The natural history of otoferlin-related hearing loss, the relationship between OTOF genotype and hearing loss phenotype, and the outcomes of clinical practices in patients with this genetic disorder are incompletely understood because most analyses have reported on small numbers of cases with homogeneous OTOF genotypes. Here, we present the first systematic, quantitative literature review of otoferlin-related hearing loss, which analyzes patient-specific data from 422 individuals across 61 publications. While most patients display a typical phenotype of severe-to-profound hearing loss with prelingual onset, 10-15% of patients display atypical phenotypes, including mild-to-moderate, progressive, and temperature-sensitive hearing loss. Patients' phenotypic presentations appear to depend on their specific genotypes. For example, non-truncating variants located in and immediately downstream of the C2E calcium-binding domain are more likely to produce atypical phenotypes. Additionally, the prevalence of certain sequence variants and their associated phenotypes varies between populations due to evolutionary founder effects. Our analyses also suggest otoacoustic emissions are less common in older patients and those with two truncating OTOF variants. Critically, our review has implications for the application and limitations of clinical practices, including newborn hearing screenings, hearing aid trials, cochlear implants, and upcoming gene therapy clinical trials. We conclude by discussing the limitations of available research and recommendations for future studies on this genetic cause of hearing loss.
Collapse
|
10
|
Cortada M, Levano S, Hall MN, Bodmer D. mTORC2 regulates auditory hair cell structure and function. iScience 2023; 26:107687. [PMID: 37694145 PMCID: PMC10484995 DOI: 10.1016/j.isci.2023.107687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/14/2023] [Accepted: 08/17/2023] [Indexed: 09/12/2023] Open
Abstract
mTOR broadly controls cell growth, but little is known about the role of mTOR complex 2 (mTORC2) in the inner ear. To investigate the role of mTORC2 in sensory hair cells (HCs), we generated HC-specific Rictor knockout (HC-RicKO) mice. HC-RicKO mice exhibited early-onset, progressive, and profound hearing loss. Increased DPOAE thresholds indicated outer HC dysfunction. HCs are lost, but this occurs after hearing loss. Ultrastructural analysis revealed stunted and absent stereocilia in outer HCs. In inner HCs, the number of synapses was significantly decreased and the remaining synapses displayed a disrupted actin cytoskeleton and disorganized Ca2+ channels. Thus, the mTORC2 signaling pathway plays an important role in regulating auditory HC structure and function via regulation of the actin cytoskeleton. These results provide molecular insights on a central regulator of cochlear HCs and thus hearing.
Collapse
Affiliation(s)
- Maurizio Cortada
- Department of Biomedicine, University of Basel, CH-4031 Basel, Switzerland
| | - Soledad Levano
- Department of Biomedicine, University of Basel, CH-4031 Basel, Switzerland
| | | | - Daniel Bodmer
- Department of Biomedicine, University of Basel, CH-4031 Basel, Switzerland
- Clinic for Otorhinolaryngology, Head and Neck Surgery, University of Basel Hospital, CH-4031 Basel, Switzerland
| |
Collapse
|
11
|
Durbin RJ, Heredia DJ, Gould TW, Renden RB. Postsynaptic Calcium Extrusion at the Mouse Neuromuscular Junction Alkalinizes the Synaptic Cleft. J Neurosci 2023; 43:5741-5752. [PMID: 37474311 PMCID: PMC10423045 DOI: 10.1523/jneurosci.0815-23.2023] [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: 05/04/2023] [Revised: 07/07/2023] [Accepted: 07/14/2023] [Indexed: 07/22/2023] Open
Abstract
Neurotransmission is shaped by extracellular pH. Alkalization enhances pH-sensitive transmitter release and receptor activation, whereas acidification inhibits these processes and can activate acid-sensitive conductances in the synaptic cleft. Previous work has shown that the synaptic cleft can either acidify because of synaptic vesicular release and/or alkalize because of Ca2+ extrusion by the plasma membrane ATPase (PMCA). The direction of change differs across synapse types. At the mammalian neuromuscular junction (NMJ), the direction and magnitude of pH transients in the synaptic cleft during transmission remain ambiguous. We set out to elucidate the extracellular pH transients that occur at this cholinergic synapse under near-physiological conditions and identify their sources. We monitored pH-dependent changes in the synaptic cleft of the mouse levator auris longus using viral expression of the pseudoratiometric probe pHusion-Ex in the muscle. Using mice from both sexes, a significant and prolonged alkalization occurred when stimulating the connected nerve for 5 s at 50 Hz, which was dependent on postsynaptic intracellular Ca2+ release. Sustained stimulation for a longer duration (20 s at 50 Hz) caused additional prolonged net acidification at the cleft. To investigate the mechanism underlying cleft alkalization, we used muscle-expressed GCaMP3 to monitor the contribution of postsynaptic Ca2+ Activity-induced liberation of intracellular Ca2+ in muscle positively correlated with alkalization of the synaptic cleft, whereas inhibiting PMCA significantly decreased the extent of cleft alkalization. Thus, cholinergic synapses of the mouse NMJ typically alkalize because of cytosolic Ca2+ liberated in muscle during activity, unless under highly strenuous conditions where acidification predominates.SIGNIFICANCE STATEMENT Changes in synaptic cleft pH alter neurotransmission, acting on receptors and channels on both sides of the synapse. Synaptic acidification has been associated with a myriad of diseases in the central and peripheral nervous system. Here, we report that in near-physiological recording conditions the cholinergic neuromuscular junction shows use-dependent bidirectional changes in synaptic cleft pH-immediate alkalinization and a long-lasting acidification under prolonged stimulation. These results provide further insight into physiologically relevant changes at cholinergic synapses that have not been defined previously. Understanding and identifying synaptic pH transients during and after neuronal activity provides insight into short-term synaptic plasticity synapses and may identify therapeutic targets for diseases.
Collapse
Affiliation(s)
- Ryan J Durbin
- Integrative Neuroscience Graduate Program, University of Nevada, Reno, Reno, Nevada 89557
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, Nevada 89557
| | - Dante J Heredia
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, Nevada 89557
| | - Thomas W Gould
- Integrative Neuroscience Graduate Program, University of Nevada, Reno, Reno, Nevada 89557
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, Nevada 89557
| | - Robert B Renden
- Integrative Neuroscience Graduate Program, University of Nevada, Reno, Reno, Nevada 89557
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, Nevada 89557
| |
Collapse
|
12
|
Gao Y, Khan YA, Mo W, White KI, Perkins M, Pfuetzner RA, Trapani JG, Brunger AT, Nicolson T. Sensory deficit screen identifies nsf mutation that differentially affects SNARE recycling and quality control. Cell Rep 2023; 42:112345. [PMID: 37027300 PMCID: PMC10524599 DOI: 10.1016/j.celrep.2023.112345] [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: 11/15/2022] [Revised: 02/24/2023] [Accepted: 03/21/2023] [Indexed: 04/08/2023] Open
Abstract
The AAA+ NSF complex is responsible for SNARE complex disassembly both before and after membrane fusion. Loss of NSF function results in pronounced developmental and degenerative defects. In a genetic screen for sensory deficits in zebrafish, we identified a mutation in nsf, I209N, that impairs hearing and balance in a dosage-dependent manner without accompanying defects in motility, myelination, and innervation. In vitro experiments demonstrate that while the I209N NSF protein recognizes SNARE complexes, the effects on disassembly are dependent upon the type of SNARE complex and I209N concentration. Higher levels of I209N protein produce a modest decrease in binary (syntaxin-SNAP-25) SNARE complex disassembly and residual ternary (syntaxin-1A-SNAP-25-synaptobrevin-2) disassembly, whereas at lower concentrations binary disassembly activity is strongly reduced and ternary disassembly activity is absent. Our study suggests that the differential effect on disassembly of SNARE complexes leads to selective effects on NSF-mediated membrane trafficking and auditory/vestibular function.
Collapse
Affiliation(s)
- Yan Gao
- Department of Otolaryngology, Head and Neck Surgery, Stanford Medical School, 300 Pasteur Drive, Stanford, CA 94303, USA
| | - Yousuf A Khan
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA; Department of Structural Biology, Stanford University, Stanford, CA, USA; Department of Photon Science, Stanford University, Stanford, CA, USA; Center for Biomedical Informatics Research, Stanford University, Stanford, CA, USA
| | - Weike Mo
- Graduate Program Biomedical Sciences, Oregon Hearing Research Center and Vollum Institute, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - K Ian White
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA; Department of Structural Biology, Stanford University, Stanford, CA, USA; Department of Photon Science, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Matthew Perkins
- Department of Biology and Neuroscience Program, Amherst College, Amherst, MA 01002, USA
| | - Richard A Pfuetzner
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA; Department of Structural Biology, Stanford University, Stanford, CA, USA; Department of Photon Science, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Josef G Trapani
- Department of Biology and Neuroscience Program, Amherst College, Amherst, MA 01002, USA
| | - Axel T Brunger
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA; Department of Structural Biology, Stanford University, Stanford, CA, USA; Department of Photon Science, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Teresa Nicolson
- Department of Otolaryngology, Head and Neck Surgery, Stanford Medical School, 300 Pasteur Drive, Stanford, CA 94303, USA.
| |
Collapse
|
13
|
Frederick CE, Zenisek D. Ribbon Synapses and Retinal Disease: Review. Int J Mol Sci 2023; 24:5090. [PMID: 36982165 PMCID: PMC10049380 DOI: 10.3390/ijms24065090] [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: 10/28/2022] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/30/2023] Open
Abstract
Synaptic ribbons are presynaptic protein complexes that are believed to be important for the transmission of sensory information in the visual system. Ribbons are selectively associated with those synapses where graded changes in membrane potential drive continuous neurotransmitter release. Defective synaptic transmission can arise as a result of the mutagenesis of a single ribbon component. Visual diseases that stem from malfunctions in the presynaptic molecular machinery of ribbon synapses in the retina are rare. In this review, we provide an overview of synaptopathies that give rise to retinal malfunction and our present understanding of the mechanisms that underlie their pathogenesis and discuss muscular dystrophies that exhibit ribbon synapse involvement in the pathology.
Collapse
Affiliation(s)
| | - David Zenisek
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, 333 Cedar Street, P.O. Box 208026, New Haven, CT 06510, USA
| |
Collapse
|
14
|
Calcium signaling and genetic rare diseases: An auditory perspective. Cell Calcium 2023; 110:102702. [PMID: 36791536 DOI: 10.1016/j.ceca.2023.102702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/07/2023]
Abstract
Deafness is a highly heterogeneous disorder which stems, for 50%, from genetic origins. Sensory transduction relies mainly on sensory hair cells of the cochlea, in the inner ear. Calcium is key for the function of these cells and acts as a fundamental signal transduction. Its homeostasis depends on three factors: the calcium influx, through the mechanotransduction channel at the apical pole of the hair cell as well as the voltage-gated calcium channel at the base of the cells; the calcium buffering via Ca2+-binding proteins in the cytoplasm, but also in organelles such as mitochondria and the reticulum endoplasmic mitochondria-associated membranes with specialized proteins; and the calcium extrusion through the Ca-ATPase pump, located all over the plasma membrane. In addition, the synaptic transmission to the central nervous system is also controlled by calcium. Genetic studies of inherited deafness have tremendously helped understand the underlying molecular pathways of calcium signaling. In this review, we discuss these different factors in light of the associated genetic diseases (syndromic and non-syndromic deafness) and the causative genes.
Collapse
|
15
|
O'Sullivan JDB, Bullen A, Mann ZF. Mitochondrial form and function in hair cells. Hear Res 2023; 428:108660. [PMID: 36525891 DOI: 10.1016/j.heares.2022.108660] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/07/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022]
Abstract
Hair cells (HCs) are specialised sensory receptors residing in the neurosensory epithelia of inner ear sense organs. The precise morphological and physiological properties of HCs allow us to perceive sound and interact with the world around us. Mitochondria play a significant role in normal HC function and are also intricately involved in HC death. They generate ATP essential for sustaining the activity of ion pumps, Ca2+ transporters and the integrity of the stereociliary bundle during transduction as well as regulating cytosolic calcium homoeostasis during synaptic transmission. Advances in imaging techniques have allowed us to study mitochondrial populations throughout the HC, and how they interact with other organelles. These analyses have identified distinct mitochondrial populations between the apical and basolateral portions of the HC, in which mitochondrial morphology appears determined by the physiological processes in the different cellular compartments. Studies in HCs across species show that ototoxic agents, ageing and noise damage directly impact mitochondrial structure and function resulting in HC death. Deciphering the molecular mechanisms underlying this mitochondrial sensitivity, and how their morphology relates to their function during HC death, requires that we first understand this relationship in the context of normal HC function.
Collapse
Affiliation(s)
- James D B O'Sullivan
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral, Craniofacial Sciences, King's College London, London SE1 9RT, U.K
| | - Anwen Bullen
- UCL Ear Institute, University College London, London WC1×8EE, U.K.
| | - Zoë F Mann
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral, Craniofacial Sciences, King's College London, London SE1 9RT, U.K.
| |
Collapse
|
16
|
The effect of time regime in noise exposure on the auditory system and behavioural stress in the zebrafish. Sci Rep 2022; 12:15353. [PMID: 36097161 PMCID: PMC9468136 DOI: 10.1038/s41598-022-19573-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 08/31/2022] [Indexed: 11/29/2022] Open
Abstract
Anthropogenic noise of variable temporal patterns is increasing in aquatic environments, causing physiological stress and sensory impairment. However, scarce information exists on exposure effects to continuous versus intermittent disturbances, which is critical for noise sustainable management. We tested the effects of different noise regimes on the auditory system and behaviour in the zebrafish (Danio rerio). Adult zebrafish were exposed for 24 h to either white noise (150 ± 10 dB re 1 μPa) or silent control. Acoustic playbacks varied in temporal patterns—continuous, fast and slow regular intermittent, and irregular intermittent. Auditory sensitivity was assessed with Auditory Evoked Potential recordings, revealing hearing loss and increased response latency in all noise-treated groups. The highest mean threshold shifts (c. 13 dB) were registered in continuous and fast intermittent treatments, and no differences were found between regular and irregular regimes. Inner ear saccule did not reveal significant hair cell loss but showed a decrease in presynaptic Ribeye b protein especially after continuous exposure. Behavioural assessment using the standardized Novel Tank Diving assay showed that all noise-treated fish spent > 98% time in the bottom within the first minute compared to 82% in control, indicating noise-induced anxiety/stress. We provide first data on how different noise time regimes impact a reference fish model, suggesting that overall acoustic energy is more important than regularity when predicting noise effects.
Collapse
|
17
|
Hou S, Zhang J, Wu Y, Junmin C, Yuyu H, He B, Yang Y, Hong Y, Chen J, Yang J, Li S. FGF22 deletion causes hidden hearing loss by affecting the function of inner hair cell ribbon synapses. Front Mol Neurosci 2022; 15:922665. [PMID: 35966010 PMCID: PMC9366910 DOI: 10.3389/fnmol.2022.922665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 07/04/2022] [Indexed: 12/04/2022] Open
Abstract
Ribbon synapses are important structures in transmitting auditory signals from the inner hair cells (IHCs) to their corresponding spiral ganglion neurons (SGNs). Over the last few decades, deafness has been primarily attributed to the deterioration of cochlear hair cells rather than ribbon synapses. Hearing dysfunction that cannot be detected by the hearing threshold is defined as hidden hearing loss (HHL). The relationship between ribbon synapses and FGF22 deletion remains unknown. In this study, we used a 6-week-old FGF22 knockout mice model (Fgf22–/–) and mainly focused on alteration in ribbon synapses by applying the auditory brainstem response (ABR) test, the immunofluorescence staining, the patch-clamp recording, and quantitative real-time PCR. In Fgf22–/– mice, we found the decreased amplitude of ABR wave I, the reduced vesicles of ribbon synapses, and the decreased efficiency of exocytosis, which was suggested by a decrease in the capacitance change. Quantitative real-time PCR revealed that Fgf22–/– led to dysfunction in ribbon synapses by downregulating SNAP-25 and Gipc3 and upregulating MEF2D expression, which was important for the maintenance of ribbon synapses’ function. Our research concluded that FGF22 deletion caused HHL by affecting the function of IHC ribbon synapses and may offer a novel therapeutic target to meet an ever-growing demand for deafness treatment.
Collapse
Affiliation(s)
- Shule Hou
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Jifang Zhang
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Yan Wu
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Chen Junmin
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Huang Yuyu
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Baihui He
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Yan Yang
- Liaoning Medical Device Test Institute, Shenyang, China
| | - Yuren Hong
- Laboratory of Electron Microscope Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiarui Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Shanghai Children’s Hospital, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Jiarui Chen,
| | - Jun Yang
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
- Jun Yang,
| | - Shuna Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
- Shuna Li,
| |
Collapse
|
18
|
Spaiardi P, Marcotti W, Masetto S, Johnson SL. Signal transmission in mature mammalian vestibular hair cells. Front Cell Neurosci 2022; 16:806913. [PMID: 35936492 PMCID: PMC9353129 DOI: 10.3389/fncel.2022.806913] [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: 11/01/2021] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
The maintenance of balance and gaze relies on the faithful and rapid signaling of head movements to the brain. In mammals, vestibular organs contain two types of sensory hair cells, type-I and type-II, which convert the head motion-induced movement of their hair bundles into a graded receptor potential that drives action potential activity in their afferent fibers. While signal transmission in both hair cell types involves Ca2+-dependent quantal release of glutamate at ribbon synapses, type-I cells appear to also exhibit a non-quantal mechanism that is believed to increase transmission speed. However, the reliance of mature type-I hair cells on non-quantal transmission remains unknown. Here we investigated synaptic transmission in mammalian utricular hair cells using patch-clamp recording of Ca2+ currents and changes in membrane capacitance (ΔCm). We found that mature type-II hair cells showed robust exocytosis with a high-order dependence on Ca2+ entry. By contrast, exocytosis was approximately 10 times smaller in type-I hair cells. Synaptic vesicle exocytosis was largely absent in mature vestibular hair cells of CaV1.3 (CaV1.3−/−) and otoferlin (Otof−/−) knockout mice. Even though Ca2+-dependent exocytosis was small in type-I hair cells of wild-type mice, or absent in CaV1.3−/− and Otof−/−mice, these cells were able to drive action potential activity in the postsynaptic calyces. This supports a functional role for non-quantal synaptic transmission in type-I cells. The large vesicle pools in type-II cells would facilitate sustained transmission of tonic or low-frequency signals. In type-I cells, the restricted vesicle pool size, together with a rapid non-quantal mechanism, could allow them to sustain high-frequency phasic signal transmission at their specialized large calyceal synapses.
Collapse
Affiliation(s)
- Paolo Spaiardi
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Walter Marcotti
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
- Sheffield Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom
| | - Sergio Masetto
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Stuart L. Johnson
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
- Sheffield Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom
- *Correspondence: Stuart L. Johnson
| |
Collapse
|
19
|
Hickox AE, Valero MD, McLaughlin JT, Robinson GS, Wellman JA, McKenna MJ, Sewell WF, Simons EJ. Genetic Medicine for Hearing Loss: OTOF as Exemplar. J Am Acad Audiol 2022; 32:646-653. [PMID: 35609591 DOI: 10.1055/s-0041-1730410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Millions of people worldwide have disabling hearing loss because one of their genes generates an incorrect version of some specific protein the ear requires for hearing. In many of these cases, delivering the correct version of the gene to a specific target cell within the inner ear has the potential to restore cochlear function to enable high-acuity physiologic hearing. Purpose: In this review, we outline our strategy for the development of genetic medicines with the potential to treat hearing loss. We will use the example of otoferlin gene (OTOF)-mediated hearing loss, a sensorineural hearing loss due to autosomal recessive mutations of the OTOF gene.
Collapse
Affiliation(s)
| | | | | | | | | | | | - William F Sewell
- Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|
20
|
Ripley S, Xia L, Zhang Z, Aiken SJ, Wang J. Animal-to-Human Translation Difficulties and Problems With Proposed Coding-in-Noise Deficits in Noise-Induced Synaptopathy and Hidden Hearing Loss. Front Neurosci 2022; 16:893542. [PMID: 35720689 PMCID: PMC9199355 DOI: 10.3389/fnins.2022.893542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/22/2022] [Indexed: 12/26/2022] Open
Abstract
Noise induced synaptopathy (NIS) and hidden hearing loss (NIHHL) have been hot topic in hearing research since a massive synaptic loss was identified in CBA mice after a brief noise exposure that did not cause permanent threshold shift (PTS) in 2009. Based upon the amount of synaptic loss and the bias of it to synapses with a group of auditory nerve fibers (ANFs) with low spontaneous rate (LSR), coding-in-noise deficit (CIND) has been speculated as the major difficult of hearing in subjects with NIS and NIHHL. This speculation is based upon the idea that the coding of sound at high level against background noise relies mainly on the LSR ANFs. However, the translation from animal data to humans for NIS remains to be justified due to the difference in noise exposure between laboratory animals and human subjects in real life, the lack of morphological data and reliable functional methods to quantify or estimate the loss of the afferent synapses by noise. Moreover, there is no clear, robust data revealing the CIND even in animals with the synaptic loss but no PTS. In humans, both positive and negative reports are available. The difficulty in verifying CINDs has led a re-examination of the hypothesis that CIND is the major deficit associated with NIS and NIHHL, and the theoretical basis of this idea on the role of LSR ANFs. This review summarized the current status of research in NIS and NIHHL, with focus on the translational difficulty from animal data to human clinicals, the technical difficulties in quantifying NIS in humans, and the problems with the SR theory on signal coding. Temporal fluctuation profile model was discussed as a potential alternative for signal coding at high sound level against background noise, in association with the mechanisms of efferent control on the cochlea gain.
Collapse
Affiliation(s)
- Sara Ripley
- School of Communication Sciences and Disorders, Dalhousie University, Halifax, NS, Canada
| | - Li Xia
- Department of Otolaryngology-Head and Neck Surgery, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Zhen Zhang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai, China
| | - Steve J. Aiken
- School of Communication Sciences and Disorders, Dalhousie University, Halifax, NS, Canada
| | - Jian Wang
- School of Communication Sciences and Disorders, Dalhousie University, Halifax, NS, Canada
- Department of Otolaryngology-Head and Neck Surgery, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| |
Collapse
|
21
|
Liu Z, Luo Y, Guo R, Yang B, Shi L, Sun J, Guo W, Gong S, Jiang X, Liu K. Head and Neck Radiotherapy Causes Significant Disruptions of Cochlear Ribbon Synapses and Consequent Sensorineural Hearing Loss. Radiother Oncol 2022; 173:207-214. [DOI: 10.1016/j.radonc.2022.05.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 05/13/2022] [Accepted: 05/25/2022] [Indexed: 10/18/2022]
|
22
|
Deng X, Hu Z. Hearing Recovery Induced by DNA Demethylation in a Chemically Deafened Adult Mouse Model. Front Cell Neurosci 2022; 16:792089. [PMID: 35250483 PMCID: PMC8891629 DOI: 10.3389/fncel.2022.792089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 01/04/2022] [Indexed: 11/13/2022] Open
Abstract
Functional hair cell regeneration in the adult mammalian inner ear remains challenging. This study aimed to study the function of new hair cells induced by a DNA demethylating agent 5-azacytidine. Adult mice were deafened chemically, followed by injection of 5-azacytidine or vehicle into the inner ear. Functionality of regenerated hair cells was evaluated by expression of hair cell proteins, auditory brainstem response (ABR), and distortion-product otoacoustic emission (DPOAE) tests for 6 weeks. In the vehicle-treated group, no cells expressed the hair cell-specific protein myosin VIIa in the cochlea, whereas numerous myosin VIIa-expressing cells were found in the 5-azacytidine-treated cochlea, suggesting the regeneration of auditory hair cells. Moreover, regenerated hair cells were co-labeled with functional proteins espin and prestin. Expression of ribbon synapse proteins suggested synapse formation between new hair cells and neurons. In hearing tests, progressive improvements in ABR [5–30 dB sound pressure level (SPL)] and DPOAE (5–20 dB) thresholds were observed in 5-azacytidine-treated mice. In vehicle-treated mice, there were <5 dB threshold changes in hearing tests. This study demonstrated the ability of 5-azacytidine to promote the functional regeneration of auditory hair cells in a mature mouse model via DNA demethylation, which may provide insights into hearing regeneration using an epigenetic approach.
Collapse
Affiliation(s)
- Xin Deng
- Department of Otolaryngology-Head and Neck Surgery (HNS), Wayne State University School of Medicine, Detroit, MI, United States
| | - Zhengqing Hu
- Department of Otolaryngology-Head and Neck Surgery (HNS), Wayne State University School of Medicine, Detroit, MI, United States
- John D. Dingell VA Medical Center, Detroit, MI, United States
- *Correspondence: Zhengqing Hu,
| |
Collapse
|
23
|
Henry KS. Animal models of hidden hearing loss: Does auditory-nerve-fiber loss cause real-world listening difficulties? Mol Cell Neurosci 2022; 118:103692. [PMID: 34883241 PMCID: PMC8928575 DOI: 10.1016/j.mcn.2021.103692] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 01/03/2023] Open
Abstract
Afferent innervation of the cochlea by the auditory nerve declines during aging and potentially after sound overexposure, producing the common pathology known as cochlear synaptopathy. Auditory-nerve-fiber loss is difficult to detect with the clinical audiogram and has been proposed to cause 'hidden hearing loss' including impaired speech-in-noise perception. While evidence that auditory-nerve-fiber loss causes hidden hearing loss in humans is controversial, behavioral animal models hold promise to rigorously test this hypothesis because neural lesions can be induced and histologically validated. Here, we review recent animal behavioral studies on the impact of auditory-nerve-fiber loss on perception in a range of species. We first consider studies of tinnitus and hyperacusis inferred from acoustic startle reflexes, followed by a review of operant-conditioning studies of the audiogram, temporal integration for tones of varying duration, temporal resolution of gaps in noise, and tone-in-noise detection. Studies quantifying the audiogram show that tone-in-quiet sensitivity is unaffected by auditory-nerve-fiber loss unless neural lesions exceed 80%, at which point large deficits are possible. Changes in other aspects of perception, which were typically investigated for moderate-to-severe auditory-nerve-fiber loss of 50-70%, appear heterogeneous across studies and might be small compared to impairment caused by hair-cell pathologies. Future studies should pursue recent findings that behavioral sensitivity to brief tones and silent gaps in noise may be particularly vulnerable to auditory-nerve-fiber loss. Furthermore, aspects of auditory perception linked to central inhibition and fine neural response timing, such as modulation masking release and spatial hearing, may be productive directions for further animal behavioral research.
Collapse
Affiliation(s)
- Kenneth S Henry
- Departments of Otolaryngology, Biomedical Engineering, and Neuroscience, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
24
|
Suthakar K, Liberman MC. Auditory-nerve responses in mice with noise-induced cochlear synaptopathy. J Neurophysiol 2021; 126:2027-2038. [PMID: 34788179 DOI: 10.1152/jn.00342.2021] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [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
|
25
|
Lee J, Kawai K, Holt JR, Géléoc GSG. Sensory transduction is required for normal development and maturation of cochlear inner hair cell synapses. eLife 2021; 10:e69433. [PMID: 34734805 PMCID: PMC8598158 DOI: 10.7554/elife.69433] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 11/01/2021] [Indexed: 12/23/2022] Open
Abstract
Acoustic overexposure and aging can damage auditory synapses in the inner ear by a process known as synaptopathy. These insults may also damage hair bundles and the sensory transduction apparatus in auditory hair cells. However, a connection between sensory transduction and synaptopathy has not been established. To evaluate potential contributions of sensory transduction to synapse formation and development, we assessed inner hair cell synapses in several genetic models of dysfunctional sensory transduction, including mice lacking transmembrane channel-like (Tmc) 1, Tmc2, or both, in Beethoven mice which carry a dominant Tmc1 mutation and in Spinner mice which carry a recessive mutation in transmembrane inner ear (Tmie). Our analyses reveal loss of synapses in the absence of sensory transduction and preservation of synapses in Tmc1-null mice following restoration of sensory transduction via Tmc1 gene therapy. These results provide insight into the requirement of sensory transduction for hair cell synapse development and maturation.
Collapse
Affiliation(s)
- John Lee
- Speech and Hearing Bioscience & Technology Program, Division of Medical Sciences, Harvard UniversityBostonUnited States
- Department of Otolaryngology, Boston Children’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Kosuke Kawai
- Department of Otolaryngology, Boston Children’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Jeffrey R Holt
- Department of Otolaryngology, Boston Children’s Hospital and Harvard Medical SchoolBostonUnited States
- Department of Neurology, Boston Children’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Gwenaëlle SG Géléoc
- Department of Otolaryngology, Boston Children’s Hospital and Harvard Medical SchoolBostonUnited States
| |
Collapse
|
26
|
Caus Capdevila MQ, Sienknecht UJ, Köppl C. Developmental maturation of presynaptic ribbon numbers in chicken basilar-papilla hair cells and its perturbation by long-term overexpression of Wnt9a. Dev Neurobiol 2021; 81:817-832. [PMID: 34309221 DOI: 10.1002/dneu.22845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/20/2021] [Accepted: 07/15/2021] [Indexed: 11/07/2022]
Abstract
The avian basilar papilla is a valuable model system for exploring the developmental determination and differentiation of sensory hair cells and their innervation. In the mature basilar papilla, hair cells form a well-known continuum between two extreme types-tall and short hair cells-that differ strikingly in their innervation. Previous work identified Wnt9a as a crucial factor in this differentiation. Here, we quantified the number and volume of immunolabelled presynaptic ribbons in tall and short hair cells of chickens, from developmental stages shortly after ribbons first appear to the mature posthatching condition. Two longitudinal locations were sampled, responding to best frequencies of approximately 1 kHz and approximately 5.5 kHz when mature. We found significant reductions of ribbon number during normal development in the tall-hair-cell domains, but stable, low numbers in the short-hair-cell domains. Exposing developing hair cells to continuous, excessive Wnt9a levels (through virus-mediated overexpression) led to transiently abnormal high numbers of ribbons and a delayed reduction of ribbon numbers at all sampled locations. Thus, (normally) short-hair-cell domains also showed tall-hair-cell like behaviour, confirming previous findings (Munnamalai et al., 2017). However, at 3 weeks posthatching, ribbon numbers had decreased to the location-specific typical values of control hair cells at all sampled locations. Furthermore, as shown previously, mature hair cells at the basal, high-frequency location harboured larger ribbons than more apically located hair cells. This was true for both normal and Wnt9a-overexposed basilar papillae.
Collapse
Affiliation(s)
- M Queralt Caus Capdevila
- Cluster of Excellence "Hearing4all" and Research Centre Neurosensory Science, Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Ulrike J Sienknecht
- Cluster of Excellence "Hearing4all" and Research Centre Neurosensory Science, Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Christine Köppl
- Cluster of Excellence "Hearing4all" and Research Centre Neurosensory Science, Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
27
|
A convolutional neural-network framework for modelling auditory sensory cells and synapses. Commun Biol 2021; 4:827. [PMID: 34211095 PMCID: PMC8249591 DOI: 10.1038/s42003-021-02341-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 06/09/2021] [Indexed: 12/02/2022] Open
Abstract
In classical computational neuroscience, analytical model descriptions are derived from neuronal recordings to mimic the underlying biological system. These neuronal models are typically slow to compute and cannot be integrated within large-scale neuronal simulation frameworks. We present a hybrid, machine-learning and computational-neuroscience approach that transforms analytical models of sensory neurons and synapses into deep-neural-network (DNN) neuronal units with the same biophysical properties. Our DNN-model architecture comprises parallel and differentiable equations that can be used for backpropagation in neuro-engineering applications, and offers a simulation run-time improvement factor of 70 and 280 on CPU or GPU systems respectively. We focussed our development on auditory neurons and synapses, and show that our DNN-model architecture can be extended to a variety of existing analytical models. We describe how our approach for auditory models can be applied to other neuron and synapse types to help accelerate the development of large-scale brain networks and DNN-based treatments of the pathological system. Drakopoulos et al developed a machine-learning and computational-neuroscience approach that transforms analytical models of sensory neurons and synapses into deep-neural-network (DNN) neuronal units with the same biophysical properties. Focusing on auditory neurons and synapses, they showed that their DNN-model architecture could be extended to a variety of existing analytical models and to other neuron and synapse types, thus potentially assisting the development of large-scale brain networks and DNN-based treatments.
Collapse
|
28
|
Abstract
Congenital hearing loss is the most common birth defect, estimated to affect 2-3 in every 1000 births. Currently there is no cure for hearing loss. Treatment options are limited to hearing aids for mild and moderate cases, and cochlear implants for severe and profound hearing loss. Here we provide a literature overview of the environmental and genetic causes of congenital hearing loss, common animal models and methods used for hearing research, as well as recent advances towards developing therapies to treat congenital deafness. © 2021 The Authors.
Collapse
Affiliation(s)
- Justine M Renauld
- Department of Otolaryngology, Head & Neck Surgery, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Martin L Basch
- Department of Otolaryngology, Head & Neck Surgery, Case Western Reserve University School of Medicine, Cleveland, Ohio.,Department of Genetics and Genome Sciences, Case Western Reserve School of Medicine, Cleveland, Ohio.,Department of Biology, Case Western Reserve University, Cleveland, Ohio.,Department of Otolaryngology, Head & Neck Surgery, University Hospitals, Cleveland, Ohio
| |
Collapse
|
29
|
Ghosh S, Stansak K, Walters BJ. Cannabinoid Signaling in Auditory Function and Development. Front Mol Neurosci 2021; 14:678510. [PMID: 34079440 PMCID: PMC8165240 DOI: 10.3389/fnmol.2021.678510] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/15/2021] [Indexed: 12/20/2022] Open
Abstract
Plants of the genus Cannabis have been used by humans for millennia for a variety of purposes. Perhaps most notable is the use of certain Cannabis strains for their psychoactive effects. More recently, several biologically active molecules within the plants of these Cannabis strains, called phytocannabinoids or simply cannabinoids, have been identified. Furthermore, within human cells, endogenous cannabinoids, or endocannabinoids, as well as the receptors and secondary messengers that give rise to their neuromodulatory effects, have also been characterized. This endocannabinoid system (ECS) is composed of two primary ligands-anandamide and 2-arachidonyl glycerol; two primary receptors-cannabinoid receptors 1 and 2; and several enzymes involved in biosynthesis and degradation of endocannabinoid ligands including diacylglycerol lipase (DAGL) and monoacylglycerol lipase (MAGL). Here we briefly summarize cannabinoid signaling and review what has been discerned to date with regard to cannabinoid signaling in the auditory system and its roles in normal physiological function as well as pathological conditions. While much has been uncovered regarding cannabinoid signaling in the central nervous system, less attention has been paid to the auditory system specifically. Still, evidence is emerging to suggest that cannabinoid signaling is critical for the development, maturation, function, and survival of cochlear hair cells (HCs) and spiral ganglion neurons (SGNs). Furthermore, cannabinoid signaling can have profound effects on synaptic connectivity in CNS structures related to auditory processing. While clinical cases demonstrate that endogenous and exogenous cannabinoids impact auditory function, this review highlights several areas, such as SGN development, where more research is warranted.
Collapse
Affiliation(s)
- Sumana Ghosh
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, United States
| | - Kendra Stansak
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, United States
| | - Bradley J Walters
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, United States.,Department of Otolaryngology-Head and Neck Surgery, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
30
|
Disruption of the autism-related gene Pak1 causes stereocilia disorganization, hair cell loss, and deafness in mice. J Genet Genomics 2021; 48:324-332. [PMID: 34049799 DOI: 10.1016/j.jgg.2021.03.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/18/2021] [Accepted: 03/12/2021] [Indexed: 01/06/2023]
Abstract
Several clinical studies have reported that hearing loss is correlated with autism in children. However, little is known about the underlying mechanism between hearing loss and autism. p21-activated kinases (PAKs) are a family of serine/threonine kinases that can be activated by multiple signaling molecules, particularly the Rho family of small GTPases. Previous studies have shown that Pak1 mutations are associated with autism. In the present study, we take advantage of Pak1 knockout (Pak1-/-) mice to investigate the role of PAK1 in hearing function. We find that PAK1 is highly expressed in the postnatal mouse cochlea and that PAK1 deficiency leads to hair cell (HC) apoptosis and severe hearing loss. Further investigation indicates that PAK1 deficiency downregulates the phosphorylation of cofilin and ezrin-radixin-moesin and the expression of βII-spectrin, which further decreases the HC synapse density in the basal turn of cochlea and disorganized the HC stereocilia in all three turns of cochlea in Pak1-/- mice. Overall, our work demonstrates that the autism-related gene Pak1 plays a crucial role in hearing function. As the first candidate gene linking autism and hearing loss, Pak1 may serve as a potential target for the clinical diagnosis of autism-related hearing loss.
Collapse
|
31
|
Colón-Cruz L, Rodriguez-Morales R, Santana-Cruz A, Cantres-Velez J, Torrado-Tapias A, Lin SJ, Yudowski G, Kensler R, Marie B, Burgess SM, Renaud O, Varshney GK, Behra M. Cnr2 Is Important for Ribbon Synapse Maturation and Function in Hair Cells and Photoreceptors. Front Mol Neurosci 2021; 14:624265. [PMID: 33958989 PMCID: PMC8093779 DOI: 10.3389/fnmol.2021.624265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/24/2021] [Indexed: 02/04/2023] Open
Abstract
The role of the cannabinoid receptor 2 (CNR2) is still poorly described in sensory epithelia. We found strong cnr2 expression in hair cells (HCs) of the inner ear and the lateral line (LL), a superficial sensory structure in fish. Next, we demonstrated that sensory synapses in HCs were severely perturbed in larvae lacking cnr2. Appearance and distribution of presynaptic ribbons and calcium channels (Cav1.3) were profoundly altered in mutant animals. Clustering of membrane-associated guanylate kinase (MAGUK) in post-synaptic densities (PSDs) was also heavily affected, suggesting a role for cnr2 for maintaining the sensory synapse. Furthermore, vesicular trafficking in HCs was strongly perturbed suggesting a retrograde action of the endocannabinoid system (ECs) via cnr2 that was modulating HC mechanotransduction. We found similar perturbations in retinal ribbon synapses. Finally, we showed that larval swimming behaviors after sound and light stimulations were significantly different in mutant animals. Thus, we propose that cnr2 is critical for the processing of sensory information in the developing larva.
Collapse
Affiliation(s)
- Luis Colón-Cruz
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | - Roberto Rodriguez-Morales
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | - Alexis Santana-Cruz
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | - Juan Cantres-Velez
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | - Aranza Torrado-Tapias
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | - Sheng-Jia Lin
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Guillermo Yudowski
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico.,School of Medicine, Institute of Neurobiology, University of Puerto Rico, San Juan, Puerto Rico
| | - Robert Kensler
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | - Bruno Marie
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico.,School of Medicine, Institute of Neurobiology, University of Puerto Rico, San Juan, Puerto Rico
| | - Shawn M Burgess
- Developmental Genomics Section, Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Olivier Renaud
- Cell and Tissue Imaging Facility (PICT-IBiSA, FranceBioImaging), Institut Curie, PSL Research University, U934/UMR3215, Paris, France
| | - Gaurav K Varshney
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Martine Behra
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| |
Collapse
|
32
|
Chen X, Abad C, Chen ZY, Young JI, Gurumurthy CB, Walz K, Liu XZ. Generation and characterization of a P2rx2 V60L mouse model for DFNA41. Hum Mol Genet 2021; 30:985-995. [PMID: 33791800 DOI: 10.1093/hmg/ddab077] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 03/06/2021] [Accepted: 03/12/2021] [Indexed: 11/13/2022] Open
Abstract
P2RX2 encodes the P2X2 receptor, which is an adenosine triphosphate (ATP) gated (purinoreceptor) ion channel. P2RX2 c. 178G > T (p.V60L) mutation was previously identified in two unrelated Chinese families, as the cause of human DFNA41, a form of dominant, early-onset and progressive sensorineural hearing loss. We generated and characterized a knock-in mouse model based on human p.V60L mutation that recapitulates the human phenotype. Heterozygous KI mice started to exhibit hearing loss at 21-day-old and progressed to deafness by 6-month-old. Vestibular dysfunction was also observed in mutant mice. Abnormal morphology of the inner hair cells and ribbon synapses was progressively observed in KI animals suggesting that P2rx2 plays a role in the membrane spatial location of the ribbon synapses. These results suggest that P2rx2 is essential for acoustic information transfer, which can be the molecular mechanism related to hearing loss.
Collapse
Affiliation(s)
- Xiaoya Chen
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL 33136, USA.,Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Clemer Abad
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Zheng-Yi Chen
- Department of Otolaryngology and Program in Neuroscience, Harvard Medical School and Eaton Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts 02114, USA
| | - Juan I Young
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA.,Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Channabasavaiah B Gurumurthy
- Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Katherina Walz
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA.,Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Xue Zhong Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL 33136, USA.,Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
33
|
Liu W, Luque M, Li H, Schrott-Fischer A, Glueckert R, Tylstedt S, Rajan G, Ladak H, Agrawal S, Rask-Andersen H. Spike Generators and Cell Signaling in the Human Auditory Nerve: An Ultrastructural, Super-Resolution, and Gene Hybridization Study. Front Cell Neurosci 2021; 15:642211. [PMID: 33796009 PMCID: PMC8008129 DOI: 10.3389/fncel.2021.642211] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/22/2021] [Indexed: 11/23/2022] Open
Abstract
Background: The human auditory nerve contains 30,000 nerve fibers (NFs) that relay complex speech information to the brain with spectacular acuity. How speech is coded and influenced by various conditions is not known. It is also uncertain whether human nerve signaling involves exclusive proteins and gene manifestations compared with that of other species. Such information is difficult to determine due to the vulnerable, "esoteric," and encapsulated human ear surrounded by the hardest bone in the body. We collected human inner ear material for nanoscale visualization combining transmission electron microscopy (TEM), super-resolution structured illumination microscopy (SR-SIM), and RNA-scope analysis for the first time. Our aim was to gain information about the molecular instruments in human auditory nerve processing and deviations, and ways to perform electric modeling of prosthetic devices. Material and Methods: Human tissue was collected during trans-cochlear procedures to remove petro-clival meningioma after ethical permission. Cochlear neurons were processed for electron microscopy, confocal microscopy (CM), SR-SIM, and high-sensitive in situ hybridization for labeling single mRNA transcripts to detect ion channel and transporter proteins associated with nerve signal initiation and conductance. Results: Transport proteins and RNA transcripts were localized at the subcellular level. Hemi-nodal proteins were identified beneath the inner hair cells (IHCs). Voltage-gated ion channels (VGICs) were expressed in the spiral ganglion (SG) and axonal initial segments (AISs). Nodes of Ranvier (NR) expressed Nav1.6 proteins, and encoding genes critical for inter-cellular coupling were disclosed. Discussion: Our results suggest that initial spike generators are located beneath the IHCs in humans. The first NRs appear at different places. Additional spike generators and transcellular communication may boost, sharpen, and synchronize afferent signals by cell clusters at different frequency bands. These instruments may be essential for the filtering of complex sounds and may be challenged by various pathological conditions.
Collapse
Affiliation(s)
- Wei Liu
- Section of Otolaryngology, Department of Surgical Sciences, Head and Neck Surgery, Uppsala University Hospital, Uppsala, Sweden
| | - Maria Luque
- Department of Otorhinolaryngology, Medical University of Innsbruck, Innsbruck, Austria
| | - Hao Li
- Section of Otolaryngology, Department of Surgical Sciences, Head and Neck Surgery, Uppsala University Hospital, Uppsala, Sweden
| | | | - Rudolf Glueckert
- Department of Otorhinolaryngology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sven Tylstedt
- Department of Olaryngology, Västerviks Hospital, Västervik, Sweden
| | - Gunesh Rajan
- Department of Otolaryngology, Head & Neck Surgery, Luzerner Kantonsspital, Luzern, Switzerland
- Department of Otolaryngology, Head & Neck Surgery, Division of Surgery, Medical School, University of Western Australia, Perth, WA, Australia
| | - Hanif Ladak
- Department of Otolaryngology-Head and Neck Surgery, Department of Medical Biophysics and Department of Electrical and Computer Engineering, Western University, London, ON, Canada
| | - Sumit Agrawal
- Department of Otolaryngology-Head and Neck Surgery, Western University, London, ON, Canada
| | - Helge Rask-Andersen
- Section of Otolaryngology, Department of Surgical Sciences, Head and Neck Surgery, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
34
|
Outer Hair Cell Glutamate Signaling through Type II Spiral Ganglion Afferents Activates Neurons in the Cochlear Nucleus in Response to Nondamaging Sounds. J Neurosci 2021; 41:2930-2943. [PMID: 33574178 DOI: 10.1523/jneurosci.0619-20.2021] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 01/24/2021] [Accepted: 02/04/2021] [Indexed: 11/21/2022] Open
Abstract
Cochlear outer hair cells (OHCs) are known to uniquely participate in auditory processing through their electromotility, and like inner hair cells, are also capable of releasing vesicular glutamate onto spiral ganglion (SG) neurons: in this case, onto the sparse Type II SG neurons. However, unlike glutamate signaling at the inner hair cell-Type I SG neuron synapse, which is robust across a wide spectrum of sound intensities, glutamate signaling at the OHC-Type II SG neuron synapse is weaker and has been hypothesized to occur only at intense, possibly damaging sound levels. Here, we tested the ability of the OHC-Type II SG pathway to signal to the brain in response to moderate, nondamaging sound (80 dB SPL) as well as to intense sound (115 dB SPL). First, we determined the VGluTs associated with OHC signaling and then confirmed the loss of glutamatergic synaptic transmission from OHCs to Type II SG neurons in KO mice using dendritic patch-clamp recordings. Next, we generated genetic mouse lines in which vesicular glutamate release occurs selectively from OHCs, and then assessed c-Fos expression in the cochlear nucleus in response to sound. From these analyses, we show, for the first time, that glutamatergic signaling at the OHC-Type II SG neuron synapse is capable of activating cochlear nucleus neurons, even at moderate sound levels.SIGNIFICANCE STATEMENT Evidence suggests that cochlear outer hair cells (OHCs) release glutamate onto Type II spiral ganglion neurons only when exposed to loud sound, and that Type II neurons are activated by tissue damage. Knowing whether moderate level sound, without tissue damage, activates this pathway has functional implications for this fundamental auditory pathway. We first determined that OHCs rely largely on VGluT3 for synaptic glutamate release. We then used a genetically modified mouse line in which OHCs, but not inner hair cells, release vesicular glutamate to demonstrate that moderate sound exposure activates cochlear nucleus neurons via the OHC-Type II spiral ganglion pathway. Together, these data indicate that glutamate signaling at the OHC-Type II afferent synapse participates in auditory function at moderate sound levels.
Collapse
|
35
|
Brill SE, Maraslioglu A, Kurz C, Kramer F, Fuhr MF, Singh A, Friauf E. Glycinergic Transmission in the Presence and Absence of Functional GlyT2: Lessons From the Auditory Brainstem. Front Synaptic Neurosci 2021; 12:560008. [PMID: 33633558 PMCID: PMC7900164 DOI: 10.3389/fnsyn.2020.560008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 12/01/2020] [Indexed: 12/20/2022] Open
Abstract
Synaptic transmission is controlled by re-uptake systems that reduce transmitter concentrations in the synaptic cleft and recycle the transmitter into presynaptic terminals. The re-uptake systems are thought to ensure cytosolic concentrations in the terminals that are sufficient for reloading empty synaptic vesicles (SVs). Genetic deletion of glycine transporter 2 (GlyT2) results in severely disrupted inhibitory neurotransmission and ultimately to death. Here we investigated the role of GlyT2 at inhibitory glycinergic synapses in the mammalian auditory brainstem. These synapses are tuned for resilience, reliability, and precision, even during sustained high-frequency stimulation when endocytosis and refilling of SVs probably contribute substantially to efficient replenishment of the readily releasable pool (RRP). Such robust synapses are formed between MNTB and LSO neurons (medial nucleus of the trapezoid body, lateral superior olive). By means of patch-clamp recordings, we assessed the synaptic performance in controls, in GlyT2 knockout mice (KOs), and upon acute pharmacological GlyT2 blockade. Via computational modeling, we calculated the reoccupation rate of empty release sites and RRP replenishment kinetics during 60-s challenge and 60-s recovery periods. Control MNTB-LSO inputs maintained high fidelity neurotransmission at 50 Hz for 60 s and recovered very efficiently from synaptic depression. During 'marathon-experiments' (30,600 stimuli in 20 min), RRP replenishment accumulated to 1,260-fold. In contrast, KO inputs featured severe impairments. For example, the input number was reduced to ~1 (vs. ~4 in controls), implying massive functional degeneration of the MNTB-LSO microcircuit and a role of GlyT2 during synapse maturation. Surprisingly, neurotransmission did not collapse completely in KOs as inputs still replenished their small RRP 80-fold upon 50 Hz | 60 s challenge. However, they totally failed to do so for extended periods. Upon acute pharmacological GlyT2 inactivation, synaptic performance remained robust, in stark contrast to KOs. RRP replenishment was 865-fold in marathon-experiments, only ~1/3 lower than in controls. Collectively, our empirical and modeling results demonstrate that GlyT2 re-uptake activity is not the dominant factor in the SV recycling pathway that imparts indefatigability to MNTB-LSO synapses. We postulate that additional glycine sources, possibly the antiporter Asc-1, contribute to RRP replenishment at these high-fidelity brainstem synapses.
Collapse
Affiliation(s)
- Sina E Brill
- Animal Physiology Group, Department of Biology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Ayse Maraslioglu
- Animal Physiology Group, Department of Biology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Catharina Kurz
- Animal Physiology Group, Department of Biology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Florian Kramer
- Animal Physiology Group, Department of Biology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Martin F Fuhr
- Animal Physiology Group, Department of Biology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Abhyudai Singh
- Electrical & Computer Engineering, University of Delaware, Newark, DE, United States
| | - Eckhard Friauf
- Animal Physiology Group, Department of Biology, University of Kaiserslautern, Kaiserslautern, Germany
| |
Collapse
|
36
|
Integrated stress response inhibition provides sex-dependent protection against noise-induced cochlear synaptopathy. Sci Rep 2020; 10:18063. [PMID: 33093490 PMCID: PMC7582887 DOI: 10.1038/s41598-020-75058-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/30/2020] [Indexed: 11/08/2022] Open
Abstract
Noise-induced hearing loss (NIHL) is a common health concern with significant social, psychological, and cognitive implications. Moderate levels of acoustic overstimulation associated with tinnitus and impaired speech perception cause cochlear synaptopathy, characterized physiologically by reduction in wave I of the suprathreshold auditory brainstem response (ABR) and reduced number of synapses between sensory hair cells and auditory neurons. The unfolded protein response (UPR), an endoplasmic reticulum stress response pathway, has been implicated in the pathogenesis and treatment of NIHL as well as neurodegeneration and synaptic damage in the brain. In this study, we used the small molecule UPR modulator Integrated Stress Response InhiBitor (ISRIB) to treat noise-induced cochlear synaptopathy in a mouse model. Mice pretreated with ISRIB prior to noise-exposure were protected against noise-induced synapse loss. Male, but not female, mice also exhibited ISRIB-mediated protection against noise-induced suprathreshold ABR wave-I amplitude reduction. Female mice had higher baseline wave-I amplitudes but greater sensitivity to noise-induced wave-I reduction. Our results suggest that the UPR is implicated in noise-induced cochlear synaptopathy, and can be targeted for treatment.
Collapse
|
37
|
The Genetics of Variation of the Wave 1 Amplitude of the Mouse Auditory Brainstem Response. J Assoc Res Otolaryngol 2020; 21:323-336. [PMID: 32757112 DOI: 10.1007/s10162-020-00762-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 07/19/2020] [Indexed: 12/13/2022] Open
Abstract
This is the first genome-wide association study with the Hybrid Mouse Diversity Panel (HDMP) to define the genetic landscape of the variation in the suprathreshold wave 1 amplitude of the auditory brainstem response (ABR) both pre- and post-noise exposure. This measure is correlated with the density of the auditory neurons (AN) and/or the compliment of synaptic ribbons within the inner hair cells of the mouse cochlea. We analyzed suprathreshold ABR for 635 mice from 102 HMDP strains pre- and post-noise exposure (108 dB 10 kHz octave band noise exposure for 2 h) using auditory brainstem response (ABR) wave 1 suprathreshold amplitudes as part of a large survey (Myint et al., Hear Res 332:113-120, 2016). Genome-wide significance levels for pre- and post-exposure wave 1 amplitude across the HMDP were performed using FaST-LMM. Synaptic ribbon counts (Ctbp2 and mGluR2) were analyzed for the extreme strains within the HMDP. ABR wave 1 amplitude varied across all strains of the HMDP with differences ranging between 2.42 and 3.82-fold pre-exposure and between 2.43 and 7.5-fold post-exposure with several tone burst stimuli (4 kHz, 8 kHz, 12 kHz, 16 kHz, 24 kHz, and 32 kHz). Immunolabeling of paired synaptic ribbons and glutamate receptors of strains with the highest and lowest wave 1 values pre- and post-exposure revealed significant differences in functional synaptic ribbon counts. Genome-wide association analysis identified genome-wide significant threshold associations on chromosome 3 (24 kHz; JAX00105429; p < 1.12E-06) and chromosome 16 (16 kHz; JAX00424604; p < 9.02E-07) prior to noise exposure and significant associations on chromosomes 2 (32 kHz; JAX00497967; p < 3.68E-08) and 13 (8 kHz; JAX00049416; 1.07E-06) after noise exposure. In order to prioritize candidate genes, we generated cis-eQTLs from microarray profiling of RNA isolated from whole cochleae in 64 of the tested strains.This is the first report of a genome-wide association analysis, controlled for population structure, to explore the genetic landscape of suprathreshold wave 1 amplitude measurements of the mouse ABR. We have defined two genomic regions associated with wave 1 amplitude variation prior to noise exposure and an additional two associated with variation after noise exposure.
Collapse
|
38
|
Spaiardi P, Marcotti W, Masetto S, Johnson SL. Exocytosis in mouse vestibular Type II hair cells shows a high-order Ca 2+ dependence that is independent of synaptotagmin-4. Physiol Rep 2020; 8:e14509. [PMID: 32691536 PMCID: PMC7371649 DOI: 10.14814/phy2.14509] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/21/2020] [Accepted: 06/22/2020] [Indexed: 11/24/2022] Open
Abstract
Mature hair cells transduce information over a wide range of stimulus intensities and frequencies for prolonged periods of time. The efficiency of such a demanding task is reflected in the characteristics of exocytosis at their specialized presynaptic ribbons. Ribbons are electron-dense structures able to tether a large number of releasable vesicles allowing them to maintain high rates of vesicle release. Calcium entry through rapidly activating, non-inactivating CaV 1.3 (L-type) Ca2+ channels in response to cell depolarization causes a local increase in Ca2+ at the ribbon synapses, which is detected by the exocytotic Ca2+ sensors. The Ca2+ dependence of vesicle exocytosis at mammalian vestibular hair cell (VHC) ribbon synapses is believed to be linear, similar to that observed in mature cochlear inner hair cells (IHCs). The linear relation has been shown to correlate with the presence of the Ca2+ sensor synaptotagmin-4 (Syt-4). Therefore, we studied the exocytotic Ca2+ dependence, and the release kinetics of different vesicle pool populations, in Type II VHCs of control and Syt-4 knockout mice using patch-clamp capacitance measurements, under physiological recording conditions. We found that exocytosis in mature control and knockout Type II VHCs displayed a high-order dependence on Ca2+ entry, rather than the linear relation previously observed. Consistent with this finding, the Ca2+ dependence and release kinetics of the ready releasable pool (RRP) of vesicles were not affected by an absence of Syt-4. However, we did find that Syt-4 could play a role in regulating the release of the secondary releasable pool (SRP) in these cells. Our findings show that the coupling between Ca2+ influx and neurotransmitter release at mature Type II VHC ribbon synapses is faithfully described by a nonlinear relation that is likely to be more appropriate for the accurate encoding of low-frequency vestibular information, consistent with that observed at low-frequency mammalian auditory receptors.
Collapse
Affiliation(s)
- Paolo Spaiardi
- Department of Brain and Behavioral SciencesUniversity of PaviaPaviaItaly
| | - Walter Marcotti
- Department of Biomedical ScienceUniversity of SheffieldSheffieldUK
| | - Sergio Masetto
- Department of Brain and Behavioral SciencesUniversity of PaviaPaviaItaly
| | | |
Collapse
|
39
|
D’Elia A, Quaranta N, Asprella Libonati G, Ralli G, Morelli A, Inchingolo F, Cialdella F, Martellucci S, Barbara F. The cochleo-vestibular secretory senescence. JOURNAL OF GERONTOLOGY AND GERIATRICS 2020. [DOI: 10.36150/2499-6564-485] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
40
|
Soto J, Castaneda-Villa N, Gil A, Gonzalez-Velez V. Simulation of the efficiency of inner hair cell secretion in the auditory pathway. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2019:2332-2335. [PMID: 31946367 DOI: 10.1109/embc.2019.8857293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Sound coding involves several stages of processing along the auditory path. Specifically, the Inner Hair Cells (IHC) act as sensory receptors and transduce acoustic information -frequency, intensity and duration of the stimulus- into neuronal signals. In this work, a stochastic model was implemented to achieve a better understanding of the IHC-auditory nerve synapse, specifically, the process of Ready Releasable Pool (RRP) vesicle exocytosis, a complicated process to study experimentally because current protocols do not provide adequate temporal resolution, in the order of milliseconds. The presented model allows predicting the efficiency of glutamate release towards explaining maturation changes or disease impacts in the auditory pathway.
Collapse
|
41
|
Okur MN, Mao B, Kimura R, Haraczy S, Fitzgerald T, Edwards-Hollingsworth K, Tian J, Osmani W, Croteau DL, Kelley MW, Bohr VA. Short-term NAD + supplementation prevents hearing loss in mouse models of Cockayne syndrome. NPJ Aging Mech Dis 2020; 6:1. [PMID: 31934345 PMCID: PMC6946667 DOI: 10.1038/s41514-019-0040-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 12/03/2019] [Indexed: 12/13/2022] Open
Abstract
Age-related hearing loss (ARHL) is one of the most common disorders affecting elderly individuals. There is an urgent need for effective preventive measures for ARHL because none are currently available. Cockayne syndrome (CS) is a premature aging disease that presents with progressive hearing loss at a young age, but is otherwise similar to ARHL. There are two human genetic complementation groups of CS, A and B. While the clinical phenotypes in patients are similar, the proteins have very diverse functions, and insight into their convergence is of great interest. Here, we use mouse models for CS (CSA -/- and CSB m/m ) that recapitulate the hearing loss in human CS patients. We previously showed that NAD+, a key metabolite with various essential functions, is reduced in CS and associated with multiple CS phenotypes. In this study, we report that NAD+ levels are reduced in the cochlea of CSB m/m mice and that short-term treatment (10 days) with the NAD+ precursor nicotinamide riboside (NR), prevents hearing loss, restores outer hair cell loss, and improves cochlear health in CSB m/m mice. Similar, but more modest effects were observed in CSA -/- mice. Remarkably, we observed a reduction in synaptic ribbon counts in the presynaptic zones of inner hair cells in both CSA -/- and CSB m/m mice, pointing to a converging mechanism for cochlear defects in CS. Ribbon synapses facilitate rapid and sustained synaptic transmission over long periods of time. Ribeye, a core protein of synaptic ribbons, possesses an NAD(H) binding pocket which regulates its activity. Intriguingly, NAD+ supplementation rescues reduced synaptic ribbon formation in both CSA -/- and CSB m/m mutant cochleae. These findings provide valuable insight into the mechanism of CS- and ARHL-associated hearing loss, and suggest a possible intervention.
Collapse
Affiliation(s)
- Mustafa N. Okur
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224 USA
| | - Beatrice Mao
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892 USA
| | - Risako Kimura
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224 USA
| | - Scott Haraczy
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892 USA
| | - Tracy Fitzgerald
- Mouse Auditory Testing Core, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892 USA
| | - Kamren Edwards-Hollingsworth
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892 USA
| | - Jane Tian
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224 USA
| | - Wasif Osmani
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224 USA
| | - Deborah L. Croteau
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224 USA
| | - Matthew W. Kelley
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892 USA
| | - Vilhelm A. Bohr
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224 USA
- Danish Center for Healthy Aging, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| |
Collapse
|
42
|
Abstract
Auditory processing in the cochlea depends on the integrity of the mechanosensory hair cells. Over a lifetime, hearing loss can be acquired from numerous etiologies such as exposure to excessive noise, the use of ototoxic medications, bacterial or viral ear infections, head injuries, and the aging process. Loss of sensory hair cells is a common pathological feature of the varieties of acquired hearing loss. Additionally, the inner hair cell synapse can be damaged by mild insults. Therefore, surface preparations of cochlear epithelia, in combination with immunolabeling techniques and confocal imagery, are a very useful tool for the investigation of cochlear pathologies, including losses of ribbon synapses and sensory hair cells, changes in protein levels in hair cells and supporting cells, hair cell regeneration, and determination of report gene expression (i.e., GFP) for verification of successful transduction and identification of transduced cell types. The cochlea, a bony spiral-shaped structure in the inner ear, holds the auditory sensory end organ, the organ of Corti (OC). Sensory hair cells and surrounding supporting cells in the OC are contained in the cochlear duct and rest on the basilar membrane, organized in a tonotopic fashion with high-frequency detection occurring in the base and low-frequency in the apex. With the availability of molecular and genetic information and the ability to manipulate genes by knockout and knock-in techniques, mice have been widely used in biological research, including in hearing science. However, the adult mouse cochlea is miniscule, and the cochlear epithelium is encapsulated in a bony labyrinth, making microdissection difficult. Although dissection techniques have been developed and used in many laboratories, this modified microdissection method using cell and tissue adhesive is easier and more convenient. It can be used in all types of adult mouse cochleae following decalcification.
Collapse
Affiliation(s)
- Qiao-Jun Fang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina; MOE Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University
| | - Fan Wu
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina
| | - Renjie Chai
- MOE Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University
| | - Su-Hua Sha
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina;
| |
Collapse
|
43
|
How to Build a Fast and Highly Sensitive Sound Detector That Remains Robust to Temperature Shifts. J Neurosci 2019; 39:7260-7276. [PMID: 31315946 DOI: 10.1523/jneurosci.2510-18.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 06/13/2019] [Accepted: 07/09/2019] [Indexed: 12/26/2022] Open
Abstract
Frogs must have sharp hearing abilities during the warm summer months to successfully find mating partners. This study aims to understand how frog hair cell ribbon-type synapses preserve both sensitivity and temporal precision during temperature changes. Under room (∼24°C) and high (∼32°C) temperature, we performed in vitro patch-clamp recordings of hair cells and their afferent fibers in amphibian papillae of either male or female bullfrogs. Afferent fibers exhibited a wide heterogeneity in membrane input resistance (Rin) from 100 mΩ to 1000 mΩ, which may contribute to variations in spike threshold and firing frequency. At higher temperatures, most fibers increased their frequency of spike firing due to an increase in spontaneous EPSC frequencies. Hair cell resting membrane potential (Vrest) remained surprisingly stable during temperature increases, because Ca2+ influx and K+ outflux increased simultaneously. This increase in Ca2+ current likely enhanced spontaneous EPSC frequencies. These larger "leak currents" at Vrest also lowered Rin and produced higher electrical resonant frequencies. Lowering Rin will reduce the hair cells receptor potential and presumably moderate the systems sensitivity. Using membrane capacitance measurements, we suggest that hair cells can partially compensate for this reduced sensitivity by increasing exocytosis efficiency and the size of the readily releasable pool of synaptic vesicles. Furthermore, paired recordings of hair cells and their afferent fibers showed that synaptic delays shortened and multivesicular release becomes more synchronous at higher temperatures, which should improve temporal precision. Together, our results explain many previous in vivo observations on the temperature dependence of spikes in auditory nerves.SIGNIFICANCE STATEMENT The vertebrate inner ear detects and transmits auditory information over a broad dynamic range of sound frequency and intensity. It achieves remarkable sensitivity to soft sounds and precise frequency selectivity. How does the ear of cold-blooded vertebrates maintain its performance level as temperature changes? More specifically, how does the hair cell to afferent fiber synapse in bullfrog amphibian papilla adjust to a wide range of physiological temperatures without losing its sensitivity and temporal fidelity to sound signals? This study uses in vitro experiments to reveal the biophysical mechanisms that explain many observations made from in vivo auditory nerve fiber recordings. We find that higher temperature facilitates vesicle exocytosis and electrical tuning to higher sound frequencies, which benefits sensitivity and selectivity.
Collapse
|
44
|
Chakrabarti R, Wichmann C. Nanomachinery Organizing Release at Neuronal and Ribbon Synapses. Int J Mol Sci 2019; 20:E2147. [PMID: 31052288 PMCID: PMC6539712 DOI: 10.3390/ijms20092147] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/26/2019] [Accepted: 04/26/2019] [Indexed: 11/17/2022] Open
Abstract
A critical aim in neuroscience is to obtain a comprehensive view of how regulated neurotransmission is achieved. Our current understanding of synapses relies mainly on data from electrophysiological recordings, imaging, and molecular biology. Based on these methodologies, proteins involved in a synaptic vesicle (SV) formation, mobility, and fusion at the active zone (AZ) membrane have been identified. In the last decade, electron tomography (ET) combined with a rapid freezing immobilization of neuronal samples opened a window for understanding the structural machinery with the highest spatial resolution in situ. ET provides significant insights into the molecular architecture of the AZ and the organelles within the presynaptic nerve terminal. The specialized sensory ribbon synapses exhibit a distinct architecture from neuronal synapses due to the presence of the electron-dense synaptic ribbon. However, both synapse types share the filamentous structures, also commonly termed as tethers that are proposed to contribute to different steps of SV recruitment and exocytosis. In this review, we discuss the emerging views on the role of filamentous structures in SV exocytosis gained from ultrastructural studies of excitatory, mainly central neuronal compared to ribbon-type synapses with a focus on inner hair cell (IHC) ribbon synapses. Moreover, we will speculate on the molecular entities that may be involved in filament formation and hence play a crucial role in the SV cycle.
Collapse
Affiliation(s)
- Rituparna Chakrabarti
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany.
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany.
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing", 37099 Göttingen, Germany.
| | - Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany.
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany.
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing", 37099 Göttingen, Germany.
- Collaborative Research Center 1286 "Quantitative Synaptology", 37099 Göttingen, Germany.
- Auditory Neuroscience Group, Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany.
| |
Collapse
|
45
|
Heeringa AN, Köppl C. The aging cochlea: Towards unraveling the functional contributions of strial dysfunction and synaptopathy. Hear Res 2019; 376:111-124. [PMID: 30862414 DOI: 10.1016/j.heares.2019.02.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 02/01/2019] [Accepted: 02/26/2019] [Indexed: 10/27/2022]
Abstract
Strial dysfunction is commonly observed as a key consequence of aging in the cochlea. A large body of animal research, especially in the quiet-aged Mongolian gerbil, shows specific histopathological changes in the cochlear stria vascularis and the putatively corresponding effects on endocochlear potential and auditory nerve responses. However, recent work suggests that synaptopathy, or the loss of inner hair cell-auditory nerve fiber synapses, also presents as a consequence of aging. It is now believed that the loss of synapses is the earliest age-related degenerative event. The present review aims to integrate classic and novel research on age-related pathologies of the inner ear. First, we summarize current knowledge on age-related strial dysfunction and synaptopathy. We describe how these cochlear pathologies fit into the categories for presbyacusis, as first defined by Schuknecht in the '70s. Further, we discuss how strial dysfunction and synaptopathy affect sound coding by the auditory nerve and how they can be experimentally induced to study their specific contributions to age-related hearing deficits. As such, we aim to give an overview of the current literature on age-related cochlear pathologies and hope to inspire further research on the role of cochlear aging in age-related hearing deficits.
Collapse
Affiliation(s)
- Amarins N Heeringa
- Cluster of Excellence 'Hearing4all' and Research Centre Neurosensory Science, Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky University Oldenburg, 26129, Oldenburg, Germany
| | - Christine Köppl
- Cluster of Excellence 'Hearing4all' and Research Centre Neurosensory Science, Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky University Oldenburg, 26129, Oldenburg, Germany.
| |
Collapse
|
46
|
Wang J, Yin S, Chen H, Shi L. Noise-Induced Cochlear Synaptopathy and Ribbon Synapse Regeneration: Repair Process and Therapeutic Target. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1130:37-57. [PMID: 30915700 DOI: 10.1007/978-981-13-6123-4_3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The synapse between the inner hair cells (IHCs) and the spiral ganglion neurons (SGNs) in mammalian cochleae is characterized as having presynaptic ribbons and therefore is called ribbon synapse. The special molecular organization is reviewed in this chapter in association with the functional feature of this synapse in signal processing. This is followed by the review on noise-induced damage to this synapse with a focus on recent reports in animal models in which the effect of brief noise exposures is observed without causing significant permanent threshold shift (PTS). In this regard, the potential mechanism of the synaptic damage by noise and the impact of this damage on hearing are summarized to clarify the concept of noise-induced hidden hearing loss, which is defined as the functional deficits in hearing without threshold elevation. A controversial issue is addressed in this review as whether the disrupted synapses can be regenerated. Moreover, the review summarizes the work of therapeutic research to protect the synapses or to promote the regeneration of the synapse after initial disruption. Lastly, several unresolved issues are raised for investigation in the future.
Collapse
Affiliation(s)
- Jian Wang
- School of Communication Science and Disorders, Dalhousie University, Halifax, NS, Canada.
| | - Shankai Yin
- Otolaryngology Research Institute, 6th Affiliated Hospital, Shanghai Jiao-Tong University, Shanghai, China
| | - Hengchao Chen
- Otolaryngology Research Institute, 6th Affiliated Hospital, Shanghai Jiao-Tong University, Shanghai, China
| | - Lijuan Shi
- Department of Physiology, Medical College of Southeast University, Nanjing, China
| |
Collapse
|
47
|
Harrus AG, Ceccato JC, Sendin G, Bourien J, Puel JL, Nouvian R. Spiking Pattern of the Mouse Developing Inner Hair Cells Is Mostly Invariant Along the Tonotopic Axis. Front Cell Neurosci 2018; 12:407. [PMID: 30524238 PMCID: PMC6262317 DOI: 10.3389/fncel.2018.00407] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 10/22/2018] [Indexed: 01/03/2023] Open
Abstract
During development, the sensory cells of the cochlea, the inner hair cells (IHCs), fire spontaneous calcium action potentials. This activity at the pre-hearing stage allows the IHCs to autonomously excite the auditory nerve fibers and hence, represents an efficient mechanism to shape the tonotopic organization along the ascending auditory pathway. Using calcium imaging, we show that the activity in the developing cochlea consists of calcium waves that propagate across the supporting and sensory cells. Both basal and apical IHCs were characterized by similar spontaneous calcium transients interspaced with silent periods, consistent with bursts of action potentials recorded in patch-clamp. In addition, adjacent auditory hair cells tend to have a synchronized [Ca2+]i activity, irrespective of their location along the base-to-apex gradient of the cochlea. Finally, we show that the mechanical ablation of the inner phalangeal cells (IPCs), a class of supporting cells, reduces the synchronized [Ca2+]i activity between neighboring sensory cells. These findings support the hypothesis that the tonotopic map refinement in higher auditory centers would depend on the synchronization of a discrete number of auditory sensory cells.
Collapse
Affiliation(s)
- Anne-Gabrielle Harrus
- Institut des Neurosciences de Montpellier (INM), Inserm, University of Montpellier, Montpellier, France
| | - Jean-Charles Ceccato
- Institut des Neurosciences de Montpellier (INM), Inserm, University of Montpellier, Montpellier, France
| | - Gaston Sendin
- Institut des Neurosciences de Montpellier (INM), Inserm, University of Montpellier, Montpellier, France
| | - Jérôme Bourien
- Institut des Neurosciences de Montpellier (INM), Inserm, University of Montpellier, Montpellier, France
| | - Jean-Luc Puel
- Institut des Neurosciences de Montpellier (INM), Inserm, University of Montpellier, Montpellier, France
| | - Régis Nouvian
- Institut des Neurosciences de Montpellier (INM), Inserm, University of Montpellier, Montpellier, France
| |
Collapse
|
48
|
Chakrabarti R, Michanski S, Wichmann C. Vesicle sub-pool organization at inner hair cell ribbon synapses. EMBO Rep 2018; 19:embr.201744937. [PMID: 30201800 DOI: 10.15252/embr.201744937] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 08/07/2018] [Accepted: 08/21/2018] [Indexed: 02/02/2023] Open
Abstract
The afferent inner hair cell synapse harbors the synaptic ribbon, which ensures a constant vesicle supply. Synaptic vesicles (SVs) are arranged in morphologically discernable pools, linked via filaments to the ribbon or the presynaptic membrane. We propose that filaments play a major role in SV resupply and exocytosis at the ribbon. Using advanced electron microscopy, we demonstrate that SVs are organized in sub-pools defined by the filament number per vesicle and its connections. Upon stimulation, SVs increasingly linked to other vesicles and to the ribbon, whereas single-tethered SVs dominated at the membrane. Mutant mice for the hair cell protein otoferlin (pachanga, Otof Pga/Pga ) are profoundly deaf with reduced sustained release, serving as a model to investigate the SV replenishment at IHCs. Upon stimulation, multiple-tethered and docked vesicles (rarely observed in wild-type) accumulated at Otof Pga/Pga active zones due to an impairment downstream of docking. Conclusively, vesicles are organized in sub-pools at ribbon-type active zones by filaments to support vesicle supply, transport, and finally release.
Collapse
Affiliation(s)
- Rituparna Chakrabarti
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany.,Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing", Göttingen, Germany.,Göttingen Graduate School for Neurosciences, Biophysics and Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Susann Michanski
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany.,Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing", Göttingen, Germany.,Georg-August University School of Science, Göttingen, Germany
| | - Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany .,Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany.,Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing", Göttingen, Germany
| |
Collapse
|
49
|
Chen J, Hong F, Zhang C, Li L, Wang C, Shi H, Fu Y, Wang J. Differentiation and transplantation of human induced pluripotent stem cell-derived otic epithelial progenitors in mouse cochlea. Stem Cell Res Ther 2018; 9:230. [PMID: 30157937 PMCID: PMC6116394 DOI: 10.1186/s13287-018-0967-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/12/2018] [Accepted: 08/01/2018] [Indexed: 12/24/2022] Open
Abstract
Background Inner ear hair cells as mechanoreceptors are extremely important for hearing. Defects in hair cells are a major cause of deafness. Induced pluripotent stem cells (iPSCs) are promising for regenerating inner ear hair cells and treating hearing loss. Here, we investigated migration, differentiation, and synaptic connections of transplanted otic epithelial progenitors (OEPs) derived from human iPSCs in mouse cochlea. Methods Human urinary cells isolated from a healthy donor were reprogramed to form iPSCs that were induced to differentiate into OEPs and hair cell-like cells. Immunocytochemistry, electrophysiological examination, and scanning electron microscopy were used to examine characteristics of induced hair cell-like cells. OEP-derived hair cell-like cells were cocultured with spiral ganglion neurons (SGNs), and the markers of synaptic connections were detected using immunocytochemistry and transmission electron microscope. In vivo, OEPs derived from iPSCs were transplanted into the cochlea of mice by injection through the round window. Migration, differentiation, and synaptic connections of transplanted cells were also examined by thin cochlear sectioning and immunohistochemistry. Results The induced hair cell-like cells displayed typical morphological characteristics and electrophysiological properties specific to inner hair cells. In vitro, OEP-derived hair cell-like cells formed synaptic connections with SGNs in coculture. In vivo, some of the transplanted cells migrated to the site of the resident hair cells in the organ of Corti, differentiated into hair cell-like cells, and formed synaptic connections with native SGNs. Conclusions We conclude that the transplantation of OEPs is feasible for the regeneration of hair cells. These results present a substantial reference for a cell-based therapy for the loss of hair cells.
Collapse
Affiliation(s)
- Jianling Chen
- Institute of Cell and Development, College of Life Sciences, Zi-Jin-Gang Campus of Zhejiang University, Room 307, No.866, Yuhangtang Road, Hangzhou, 310058, Zhejiang, China
| | - Fanfan Hong
- Institute of Cell and Development, College of Life Sciences, Zi-Jin-Gang Campus of Zhejiang University, Room 307, No.866, Yuhangtang Road, Hangzhou, 310058, Zhejiang, China
| | - Cui Zhang
- Institute of Cell and Development, College of Life Sciences, Zi-Jin-Gang Campus of Zhejiang University, Room 307, No.866, Yuhangtang Road, Hangzhou, 310058, Zhejiang, China
| | - Liang Li
- Institute of Cell and Development, College of Life Sciences, Zi-Jin-Gang Campus of Zhejiang University, Room 307, No.866, Yuhangtang Road, Hangzhou, 310058, Zhejiang, China
| | - Cuicui Wang
- Institute of Cell and Development, College of Life Sciences, Zi-Jin-Gang Campus of Zhejiang University, Room 307, No.866, Yuhangtang Road, Hangzhou, 310058, Zhejiang, China
| | - Haosong Shi
- Department of Otorhinolaryngology, the Sixth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yong Fu
- Department of ENT, Head and Neck Surgery, the Children's Hospital, Zhejiang University School of Medicine, Zhejiang, China. .,Department of Otolaryngology, the Children Hospital, School of Medicine, Bin-Jiang Campus of Zhejiang University, No. 3333, Binsheng Road, Hangzhou, 310051, Zhejiang, China.
| | - Jinfu Wang
- Institute of Cell and Development, College of Life Sciences, Zi-Jin-Gang Campus of Zhejiang University, Room 307, No.866, Yuhangtang Road, Hangzhou, 310058, Zhejiang, China.
| |
Collapse
|
50
|
N-Methyl-D-Aspartate Receptors Involvement in the Gentamicin-Induced Hearing Loss and Pathological Changes of Ribbon Synapse in the Mouse Cochlear Inner Hair Cells. Neural Plast 2018; 2018:3989201. [PMID: 30123246 PMCID: PMC6079453 DOI: 10.1155/2018/3989201] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 05/17/2018] [Indexed: 11/17/2022] Open
Abstract
Cochlear inner hair cell (IHC) ribbon synapses play an important role in sound encoding and neurotransmitter release. Previous reports show that both noise and aminoglycoside exposures lead to reduced numbers and morphologic changes of synaptic ribbons. In this work, we determined the distribution of N-methyl-D-aspartate receptors (NMDARs) and their role in the gentamicin-induced pathological changes of cochlear IHC ribbon synaptic elements. In normal mature mouse cochleae, the majority of NMDARs were distributed on the modiolar side of IHCs and close to the IHC nuclei region, while most of synaptic ribbons and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) were located on neural terminals closer to the IHC basal poles. After gentamicin exposure, the NMDARs increased and moved towards the IHC basal poles. At the same time, synaptic ribbons and AMPARs moved toward the IHC bundle poles on the afferent dendrites. The number of ribbon synapse decreased, and this was accompanied by increased auditory brainstem response thresholds and reduced wave I amplitudes. NMDAR antagonist MK801 treatment reduced the gentamicin-induced hearing loss and the pathological changes of IHC ribbon synapse, suggesting that NMDARs were involved in gentamicin-induced ototoxicity by regulating the number and distribution of IHC ribbon synapses.
Collapse
|