1
|
Giunta R, Cheli G, Rispoli G, Russo G, Masetto S. Pimozide Inhibits Type II but Not Type I Hair Cells in Chicken Embryo and Adult Mouse Vestibular Organs. Biomedicines 2024; 12:2879. [PMID: 39767785 PMCID: PMC11673355 DOI: 10.3390/biomedicines12122879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Pimozide is a conventional antipsychotic drug of the diphenylbutylpiperidine class, widely used for treating schizophrenia and delusional disorders and for managing motor and phonic tics in Tourette's syndrome. Pimozide is known to block dopaminergic D2 receptors and various types of voltage-gated ion channels. Among its side effects, dizziness and imbalance are the most frequently observed, which may imply an effect of the drug on the vestibular sensory receptors, the hair cells. Amniotes possess two classes of vestibular hair cells, named type I and type II hair cells, which differ in terms of signal processing and transmission. We previously reported that Pimozide [3 μM] significantly increased a delayed outward rectifying K+ current (IK,V). METHODS AND RESULTS In the present study, using the whole-cell patch-clamp technique we additionally show that Pimozide decreases the inward rectifying K+ current (IK,1) and the mixed Na+/K+ current (Ih) of chicken embryo type II hair cells, whereas it does not affect type I hair cells' ionic currents. Since ion channels' expression can vary depending on age and animal species, in the present study, we also tested Pimozide in adult mouse vestibular hair cells. We found that, like in the chicken embryo, Pimozide significantly increases IK,V and decreases IK,1 and Ih in type II hair cells. However, in the adult mouse, Pimozide also slightly increased the outward rectifying K+ current in type I hair cells. CONCLUSIONS While providing a possible explanation for the vestibular side effects of Pimozide in humans, its inhibitory action on mammalian hair cells might be of interest for the local treatment of vestibular disorders characterized by altered vestibular input, like Ménière's disease.
Collapse
Affiliation(s)
- Roberta Giunta
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (R.G.); (G.C.); (G.R.)
| | - Giulia Cheli
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (R.G.); (G.C.); (G.R.)
| | - Giorgio Rispoli
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy;
| | - Giancarlo Russo
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (R.G.); (G.C.); (G.R.)
| | - Sergio Masetto
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (R.G.); (G.C.); (G.R.)
| |
Collapse
|
2
|
Martin HR, Lysakowski A, Eatock RA. The potassium channel subunit K V1.8 ( Kcna10) is essential for the distinctive outwardly rectifying conductances of type I and II vestibular hair cells. eLife 2024; 13:RP94342. [PMID: 39625061 PMCID: PMC11614384 DOI: 10.7554/elife.94342] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2024] Open
Abstract
In amniotes, head motions and tilt are detected by two types of vestibular hair cells (HCs) with strikingly different morphology and physiology. Mature type I HCs express a large and very unusual potassium conductance, gK,L, which activates negative to resting potential, confers very negative resting potentials and low input resistances, and enhances an unusual non-quantal transmission from type I cells onto their calyceal afferent terminals. Following clues pointing to KV1.8 (Kcna10) in the Shaker K channel family as a candidate gK,L subunit, we compared whole-cell voltage-dependent currents from utricular HCs of KV1.8-null mice and littermate controls. We found that KV1.8 is necessary not just for gK,L but also for fast-inactivating and delayed rectifier currents in type II HCs, which activate positive to resting potential. The distinct properties of the three KV1.8-dependent conductances may reflect different mixing with other KV subunits that are reported to be differentially expressed in type I and II HCs. In KV1.8-null HCs of both types, residual outwardly rectifying conductances include KV7 (Knq) channels. Current clamp records show that in both HC types, KV1.8-dependent conductances increase the speed and damping of voltage responses. Features that speed up vestibular receptor potentials and non-quantal afferent transmission may have helped stabilize locomotion as tetrapods moved from water to land.
Collapse
Affiliation(s)
- Hannah R Martin
- Department of Neurobiology, University of ChicagoChicagoUnited States
| | - Anna Lysakowski
- Department of Anatomy and Cell Biology, University of Illinois at ChicagoChicagoUnited States
| | - Ruth Anne Eatock
- Department of Neurobiology, University of ChicagoChicagoUnited States
| |
Collapse
|
3
|
Ono K, Jarysta A, Hughes NC, Jukic A, Chang HHV, Deans MR, Eatock RA, Cullen KE, Kindt KS, Tarchini B. Contributions of mirror-image hair cell orientation to mouse otolith organ and zebrafish neuromast function. eLife 2024; 13:RP97674. [PMID: 39531034 PMCID: PMC11556791 DOI: 10.7554/elife.97674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Otolith organs in the inner ear and neuromasts in the fish lateral-line harbor two populations of hair cells oriented to detect stimuli in opposing directions. The underlying mechanism is highly conserved: the transcription factor EMX2 is regionally expressed in just one hair cell population and acts through the receptor GPR156 to reverse cell orientation relative to the other population. In mouse and zebrafish, loss of Emx2 results in sensory organs that harbor only one hair cell orientation and are not innervated properly. In zebrafish, Emx2 also confers hair cells with reduced mechanosensory properties. Here, we leverage mouse and zebrafish models lacking GPR156 to determine how detecting stimuli of opposing directions serves vestibular function, and whether GPR156 has other roles besides orienting hair cells. We find that otolith organs in Gpr156 mouse mutants have normal zonal organization and normal type I-II hair cell distribution and mechano-electrical transduction properties. In contrast, gpr156 zebrafish mutants lack the smaller mechanically evoked signals that characterize Emx2-positive hair cells. Loss of GPR156 does not affect orientation-selectivity of afferents in mouse utricle or zebrafish neuromasts. Consistent with normal otolith organ anatomy and afferent selectivity, Gpr156 mutant mice do not show overt vestibular dysfunction. Instead, performance on two tests that engage otolith organs is significantly altered - swimming and off-vertical-axis rotation. We conclude that GPR156 relays hair cell orientation and transduction information downstream of EMX2, but not selectivity for direction-specific afferents. These results clarify how molecular mechanisms that confer bi-directionality to sensory organs contribute to function, from single hair cell physiology to animal behavior.
Collapse
Affiliation(s)
- Kazuya Ono
- Department of Neurobiology, University of ChicagoChicagoUnited States
| | | | - Natasha C Hughes
- Department of Biomedical Engineering, Johns Hopkins UniversityBaltimoreUnited States
| | - Alma Jukic
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Hui Ho Vanessa Chang
- Department of Biomedical Engineering, Johns Hopkins UniversityBaltimoreUnited States
| | - Michael R Deans
- Department of Neurobiology, Spencer Fox Eccles School of Medicine, University of UtahSalt Lake CityUnited States
- Department of Otolaryngology - Head & Neck Surgery, Spencer Fox Eccles School of Medicine at the University of UtahSalt Lake CityUnited States
| | - Ruth Anne Eatock
- Department of Neurobiology, University of ChicagoChicagoUnited States
| | - Kathleen E Cullen
- Department of Biomedical Engineering, Johns Hopkins UniversityBaltimoreUnited States
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins UniversityBaltimoreUnited States
- Department of Neuroscience, Johns Hopkins UniversityBaltimoreUnited States
- Kavli Neuroscience Discovery Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Katie S Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Basile Tarchini
- The Jackson LaboratoryBar HarborUnited States
- Tufts University School of MedicineBostonUnited States
| |
Collapse
|
4
|
Ono K, Jarysta A, Hughes NC, Jukic A, Chang HHV, Deans MR, Eatock RA, Cullen KE, Kindt K, Tarchini B. Contributions of mirror-image hair cell orientation to mouse otolith organ and zebrafish neuromast function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.26.586740. [PMID: 39282410 PMCID: PMC11398332 DOI: 10.1101/2024.03.26.586740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
Otolith organs in the inner ear and neuromasts in the fish lateral-line harbor two populations of hair cells oriented to detect stimuli in opposing directions. The underlying mechanism is highly conserved: the transcription factor EMX2 is regionally expressed in just one hair cell population and acts through the receptor GPR156 to reverse cell orientation relative to the other population. In mouse and zebrafish, loss of Emx2 results in sensory organs that harbor only one hair cell orientation and are not innervated properly. In zebrafish, Emx2 also confers hair cells with reduced mechanosensory properties. Here, we leverage mouse and zebrafish models lacking GPR156 to determine how detecting stimuli of opposing directions serves vestibular function, and whether GPR156 has other roles besides orienting hair cells. We find that otolith organs in Gpr156 mouse mutants have normal zonal organization and normal type I-II hair cell distribution and mechano-electrical transduction properties. In contrast, gpr156 zebrafish mutants lack the smaller mechanically-evoked signals that characterize Emx2-positive hair cells. Loss of GPR156 does not affect orientation-selectivity of afferents in mouse utricle or zebrafish neuromasts. Consistent with normal otolith organ anatomy and afferent selectivity, Gpr156 mutant mice do not show overt vestibular dysfunction. Instead, performance on two tests that engage otolith organs is significantly altered - swimming and off-vertical-axis rotation. We conclude that GPR156 relays hair cell orientation and transduction information downstream of EMX2, but not selectivity for direction-specific afferents. These results clarify how molecular mechanisms that confer bi-directionality to sensory organs contribute to function, from single hair cell physiology to animal behavior.
Collapse
Affiliation(s)
- Kazuya Ono
- Department of Neurobiology, University of Chicago, Chicago, IL, 60637, USA
| | | | - Natasha C Hughes
- Dept. of Biomedical Engineering, Johns Hopkins University, Baltimore, 21205 MD, USA
| | - Alma Jukic
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MA, USA
| | - Hui Ho Vanessa Chang
- Dept. of Biomedical Engineering, Johns Hopkins University, Baltimore, 21205 MD, USA
| | - Michael R Deans
- Department of Neurobiology, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, Utah, USA
- Department of Otolaryngology - Head & Neck Surgery, Spencer Fox Eccles School of Medicine at the University of Utah, Salt Lake City, Utah, USA
| | - Ruth Anne Eatock
- Department of Neurobiology, University of Chicago, Chicago, IL, 60637, USA
| | - Kathleen E Cullen
- Dept. of Biomedical Engineering, Johns Hopkins University, Baltimore, 21205 MD, USA
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, Baltimore 21205 MD, USA
- Department of Neuroscience, Johns Hopkins University, Baltimore 21205 MD, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore 21205 MD, USA
| | - Katie Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MA, USA
| | - Basile Tarchini
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
- Tufts University School of Medicine, Boston, MA 02111, USA
| |
Collapse
|
5
|
Martin HR, Lysakowski A, Eatock RA. The potassium channel subunit K V1.8 ( Kcna10) is essential for the distinctive outwardly rectifying conductances of type I and II vestibular hair cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.21.563853. [PMID: 38045305 PMCID: PMC10690164 DOI: 10.1101/2023.11.21.563853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
In amniotes, head motions and tilt are detected by two types of vestibular hair cells (HCs) with strikingly different morphology and physiology. Mature type I HCs express a large and very unusual potassium conductance, gK,L, which activates negative to resting potential, confers very negative resting potentials and low input resistances, and enhances an unusual non-quantal transmission from type I cells onto their calyceal afferent terminals. Following clues pointing to KV1.8 (KCNA10) in the Shaker K channel family as a candidate gK,L subunit, we compared whole-cell voltage-dependent currents from utricular hair cells of KV1.8-null mice and littermate controls. We found that KV1.8 is necessary not just for gK,L but also for fast-inactivating and delayed rectifier currents in type II HCs, which activate positive to resting potential. The distinct properties of the three KV1.8-dependent conductances may reflect different mixing with other KV subunits that are reported to be differentially expressed in type I and II HCs. In KV1.8-null HCs of both types, residual outwardly rectifying conductances include KV7 (KCNQ) channels. Current clamp records show that in both HC types, KV1.8-dependent conductances increase the speed and damping of voltage responses. Features that speed up vestibular receptor potentials and non-quantal afferent transmission may have helped stabilize locomotion as tetrapods moved from water to land.
Collapse
Affiliation(s)
| | - Anna Lysakowski
- University of Illinois at Chicago, Department of Anatomy and Cell Biology
| | | |
Collapse
|
6
|
Lipovsek M. Comparative biology of the amniote vestibular utricle. Hear Res 2024; 448:109035. [PMID: 38763033 DOI: 10.1016/j.heares.2024.109035] [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: 03/31/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/21/2024]
Abstract
The sensory epithelia of the auditory and vestibular systems of vertebrates have shared developmental and evolutionary histories. However, while the auditory epithelia show great variation across vertebrates, the vestibular sensory epithelia appear seemingly more conserved. An exploration of the current knowledge of the comparative biology of the amniote utricle, a vestibular sensory epithelium that senses linear acceleration, shows interesting instances of variability between birds and mammals. The distribution of sensory hair cell types, the position of the line of hair bundle polarity reversal and the properties of supporting cells show marked differences, likely impacting vestibular function and hair cell regeneration potential.
Collapse
Affiliation(s)
- Marcela Lipovsek
- Ear Institute, Faculty of Brain Sciences, University College London, London, UK.
| |
Collapse
|
7
|
Heffer A, Lee C, Holt JC, Kiernan AE. Notch1 is required to maintain supporting cell identity and vestibular function during maturation of the mammalian balance organs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.21.600098. [PMID: 38948821 PMCID: PMC11212955 DOI: 10.1101/2024.06.21.600098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The inner ear houses two sensory modalities: the hearing organ, located in the cochlea, and the balance organs, located throughout the vestibular regions of the ear. Both hearing and vestibular sensory regions are composed of similar cell types, including hair cells and associated supporting cells. Recently, we showed that Notch1 is required for maintaining supporting cell survival postnatally during cochlear maturation. However, it is not known whether Notch1 plays a similar role in the balance organs of the inner ear. To characterize the role of Notch during vestibular maturation, we conditionally deleted Notch1 from Sox2-expressing cells of the vestibular organs in the mouse at P0/P1. Histological analyses showed a dramatic loss of supporting cells accompanied by an increase in type II hair cells without cell death, indicating the supporting cells are converting to hair cells in the maturing vestibular regions. Analysis of 6-week old animals indicate that the converted hair cells survive, despite the reduction of supporting cells. Interestingly, measurements of vestibular sensory evoked potentials (VsEPs), known to be generated in the striolar regions of the vestibular afferents in the maculae, failed to show a response, indicating that NOTCH1 expression is critical for striolar function postnatally. Consistent with this, we find that the specialized type I hair cells in the striola fail to develop the complex calyces typical of these cells. These defects are likely due to the reduction in supporting cells, which have previously been shown to express factors critical for the striolar region. Similar to other mutants that lack proper striolar development, Notch1 mutants do not exhibit typical vestibular behaviors such as circling and head shaking, but do show difficulties in some vestibular tests, including the balance beam and forced swim test. These results indicate that, unlike the hearing organ in which the supporting cells undergo cell death, supporting cells in the balance regions retain the ability to convert to hair cells during maturation, which survive into adulthood despite the reduction in supporting cells.
Collapse
Affiliation(s)
- Alison Heffer
- Flaum Eye Institute, Department of Ophthalmology, University of Rochester Medical Center, Rochester, New York, 14642, USA
| | - Choongheon Lee
- Department of Otolaryngology, University of Rochester, Rochester, NY, 14642, USA
| | - Joseph C. Holt
- Department of Otolaryngology, University of Rochester, Rochester, NY, 14642, USA
- Dept. of Neuroscience, University of Rochester, Rochester, New York 14642, USA
| | - Amy E. Kiernan
- Flaum Eye Institute, Department of Ophthalmology, University of Rochester Medical Center, Rochester, New York, 14642, USA
| |
Collapse
|
8
|
Wang T, Ling AH, Billings SE, Hosseini DK, Vaisbuch Y, Kim GS, Atkinson PJ, Sayyid ZN, Aaron KA, Wagh D, Pham N, Scheibinger M, Zhou R, Ishiyama A, Moore LS, Maria PS, Blevins NH, Jackler RK, Alyono JC, Kveton J, Navaratnam D, Heller S, Lopez IA, Grillet N, Jan TA, Cheng AG. Single-cell transcriptomic atlas reveals increased regeneration in diseased human inner ear balance organs. Nat Commun 2024; 15:4833. [PMID: 38844821 PMCID: PMC11156867 DOI: 10.1038/s41467-024-48491-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 04/29/2024] [Indexed: 06/09/2024] Open
Abstract
Mammalian inner ear hair cell loss leads to permanent hearing and balance dysfunction. In contrast to the cochlea, vestibular hair cells of the murine utricle have some regenerative capacity. Whether human utricular hair cells regenerate in vivo remains unknown. Here we procured live, mature utricles from organ donors and vestibular schwannoma patients, and present a validated single-cell transcriptomic atlas at unprecedented resolution. We describe markers of 13 sensory and non-sensory cell types, with partial overlap and correlation between transcriptomes of human and mouse hair cells and supporting cells. We further uncover transcriptomes unique to hair cell precursors, which are unexpectedly 14-fold more abundant in vestibular schwannoma utricles, demonstrating the existence of ongoing regeneration in humans. Lastly, supporting cell-to-hair cell trajectory analysis revealed 5 distinct patterns of dynamic gene expression and associated pathways, including Wnt and IGF-1 signaling. Our dataset constitutes a foundational resource, accessible via a web-based interface, serving to advance knowledge of the normal and diseased human inner ear.
Collapse
Affiliation(s)
- Tian Wang
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Otolaryngology - Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, 410011, PR China
| | - Angela H Ling
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Otolaryngology - Head and Neck Surgery, Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Sara E Billings
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Davood K Hosseini
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Yona Vaisbuch
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Grace S Kim
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Patrick J Atkinson
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Zahra N Sayyid
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Ksenia A Aaron
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Dhananjay Wagh
- Stanford Genomics Facility, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Nicole Pham
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Mirko Scheibinger
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Ruiqi Zhou
- Department of Otolaryngology - Head and Neck Surgery, Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Akira Ishiyama
- Department of Head and Neck Surgery, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Lindsay S Moore
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Peter Santa Maria
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Nikolas H Blevins
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Robert K Jackler
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jennifer C Alyono
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - John Kveton
- Department of Surgery, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Dhasakumar Navaratnam
- Department of Surgery, Yale University School of Medicine, New Haven, CT, 06510, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Stefan Heller
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Ivan A Lopez
- Department of Head and Neck Surgery, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Nicolas Grillet
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Taha A Jan
- Department of Otolaryngology - Head and Neck Surgery, Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| | - Alan G Cheng
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
9
|
Borrajo M, Sedano D, Palou A, Giménez-Esbrí V, Barrallo-Gimeno A, Llorens J. Maturation of type I and type II rat vestibular hair cells in vivo and in vitro. Front Cell Dev Biol 2024; 12:1404894. [PMID: 38895157 PMCID: PMC11183282 DOI: 10.3389/fcell.2024.1404894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024] Open
Abstract
Vestibular sensory epithelia contain type I and type II sensory hair cells (HCI and HCII). Recent studies have revealed molecular markers for the identification of these cells, but the precise composition of each vestibular epithelium (saccule, utricle, lateral crista, anterior crista, posterior crista) and their postnatal maturation have not been described in detail. Moreover, in vitro methods to study this maturation are not well developed. We obtained total HCI and HCII counts in adult rats and studied the maturation of the epithelia from birth (P0) to postnatal day 28 (P28). Adult vestibular epithelia hair cells were found to comprise ∼65% HCI expressing osteopontin and PMCA2, ∼30% HCII expressing calretinin, and ∼4% HCII expressing SOX2 but neither osteopontin nor calretinin. At birth, immature HCs express both osteopontin and calretinin. P28 epithelia showed an almost adult-like composition but still contained 1.3% of immature HCs. In addition, we obtained free-floating 3D cultures of the epithelia at P1, which formed a fluid-filled cyst, and studied their survival and maturation in vitro up to day 28 (28 DIV). These cultures showed good HC resiliency and maturation. Using an enriched medium for the initial 4 days, a HCI/calretinin+-HCII ratio close to the in vivo ratio was obtained. These cultures are suitable to study HC maturation and mature HCs in pharmacological, toxicological and molecular research.
Collapse
Affiliation(s)
- Mireia Borrajo
- Departament de Ciències Fisiològiques, Universitat de Barcelona (UB), Hospitalet de Llobregat, Catalunya, Spain
- Institut de Neurociències, Universitat de Barcelona (UB), Barcelona, Catalunya, Spain
- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Catalunya, Spain
| | - David Sedano
- Departament de Ciències Fisiològiques, Universitat de Barcelona (UB), Hospitalet de Llobregat, Catalunya, Spain
| | - Aïda Palou
- Departament de Ciències Fisiològiques, Universitat de Barcelona (UB), Hospitalet de Llobregat, Catalunya, Spain
- Institut de Neurociències, Universitat de Barcelona (UB), Barcelona, Catalunya, Spain
- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Catalunya, Spain
| | - Víctor Giménez-Esbrí
- Departament de Ciències Fisiològiques, Universitat de Barcelona (UB), Hospitalet de Llobregat, Catalunya, Spain
- Institut de Neurociències, Universitat de Barcelona (UB), Barcelona, Catalunya, Spain
- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Catalunya, Spain
| | - Alejandro Barrallo-Gimeno
- Departament de Ciències Fisiològiques, Universitat de Barcelona (UB), Hospitalet de Llobregat, Catalunya, Spain
- Institut de Neurociències, Universitat de Barcelona (UB), Barcelona, Catalunya, Spain
- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Catalunya, Spain
| | - Jordi Llorens
- Departament de Ciències Fisiològiques, Universitat de Barcelona (UB), Hospitalet de Llobregat, Catalunya, Spain
- Institut de Neurociències, Universitat de Barcelona (UB), Barcelona, Catalunya, Spain
- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Catalunya, Spain
| |
Collapse
|
10
|
You D, Ni W, Huang Y, Zhou Q, Zhang Y, Jiang T, Chen Y, Li W. The proper timing of Atoh1 expression is pivotal for hair cell subtype differentiation and the establishment of inner ear function. Cell Mol Life Sci 2023; 80:349. [PMID: 37930405 PMCID: PMC10628023 DOI: 10.1007/s00018-023-04947-w] [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: 07/05/2023] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 11/07/2023]
Abstract
Atoh1 overexpression is essential for hair cell (HC) regeneration in the sensory epithelium of mammalian auditory and vestibular organs. However, Atoh1 overexpression alone cannot induce fully mature and functional HCs in the mammalian inner ear. In the current study, we investigated the effect of Atoh1 constitutive overexpression in native HCs by manipulating Atoh1 expression at different developmental stages. We demonstrated that constitutive overexpression of Atoh1 in native vestibular HCs did not affect cell survival but did impair vestibular function by interfering with the subtype differentiation of HCs and hair bundle development. In contrast, Atoh1 overexpression in cochlear HCs impeded their maturation, eventually leading to gradual HC loss in the cochlea and hearing dysfunction. Our study suggests that time-restricted Atoh1 expression is essential for the differentiation and survival of HCs in the inner ear, and this is pivotal for both hearing and vestibular function re-establishment through Atoh1 overexpression-induced HC regeneration strategies.
Collapse
Affiliation(s)
- Dan You
- ENT Institute, Department of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200031, People's Republic of China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031, People's Republic of China
| | - Wenli Ni
- ENT Institute, Department of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200031, People's Republic of China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031, People's Republic of China
| | - Yikang Huang
- ENT Institute, Department of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200031, People's Republic of China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031, People's Republic of China
| | - Qin Zhou
- ENT Institute, Department of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200031, People's Republic of China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031, People's Republic of China
| | - Yanping Zhang
- ENT Institute, Department of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200031, People's Republic of China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031, People's Republic of China
| | - Tao Jiang
- ENT Institute, Department of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200031, People's Republic of China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031, People's Republic of China
| | - Yan Chen
- ENT Institute, Department of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200031, People's Republic of China.
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031, People's Republic of China.
| | - Wenyan Li
- ENT Institute, Department of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200031, People's Republic of China.
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031, People's Republic of China.
| |
Collapse
|
11
|
Michanski S, Henneck T, Mukhopadhyay M, Steyer AM, Gonzalez PA, Grewe K, Ilgen P, Gültas M, Fornasiero EF, Jakobs S, Möbius W, Vogl C, Pangršič T, Rizzoli SO, Wichmann C. Age-dependent structural reorganization of utricular ribbon synapses. Front Cell Dev Biol 2023; 11:1178992. [PMID: 37635868 PMCID: PMC10447907 DOI: 10.3389/fcell.2023.1178992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 07/17/2023] [Indexed: 08/29/2023] Open
Abstract
In mammals, spatial orientation is synaptically-encoded by sensory hair cells of the vestibular labyrinth. Vestibular hair cells (VHCs) harbor synaptic ribbons at their presynaptic active zones (AZs), which play a critical role in molecular scaffolding and facilitate synaptic release and vesicular replenishment. With advancing age, the prevalence of vestibular deficits increases; yet, the underlying mechanisms are not well understood and the possible accompanying morphological changes in the VHC synapses have not yet been systematically examined. We investigated the effects of maturation and aging on the ultrastructure of the ribbon-type AZs in murine utricles using various electron microscopic techniques and combined them with confocal and super-resolution light microscopy as well as metabolic imaging up to 1 year of age. In older animals, we detected predominantly in type I VHCs the formation of floating ribbon clusters, mostly consisting of newly synthesized ribbon material. Our findings suggest that VHC ribbon-type AZs undergo dramatic structural alterations upon aging.
Collapse
Affiliation(s)
- Susann Michanski
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, InnerEarLab and Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
- Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, Göttingen, Germany
| | - Timo Henneck
- Biology Bachelor Program, University of Göttingen, Göttingen, Germany
| | - Mohona Mukhopadhyay
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany
| | - Anna M. Steyer
- Electron Microscopy-City Campus, Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University of Göttingen, Göttingen, Germany
| | - Paola Agüi Gonzalez
- Department for Neuro-and Sensory Physiology, University Medical Center Göttingen, Center for Biostructural Imaging of Neurodegeneration (BIN), Göttingen, Germany
| | - Katharina Grewe
- Department for Neuro-and Sensory Physiology, University Medical Center Göttingen, Center for Biostructural Imaging of Neurodegeneration (BIN), Göttingen, Germany
| | - Peter Ilgen
- Clinic of Neurology, University Medical Center Göttingen, Göttingen, Germany
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Translational Neuroinflammation and Automated Microscopy TNM, Göttingen, Germany
| | - Mehmet Gültas
- Faculty of Agriculture, South Westphalia University of Applied Sciences, Soest, Germany
| | - Eugenio F. Fornasiero
- Department for Neuro-and Sensory Physiology, University Medical Center Göttingen, Center for Biostructural Imaging of Neurodegeneration (BIN), Göttingen, Germany
| | - Stefan Jakobs
- Clinic of Neurology, University Medical Center Göttingen, Göttingen, Germany
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Translational Neuroinflammation and Automated Microscopy TNM, Göttingen, Germany
| | - Wiebke Möbius
- Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, Göttingen, Germany
- Electron Microscopy-City Campus, Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Christian Vogl
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
- Presynaptogenesis and Intracellular Transport in Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
| | - Tina Pangršič
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
- Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, Göttingen, Germany
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany
| | - Silvio O. Rizzoli
- Department for Neuro-and Sensory Physiology, University Medical Center Göttingen, Center for Biostructural Imaging of Neurodegeneration (BIN), Göttingen, Germany
| | - Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, InnerEarLab and Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
- Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
12
|
van der Valk WH, van Beelen ESA, Steinhart MR, Nist-Lund C, Osorio D, de Groot JCMJ, Sun L, van Benthem PPG, Koehler KR, Locher H. A single-cell level comparison of human inner ear organoids with the human cochlea and vestibular organs. Cell Rep 2023; 42:112623. [PMID: 37289589 PMCID: PMC10592453 DOI: 10.1016/j.celrep.2023.112623] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 02/21/2023] [Accepted: 05/23/2023] [Indexed: 06/10/2023] Open
Abstract
Inner ear disorders are among the most common congenital abnormalities; however, current tissue culture models lack the cell type diversity to study these disorders and normal otic development. Here, we demonstrate the robustness of human pluripotent stem cell-derived inner ear organoids (IEOs) and evaluate cell type heterogeneity by single-cell transcriptomics. To validate our findings, we construct a single-cell atlas of human fetal and adult inner ear tissue. Our study identifies various cell types in the IEOs including periotic mesenchyme, type I and type II vestibular hair cells, and developing vestibular and cochlear epithelium. Many genes linked to congenital inner ear dysfunction are confirmed to be expressed in these cell types. Additional cell-cell communication analysis within IEOs and fetal tissue highlights the role of endothelial cells on the developing sensory epithelium. These findings provide insights into this organoid model and its potential applications in studying inner ear development and disorders.
Collapse
Affiliation(s)
- Wouter H van der Valk
- OtoBiology Leiden, Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, 2333 ZA Leiden, the Netherlands; Department of Otolaryngology, Boston Children's Hospital, Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02115, USA.
| | - Edward S A van Beelen
- OtoBiology Leiden, Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Matthew R Steinhart
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Carl Nist-Lund
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel Osorio
- Research Computing, Department of Information Technology, Boston Children's Hospital, Boston, MA 02115, USA
| | - John C M J de Groot
- OtoBiology Leiden, Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Liang Sun
- Research Computing, Department of Information Technology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Peter Paul G van Benthem
- OtoBiology Leiden, Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Karl R Koehler
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02115, USA; Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA 02115, USA.
| | - Heiko Locher
- OtoBiology Leiden, Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands; The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, 2333 ZA Leiden, the Netherlands.
| |
Collapse
|
13
|
Giunta R, Cheli G, Spaiardi P, Russo G, Masetto S. Pimozide Increases a Delayed Rectifier K + Conductance in Chicken Embryo Vestibular Hair Cells. Biomedicines 2023; 11:biomedicines11020488. [PMID: 36831024 PMCID: PMC9953418 DOI: 10.3390/biomedicines11020488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Pimozide is a conventional antipsychotic drug largely used in the therapy for schizophrenia and Tourette's syndrome. Pimozide is assumed to inhibit synaptic transmission at the CNS by acting as a dopaminergic D2 receptor antagonist. Moreover, pimozide has been shown to block voltage-gated Ca2+ and K+ channels in different cells. Despite its widespread clinical use, pimozide can cause several adverse effects, including extrapyramidal symptoms and cardiac arrhythmias. Dizziness and loss of balance are among the most common side effects of pimozide. By using the patch-clamp whole-cell technique, we investigated the effect of pimozide [3 μM] on K+ channels expressed by chicken embryo vestibular type-II hair cells. We found that pimozide slightly blocks a transient outward rectifying A-type K+ current but substantially increases a delayed outward rectifying K+ current. The net result was a significant hyperpolarization of type-II hair cells at rest and a strong reduction of their response to depolarizing stimuli. Our findings are consistent with an inhibitory effect of pimozide on the afferent synaptic transmission by type-II hair cells. Moreover, they provide an additional key to understanding the beneficial/collateral pharmacological effects of pimozide. The finding that pimozide can act as a K+ channel opener provides a new perspective for the use of this drug.
Collapse
Affiliation(s)
- Roberta Giunta
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| | - Giulia Cheli
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| | - Paolo Spaiardi
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
| | - Giancarlo Russo
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| | - Sergio Masetto
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
- Correspondence:
| |
Collapse
|
14
|
Shi T, Beaulieu MO, Saunders LM, Fabian P, Trapnell C, Segil N, Crump JG, Raible DW. Single-cell transcriptomic profiling of the zebrafish inner ear reveals molecularly distinct hair cell and supporting cell subtypes. eLife 2023; 12:82978. [PMID: 36598134 PMCID: PMC9851615 DOI: 10.7554/elife.82978] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 01/04/2023] [Indexed: 01/05/2023] Open
Abstract
A major cause of human deafness and vestibular dysfunction is permanent loss of the mechanosensory hair cells of the inner ear. In non-mammalian vertebrates such as zebrafish, regeneration of missing hair cells can occur throughout life. While a comparative approach has the potential to reveal the basis of such differential regenerative ability, the degree to which the inner ears of fish and mammals share common hair cells and supporting cell types remains unresolved. Here, we perform single-cell RNA sequencing of the zebrafish inner ear at embryonic through adult stages to catalog the diversity of hair cells and non-sensory supporting cells. We identify a putative progenitor population for hair cells and supporting cells, as well as distinct hair and supporting cell types in the maculae versus cristae. The hair cell and supporting cell types differ from those described for the lateral line system, a distributed mechanosensory organ in zebrafish in which most studies of hair cell regeneration have been conducted. In the maculae, we identify two subtypes of hair cells that share gene expression with mammalian striolar or extrastriolar hair cells. In situ hybridization reveals that these hair cell subtypes occupy distinct spatial domains within the three macular organs, the utricle, saccule, and lagena, consistent with the reported distinct electrophysiological properties of hair cells within these domains. These findings suggest that primitive specialization of spatially distinct striolar and extrastriolar hair cells likely arose in the last common ancestor of fish and mammals. The similarities of inner ear cell type composition between fish and mammals validate zebrafish as a relevant model for understanding inner ear-specific hair cell function and regeneration.
Collapse
Affiliation(s)
- Tuo Shi
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Marielle O Beaulieu
- Department of Otolaryngology-Head and Neck Surgery, University of WashingtonSeattleUnited States
| | - Lauren M Saunders
- Department of Genome Sciences, University of WashingtonSeattleUnited States
| | - Peter Fabian
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Cole Trapnell
- Department of Genome Sciences, University of WashingtonSeattleUnited States
| | - Neil Segil
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - J Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - David W Raible
- Department of Otolaryngology-Head and Neck Surgery, University of WashingtonSeattleUnited States
- Department of Genome Sciences, University of WashingtonSeattleUnited States
- Department of Biological Structure, University of WashingtonSeattleUnited States
| |
Collapse
|
15
|
Ciani Berlingeri AN, Pujol R, Cox BC, Stone JS. Sox2 is required in supporting cells for normal levels of vestibular hair cell regeneration in adult mice. Hear Res 2022; 426:108642. [PMID: 36334348 DOI: 10.1016/j.heares.2022.108642] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 09/16/2022] [Accepted: 10/19/2022] [Indexed: 11/04/2022]
Abstract
Sox2 is a transcription factor that is necessary in the mammalian inner ear for development of sensory hair cells and supporting cells. Sox2 is expressed in supporting cells of adult mammals, but its function in this context is poorly understood. Given its role in the developing inner ear, we hypothesized that Sox2 is required in vestibular supporting cells for regeneration of type II hair cells after damage. Using adult mice, we deleted Sox2 from Sox9-CreER-expressing supporting cells prior to diphtheria toxin-mediated hair cell destruction and used fate-mapping to assess regeneration. In utricles of control mice with normal Sox2 expression, supporting cells regenerated nearly 200 hair cells by 3 weeks post-damage, which doubled by 12 weeks. In contrast, mice with Sox2 deletion from supporting cells had approximately 20 fate-mapped hair cells at 3 weeks post-damage, and this number did not change significantly by 12 weeks, indicating regeneration was dramatically curtailed. We made similar observations for saccules and ampullae. We found no evidence that supporting cells lacking Sox2 had altered cellular density, morphology, or ultrastructure. However, some Sox2-negative supporting cell nuclei appeared to migrate apically but did not turn on hair cell markers, and type I hair cell survival was higher. Sox2 heterozygotes also had reduced regeneration in utricles, but more hair cells were replaced than mice with Sox2 deletion. Our study determined that Sox2 is required in supporting cells for normal levels of vestibular hair cell regeneration but found no other major requirements for Sox2 in adult supporting cells.
Collapse
Affiliation(s)
- Amanda N Ciani Berlingeri
- Department of Speech and Hearing Sciences, University of Washington, Seattle, Washington, United States; Department of Otolaryngology-Head and Neck Surgery and the Virginia Merrill Bloedel Research Center, University of Washington School of Medicine, Seattle, Washington, United States
| | - Rémy Pujol
- University of Montpellier, INM-INSERM Unit 1298, Montpellier, France
| | - Brandon C Cox
- Departments of Pharmacology and Otolaryngology, Southern Illinois University School of Medicine, Springfield, Illinois, United States
| | - Jennifer S Stone
- Department of Otolaryngology-Head and Neck Surgery and the Virginia Merrill Bloedel Research Center, University of Washington School of Medicine, Seattle, Washington, United States.
| |
Collapse
|
16
|
You D, Guo J, Zhang Y, Guo L, Lu X, Huang X, Sun S, Li H. The heterogeneity of mammalian utricular cells over the course of development. Clin Transl Med 2022; 12:e1052. [PMID: 36178017 PMCID: PMC9523683 DOI: 10.1002/ctm2.1052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 08/19/2022] [Accepted: 08/25/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND The inner ear organ is a delicate tissue consisting of hair cells (HCs) and supporting cells (SCs).The mammalian inner ear HCs are terminally differentiated cells that cannot spontaneously regenerate in adults. Epithelial non-hair cells (ENHCs) in the utricle include HC progenitors and SCs, and the progenitors share similar characteristics with SCs in the neonatal inner ear. METHODS We applied single-cell sequencing to whole mouse utricles from the neonatal period to adulthood, including samples from postnatal day (P)2, P7 and P30 mice. Furthermore, using transgenic mice and immunostaining, we traced the source of new HC generation. RESULTS We identified several sensory epithelial cell clusters and further found that new HCs arose mainly through differentiation from Sox9+ progenitor cells and that only a few cells were produced by mitotic proliferation in both neonatal and adult mouse utricles. In addition, we identified the proliferative cells using the marker UbcH10 and demonstrated that in adulthood the mitotically generated HCs were primarily found in the extrastriola. Moreover, we observed that not only Type II, but also Type I HCs could be regenerated by either mitotic cell proliferation or progenitor cell differentiation. CONCLUSIONS Overall, our findings expand our understanding of ENHC cell fate and the characteristics of the vestibular organs in mammals over the course of development.
Collapse
Affiliation(s)
- Dan You
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina,Department of Otorhinolaryngology‐Head and Neck SurgeryZhongshan HospitalFudan UniversityShanghaiChina
| | - Jin Guo
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
| | - Yunzhong Zhang
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
| | - Luo Guo
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
| | - Xiaoling Lu
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
| | - Xinsheng Huang
- Department of Otorhinolaryngology‐Head and Neck SurgeryZhongshan HospitalFudan UniversityShanghaiChina
| | - Shan Sun
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
| | - Huawei Li
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina,Institutes of Biomedical SciencesFudan UniversityShanghaiChina,NHC Key Laboratory of Hearing Medicine, Fudan UniversityShanghaiChina,The Institutes of Brain Science and the Collaborative Innovation Center for Brain ScienceFudan UniversityShanghaiChina
| |
Collapse
|
17
|
Mukhopadhyay M, Pangrsic T. Synaptic transmission at the vestibular hair cells of amniotes. Mol Cell Neurosci 2022; 121:103749. [PMID: 35667549 DOI: 10.1016/j.mcn.2022.103749] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 05/09/2022] [Accepted: 06/01/2022] [Indexed: 11/19/2022] Open
Abstract
A harmonized interplay between the central nervous system and the five peripheral end organs is how the vestibular system helps organisms feel a sense of balance and motion in three-dimensional space. The receptor cells of this system, much like their cochlear equivalents, are the specialized hair cells. However, research over the years has shown that the vestibular endorgans and hair cells evolved very differently from their cochlear counterparts. The structurally unique calyceal synapse, which appeared much later in the evolutionary time scale, and continues to intrigue researchers, is now known to support several forms of synaptic neurotransmission. The conventional quantal transmission is believed to employ the ribbon structures, which carry several tethered vesicles filled with neurotransmitters. However, the field of vestibular hair cell synaptic molecular anatomy is still at a nascent stage and needs further work. In this review, we will touch upon the basic structure and function of the peripheral vestibular system, with the focus on the various modes of neurotransmission at the type I vestibular hair cells. We will also shed light on the current knowledge about the molecular anatomy of the vestibular hair cell synapses and vestibular synaptopathy.
Collapse
Affiliation(s)
- Mohona Mukhopadhyay
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, and Institute for Auditory Neuroscience, 37075 Göttingen, Germany
| | - Tina Pangrsic
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, and Institute for Auditory Neuroscience, 37075 Göttingen, Germany; Auditory Neuroscience Group, Max Planck Institute for Multidisciplinary Sciences, 37075 Göttingen, Germany; Collaborative Research Center 889, University of Göttingen, Göttingen, Germany; Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, 37075 Göttingen, Germany.
| |
Collapse
|
18
|
Kim GS, Wang T, Sayyid ZN, Fuhriman J, Jones SM, Cheng AG. Repair of surviving hair cells in the damaged mouse utricle. Proc Natl Acad Sci U S A 2022; 119:e2116973119. [PMID: 35380897 PMCID: PMC9169652 DOI: 10.1073/pnas.2116973119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 02/21/2022] [Indexed: 11/18/2022] Open
Abstract
Sensory hair cells (HCs) in the utricle are mechanoreceptors required to detect linear acceleration. After damage, the mammalian utricle partially restores the HC population and organ function, although regenerated HCs are primarily type II and immature. Whether native, surviving HCs can repair and contribute to this recovery is unclear. Here, we generated the Pou4f3DTR/+; Atoh1CreERTM/+; Rosa26RtdTomato/+ mouse to fate map HCs prior to ablation. After HC ablation, vestibular evoked potentials were abolished in all animals, with ∼57% later recovering responses. Relative to nonrecovery mice, recovery animals harbored more Atoh1-tdTomato+ surviving HCs. In both groups, surviving HCs displayed markers of both type I and type II subtypes and afferent synapses, despite distorted lamination and morphology. Surviving type II HCs remained innervated in both groups, whereas surviving type I HCs first lacked and later regained calyces in the recovery, but not the nonrecovery, group. Finally, surviving HCs initially displayed immature and subsequently mature-appearing bundles in the recovery group. These results demonstrate that surviving HCs are capable of self-repair and may contribute to the recovery of vestibular function.
Collapse
Affiliation(s)
- Grace S. Kim
- Department of Otolaryngology–Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305
| | - Tian Wang
- Department of Otolaryngology–Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305
| | - Zahra N. Sayyid
- Department of Otolaryngology–Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305
| | - Jessica Fuhriman
- Department of Otolaryngology–Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305
| | - Sherri M. Jones
- Department of Special Education and Communication Disorders, College of Education and Human Sciences, University of Nebraska, Lincoln, NE 68583
| | - Alan G. Cheng
- Department of Otolaryngology–Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
19
|
Maudoux A, Vitry S, El-Amraoui A. Vestibular Deficits in Deafness: Clinical Presentation, Animal Modeling, and Treatment Solutions. Front Neurol 2022; 13:816534. [PMID: 35444606 PMCID: PMC9013928 DOI: 10.3389/fneur.2022.816534] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
The inner ear is responsible for both hearing and balance. These functions are dependent on the correct functioning of mechanosensitive hair cells, which convert sound- and motion-induced stimuli into electrical signals conveyed to the brain. During evolution of the inner ear, the major changes occurred in the hearing organ, whereas the structure of the vestibular organs remained constant in all vertebrates over the same period. Vestibular deficits are highly prevalent in humans, due to multiple intersecting causes: genetics, environmental factors, ototoxic drugs, infections and aging. Studies of deafness genes associated with balance deficits and their corresponding animal models have shed light on the development and function of these two sensory systems. Bilateral vestibular deficits often impair individual postural control, gaze stabilization, locomotion and spatial orientation. The resulting dizziness, vertigo, and/or falls (frequent in elderly populations) greatly affect patient quality of life. In the absence of treatment, prosthetic devices, such as vestibular implants, providing information about the direction, amplitude and velocity of body movements, are being developed and have given promising results in animal models and humans. Novel methods and techniques have led to major progress in gene therapies targeting the inner ear (gene supplementation and gene editing), 3D inner ear organoids and reprograming protocols for generating hair cell-like cells. These rapid advances in multiscale approaches covering basic research, clinical diagnostics and therapies are fostering interdisciplinary research to develop personalized treatments for vestibular disorders.
Collapse
Affiliation(s)
- Audrey Maudoux
- Unit Progressive Sensory Disorders, Pathophysiology and Therapy, Institut Pasteur, Institut de l'Audition, Université de Paris, INSERM-UMRS1120, Paris, France
- Center for Balance Evaluation in Children (EFEE), Otolaryngology Department, Assistance Publique des Hôpitaux de Paris, Robert-Debré University Hospital, Paris, France
| | - Sandrine Vitry
- Unit Progressive Sensory Disorders, Pathophysiology and Therapy, Institut Pasteur, Institut de l'Audition, Université de Paris, INSERM-UMRS1120, Paris, France
| | - Aziz El-Amraoui
- Unit Progressive Sensory Disorders, Pathophysiology and Therapy, Institut Pasteur, Institut de l'Audition, Université de Paris, INSERM-UMRS1120, Paris, France
| |
Collapse
|
20
|
Huang Y, Mao H, Chen Y. Regeneration of Hair Cells in the Human Vestibular System. Front Mol Neurosci 2022; 15:854635. [PMID: 35401109 PMCID: PMC8987309 DOI: 10.3389/fnmol.2022.854635] [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: 01/14/2022] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
The vestibular system is a critical part of the human balance system, malfunction of this system will lead to balance disorders, such as vertigo. Mammalian vestibular hair cells, the mechanical receptors for vestibular function, are sensitive to ototoxic drugs and virus infection, and have a limited restorative capacity after damage. Considering that no artificial device can be used to replace vestibular hair cells, promoting vestibular hair cell regeneration is an ideal way for vestibular function recovery. In this manuscript, the development of human vestibular hair cells during the whole embryonic stage and the latest research on human vestibular hair cell regeneration is summarized. The limitations of current studies are emphasized and future directions are discussed.
Collapse
Affiliation(s)
- Yikang Huang
- State Key Laboratory of Medical Neurobiology, Department of Otorhinolaryngology, Eye and ENT Hospital, MOE Frontiers Center for Brain Science, ENT Institute, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Huanyu Mao
- State Key Laboratory of Medical Neurobiology, Department of Otorhinolaryngology, Eye and ENT Hospital, MOE Frontiers Center for Brain Science, ENT Institute, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Yan Chen
- State Key Laboratory of Medical Neurobiology, Department of Otorhinolaryngology, Eye and ENT Hospital, MOE Frontiers Center for Brain Science, ENT Institute, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
- *Correspondence: Yan Chen,
| |
Collapse
|
21
|
González-Garrido A, Pujol R, López-Ramírez O, Finkbeiner C, Eatock RA, Stone JS. The Differentiation Status of Hair Cells That Regenerate Naturally in the Vestibular Inner Ear of the Adult Mouse. J Neurosci 2021; 41:7779-7796. [PMID: 34301830 PMCID: PMC8445055 DOI: 10.1523/jneurosci.3127-20.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 07/07/2021] [Accepted: 07/19/2021] [Indexed: 11/21/2022] Open
Abstract
Aging, disease, and trauma can lead to loss of vestibular hair cells and permanent vestibular dysfunction. Previous work showed that, following acute destruction of ∼95% of vestibular hair cells in adult mice, ∼20% regenerate naturally (without exogenous factors) through supporting cell transdifferentiation. There is, however, no evidence for the recovery of vestibular function. To gain insight into the lack of functional recovery, we assessed functional differentiation in regenerated hair cells for up to 15 months, focusing on key stages in stimulus transduction and transmission: hair bundles, voltage-gated conductances, and synaptic contacts. Regenerated hair cells had many features of mature type II vestibular hair cells, including polarized mechanosensitive hair bundles with zone-appropriate stereocilia heights, large voltage-gated potassium currents, basolateral processes, and afferent and efferent synapses. Regeneration failed, however, to recapture the full range of properties of normal populations, and many regenerated hair cells had some properties of immature hair cells, including small transduction currents, voltage-gated sodium currents, and small or absent HCN (hyperpolarization-activated cyclic nucleotide-gated) currents. Furthermore, although mouse vestibular epithelia normally have slightly more type I hair cells than type II hair cells, regenerated hair cells acquired neither the low-voltage-activated potassium channels nor the afferent synaptic calyces that distinguish mature type I hair cells from type II hair cells and confer distinctive physiology. Thus, natural regeneration of vestibular hair cells in adult mice is limited in total cell number, cell type diversity, and extent of cellular differentiation, suggesting that manipulations are needed to promote full regeneration with the potential for recovery of vestibular function.SIGNIFICANCE STATEMENT Death of inner ear hair cells in adult mammals causes permanent loss of hearing and balance. In adult mice, the sudden death of most vestibular hair cells stimulates the production of new hair cells but does not restore balance. We investigated whether the lack of systems-level function reflects functional deficiencies in the regenerated hair cells. The regenerated population acquired mechanosensitivity, voltage-gated channels, and afferent synapses, but did not reproduce the full range of hair cell types. Notably, no regenerated cells acquired the distinctive properties of type I hair cells, a major functional class in amniote vestibular organs. To recover vestibular system function in adults, we may need to solve how to regenerate the normal variety of mature hair cells.
Collapse
Affiliation(s)
| | - Rémy Pujol
- The Virginia Merrill Bloedel Hearing Research Center and the Department of Otolaryngology Head and Neck Surgery, University of Washington, Seattle, Washington 98195
- Institute for Neurosciences of Montpellier-Institut National de la Santé et de la Recherche Médicale Unit 1052, University of Montpellier, 34091 Montpellier, France
| | - Omar López-Ramírez
- Department of Neurobiology, University of Chicago, Chicago, Illinois 60637
| | - Connor Finkbeiner
- The Virginia Merrill Bloedel Hearing Research Center and the Department of Otolaryngology Head and Neck Surgery, University of Washington, Seattle, Washington 98195
| | - Ruth Anne Eatock
- Department of Neurobiology, University of Chicago, Chicago, Illinois 60637
| | - Jennifer S Stone
- The Virginia Merrill Bloedel Hearing Research Center and the Department of Otolaryngology Head and Neck Surgery, University of Washington, Seattle, Washington 98195
| |
Collapse
|
22
|
Jan TA, Eltawil Y, Ling AH, Chen L, Ellwanger DC, Heller S, Cheng AG. Spatiotemporal dynamics of inner ear sensory and non-sensory cells revealed by single-cell transcriptomics. Cell Rep 2021; 36:109358. [PMID: 34260939 PMCID: PMC8378666 DOI: 10.1016/j.celrep.2021.109358] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 12/25/2020] [Accepted: 06/17/2021] [Indexed: 11/28/2022] Open
Abstract
The utricle is a vestibular sensory organ that requires mechanosensitive hair cells to detect linear acceleration. In neonatal mice, new hair cells are derived from non-sensory supporting cells, yet cell type diversity and mechanisms of cell addition remain poorly characterized. Here, we perform computational analyses on single-cell transcriptomes to categorize cell types and resolve 14 individual sensory and non-sensory subtypes. Along the periphery of the sensory epithelium, we uncover distinct groups of transitional epithelial cells, marked by Islr, Cnmd, and Enpep expression. By reconstructing de novo trajectories and gene dynamics, we show that as the utricle expands, Islr+ transitional epithelial cells exhibit a dynamic and proliferative phase to generate new supporting cells, followed by coordinated differentiation into hair cells. Taken together, our study reveals a sequential and coordinated process by which non-sensory epithelial cells contribute to growth of the postnatal mouse sensory epithelium. The postnatal mouse utricle expands by more than 35% and doubles its number of hair cells during the first 8 days. Using single-cell transcriptomics, Jan et al. show that the surrounding transitional epithelial cells proliferate and contribute to the expansion of the sensory epithelium through a stepwise differentiation mechanism.
Collapse
Affiliation(s)
- Taha A Jan
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA; Department of Otolaryngology-Head and Neck Surgery, University of California San Francisco, San Francisco, CA 94115, USA
| | - Yasmin Eltawil
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Angela H Ling
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA; Department of Otolaryngology-Head and Neck Surgery, University of California San Francisco, San Francisco, CA 94115, USA
| | - Leon Chen
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Daniel C Ellwanger
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA; Genome Analysis Unit, Amgen Research, Amgen Inc., South San Francisco, CA 94080, USA
| | - Stefan Heller
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA.
| | - Alan G Cheng
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA.
| |
Collapse
|
23
|
Wilkerson BA, Zebroski HL, Finkbeiner CR, Chitsazan AD, Beach KE, Sen N, Zhang RC, Bermingham-McDonogh O. Novel cell types and developmental lineages revealed by single-cell RNA-seq analysis of the mouse crista ampullaris. eLife 2021; 10:e60108. [PMID: 34003106 PMCID: PMC8189719 DOI: 10.7554/elife.60108] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 05/17/2021] [Indexed: 12/16/2022] Open
Abstract
This study provides transcriptomic characterization of the cells of the crista ampullaris, sensory structures at the base of the semicircular canals that are critical for vestibular function. We performed single-cell RNA-seq on ampullae microdissected from E16, E18, P3, and P7 mice. Cluster analysis identified the hair cells, support cells and glia of the crista as well as dark cells and other nonsensory epithelial cells of the ampulla, mesenchymal cells, vascular cells, macrophages, and melanocytes. Cluster-specific expression of genes predicted their spatially restricted domains of gene expression in the crista and ampulla. Analysis of cellular proportions across developmental time showed dynamics in cellular composition. The new cell types revealed by single-cell RNA-seq could be important for understanding crista function and the markers identified in this study will enable the examination of their dynamics during development and disease.
Collapse
Affiliation(s)
- Brent A Wilkerson
- Department of Biological Structure, University of WashingtonSeattleUnited States
- Institute for Stem Cells and Regenerative Medicine, University of WashingtonSeattleUnited States
| | - Heather L Zebroski
- Department of Biological Structure, University of WashingtonSeattleUnited States
- Institute for Stem Cells and Regenerative Medicine, University of WashingtonSeattleUnited States
| | - Connor R Finkbeiner
- Department of Biological Structure, University of WashingtonSeattleUnited States
- Institute for Stem Cells and Regenerative Medicine, University of WashingtonSeattleUnited States
| | - Alex D Chitsazan
- Department of Biological Structure, University of WashingtonSeattleUnited States
- Institute for Stem Cells and Regenerative Medicine, University of WashingtonSeattleUnited States
- Department of Biochemistry, University of WashingtonSeattleUnited States
| | - Kylie E Beach
- Department of Biological Structure, University of WashingtonSeattleUnited States
- Institute for Stem Cells and Regenerative Medicine, University of WashingtonSeattleUnited States
| | - Nilasha Sen
- Department of Biological Structure, University of WashingtonSeattleUnited States
| | - Renee C Zhang
- Department of Biological Structure, University of WashingtonSeattleUnited States
| | - Olivia Bermingham-McDonogh
- Department of Biological Structure, University of WashingtonSeattleUnited States
- Institute for Stem Cells and Regenerative Medicine, University of WashingtonSeattleUnited States
| |
Collapse
|
24
|
Qian X, He Z, Wang Y, Chen B, Hetrick A, Dai C, Chi F, Li H, Ren D. Hair cell uptake of gentamicin in the developing mouse utricle. J Cell Physiol 2020; 236:5235-5252. [PMID: 33368220 DOI: 10.1002/jcp.30228] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 11/27/2020] [Accepted: 12/08/2020] [Indexed: 12/27/2022]
Abstract
Intratympanic injection of gentamicin has proven to be an effective therapy for intractable vestibular dysfunction. However, most studies to date have focused on the cochlea, so little is known about the distribution and uptake of gentamicin by the counterpart of the auditory system, specifically vestibular hair cells (HCs). Here, with a combination of in vivo and in vitro approaches, we used a gentamicin-Texas Red (GTTR) conjugate to investigate the mechanisms of gentamicin vestibulotoxicity in the developing mammalian utricular HCs. In vivo, GTTR fluorescence was concentrated in the apical cytoplasm and the cellular membrane of neonatal utricular HCs, but scarce in the nucleus of HCs and supporting cells. Quantitative analysis showed the GTTR uptake by striolar HCs was significantly higher than that in the extrastriola. In addition, the GTTR fluorescence intensity in the striola was increased gradually from 1 to 8 days, peaking at 8-9 days postnatally. In vitro, utricle explants were incubated with GTTR and candidate uptake conduits, including mechanotransduction (MET) channels and endocytosis in the HC, were inhibited separately. GTTR uptake by HCs could be inhibited by quinine, a blocker of MET channels, under both normal and stressed conditions. Meanwhile, endocytic inhibition only reduced GTTR uptake in the CoCl2 hypoxia model. In sum, the maturation of MET channels mediated uptake of GTTR into vestibular HCs. Under stressed conditions, MET channels play a pronounced role, manifested by channel-dependent stress enhanced GTTR permeation, while endocytosis participates in GTTR entry in a more selective manner.
Collapse
Affiliation(s)
- Xiaoqing Qian
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, China
| | - Ziyu He
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, China
| | - Yanmei Wang
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, China
| | - Binjun Chen
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, China
| | - Alisa Hetrick
- Research Service, VA Loma Linda Healthcare System, Loma Linda, California, USA
| | - Chunfu Dai
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, China
| | - Fanglu Chi
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, China
| | - Hongzhe Li
- Research Service, VA Loma Linda Healthcare System, Loma Linda, California, USA.,Department of Otolaryngology-Head and Neck Surgery, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Dongdong Ren
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, China
| |
Collapse
|
25
|
Qian X, Ma R, Wang X, Xu X, Yang J, Chi F, Ren D. Simultaneous gentamicin-mediated damage and Atoh1 overexpression promotes hair cell regeneration in the neonatal mouse utricle. Exp Cell Res 2020; 398:112395. [PMID: 33279477 DOI: 10.1016/j.yexcr.2020.112395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 11/17/2020] [Accepted: 11/21/2020] [Indexed: 01/24/2023]
Abstract
Loss of hair cells from vestibular epithelium results in balance dysfunction. The current therapeutic regimen for vestibular diseases is limited. Upon injury or Atoh1 overexpression, hair cell replacement occurs rapidly in the mammalian utricle, suggesting a promising approach to induce vestibular hair cell regeneration. In this study, we applied simultaneous gentamicin-mediated hair cell ablation and Atoh1 overexpression to induce neonatal utricular hair cell formation in vitro. We confirmed that type I hair cells were the primary targets of gentamicin. Furthermore, injury and Atoh1 overexpression promoted hair cell regeneration in a timely and efficient manner through robust viral transfection. Hair cells regenerated with type II characteristics in the striola and type I/II characteristics in non-sensory regions. Rare EdU+/myosin7a+ cells in sensory regions and robust EdU+/myosin7a+ signals in ectopic regions indicate that transdifferentiation of supporting cells in situ, and mitosis and differentiation of non-sensory epithelial cells in ectopic regions, are sources of regenerative hair cells. Distinct regeneration patterns in in situ and ectopic regions suggested robust plasticity of vestibular non-sensory epithelium, generating more developed hair cell subtypes and thus providing a promising stem cell-like source of hair cells. These findings suggest that simultaneously causing injury and overexpressing Atoh1 promotes hair cell regeneration efficacy and maturity, thus expanding the understanding of ectopic plasticity in neonatal vestibular organs.
Collapse
Affiliation(s)
- Xiaoqing Qian
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, China
| | - Rui Ma
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, China
| | - Xinwei Wang
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, China
| | - Xinda Xu
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, China
| | - Juanmei Yang
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, China.
| | - Fanglu Chi
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, China.
| | - Dongdong Ren
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, China.
| |
Collapse
|
26
|
Ratzan EM, Moon AM, Deans MR. Fgf8 genetic labeling reveals the early specification of vestibular hair cell type in mouse utricle. Development 2020; 147:dev.192849. [PMID: 33046506 DOI: 10.1242/dev.192849] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 10/07/2020] [Indexed: 01/16/2023]
Abstract
FGF8 signaling plays diverse roles in inner ear development, acting at multiple stages from otic placode induction to cellular differentiation in the organ of Corti. As a secreted morphogen with diverse functions, Fgf8 expression is likely to be spatially restricted and temporally dynamic throughout inner ear development. We evaluated these characteristics using genetic labeling mediated by Fgf8 mcm gene-targeted mice and determined that Fgf8 expression is a specific and early marker of Type-I vestibular hair cell identity. Fgf8 mcm expression initiates at E11.5 in the future striolar region of the utricle, labeling hair cells following EdU birthdating, and demonstrates that sub-type identity is determined shortly after terminal mitosis. This early fate specification is not apparent using markers or morphological criteria that are not present before birth in the mouse. Although analyses of Fgf8 conditional knockout mice did not reveal developmental phenotypes, the restricted pattern of Fgf8 expression suggests that functionally redundant FGF ligands may contribute to vestibular hair cell differentiation and supports a developmental model in which Type-I and Type-II hair cells develop in parallel rather than from an intermediate precursor.
Collapse
Affiliation(s)
- Evan M Ratzan
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.,Interdepartmental Program in Neuroscience, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Anne M Moon
- Departments of Molecular and Functional Genomics and Pediatrics, Weis Center for Research, Geisinger Clinic and Geisinger Commonwealth School of Medicine, Danville, PA 17822, USA.,Departments of Pediatrics and Human Genetics, University of Utah, Salt Lake City, UT 84112 USA
| | - Michael R Deans
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA .,Department of Surgery, Division of Otolaryngology, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| |
Collapse
|
27
|
Ramakrishna Y, Manca M, Glowatzki E, Sadeghi SG. Cholinergic Modulation of Membrane Properties of Calyx Terminals in the Vestibular Periphery. Neuroscience 2020; 452:98-110. [PMID: 33197502 DOI: 10.1016/j.neuroscience.2020.10.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 01/10/2023]
Abstract
Vestibular nerve afferents are divided into regular and irregular groups based on the variability of interspike intervals in their resting discharge. Most afferents receive inputs from bouton terminals that contact type II hair cells as well as from calyx terminals that cover the basolateral walls of type I hair cells. Calyces have an abundance of different subtypes of KCNQ (Kv7) potassium channels and muscarinic acetylcholine receptors (mAChRs) and receive cholinergic efferent inputs from neurons in the brainstem. We investigated whether mAChRs affected membrane properties and firing patterns of calyx terminals through modulation of KCNQ channel activity. Patch clamp recordings were performed from calyx terminals in central regions of the cristae of the horizontal and anterior canals in 13-26 day old Sprague-Dawley rats. KCNQ mediated currents were observed as voltage sensitive currents with slow kinetics (activation and deactivation), resulting in spike frequency adaptation so that calyces at best fired a single action potential at the beginning of a depolarizing step. Activation of mAChRs by application of oxotremorine methiodide or inhibition of KCNQ channels by linopirdine dihydrochloride decreased voltage activated currents by ∼30%, decreased first spike latencies by ∼40%, resulted in action potential generation in response to smaller current injections and at lower (i.e., more hyperpolarized) membrane potentials, and increased the number of spikes fired during depolarizing steps. Interestingly, some of the calyces showed spontaneous discharge in the presence of these drugs. Together, these findings suggest that cholinergic efferents can modulate the response properties and encoding of head movements by afferents.
Collapse
Affiliation(s)
- Yugandhar Ramakrishna
- Center for Hearing and Deafness, Department of Communicative Disorders and Sciences, State University of New York at Buffalo, Buffalo, NY, United States; Department of Communication Disorders and Sciences, California State University, Northridge, CA, United States
| | - Marco Manca
- Department of Otolaryngology - Head and Neck Surgery, The Center for Hearing and Balance, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Elisabeth Glowatzki
- Department of Otolaryngology - Head and Neck Surgery, The Center for Hearing and Balance, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Soroush G Sadeghi
- Center for Hearing and Deafness, Department of Communicative Disorders and Sciences, State University of New York at Buffalo, Buffalo, NY, United States; Neuroscience Program, State University of New York at Buffalo, Buffalo, NY, United States.
| |
Collapse
|
28
|
Ramakrishna Y, Sadeghi SG. Activation of GABA B receptors results in excitatory modulation of calyx terminals in rat semicircular canal cristae. J Neurophysiol 2020; 124:962-972. [PMID: 32816581 PMCID: PMC7509296 DOI: 10.1152/jn.00243.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/15/2022] Open
Abstract
Previous studies have found GABA in vestibular end organs. However, existence of GABA receptors or possible GABAergic effects on vestibular nerve afferents has not been investigated. The current study was conducted to determine whether activation of GABAB receptors affects calyx afferent terminals in the central region of the cristae of semicircular canals. We used patch-clamp recording in postnatal day 13-18 (P13-P18) Sprague-Dawley rats of either sex. Application of GABAB receptor agonist baclofen inhibited voltage-sensitive potassium currents. This effect was blocked by selective GABAB receptor antagonist CGP 35348. Application of antagonists of small (SK)- and large-conductance potassium (BK) channels almost completely blocked the effects of baclofen. The remaining baclofen effect was blocked by cadmium chloride, suggesting that it could be due to inhibition of voltage-gated calcium channels. Furthermore, baclofen had no effect in the absence of calcium in the extracellular fluid. Inhibition of potassium currents by GABAB activation resulted in an excitatory effect on calyx terminal action potential firing. While in the control condition calyces could only fire a single action potential during step depolarizations, in the presence of baclofen they fired continuously during steps and a few even showed repetitive discharge. We also found a decrease in threshold for action potential generation and a decrease in first-spike latency during step depolarization. These results provide the first evidence for the presence of GABAB receptors on calyx terminals, showing that their activation results in an excitatory effect and that GABA inputs could be used to modulate calyx response properties.NEW & NOTEWORTHY Using in vitro whole cell patch-clamp recordings from calyx terminals in the vestibular end organs, we show that activation of GABAB receptors result in an excitatory effect, with decreased spike-frequency adaptation and shortened first-spike latencies. Our results suggest that these effects are mediated through inhibition of calcium-sensitive potassium channels.
Collapse
Affiliation(s)
- Yugandhar Ramakrishna
- Center for Hearing and Deafness, Department of Communicative Disorders and Sciences, State University of New York at Buffalo, Buffalo, New York
- Department of Communication Disorders and Sciences, California State University, Northridge, Northridge, California
| | - Soroush G Sadeghi
- Center for Hearing and Deafness, Department of Communicative Disorders and Sciences, State University of New York at Buffalo, Buffalo, New York
- Neuroscience Program, State University of New York at Buffalo, Buffalo, New York
| |
Collapse
|
29
|
Lentz JJ, Pan B, Ponnath A, Tran CM, Nist-Lund C, Galvin A, Goldberg H, Robillard KN, Jodelka FM, Farris HE, Huang J, Chen T, Zhu H, Zhou W, Rigo F, Hastings ML, Géléoc GSG. Direct Delivery of Antisense Oligonucleotides to the Middle and Inner Ear Improves Hearing and Balance in Usher Mice. Mol Ther 2020; 28:2662-2676. [PMID: 32818431 PMCID: PMC7704764 DOI: 10.1016/j.ymthe.2020.08.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/05/2020] [Accepted: 08/02/2020] [Indexed: 12/16/2022] Open
Abstract
Usher syndrome is a syndromic form of hereditary hearing impairment that includes sensorineural hearing loss and delayed-onset retinitis pigmentosa (RP). Type 1 Usher syndrome (USH1) is characterized by congenital profound sensorineural hearing impairment and vestibular areflexia, with adolescent-onset RP. Systemic treatment with antisense oligonucleotides (ASOs) targeting the human USH1C c.216G>A splicing mutation in a knockin mouse model of USH1 restores hearing and balance. Herein, we explore the effect of delivering ASOs locally to the ear to treat hearing and vestibular dysfunction associated with Usher syndrome. Three localized delivery strategies were investigated in USH1C mice: inner ear injection, trans-tympanic membrane injection, and topical tympanic membrane application. We demonstrate, for the first time, that ASOs delivered directly to the ear correct Ush1c expression in inner ear tissue, improve cochlear hair cell transduction currents, restore vestibular afferent irregularity, spontaneous firing rate, and sensitivity to head rotation, and successfully recover hearing thresholds and balance behaviors in USH1C mice. We conclude that local delivery of ASOs to the middle and inner ear reach hair cells and can rescue both hearing and balance. These results also demonstrate the therapeutic potential of ASOs to treat hearing and balance deficits associated with Usher syndrome and other ear diseases.
Collapse
Affiliation(s)
- Jennifer J Lentz
- Department of Otorhinolaryngology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| | - Bifeng Pan
- Department of Otolaryngology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Abhilash Ponnath
- Department of Otorhinolaryngology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Christopher M Tran
- Department of Otorhinolaryngology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Carl Nist-Lund
- Department of Otolaryngology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Alice Galvin
- Department of Otolaryngology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hannah Goldberg
- Department of Otolaryngology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Katelyn N Robillard
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Francine M Jodelka
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Hamilton E Farris
- Department of Otorhinolaryngology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Jun Huang
- Department of Otolaryngology and Communicative Sciences, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Tianwen Chen
- Department of Otolaryngology and Communicative Sciences, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Hong Zhu
- Department of Otolaryngology and Communicative Sciences, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Wu Zhou
- Department of Otolaryngology and Communicative Sciences, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Frank Rigo
- Ionis Pharmaceuticals, Inc., Carlsbad, CA 92008, USA
| | - Michelle L Hastings
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Gwenaëlle S G Géléoc
- Department of Otolaryngology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
30
|
Meredith FL, Rennie KJ. Persistent and resurgent Na + currents in vestibular calyx afferents. J Neurophysiol 2020; 124:510-524. [PMID: 32667253 DOI: 10.1152/jn.00124.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Vestibular afferent neurons convey information from hair cells in the peripheral vestibular end organs to central nuclei. Primary vestibular afferent neurons can fire action potentials at high rates and afferent firing patterns vary with the position of nerve terminal endings in vestibular neuroepithelia. Terminals contact hair cells as small bouton or large calyx endings. To investigate the role of Na+ currents (INa) in firing mechanisms, we investigated biophysical properties of INa in calyx-bearing afferents. Whole cell patch-clamp recordings were made from calyx terminals in thin slices of gerbil crista at different postnatal ages: immature [postnatal day (P)5-P8, young (P13-P15), and mature (P30-P45)]. A large transient Na+ current (INaT) was completely blocked by 300 nM tetrodotoxin (TTX) in mature calyces. In addition, INaT was accompanied by much smaller persistent Na+ currents (INaP) and distinctive resurgent Na+ currents (INaR), which were also blocked by TTX. ATX-II, a toxin that slows Na+ channel inactivation, enhanced INaP in immature and mature calyces. 4,9-Anhydro-TTX (4,9-ah-TTX), which selectively blocks Nav1.6 channels, abolished the enhanced INa in mature, but not immature, calyces. Therefore, Nav1.6 channels mediate a component of INaT and INaP in mature calyces, but are minimally expressed at early postnatal days. INaR was expressed in less than one-third of calyces at P6-P8, but expression increased with development, and in mature cristae INaR was frequently found in peripheral calyces. INaR served to increase the availability of Na+ channels following brief membrane depolarizations. In current clamp, the rate and regularity of action potential firing decreased in mature peripheral calyces following 4,9-ah-TTX application. Therefore, Nav1.6 channels are upregulated during development, contribute to INaT, INaP, and INaR, and may regulate excitability by enabling higher mean discharge rates in a subpopulation of mature calyx afferents.NEW & NOTEWORTHY Action potential firing patterns differ between groups of afferent neurons innervating vestibular epithelia. We investigated the biophysical properties of Na+ currents in specialized vestibular calyx afferent terminals during postnatal development. Mature calyces express Na+ currents with transient, persistent, and resurgent components. Nav1.6 channels contribute to resurgent Na+ currents and may enhance firing in peripheral calyx afferents. Understanding Na+ channels that contribute to vestibular nerve responses has implications for developing new treatments for vestibular dysfunction.
Collapse
Affiliation(s)
- Frances L Meredith
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado
| | - Katherine J Rennie
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Physiology & Biophysics, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
31
|
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
|
32
|
Yu Z, McIntosh JM, Sadeghi SG, Glowatzki E. Efferent synaptic transmission at the vestibular type II hair cell synapse. J Neurophysiol 2020; 124:360-374. [PMID: 32609559 DOI: 10.1152/jn.00143.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In the vestibular peripheral organs, type I and type II hair cells (HCs) transmit incoming signals via glutamatergic quantal transmission onto afferent nerve fibers. Additionally, type I HCs transmit via "non-quantal" transmission to calyx afferent fibers, by accumulation of glutamate and potassium in the synaptic cleft. Vestibular efferent inputs originating in the brainstem contact type II HCs and vestibular afferents. Here, synaptic inputs to type II HCs were characterized by using electrical and optogenetic stimulation of efferent fibers combined with in vitro whole cell patch-clamp recording from type II HCs in the rodent vestibular crista. Properties of efferent synaptic currents in type II HCs were similar to those found in cochlear HCs and mediated by activation of α9-containing nicotinic acetylcholine receptors (nAChRs) and small-conductance calcium-activated potassium (SK) channels. While efferents showed a low probability of release at low frequencies of stimulation, repetitive stimulation resulted in facilitation and increased probability of release. Notably, the membrane potential of type II HCs during optogenetic stimulation of efferents showed a strong hyperpolarization in response to single pulses and was further enhanced by repetitive stimulation. Such efferent-mediated inhibition of type II HCs can provide a mechanism to adjust the contribution of signals from type I and type II HCs to vestibular nerve fibers, with a shift of the response to be more like that of calyx-only afferents with faster non-quantal responses.NEW & NOTEWORTHY Type II vestibular hair cells (HCs) receive inputs from efferent neurons in the brain stem. We used in vitro optogenetic and electrical stimulation of vestibular efferent fibers to study their synaptic inputs to type II HCs. Stimulation of efferents inhibited type II HCs, similar to efferent effects on cochlear HCs. We propose that efferent inputs adjust the contribution of signals from type I and II HCs to vestibular nerve fibers.
Collapse
Affiliation(s)
- Zhou Yu
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Otolaryngology-Head and Neck Surgery, The Center for Hearing and Balance, and The Center for Sensory Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Soroush G Sadeghi
- Department of Communicative Disorders and Sciences, and Center for Hearing and Deafness, State University of New York at Buffalo, Buffalo, New York.,Neuroscience Program, State University of New York at Buffalo, Buffalo, New York
| | - Elisabeth Glowatzki
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Otolaryngology-Head and Neck Surgery, The Center for Hearing and Balance, and The Center for Sensory Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
33
|
Lu J, Hu L, Ye B, Hu H, Tao Y, Shu Y, Hao Chiang, Borse V, Xiang M, Wu H, Edge ASB, Shi F. Increased Type I and Decreased Type II Hair Cells after Deletion of Sox2 in the Developing Mouse Utricle. Neuroscience 2019; 422:146-160. [PMID: 31678344 PMCID: PMC10858341 DOI: 10.1016/j.neuroscience.2019.09.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 09/21/2019] [Accepted: 09/23/2019] [Indexed: 12/13/2022]
Abstract
The vestibular system of the inner ear contains Type I and Type II hair cells (HCs) generated from sensory progenitor cells; however, little is known about how the HC subtypes are formed. Sox2 (encoding SRY-box 2) is expressed in Type II, but not in Type I, HCs. The present study aimed to investigate the role of SOX2 in cell fate determination in Type I vs. Type II HCs. First, we confirmed that Type I HCs developed from Sox2-expressing cells through lineage tracing of Sox2-positive cells using a CAG-tdTomato reporter mouse crossed with a Sox2-CreER mouse. Then, Sox2 loss of function was induced in HCs, using Sox2flox transgenic mice crossed with a Gfi1-Cre driver mouse. Knockout of Sox2 in HCs increased the number of Type I HCs and decreased the number of Type II HCs, while the total number of HCs and Sox2-positive supporting cells did not change. In addition, the effect of Sox2-knockout persisted into adulthood, resulting in an increased number of Type I HCs. These results demonstrate that SOX2 plays a critical role in the determination of Type II vs. Type I HC fate. The results suggested that Sox2 is a potential target for generating Type I HCs, which may be important for regenerative strategies for balance disorders.
Collapse
Affiliation(s)
- Jingrong Lu
- Department of Otolaryngology-Head & Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China; Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, China
| | - Lingxiang Hu
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, China; Department of Otolaryngology Head & Neck Surgery, Shanghai 9th People's Hospital/Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Bin Ye
- Department of Otolaryngology-Head & Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China; Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, China
| | - Haixia Hu
- Department of Otolaryngology-Head & Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China; Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, China
| | - Yong Tao
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, China; Department of Otolaryngology Head & Neck Surgery, Shanghai 9th People's Hospital/Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yilai Shu
- ENT Institute and Otorhinolaryngology Department, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China; Key Laboratory of Hearing Medicine of National Health and Family Planning Commission (NHFPC), Shanghai, China
| | - Hao Chiang
- Department of Otolaryngology, Harvard Medical School, Boston, MA 02115, USA; Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Vikrant Borse
- Department of Otolaryngology, Harvard Medical School, Boston, MA 02115, USA; Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Mingliang Xiang
- Department of Otolaryngology-Head & Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China; Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, China
| | - Hao Wu
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, China; Department of Otolaryngology Head & Neck Surgery, Shanghai 9th People's Hospital/Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China.
| | - Albert S B Edge
- Department of Otolaryngology, Harvard Medical School, Boston, MA 02115, USA; Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Fuxin Shi
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China; Decibel Therapeutics, Boston, MA 02215, USA.
| |
Collapse
|
34
|
Curthoys IS, Grant JW, Pastras CJ, Brown DJ, Burgess AM, Brichta AM, Lim R. A review of mechanical and synaptic processes in otolith transduction of sound and vibration for clinical VEMP testing. J Neurophysiol 2019; 122:259-276. [DOI: 10.1152/jn.00031.2019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Older studies of mammalian otolith physiology have focused mainly on sustained responses to low-frequency (<50 Hz) or maintained linear acceleration. So the otoliths have been regarded as accelerometers. Thus evidence of otolithic activation and high-precision phase locking to high-frequency sound and vibration appears to be very unusual. However, those results are exactly in accord with a substantial body of knowledge of otolith function in fish and frogs. It is likely that phase locking of otolith afferents to vibration is a general property of all vertebrates. This review examines the literature about the activation and phase locking of single otolithic neurons to air-conducted sound and bone-conducted vibration, in particular the high precision of phase locking shown by mammalian irregular afferents that synapse on striolar type I hair cells by calyx endings. Potassium in the synaptic cleft between the type I hair cell receptor and the calyx afferent ending may be responsible for the tight phase locking of these afferents even at very high discharge rates. Since frogs and fish do not possess full calyx endings, it is unlikely that they show phase locking with such high precision and to such high frequencies as has been found in mammals. The high-frequency responses have been modeled as the otoliths operating in a seismometer mode rather than an accelerometer mode. These high-frequency otolithic responses constitute the neural basis for clinical vestibular-evoked myogenic potential tests of otolith function.
Collapse
Affiliation(s)
- Ian S. Curthoys
- Vestibular Research Laboratory, School of Psychology, the University of Sydney, New South Wales, Australia
| | - J. Wally Grant
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, Virginia
| | - Christopher J. Pastras
- The Meniere’s Laboratory, Sydney Medical School, University of Sydney, New South Wales, Australia
| | - Daniel J. Brown
- The Meniere’s Laboratory, Sydney Medical School, University of Sydney, New South Wales, Australia
| | - Ann M. Burgess
- Vestibular Research Laboratory, School of Psychology, the University of Sydney, New South Wales, Australia
| | - Alan M. Brichta
- School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute. Newcastle, New South Wales, Australia
| | - Rebecca Lim
- School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute. Newcastle, New South Wales, Australia
| |
Collapse
|
35
|
Wang T, Niwa M, Sayyid ZN, Hosseini DK, Pham N, Jones SM, Ricci AJ, Cheng AG. Uncoordinated maturation of developing and regenerating postnatal mammalian vestibular hair cells. PLoS Biol 2019; 17:e3000326. [PMID: 31260439 PMCID: PMC6602158 DOI: 10.1371/journal.pbio.3000326] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 05/30/2019] [Indexed: 11/18/2022] Open
Abstract
Sensory hair cells are mechanoreceptors required for hearing and balance functions. From embryonic development, hair cells acquire apical stereociliary bundles for mechanosensation, basolateral ion channels that shape receptor potential, and synaptic contacts for conveying information centrally. These key maturation steps are sequential and presumed coupled; however, whether hair cells emerging postnatally mature similarly is unknown. Here, we show that in vivo postnatally generated and regenerated hair cells in the utricle, a vestibular organ detecting linear acceleration, acquired some mature somatic features but hair bundles appeared nonfunctional and short. The utricle consists of two hair cell subtypes with distinct morphological, electrophysiological and synaptic features. In both the undamaged and damaged utricle, fate-mapping and electrophysiology experiments showed that Plp1+ supporting cells took on type II hair cell properties based on molecular markers, basolateral conductances and synaptic properties yet stereociliary bundles were absent, or small and nonfunctional. By contrast, Lgr5+ supporting cells regenerated hair cells with type I and II properties, representing a distinct hair cell precursor subtype. Lastly, direct physiological measurements showed that utricular function abolished by damage was partially regained during regeneration. Together, our data reveal a previously unrecognized aberrant maturation program for hair cells generated and regenerated postnatally and may have broad implications for inner ear regenerative therapies. During development, sensory hair cells undergo a series of critical maturation steps that are sequential and presumed coupled, but whether regenerated hair cells mature similarly is unknown. This study shows that regenerated vestibular hair cells acquired some mature somatic features, but the apical bundles remained immature.
Collapse
Affiliation(s)
- Tian Wang
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mamiko Niwa
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Zahra N. Sayyid
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Davood K. Hosseini
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Nicole Pham
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Sherri M. Jones
- Department of Special Education and Communication Disorders, College of Education and Human Sciences, University of Nebraska, Lincoln, Nebraska, United States of America
| | - Anthony J. Ricci
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail: (AGC); (AJR)
| | - Alan G. Cheng
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail: (AGC); (AJR)
| |
Collapse
|
36
|
Holman HA, Poppi LA, Frerck M, Rabbitt RD. Spontaneous and Acetylcholine Evoked Calcium Transients in the Developing Mouse Utricle. Front Cell Neurosci 2019; 13:186. [PMID: 31133810 PMCID: PMC6514437 DOI: 10.3389/fncel.2019.00186] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 04/12/2019] [Indexed: 11/13/2022] Open
Abstract
Spontaneous calcium transients are present during early postnatal development in the mouse retina and cochlea, and play an important role in maturation of the sensory organs and neural circuits in the central nervous system (CNS). It is not known whether similar calcium transients occur during postnatal development in the vestibular sensory organs. Here we demonstrate spontaneous intracellular calcium transients in sensory hair cells (HCs) and supporting cells (SCs) in the murine utricular macula during the first two postnatal weeks. Calcium transients were monitored using a genetically encoded calcium indicator, GCaMP5G (G5), at 100 ms-frame−1 in excised utricle sensory epithelia, including HCs, SCs, and neurons. The reporter line expressed G5 and tdTomato (tdT) in a Gad2-Cre dependent manner within a subset of utricular HCs, SCs and neurons. Kinetics of the G5 reporter limited temporal resolution to calcium events lasting longer than 200 ms. Spontaneous calcium transients lasting 1-2 s were observed in the expressing population of HCs at birth and slower spontaneous transients lasting 10-30 s appeared in SCs by P3. Beginning at P5, calcium transients could be modulated by application of the efferent neurotransmitter acetylcholine (ACh). In mature mice, calcium transients in the utricular macula occurred spontaneously, had a duration 1-2 s, and could be modulated by the exogenous application of acetylcholine (ACh) or muscarine. Long-lasting calcium transients evoked by ACh in mature mice were blocked by atropine, consistent with previous reports describing the role of muscarinic receptors expressed in calyx bearing afferents in efferent control of vestibular sensation. Large spontaneous and ACh evoked transients were reversibly blocked by the inositol trisphosphate receptor (IP3R) antagonist aminoethoxydiphenyl borate (2-APB). Results demonstrate long-lasting calcium transients are present in the utricular macula during the first postnatal week, and that responses to ACh mature over this same time period.
Collapse
Affiliation(s)
- Holly A Holman
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Lauren A Poppi
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States.,School of Biomedical Science and Pharmacy, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Micah Frerck
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Richard D Rabbitt
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States.,Neuroscience Program, University of Utah, Salt Lake City, UT, United States.,Otolaryngology-Head and Neck Surgery, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
37
|
Warchol ME, Massoodnia R, Pujol R, Cox BC, Stone JS. Development of hair cell phenotype and calyx nerve terminals in the neonatal mouse utricle. J Comp Neurol 2019; 527:1913-1928. [PMID: 30724338 DOI: 10.1002/cne.24658] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 01/26/2019] [Accepted: 02/01/2019] [Indexed: 01/03/2023]
Abstract
The vestibular organs of reptiles, birds, and mammals possess Type I and Type II sensory hair cells, which have distinct morphologies, physiology, and innervation. Little is known about how vestibular hair cells adopt a Type I or Type II identity or acquire proper innervation. One distinguishing marker is the transcription factor Sox2, which is expressed in all developing hair cells but persists only in Type II hair cells in maturity. We examined Sox2 expression and formation of afferent nerve terminals in mouse utricles between postnatal days 0 (P0) and P17. Between P3 and P14, many hair cells lost Sox2 immunoreactivity and the density of calyceal afferent nerve terminals (specific to Type I hair cells) increased in all regions of the utricle. At early time points, many calyces enclosed Sox2-labeled hair cells, while some Sox2-negative hair cells within the striola had not yet developed a calyx. These observations indicate that calyx maturation is not temporally correlated with loss of Sox2 expression in Type I hair cells. To determine which type(s) of hair cells are formed postnatally, we fate-mapped neonatal supporting cells by injecting Plp-CreER T2 :Rosa26 tdTomato mice with tamoxifen at P2 and P3. At P9, tdTomato-positive hair cells were immature and not classifiable by type. At P30, tdTomato-positive hair cells increased 1.8-fold compared to P9, and 91% of tdTomato-labeled hair cells were Type II. Our findings show that most neonatally-derived hair cells become Type II, and many Type I hair cells (formed before P2) downregulate Sox2 and acquire calyces between P0 and P14.
Collapse
Affiliation(s)
- Mark E Warchol
- Department of Otolaryngology, Washington University, St Louis, Missouri
| | - Roxanna Massoodnia
- Department of Otolaryngology-Head and Neck Surgery and the Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, Washington
| | - Remy Pujol
- Department of Otolaryngology-Head and Neck Surgery and the Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, Washington.,INSERM Unit 1051, Institute of Neuroscience, University of Montpellier, Montpellier, France
| | - Brandon C Cox
- Departments of Pharmacology and Surgery, Division of Otolaryngology, Southern Illinois University School of Medicine, Springfield, Illinois
| | - Jennifer S Stone
- Department of Otolaryngology-Head and Neck Surgery and the Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, Washington
| |
Collapse
|
38
|
Eatock RA. Specializations for Fast Signaling in the Amniote Vestibular Inner Ear. Integr Comp Biol 2019; 58:341-350. [PMID: 29920589 DOI: 10.1093/icb/icy069] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
During rapid locomotion, the vestibular inner ear provides head-motion signals that stabilize posture, gaze, and heading. Afferent nerve fibers from central and peripheral zones of vestibular sensory epithelia use temporal and rate encoding, respectively, to emphasize different aspects of head motion: central afferents adapt faster to sustained head position and favor higher stimulus frequencies, reflecting specializations at each stage from motion of the accessory structure to spike propagation to the brain. One specialization in amniotes is an unusual nonquantal synaptic mechanism by which type I hair cells transmit to large calyceal terminals of afferent neurons. The reduced synaptic delay of this mechanism may have evolved to serve reliable and fast input to reflex pathways that ensure stable locomotion on land.
Collapse
Affiliation(s)
- Ruth Anne Eatock
- Department of Neurobiology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
39
|
Pickett SB, Thomas ED, Sebe JY, Linbo T, Esterberg R, Hailey DW, Raible DW. Cumulative mitochondrial activity correlates with ototoxin susceptibility in zebrafish mechanosensory hair cells. eLife 2018; 7:38062. [PMID: 30596476 PMCID: PMC6345563 DOI: 10.7554/elife.38062] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 12/31/2018] [Indexed: 12/11/2022] Open
Abstract
Mitochondria play a prominent role in mechanosensory hair cell damage and death. Although hair cells are thought to be energetically demanding cells, how mitochondria respond to these demands and how this might relate to cell death is largely unexplored. Using genetically encoded indicators, we found that mitochondrial calcium flux and oxidation are regulated by mechanotransduction and demonstrate that hair cell activity has both acute and long-term consequences on mitochondrial function. We tested whether variation in mitochondrial activity reflected differences in the vulnerability of hair cells to the toxic drug neomycin. We observed that susceptibility did not correspond to the acute level of mitochondrial activity but rather to the cumulative history of that activity.
Collapse
Affiliation(s)
- Sarah B Pickett
- Department of Biological Structure, University of Washington, Seattle, United States.,Graduate Program in Neuroscience, University of Washington, Seattle, United States
| | - Eric D Thomas
- Department of Biological Structure, University of Washington, Seattle, United States.,Graduate Program in Neuroscience, University of Washington, Seattle, United States
| | - Joy Y Sebe
- Department of Biological Structure, University of Washington, Seattle, United States
| | - Tor Linbo
- Department of Biological Structure, University of Washington, Seattle, United States
| | - Robert Esterberg
- Department of Biological Structure, University of Washington, Seattle, United States.,Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, United States
| | - Dale W Hailey
- Department of Biological Structure, University of Washington, Seattle, United States.,Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, United States
| | - David W Raible
- Department of Biological Structure, University of Washington, Seattle, United States.,Graduate Program in Neuroscience, University of Washington, Seattle, United States.,Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, United States
| |
Collapse
|
40
|
McInturff S, Burns JC, Kelley MW. Characterization of spatial and temporal development of Type I and Type II hair cells in the mouse utricle using new cell-type-specific markers. Biol Open 2018; 7:bio038083. [PMID: 30455179 PMCID: PMC6262869 DOI: 10.1242/bio.038083] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 09/14/2018] [Indexed: 12/16/2022] Open
Abstract
The utricle of the inner ear, a vestibular sensory structure that mediates perception of linear acceleration, is comprised of two morphologically and physiologically distinct types of mechanosensory hair cells, referred to as Type Is and Type IIs. While these cell types are easily discriminated in an adult utricle, understanding their development has been hampered by a lack of molecular markers that can be used to identify each cell type prior to maturity. Therefore, we collected single hair cells at three different ages and used single cell RNAseq to characterize the transcriptomes of those cells. Analysis of differential gene expression identified Spp1 as a specific marker for Type I hair cells and Mapt and Anxa4 as specific markers for Type II hair cells. Antibody labeling confirmed the specificity of these markers which were then used to examine the temporal and spatial development of utricular hair cells. While Type I hair cells develop in a gradient that extends across the utricle from posterior-medial to anterior-lateral, Type II hair cells initially develop in the central striolar region and then extend uniformly towards the periphery. Finally, by combining these markers with genetic fate mapping, we demonstrate that over 98% of all Type I hair cells develop prior to birth while over 98% of Type II hair cells develop post-natally. These results are consistent with previous findings suggesting that Type I hair cells develop first and refute the hypothesis that Type II hair cells represent a transitional form between immature and Type I hair cells.
Collapse
Affiliation(s)
- Stephen McInturff
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, MD 20892, USA
| | - Joseph C Burns
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, MD 20892, USA
| | - Matthew W Kelley
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Porter Neuroscience Research Center, Bethesda, MD 20892, USA
| |
Collapse
|
41
|
Meredith FL, Rennie KJ. Regional and Developmental Differences in Na + Currents in Vestibular Primary Afferent Neurons. Front Cell Neurosci 2018; 12:423. [PMID: 30487736 PMCID: PMC6246661 DOI: 10.3389/fncel.2018.00423] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/29/2018] [Indexed: 02/04/2023] Open
Abstract
The vestibular system relays information about head position via afferent nerve fibers to the brain in the form of action potentials. Voltage-gated Na+ channels in vestibular afferents drive the initiation and propagation of action potentials, but their expression during postnatal development and their contributions to firing in diverse mature afferent populations are unknown. Electrophysiological techniques were used to determine Na+ channel subunit types in vestibular calyx-bearing afferents at different stages of postnatal development. We used whole cell patch clamp recordings in thin slices of gerbil crista neuroepithelium to investigate Na+ channels and firing patterns in central zone (CZ) and peripheral zone (PZ) afferents. PZ afferents are exclusively dimorphic, innervating type I and type II hair cells, whereas CZ afferents can form dimorphs or calyx-only terminals which innervate type I hair cells alone. All afferents expressed tetrodotoxin (TTX)-sensitive Na+ currents, but TTX-sensitivity varied with age. During the fourth postnatal week, 200–300 nM TTX completely blocked sodium currents in PZ and CZ calyces. By contrast, in immature calyces [postnatal day (P) 5–11], a small component of peak sodium current remained in 200 nM TTX. Application of 1 μM TTX, or Jingzhaotoxin-III plus 200 nM TTX, abolished sodium current in immature calyces, suggesting the transient expression of voltage-gated sodium channel 1.5 (Nav1.5) during development. A similar TTX-insensitive current was found in early postnatal crista hair cells (P5–9) and constituted approximately one third of the total sodium current. The Nav1.6 channel blocker, 4,9-anhydrotetrodotoxin, reduced a component of sodium current in immature and mature calyces. At 100 nM 4,9-anhydrotetrodotoxin, peak sodium current was reduced on average by 20% in P5–14 calyces, by 37% in mature dimorphic PZ calyces, but by less than 15% in mature CZ calyx-only terminals. In mature PZ calyces, action potentials became shorter and broader in the presence of 4,9-anhydrotetrodotoxin implicating a role for Nav1.6 channels in firing in dimorphic afferents.
Collapse
Affiliation(s)
- Frances L Meredith
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Katherine J Rennie
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, CO, United States.,Department of Physiology & Biophysics, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
42
|
Stone JS, Wisner SR, Bucks SA, Mellado Lagarde MM, Cox BC. Characterization of Adult Vestibular Organs in 11 CreER Mouse Lines. J Assoc Res Otolaryngol 2018; 19:381-399. [PMID: 29869046 DOI: 10.1007/s10162-018-0676-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/07/2018] [Indexed: 10/14/2022] Open
Abstract
Utricles are vestibular sense organs that encode linear head movements. They are composed of a sensory epithelium with type I and type II hair cells and supporting cells, sitting atop connective tissue, through which vestibular nerves project. We characterized utricular Cre expression in 11 murine CreER lines using the ROSA26tdTomato reporter line and tamoxifen induction at 6 weeks of age. This characterization included Calbindin2CreERT2, Fgfr3-iCreERT2, GFAP-A-CreER™, GFAP-B-CreER™, GLAST-CreERT2, Id2CreERT2, OtoferlinCreERT2, ParvalbuminCreERT2, Prox1CreERT2, Sox2CreERT2, and Sox9-CreERT2. OtoferlinCreERT2 mice had inducible Cre activity specific to hair cells. GLAST-CreERT2, Id2CreERT2, and Sox9-CreERT2 had inducible Cre activity specific to supporting cells. Sox2CreERT2 had inducible Cre activity in supporting cells and most type II hair cells. ParvalbuminCreERT2 mice had small numbers of labeled vestibular nerve afferents. Calbindin2CreERT2 mice had labeling of most type II hair cells and some type I hair cells and supporting cells. Only rare (or no) tdTomato-positive cells were detected in utricles of Fgfr3-iCreERT2, GFAP-A-CreER™, GFAP-B-CreER™, and Prox1CreERT2 mice. No Cre leakiness (tdTomato expression in the absence of tamoxifen) was observed in OtoferlinCreERT2 mice. A small degree of leakiness was seen in GLAST-CreERT2, Id2CreERT2, Sox2CreERT2, and Sox9-CreERT2 lines. Calbindin2CreERT2 mice had similar tdTomato expression with or without tamoxifen, indicating lack of inducible control under the conditions tested. In conclusion, 5 lines-GLAST-CreERT2, Id2CreERT2, OtoferlinCreERT2, Sox2CreERT2, and Sox9-CreERT2-showed cell-selective, inducible Cre activity with little leakiness, providing new genetic tools for researchers studying the vestibular periphery.
Collapse
Affiliation(s)
- Jennifer S Stone
- Department of Otolaryngology-Head and Neck Surgery, Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, WA, USA
| | - Serena R Wisner
- Department of Otolaryngology-Head and Neck Surgery, Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, WA, USA
| | - Stephanie A Bucks
- Department of Otolaryngology-Head and Neck Surgery, Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, WA, USA
| | - Marcia M Mellado Lagarde
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Brandon C Cox
- Departments of Pharmacology and Surgery, Division of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, USA.
| |
Collapse
|
43
|
Li Y, Jia S, Liu H, Tateya T, Guo W, Yang S, Beisel KW, He DZZ. Characterization of Hair Cell-Like Cells Converted From Supporting Cells After Notch Inhibition in Cultures of the Organ of Corti From Neonatal Gerbils. Front Cell Neurosci 2018; 12:73. [PMID: 29662441 PMCID: PMC5890164 DOI: 10.3389/fncel.2018.00073] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 03/02/2018] [Indexed: 12/11/2022] Open
Abstract
The senses of hearing and balance depend upon hair cells, the sensory receptors of the inner ear. Hair cells transduce mechanical stimuli into electrical activity. Loss of hair cells as a result of aging or exposure to noise and ototoxic drugs is the major cause of noncongenital hearing and balance deficits. In the ear of non-mammals, lost hair cells can spontaneously be replaced by production of new hair cells from conversion of supporting cells. Although supporting cells in adult mammals have lost that capability, neonatal supporting cells are able to convert to hair cells after inhibition of Notch signaling. We questioned whether Notch inhibition is sufficient to convert supporting cells to functional hair cells using electrophysiology and electron microscopy. We showed that pharmacological inhibition of the canonical Notch pathway in the cultured organ of Corti prepared from neonatal gerbils induced stereocilia formation in supporting cells (defined as hair cell-like cells or HCLCs) and supernumerary stereocilia in hair cells. The newly emerged stereocilia bundles of HCLCs were functional, i.e., able to respond to mechanical stimulation with mechanotransduction (MET) current. Transmission electron microscopy (TEM) showed that HCLCs converted from pillar cells maintained the pillar cell shape and that subsurface cisternae, normally observed underneath the cytoskeleton in outer hair cells (OHCs), was not present in Deiters’ cells-derived HCLCs. Voltage-clamp recordings showed that whole-cell currents from Deiters’ cells-derived HCLCs retained the same kinetics and magnitude seen in normal Deiters’ cells and that nonlinear capacitance (NLC), an electrical hallmark of OHC electromotility, was not detected from any HCLCs measured. Taken together, these results suggest that while Notch inhibition is sufficient for promoting stereocilia bundle formation, it is insufficient to convert neonatal supporting cells to mature hair cells. The fact that Notch inhibition led to stereocilia formation in supporting cells and supernumerary stereocilia in existing hair cells appears to suggest that Notch signaling may regulate stereocilia formation and stability during development.
Collapse
Affiliation(s)
- Yi Li
- Department of Otorhinolaryngology, Beijing Tongren Hospital, Beijing Capital Medical University, Beijing, China.,Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| | - Shuping Jia
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| | - Huizhan Liu
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| | - Tomoko Tateya
- Institute for Virus Research, Kyoto University, Kyoto, Japan
| | - Weiwei Guo
- Department of Otorhinolaryngology, PLA General Hospital, Beijing, China
| | - Shiming Yang
- Department of Otorhinolaryngology, PLA General Hospital, Beijing, China
| | - Kirk W Beisel
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| | - David Z Z He
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| |
Collapse
|
44
|
Vijayakumar S, Depreux FF, Jodelka FM, Lentz JJ, Rigo F, Jones TA, Hastings ML. Rescue of peripheral vestibular function in Usher syndrome mice using a splice-switching antisense oligonucleotide. Hum Mol Genet 2018. [PMID: 28633508 DOI: 10.1093/hmg/ddx234] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Usher syndrome type 1C (USH1C/harmonin) is associated with profound retinal, auditory and vestibular dysfunction. We have previously reported on an antisense oligonucleotide (ASO-29) that dramatically improves auditory function and balance behavior in mice homozygous for the harmonin mutation Ush1c c.216G > A following a single systemic administration. The findings were suggestive of improved vestibular function; however, no direct vestibular assessment was made. Here, we measured vestibular sensory evoked potentials (VsEPs) to directly assess vestibular function in Usher mice. We report that VsEPs are absent or abnormal in Usher mice, indicating profound loss of vestibular function. Strikingly, Usher mice receiving ASO-29 treatment have normal or elevated vestibular response thresholds when treated during a critical period between postnatal day 1 and 5, respectively. In contrast, treatment of mice with ASO-29 treatment at P15 was minimally effective at rescuing vestibular function. Interestingly, ASO-29 treatment at P1, P5 or P15 resulted in sufficient vestibular recovery to support normal balance behaviors, suggesting a therapeutic benefit to balance with ASO-29 treatment at P15 despite the profound vestibular functional deficits that persist with treatment at this later time. These findings provide the first direct evidence of an effective treatment of peripheral vestibular function in a mouse model of USH1C and reveal the potential for using antisense technology to treat vestibular dysfunction.
Collapse
Affiliation(s)
- Sarath Vijayakumar
- Department of Special Education and Communication Disorders, University of Nebraska-Lincoln, 304 Barkley Memorial Center, Lincoln, NE 68583, USA
| | - Frederic F Depreux
- Department of Cell Biology and Anatomy, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Francine M Jodelka
- Department of Cell Biology and Anatomy, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Jennifer J Lentz
- Department of Otorhinolaryngology, Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, CA 92010, USA
| | - Timothy A Jones
- Department of Special Education and Communication Disorders, University of Nebraska-Lincoln, 304 Barkley Memorial Center, Lincoln, NE 68583, USA
| | - Michelle L Hastings
- Department of Cell Biology and Anatomy, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| |
Collapse
|
45
|
Spaiardi P, Tavazzani E, Manca M, Milesi V, Russo G, Prigioni I, Marcotti W, Magistretti J, Masetto S. An allosteric gating model recapitulates the biophysical properties of I K,L expressed in mouse vestibular type I hair cells. J Physiol 2017; 595:6735-6750. [PMID: 28862328 PMCID: PMC5663832 DOI: 10.1113/jp274202] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 08/30/2017] [Indexed: 12/11/2022] Open
Abstract
Key points Vestibular type I and type II hair cells and their afferent fibres send information to the brain regarding the position and movement of the head. The characteristic feature of type I hair cells is the expression of a low‐voltage‐activated outward rectifying K+ current, IK,L, whose biophysical properties and molecular identity are still largely unknown. In vitro, the afferent nerve calyx surrounding type I hair cells causes unstable intercellular K+ concentrations, altering the biophysical properties of IK,L. We found that in the absence of the calyx, IK,L in type I hair cells exhibited unique biophysical activation properties, which were faithfully reproduced by an allosteric channel gating scheme. These results form the basis for a molecular and pharmacological identification of IK,L.
Abstract Type I and type II hair cells are the sensory receptors of the mammalian vestibular epithelia. Type I hair cells are characterized by their basolateral membrane being enveloped in a single large afferent nerve terminal, named the calyx, and by the expression of a low‐voltage‐activated outward rectifying K+ current, IK,L. The biophysical properties and molecular profile of IK,L are still largely unknown. By using the patch‐clamp whole‐cell technique, we examined the voltage‐ and time‐dependent properties of IK,L in type I hair cells of the mouse semicircular canal. We found that the biophysical properties of IK,L were affected by an unstable K+ equilibrium potential (VeqK+). Both the outward and inward K+ currents shifted VeqK+ consistent with K+ accumulation or depletion, respectively, in the extracellular space, which we attributed to a residual calyx attached to the basolateral membrane of the hair cells. We therefore optimized the hair cell dissociation protocol in order to isolate mature type I hair cells without their calyx. In these cells, the uncontaminated IK,L showed a half‐activation at –79.6 mV and a steep voltage dependence (2.8 mV). IK,L also showed complex activation and deactivation kinetics, which we faithfully reproduced by an allosteric channel gating scheme where the channel is able to open from all (five) closed states. The ‘early’ open states substantially contribute to IK,L activation at negative voltages. This study provides the first complete description of the ‘native’ biophysical properties of IK,L in adult mouse vestibular type I hair cells. Vestibular type I and type II hair cells and their afferent fibres send information to the brain regarding the position and movement of the head. The characteristic feature of type I hair cells is the expression of a low‐voltage‐activated outward rectifying K+ current, IK,L, whose biophysical properties and molecular identity are still largely unknown. In vitro, the afferent nerve calyx surrounding type I hair cells causes unstable intercellular K+ concentrations, altering the biophysical properties of IK,L. We found that in the absence of the calyx, IK,L in type I hair cells exhibited unique biophysical activation properties, which were faithfully reproduced by an allosteric channel gating scheme. These results form the basis for a molecular and pharmacological identification of IK,L.
Collapse
Affiliation(s)
- Paolo Spaiardi
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, 27100, Italy
| | - Elisa Tavazzani
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, 27100, Italy
| | - Marco Manca
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, 27100, Italy
| | - Veronica Milesi
- Instituto de Estudios Inmunológios y Fisiopatológicos (IIFP) - CONICET, Universidad Nacional de La Plata, La Plata, 1900, Argentina
| | - Giancarlo Russo
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, 27100, Italy
| | - Ivo Prigioni
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, 27100, Italy
| | - Walter Marcotti
- Department of Biomedical Science, University of Sheffield, Sheffield, S10 2TN, UK
| | - Jacopo Magistretti
- Department of Biology and Biotechnology, University of Pavia, Pavia, 27100, Italy
| | - Sergio Masetto
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, 27100, Italy
| |
Collapse
|
46
|
Chaves PP, Valdoria CM, Amorim MCP, Vasconcelos RO. Ontogenetic development of the inner ear saccule and utricle in the Lusitanian toadfish: Potential implications for auditory sensitivity. Hear Res 2017; 353:112-121. [DOI: 10.1016/j.heares.2017.06.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 06/01/2017] [Accepted: 06/13/2017] [Indexed: 10/19/2022]
|
47
|
Core Body Temperature Effects on the Mouse Vestibulo-ocular Reflex. J Assoc Res Otolaryngol 2017; 18:827-835. [PMID: 28755310 DOI: 10.1007/s10162-017-0639-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 07/17/2017] [Indexed: 01/12/2023] Open
Abstract
Core body temperature has been shown to affect vestibular end-organ and nerve afferents so that their resting discharge rate and sensitivity increase with temperature. Our aim was to determine whether these changes observed in extracellular nerve recordings of anaesthetized C57BL/6 mice corresponded to changes in the behavioural vestibulo-ocular reflex (VOR) of alert mice. The VOR drives eye rotations to keep images stable on the retina during head movements. We measured the VOR gain (eye velocity/head velocity) and phase (delay between vestibular stimulus and response) during whole-body sinusoidal rotations ranging 0.5-12 Hz with peak velocity 50 or 100 °/s in nine adult C57BL/6 mice. We also measured the VOR during whole-body transient rotations with acceleration 3000 or 6000 °/s2 reaching a plateau of 150 or 300 °/s. These measures were obtained while the mouse's core body temperature was held at either 32 or 37 °C for at least 35 min before recording. The temperature presentation order and timing were pseudo-randomized. We found that a temperature increase from 32 to 37 °C caused a significant increase in sinusoidal VOR gain of 17 % (P < 0.001). Temperature had no other effects on the behavioural VOR. Our data suggest that temperature effects on regularly firing afferents best correspond to the changes that we observed in the VOR gain.
Collapse
|
48
|
Burns JC, Stone JS. Development and regeneration of vestibular hair cells in mammals. Semin Cell Dev Biol 2017; 65:96-105. [PMID: 27864084 PMCID: PMC5423856 DOI: 10.1016/j.semcdb.2016.11.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 11/03/2016] [Indexed: 10/20/2022]
Abstract
Vestibular sensation is essential for gaze stabilization, balance, and perception of gravity. The vestibular receptors in mammals, Type I and Type II hair cells, are located in five small organs in the inner ear. Damage to hair cells and their innervating neurons can cause crippling symptoms such as vertigo, visual field oscillation, and imbalance. In adult rodents, some Type II hair cells are regenerated and become re-innervated after damage, presenting opportunities for restoring vestibular function after hair cell damage. This article reviews features of vestibular sensory cells in mammals, including their basic properties, how they develop, and how they are replaced after damage. We discuss molecules that control vestibular hair cell regeneration and highlight areas in which our understanding of development and regeneration needs to be deepened.
Collapse
Affiliation(s)
- Joseph C Burns
- Decibel Therapeutics, 215 First St., Suite 430, Cambridge, MA 02142, USA.
| | - Jennifer S Stone
- Department of Otolaryngology/Head and Neck Surgery and The Virginia Merrill Bloedel Hearing Research Center, University of Washington School of Medicine, Box 357923, Seattle, WA 98195-7923, USA.
| |
Collapse
|
49
|
Kirk ME, Meredith FL, Benke TA, Rennie KJ. AMPA receptor-mediated rapid EPSCs in vestibular calyx afferents. J Neurophysiol 2017; 117:2312-2323. [PMID: 28298303 DOI: 10.1152/jn.00394.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 03/10/2017] [Accepted: 03/10/2017] [Indexed: 01/21/2023] Open
Abstract
In the vestibular periphery neurotransmission between hair cells and primary afferent nerves occurs via specialized ribbon synapses. Type I vestibular hair cells (HCIs) make synaptic contacts with calyx terminals, which enclose most of the HCI basolateral surface. To probe synaptic transmission, whole cell patch-clamp recordings were made from calyx afferent terminals isolated together with their mature HCIs from gerbil crista. Neurotransmitter release was measured as excitatory postsynaptic currents (EPSCs) in voltage clamp. Spontaneous EPSCs were classified as simple or complex. Simple events exhibited a rapid rise time and a fast monoexponential decay (time constant < 1 ms). The remaining events, constituting ~40% of EPSCs, showed more complex characteristics. Extracellular Sr2+ greatly increased EPSC frequency, and EPSCs were blocked by the AMPA receptor blocker NBQX. The role of presynaptic Ca2+ channels was assessed by application of the L-type Ca2+ channel blocker nifedipine (20 µM), which reduced EPSC frequency. In contrast, the L-type Ca2+ channel opener BAY K 8644 increased EPSC frequency. Cyclothiazide increased the decay time constant of averaged simple EPSCs by approximately twofold. The low-affinity AMPA receptor antagonist γ-d-glutamylglycine (2 mM) reduced the proportion of simple EPSCs relative to complex events, indicating glutamate accumulation in the restricted cleft between HCI and calyx. In crista slices EPSC frequency was greater in central compared with peripheral calyces, which may be due to greater numbers of presynaptic ribbons in central hair cells. Our data support a role for L-type Ca2+ channels in spontaneous release and demonstrate regional variations in AMPA-mediated quantal transmission at the calyx synapse.NEW & NOTEWORTHY In vestibular calyx terminals of mature cristae we find that the majority of excitatory postsynaptic currents (EPSCs) are rapid monophasic events mediated by AMPA receptors. Spontaneous EPSCs are reduced by an L-type Ca2+ channel blocker and notably enhanced in extracellular Sr2+ EPSC frequency is greater in central areas of the crista compared with peripheral areas and may be associated with more numerous presynaptic ribbons in central hair cells.
Collapse
Affiliation(s)
- Matthew E Kirk
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado
| | - Frances L Meredith
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado
| | - Timothy A Benke
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.,Departments of Pediatrics, Neurology, and Pharmacology, University of Colorado School of Medicine, Aurora, Colorado
| | - Katherine J Rennie
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado; .,Department of Physiology and Biophysics, University of Colorado School of Medicine, Aurora, Colorado; and
| |
Collapse
|
50
|
Sultemeier DR, Choy KR, Schweizer FE, Hoffman LF. Spaceflight-induced synaptic modifications within hair cells of the mammalian utricle. J Neurophysiol 2017; 117:2163-2178. [PMID: 28228581 DOI: 10.1152/jn.00240.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 02/21/2017] [Accepted: 02/21/2017] [Indexed: 02/08/2023] Open
Abstract
Exposure to the microgravity conditions of spaceflight alleviates the load normally imposed by the Earth's gravitational field on the inner ear utricular epithelia. Previous ultrastructural investigations have shown that spaceflight induces an increase in synapse density within hair cells of the rat utricle. However, the utricle exhibits broad physiological heterogeneity across different epithelial regions, and it is unknown whether capabilities for synaptic plasticity generalize to hair cells across its topography. To achieve systematic and broader sampling of the epithelium than was previously conducted, we used immunohistochemistry and volumetric image analyses to quantify synapse distributions across representative utricular regions in specimens from mice exposed to spaceflight (a 15-day mission of the space shuttle Discovery). These measures were compared with similarly sampled Earth-bound controls. Following paraformaldehyde fixation and microdissection, immunohistochemistry was performed on intact specimens to label presynaptic ribbons (anti-CtBP2) and postsynaptic receptor complexes (anti-Shank1A). Synapses were identified as closely apposed pre- and postsynaptic puncta. Epithelia from horizontal semicircular canal cristae served as "within-specimen" controls, whereas utricles and cristae from Earth-bound cohorts served as experimental controls. We found that synapse densities decreased in the medial extrastriolae of microgravity specimens compared with experimental controls, whereas they were unchanged in the striolae and horizontal cristae from the two conditions. These data demonstrate that structural plasticity was topographically localized to the utricular region that encodes very low frequency and static changes in linear acceleration, and illuminates the remarkable capabilities of utricular hair cells for synaptic plasticity in adapting to novel gravitational environments.NEW & NOTEWORTHY Spaceflight imposes a radically different sensory environment from that in which the inner ear utricle normally operates. We investigated synaptic modifications in utricles from mice flown aboard a space shuttle mission. Structural synaptic plasticity was detected in the medial extrastriola, a region associated with encoding static head position, as decreased synapse density. These results are remarkably congruent with a recent report of decreased utricular function in astronauts immediately after returning from the International Space Station.
Collapse
Affiliation(s)
- David R Sultemeier
- Department of Head & Neck Surgery, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Kristel R Choy
- Department of Head & Neck Surgery, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Felix E Schweizer
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, California; and.,Brain Research Institute, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Larry F Hoffman
- Department of Head & Neck Surgery, David Geffen School of Medicine, University of California, Los Angeles, California; .,Brain Research Institute, David Geffen School of Medicine, University of California, Los Angeles, California
| |
Collapse
|