1
|
Wang T, Yang T, Kedaigle A, Pregernig G, McCarthy R, Holmes B, Wu X, Becker L, Pan N, So K, Chen L, He J, Mahmoudi A, Negi S, Kowalczyk M, Gibson T, Druckenbrod N, Cheng AG, Burns J. Precise genetic control of ATOH1 enhances maturation of regenerated hair cells in the mature mouse utricle. Nat Commun 2024; 15:9166. [PMID: 39448563 PMCID: PMC11502789 DOI: 10.1038/s41467-024-53153-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Abstract
Vestibular hair cells are mechanoreceptors critical for detecting head position and motion. In mammals, hair cell loss causes vestibular dysfunction as spontaneous regeneration is nearly absent. Constitutive expression of exogenous ATOH1, a hair cell transcription factor, increases hair cell regeneration, however, these cells fail to fully mature. Here, we profiled mouse utricles at 14 time points, and defined transcriptomes of developing and mature vestibular hair cells. To mimic native hair cells which downregulate endogenous ATOH1 as they mature, we engineered viral vectors carrying the supporting cell promoters GFAP and RLBP1. In utricles damaged ex vivo, both CMV-ATOH1 and GFAP-ATOH1 increased regeneration more effectively than RLBP1-ATOH1, while GFAP-ATOH1 and RLBP1-ATOH1 induced hair cells with more mature transcriptomes. In utricles damaged in vivo, GFAP-ATOH1 induced regeneration of hair cells expressing genes indicative of maturing type II hair cells, and more hair cells with bundles and synapses than untreated organs. Together our results demonstrate the efficacy of spatiotemporal control of ATOH1 overexpression in inner ear hair cell regeneration.
Collapse
Affiliation(s)
- Tian Wang
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, 94305, USA
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Tian Yang
- Decibel Therapeutics, Boston, MA, 02215, USA
| | | | - Gabriela Pregernig
- Decibel Therapeutics, Boston, MA, 02215, USA
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Ryan McCarthy
- Decibel Therapeutics, Boston, MA, 02215, USA
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Ben Holmes
- Decibel Therapeutics, Boston, MA, 02215, USA
| | - Xudong Wu
- Decibel Therapeutics, Boston, MA, 02215, USA
| | - Lars Becker
- Decibel Therapeutics, Boston, MA, 02215, USA
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Ning Pan
- Decibel Therapeutics, Boston, MA, 02215, USA
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Kathy So
- Decibel Therapeutics, Boston, MA, 02215, USA
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Leon Chen
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, 94305, USA
| | - Jun He
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, 94305, USA
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Ahmad Mahmoudi
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, 94305, USA
| | - Soumya Negi
- Decibel Therapeutics, Boston, MA, 02215, USA
| | | | | | | | - Alan G Cheng
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, 94305, USA.
| | | |
Collapse
|
2
|
Singh K, Jayaram M, Hanumantharaju A, Tõnissoo T, Jagomäe T, Mikheim K, Muthuraman S, Gilbert SF, Plaas M, Schäfer MKE, Innos J, Lilleväli K, Philips MA, Vasar E. The IgLON family of cell adhesion molecules expressed in developing neural circuits ensure the proper functioning of the sensory system in mice. Sci Rep 2024; 14:22593. [PMID: 39349721 PMCID: PMC11442611 DOI: 10.1038/s41598-024-73358-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024] Open
Abstract
Deletions and malfunctions of the IgLON family of cell adhesion molecules are associated with anatomical, behavioral, and metabolic manifestations of neuropsychiatric disorders. We have previously shown that IgLON genes are expressed in sensory nuclei/pathways and that IgLON proteins modulate sensory processing. Here, we examined the expression of IgLON alternative promoter-specific isoforms during embryonic development and studied the sensory consequences of the anatomical changes when one of the IgLON genes, Negr1, is knocked out. At the embryonal age of E12.5 and E13.5, various IgLONs were distributed differentially and dynamically in the developing sensory areas within the central and peripheral nervous system, as well as in limbs and mammary glands. Sensory tests showed that Negr1 deficiency causes differences in vestibular function and temperature sensitivity in the knockout mice. Sex-specific differences were noted across olfaction, vestibular functioning, temperature regulation, and mechanical sensitivity. Our findings highlight the involvement of IgLON molecules during sensory circuit formation and suggest Negr1's critical role in somatosensory processing.
Collapse
Affiliation(s)
- Katyayani Singh
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia.
| | - Mohan Jayaram
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Arpana Hanumantharaju
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Tambet Tõnissoo
- Institute of Molecular and Cell Biology, University of Tartu, Vanemuise 46-221, Ria 23-204, 51010, Tartu, Estonia
| | - Toomas Jagomäe
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14B Ravila Street, 50411, Tartu, Estonia
| | - Kaie Mikheim
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Srirathi Muthuraman
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Scott F Gilbert
- Department of Biology, Swarthmore College, Swarthmore, PA, USA
| | - Mario Plaas
- Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, 14B Ravila Street, 50411, Tartu, Estonia
| | - Michael K E Schäfer
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, 55131, Mainz, Germany
- Focus Program Translational Neurosciences, Johannes Gutenberg-University Mainz, 55131, Mainz, Germany
- Research Center for Immunotherapy, Johannes Gutenberg-University Mainz, 55131, Mainz, Germany
| | - Jürgen Innos
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Kersti Lilleväli
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
- The Centre of Estonian Rural Research and Knowledge, 48309, Jõgeva Alevik, Estonia
| | - Mari-Anne Philips
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Eero Vasar
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| |
Collapse
|
3
|
Beaulieu MO, Thomas ED, Raible DW. Transdifferentiation is temporally uncoupled from progenitor pool expansion during hair cell regeneration in the zebrafish inner ear. Development 2024; 151:dev202944. [PMID: 39045613 PMCID: PMC11361639 DOI: 10.1242/dev.202944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/15/2024] [Indexed: 07/25/2024]
Abstract
Death of mechanosensory hair cells in the inner ear is a common cause of auditory and vestibular impairment in mammals, which have a limited ability to regrow these cells after damage. In contrast, non-mammalian vertebrates, including zebrafish, can robustly regenerate hair cells after severe organ damage. The zebrafish inner ear provides an understudied model system for understanding hair cell regeneration in organs that are highly conserved with their mammalian counterparts. Here, we quantitatively examine hair cell addition during growth and regeneration of the larval zebrafish inner ear. We used a genetically encoded ablation method to induce hair cell death and we observed gradual regeneration with correct spatial patterning over a 2-week period following ablation. Supporting cells, which surround and are a source of new hair cells, divide in response to hair cell ablation, expanding the possible progenitor pool. In parallel, nascent hair cells arise from direct transdifferentiation of progenitor pool cells temporally uncoupled from supporting cell division. These findings reveal a previously unrecognized mechanism of hair cell regeneration with implications for how hair cells may be encouraged to regenerate in the mammalian ear.
Collapse
Affiliation(s)
- Marielle O. Beaulieu
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA 98195, USA
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology - Head and Neck Surgery, University of Washington, Seattle, WA 98195, USA
| | - Eric D. Thomas
- Neuroscience Graduate Program, University of Washington, Seattle, WA 98195, USA
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - David W. Raible
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA 98195, USA
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology - Head and Neck Surgery, University of Washington, Seattle, WA 98195, USA
- Neuroscience Graduate Program, University of Washington, Seattle, WA 98195, USA
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
4
|
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
|
5
|
Beaulieu MO, Thomas ED, Raible DW. Transdifferentiation is uncoupled from progenitor pool expansion during hair cell regeneration in the zebrafish inner ear. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.09.588777. [PMID: 38645220 PMCID: PMC11030336 DOI: 10.1101/2024.04.09.588777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Death of mechanosensory hair cells in the inner ear is a common cause of auditory and vestibular impairment in mammals, which have a limited ability to regrow these cells after damage. In contrast, non-mammalian vertebrates including zebrafish can robustly regenerate hair cells following severe organ damage. The zebrafish inner ear provides an understudied model system for understanding hair cell regeneration in organs that are highly conserved with their mammalian counterparts. Here we quantitatively examine hair cell addition during growth and regeneration of the larval zebrafish inner ear. We used a genetically encoded ablation method to induce hair cell death and observed gradual regeneration with correct spatial patterning over two weeks following ablation. Supporting cells, which surround and are a source of new hair cells, divide in response to hair cell ablation, expanding the possible progenitor pool. In parallel, nascent hair cells arise from direct transdifferentiation of progenitor pool cells uncoupled from progenitor division. These findings reveal a previously unrecognized mechanism of hair cell regeneration with implications for how hair cells may be encouraged to regenerate in the mammalian ear.
Collapse
Affiliation(s)
- Marielle O. Beaulieu
- Molecular and Cellular Biology Graduate Program, Seattle, WA
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology - Head and Neck Surgery, Seattle, WA
| | - Eric D. Thomas
- Neuroscience Graduate Program, Seattle, WA
- Department of Biological Structure University of Washington, Seattle, WA
| | - David W. Raible
- Molecular and Cellular Biology Graduate Program, Seattle, WA
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology - Head and Neck Surgery, Seattle, WA
- Neuroscience Graduate Program, Seattle, WA
- Department of Biological Structure University of Washington, Seattle, WA
| |
Collapse
|
6
|
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
|
7
|
Kwak M, Southard KM, Kim WR, Lin A, Kim NH, Gopalappa R, Lee HJ, An M, Choi SH, Jung Y, Noh K, Farlow J, Georgakopoulos A, Robakis NK, Kang MK, Kutys ML, Seo D, Kim HH, Kim YH, Cheon J, Gartner ZJ, Jun YW. Adherens junctions organize size-selective proteolytic hotspots critical for Notch signalling. Nat Cell Biol 2022; 24:1739-1753. [PMID: 36456828 PMCID: PMC10665132 DOI: 10.1038/s41556-022-01031-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 10/19/2022] [Indexed: 12/02/2022]
Abstract
Adherens junctions (AJs) create spatially, chemically and mechanically discrete microdomains at cellular interfaces. Here, using a mechanogenetic platform that generates artificial AJs with controlled protein localization, clustering and mechanical loading, we find that AJs also organize proteolytic hotspots for γ-secretase with a spatially regulated substrate selectivity that is critical in the processing of Notch and other transmembrane proteins. Membrane microdomains outside of AJs exclusively organize Notch ligand-receptor engagement (LRE microdomains) to initiate receptor activation. Conversely, membrane microdomains within AJs exclusively serve to coordinate regulated intramembrane proteolysis (RIP microdomains). They do so by concentrating γ-secretase and primed receptors while excluding full-length Notch. AJs induce these functionally distinct microdomains by means of lipid-dependent γ-secretase recruitment and size-dependent protein segregation. By excluding full-length Notch from RIP microdomains, AJs prevent inappropriate enzyme-substrate interactions and suppress spurious Notch activation. Ligand-induced ectodomain shedding eliminates size-dependent segregation, releasing Notch to translocate into AJs for processing by γ-secretase. This mechanism directs radial differentiation of ventricular zone-neural progenitor cells in vivo and more broadly regulates the proteolysis of other large cell-surface receptors such as amyloid precursor protein. These findings suggest an unprecedented role of AJs in creating size-selective spatial switches that choreograph γ-secretase processing of multiple transmembrane proteins regulating development, homeostasis and disease.
Collapse
Affiliation(s)
- Minsuk Kwak
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, Republic of Korea
| | - Kaden M Southard
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Woon Ryoung Kim
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA
| | - Annie Lin
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA
| | - Nam Hyeong Kim
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, Republic of Korea
- Department of Nano Engineering, Sungkyunkwan University, Suwon, Republic of Korea
- Imnewrun Inc., Suwon, Republic of Korea
| | - Ramu Gopalappa
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyun Jung Lee
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA
| | - Minji An
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Seo Hyun Choi
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Yunmin Jung
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Kunwoo Noh
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Justin Farlow
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Anastasios Georgakopoulos
- Department of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nikolaos K Robakis
- Department of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Min K Kang
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Matthew L Kutys
- Department of Cell and Tissue Biology, University of California, San Francisco, CA, USA
| | - Daeha Seo
- Department of Physics and Chemistry, DGIST, Daegu, Republic of Korea
| | - Hyongbum Henry Kim
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Plus Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong Ho Kim
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, Republic of Korea
- Department of Nano Engineering, Sungkyunkwan University, Suwon, Republic of Korea
- Imnewrun Inc., Suwon, Republic of Korea
| | - Jinwoo Cheon
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul, Republic of Korea
| | - Zev J Gartner
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| | - Young-Wook Jun
- Department of Otolaryngology, University of California, San Francisco, CA, USA.
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA.
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA.
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea.
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Abstract
It is well established that humans and other mammals are minimally regenerative compared with organisms such as zebrafish, salamander or amphibians. In recent years, however, the identification of regenerative potential in neonatal mouse tissues that normally heal poorly in adults has transformed our understanding of regenerative capacity in mammals. In this Review, we survey the mammalian tissues for which regenerative or improved neonatal healing has been established, including the heart, cochlear hair cells, the brain and spinal cord, and dense connective tissues. We also highlight common and/or tissue-specific mechanisms of neonatal regeneration, which involve cells, signaling pathways, extracellular matrix, immune cells and other factors. The identification of such common features across neonatal tissues may direct therapeutic strategies that will be broadly applicable to multiple adult tissues.
Collapse
Affiliation(s)
| | - Alice H. Huang
- Department of Orthopedic Surgery, Columbia University, New York, NY 10032, USA
| |
Collapse
|
9
|
Rudolf MA, Andreeva A, Kim CE, DeNovio ACJ, Koshar AN, Baker W, Cartagena-Rivera AX, Corwin JT. Stiffening of Circumferential F-Actin Bands Correlates With Regenerative Failure and May Act as a Biomechanical Brake in the Mammalian Inner Ear. Front Cell Neurosci 2022; 16:859882. [PMID: 35602553 PMCID: PMC9114303 DOI: 10.3389/fncel.2022.859882] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
The loss of inner ear hair cells causes permanent hearing and balance deficits in humans and other mammals, but non-mammals recover after supporting cells (SCs) divide and replace hair cells. The proliferative capacity of mammalian SCs declines as exceptionally thick circumferential F-actin bands develop at their adherens junctions. We hypothesized that the reinforced junctions were limiting regenerative responses of mammalian SCs by impeding changes in cell shape and epithelial tension. Using micropipette aspiration and atomic force microscopy, we measured mechanical properties of utricles from mice and chickens. Our data show that the epithelial surface of the mouse utricle stiffens significantly during postnatal maturation. This stiffening correlates with and is dependent on the postnatal accumulation of F-actin and the cross-linker Alpha-Actinin-4 at SC-SC junctions. In chicken utricles, where SCs lack junctional reinforcement, the epithelial surface remains compliant. There, SCs undergo oriented cell divisions and their apical surfaces progressively elongate throughout development, consistent with anisotropic intraepithelial tension. In chicken utricles, inhibition of actomyosin contractility led to drastic SC shape change and epithelial buckling, but neither occurred in mouse utricles. These findings suggest that species differences in the capacity for hair cell regeneration may be attributable in part to the differences in the stiffness and contractility of the actin cytoskeletal elements that reinforce adherens junctions and participate in regulation of the cell cycle.
Collapse
Affiliation(s)
- Mark A. Rudolf
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Anna Andreeva
- School of Sciences and Humanities, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Christina E. Kim
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Anthony C.-J. DeNovio
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Antoan N. Koshar
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Wendy Baker
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Alexander X. Cartagena-Rivera
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, United States
| | - Jeffrey T. Corwin
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, United States
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
10
|
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: 3.0] [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
|
11
|
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
|
12
|
Bassani S, Beelen E, Rossel M, Voisin N, Morgan A, Arribat Y, Chatron N, Chrast J, Cocca M, Delprat B, Faletra F, Giannuzzi G, Guex N, Machavoine R, Pradervand S, Smits JJ, van de Kamp JM, Ziegler A, Amati F, Marlin S, Kremer H, Locher H, Maurice T, Gasparini P, Girotto G, Reymond A. Variants in USP48 encoding ubiquitin hydrolase are associated with autosomal dominant non-syndromic hereditary hearing loss. Hum Mol Genet 2021; 30:1785-1796. [PMID: 34059922 DOI: 10.1093/hmg/ddab145] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/25/2021] [Accepted: 05/25/2021] [Indexed: 02/07/2023] Open
Abstract
Non-Syndromic Hereditary Hearing Loss (NSHHL) is a genetically heterogeneous sensory disorder with about 120 genes already associated. Through exome sequencing and data aggregation, we identified a family with six affected individuals and one unrelated NSHHL patient with predicted-to-be deleterious missense variants in USP48. We also uncovered an eighth patient presenting unilateral cochlear nerve aplasia and a de novo splice variant in the same gene. USP48 encodes a ubiquitin carboxyl-terminal hydrolase under evolutionary constraint. Pathogenicity of the variants is supported by in vitro assays that showed that the mutated proteins are unable to hydrolyze tetra-ubiquitin. Correspondingly, three-dimensional representation of the protein containing the familial missense variant affects a loop that controls binding to ubiquitin. Consistent with a contribution of USP48 to auditory function, immunohistology showed that the encoded protein is expressed in the developing human inner ear, specifically in the spiral ganglion neurons, outer sulcus, interdental cells of the spiral limbus, stria vascularis, Reissner's membrane, and in the transient Kolliker's organ that is essential for auditory development. Engineered zebrafish knocked-down for usp48, the USP48 ortholog, presented with a delayed development of primary motor neurons, less developed statoacoustic neurons innervating the ears, decreased swimming velocity and circling swimming behavior indicative of vestibular dysfunction and hearing impairment. Corroboratingly, acoustic startle response assays revealed a significant decrease of auditory response of zebrafish lacking usp48 at 600 Hz and 800 Hz wavelengths. In conclusion, we describe a novel autosomal dominant NSHHL gene through a multipronged approach combining exome sequencing, animal modeling, immunohistology and molecular assays.
Collapse
Affiliation(s)
- Sissy Bassani
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.,Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Edward Beelen
- Department of Otorhinolaryngology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Norine Voisin
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Anna Morgan
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy.,Institute for Maternal and Child Health, IRCCS, Burlo Garofolo, Trieste, Italy
| | - Yoan Arribat
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Nicolas Chatron
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.,Service de Génétique, Hospices Civils de Lyon, Lyon, France
| | - Jacqueline Chrast
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Massimiliano Cocca
- Institute for Maternal and Child Health, IRCCS, Burlo Garofolo, Trieste, Italy
| | | | - Flavio Faletra
- Institute for Maternal and Child Health, IRCCS, Burlo Garofolo, Trieste, Italy
| | - Giuliana Giannuzzi
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Nicolas Guex
- Bioinformatics Competence Center, University of Lausanne, Lausanne, Switzerland
| | - Roxane Machavoine
- Centre de référence Surdités Génétiques, Hôpital Necker, Institut Imagine, Paris, France
| | - Sylvain Pradervand
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Jeroen J Smits
- Department of Otorhinolaryngology and Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jiddeke M van de Kamp
- Department of Clinical Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Alban Ziegler
- Centre de référence Surdités Génétiques, Hôpital Necker, Institut Imagine, Paris, France
| | - Francesca Amati
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Sandrine Marlin
- Centre de référence Surdités Génétiques, Hôpital Necker, Institut Imagine, Paris, France
| | - Hannie Kremer
- Department of Otorhinolaryngology and Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Heiko Locher
- Department of Otorhinolaryngology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tangui Maurice
- MMDN, Univ Montpellier, EPHE, INSERM, Montpellier, France
| | - Paolo Gasparini
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy.,Institute for Maternal and Child Health, IRCCS, Burlo Garofolo, Trieste, Italy
| | - Giorgia Girotto
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy.,Institute for Maternal and Child Health, IRCCS, Burlo Garofolo, Trieste, Italy
| | - Alexandre Reymond
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
13
|
Borse V, Barton M, Arndt H, Kaur T, Warchol ME. Dynamic patterns of YAP1 expression and cellular localization in the developing and injured utricle. Sci Rep 2021; 11:2140. [PMID: 33495483 PMCID: PMC7835353 DOI: 10.1038/s41598-020-77775-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 11/12/2020] [Indexed: 12/25/2022] Open
Abstract
The Hippo signaling pathway is a key regulator of tissue development and regeneration. Activation of the Hippo pathway leads to nuclear translocation of the YAP1 transcriptional coactivator, resulting in changes in gene expression and cell cycle entry. Recent studies have demonstrated the nuclear translocation of YAP1 during the development of the sensory organs of the inner ear, but the possible role of YAP1 in sensory regeneration of the inner ear is unclear. The present study characterized the cellular localization of YAP1 in the utricles of mice and chicks, both under normal conditions and after HC injury. During neonatal development, YAP1 expression was observed in the cytoplasm of supporting cells, and was transiently expressed in the cytoplasm of some differentiating hair cells. We also observed temporary nuclear translocation of YAP1 in supporting cells of the mouse utricle after short periods in organotypic culture. However, little or no nuclear translocation of YAP1 was observed in the utricles of neonatal or mature mice after ototoxic injury. In contrast, substantial YAP1 nuclear translocation was observed in the chicken utricle after streptomycin treatment in vitro and in vivo. Together, these data suggest that differences in YAP1 signaling may partially account for the differing regenerative abilities of the avian vs. mammalian inner ear.
Collapse
Affiliation(s)
- Vikrant Borse
- Department of Otolaryngology, School of Medicine, Washington University in Saint Louis, 660 South Euclid Ave, Box 8115, St Louis, MO, 63110, USA.
| | - Matthew Barton
- Department of Otolaryngology, School of Medicine, Washington University in Saint Louis, 660 South Euclid Ave, Box 8115, St Louis, MO, 63110, USA
| | - Harry Arndt
- Department of Otolaryngology, School of Medicine, Washington University in Saint Louis, 660 South Euclid Ave, Box 8115, St Louis, MO, 63110, USA
| | - Tejbeer Kaur
- Department of Biomedical Sciences, Creighton University School of Medicine, Nebraska, USA
| | - Mark E Warchol
- Department of Otolaryngology, School of Medicine, Washington University in Saint Louis, 660 South Euclid Ave, Box 8115, St Louis, MO, 63110, USA.
| |
Collapse
|
14
|
The integrity of cochlear hair cells is established and maintained through the localization of Dia1 at apical junctional complexes and stereocilia. Cell Death Dis 2020; 11:536. [PMID: 32678080 PMCID: PMC7366933 DOI: 10.1038/s41419-020-02743-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 07/01/2020] [Accepted: 07/03/2020] [Indexed: 02/07/2023]
Abstract
Dia1, which belongs to the diaphanous-related formin family, influences a variety of cellular processes through straight actin elongation activity. Recently, novel DIA1 mutants such as p.R1213X (p.R1204X) and p.A265S, have been reported to cause an autosomal dominant sensorineural hearing loss (DFNA1). Additionally, active DIA1 mutants induce progressive hearing loss in a gain-of-function manner. However, the subcellular localization and pathological function of DIA1(R1213X/R1204X) remains unknown. In the present study, we demonstrated the localization of endogenous Dia1 and the constitutively active DIA1 mutant in the cochlea, using transgenic mice expressing FLAG-tagged DIA1(R1204X) (DIA1-TG). Endogenous Dia1 and the DIA1 mutant were regionally expressed at the organ of Corti and the spiral ganglion from early life; alongside cochlear maturation, they became localized at the apical junctional complexes (AJCs) between hair cells (HCs) and supporting cells (SCs). To investigate HC vulnerability in the DIA1-TG mice, we exposed 4-week-old mice to moderate noise, which induced temporary threshold shifts with cochlear synaptopathy and ultrastructural changes in stereocilia 4 weeks post noise exposure. Furthermore, we established a knock-in (KI) mouse line expressing AcGFP-tagged DIA1(R1213X) (DIA1-KI) and confirmed mutant localization at AJCs and the tips of stereocilia in HCs. In MDCKAcGFP-DIA1(R1213X) cells with stable expression of AcGFP-DIA1(R1213X), AcGFP-DIA1(R1213X) revealed marked localization at microvilli on the apical surface of cells and decreased localization at cell-cell junctions. The DIA1-TG mice demonstrated hazy and ruffled circumferential actin belts at AJCs and abnormal stereocilia accompanied with HC loss at 5 months of age. In conclusion, Dia1 plays a pivotal role in the development and maintenance of AJCs and stereocilia, ensuring cochlear and HC integrity. Subclinical/latent vulnerability of HCs may be the cause of progressive hearing loss in DFNA1 patients, thus suggesting new therapeutic targets for preventing HC degeneration and progressive hearing loss associated with DFNA1.
Collapse
|
15
|
Rudolf MA, Andreeva A, Kozlowski MM, Kim CE, Moskowitz BA, Anaya-Rocha A, Kelley MW, Corwin JT. YAP Mediates Hair Cell Regeneration in Balance Organs of Chickens, But LATS Kinases Suppress Its Activity in Mice. J Neurosci 2020; 40:3915-3932. [PMID: 32341094 PMCID: PMC7219294 DOI: 10.1523/jneurosci.0306-20.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/07/2020] [Accepted: 04/13/2020] [Indexed: 02/06/2023] Open
Abstract
Loss of sensory hair cells causes permanent hearing and balance deficits in humans and other mammals, but for nonmammals such deficits are temporary. Nonmammals recover hearing and balance sensitivity after supporting cells proliferate and differentiate into replacement hair cells. Evidence of mechanical differences between those sensory epithelia and their supporting cells prompted us to investigate whether the capacity to activate YAP, an effector in the mechanosensitive Hippo pathway, correlates with regenerative capacity in acceleration-sensing utricles of chickens and mice of both sexes. After hair cell ablation, YAP accumulated in supporting cell nuclei in chicken utricles and promoted regenerative proliferation, but YAP remained cytoplasmic and little proliferation occurred in mouse utricles. YAP localization in supporting cells was also more sensitive to shape change and inhibition of MST1/2 in chicken utricles than in mouse utricles. Genetic manipulations showed that in vivo expression of the YAP-S127A variant caused robust proliferation of neonatal mouse supporting cells, which produced progeny that expressed hair cell markers, but proliferative responses declined postnatally. Expression of YAP-5SA, which more effectively evades inhibitory phosphorylation, resulted in TEAD-dependent proliferation of striolar supporting cells, even in adult utricles. Conditional deletion of LATS1/2 kinases abolished the inhibitory phosphorylation of endogenous YAP and led to striolar proliferation in adult mouse utricles. The findings suggest that damage overcomes inhibitory Hippo signaling and facilitates regenerative proliferation in nonmammalian utricles, whereas constitutive LATS1/2 kinase activity suppresses YAP-TEAD signaling in mammalian utricles and contributes to maintaining the proliferative quiescence that appears to underlie the permanence of sensory deficits.SIGNIFICANCE STATEMENT Loud sounds, ototoxic drugs, infections, and aging kill sensory hair cells in the ear, causing irreversible hearing loss and balance deficits for millions. In nonmammals, damage evokes shape changes in supporting cells, which can divide and regenerate hair cells. Such shape changes are limited in mammalian ears, where supporting cells develop E-cadherin-rich apical junctions reinforced by robust F-actin bands, and the cells fail to divide. Here, we find that damage readily activates YAP in supporting cells within balance epithelia of chickens, but not mice. Deleting LATS kinases or expressing YAP variants that evade LATS-mediated inhibitory phosphorylation induces proliferation in supporting cells of adult mice. YAP signaling eventually may be harnessed to overcome proliferative quiescence that limits regeneration in mammalian ears.
Collapse
Affiliation(s)
- Mark A Rudolf
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, Virginia 22908
| | - Anna Andreeva
- School of Sciences and Humanities, Nazarbayev University, Nursultan 010000, Republic of Kazakhstan
| | - Mikolaj M Kozlowski
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, Virginia 22908
| | - Christina E Kim
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, Virginia 22908
| | - Bailey A Moskowitz
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, Virginia 22908
| | - Alejandro Anaya-Rocha
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland 20892
| | - Matthew W Kelley
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland 20892
| | - Jeffrey T Corwin
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, Virginia 22908
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia 22908
| |
Collapse
|
16
|
Prins TJ, Myers ZA, Saldate JJ, Hoffman LF. Calbindin expression in adult vestibular epithelia. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2020; 206:623-637. [PMID: 32350587 DOI: 10.1007/s00359-020-01418-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 03/28/2020] [Accepted: 04/01/2020] [Indexed: 01/11/2023]
Abstract
The mammalian vestibular epithelia exhibit a remarkably stereotyped organization featuring cellular characteristics under planar cell polarity (PCP) control. PCP mechanisms are responsible for the organization of hair cell morphologic polarization vectors, and are thought to be responsible for the postsynaptic expression of the calcium-binding protein calretinin that defines the utricular striola and cristae central zone. However, recent analyses revealed that subtle differences in the topographic expression of oncomodulin, another calcium-binding protein, reflects heterogeneous factors driving the subtle variations in expression. Calbindin represents a third calcium-binding protein that has been previously described to be expressed in both hair cells and afferent calyces in proximity to the utricular striola and crista central zone. The objective of the present investigation was to determine calbindin's topographic pattern of expression to further elucidate the extent to which PCP mechanisms might exert control over the organization of vestibular neuroepithelia. The findings revealed that calbindin exhibited an expression pattern strikingly similar to oncomodulin. However, within calyces of the central zone calbindin was colocalized with calretinin. These results indicate that organizational features of vestibular epithelia are governed by a suite of factors that include PCP mechanisms as well others yet to be defined.
Collapse
Affiliation(s)
- Terry J Prins
- Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA, Box 951624, Los Angeles, CA, 90095-1624, USA.,Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA, 90095, USA
| | - Zachary A Myers
- Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA, Box 951624, Los Angeles, CA, 90095-1624, USA
| | - Johnny J Saldate
- Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA, Box 951624, Los Angeles, CA, 90095-1624, USA
| | - Larry F Hoffman
- Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA, Box 951624, Los Angeles, CA, 90095-1624, USA. .,Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
17
|
EGF and a GSK3 Inhibitor Deplete Junctional E-cadherin and Stimulate Proliferation in the Mature Mammalian Ear. J Neurosci 2020; 40:2618-2632. [PMID: 32079647 DOI: 10.1523/jneurosci.2630-19.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 02/02/2020] [Accepted: 02/09/2020] [Indexed: 11/21/2022] Open
Abstract
Sensory hair cell losses underlie the vast majority of permanent hearing and balance deficits in humans, but many nonmammalian vertebrates can fully recover from hearing impairments and balance dysfunctions because supporting cells (SCs) in their ears retain lifelong regenerative capacities that depend on proliferation and differentiation as replacement hair cells. Most SCs in vertebrate ears stop dividing during embryogenesis; and soon after birth, vestibular SCs in mammals transition to lasting quiescence as they develop massively thickened circumferential F-actin bands at their E-cadherin-rich adherens junctions. Here, we report that treatment with EGF and a GSK3 inhibitor thinned the circumferential F-actin bands throughout the sensory epithelium of cultured utricles that were isolated from adult mice of either sex. That treatment also caused decreases in E-cadherin, β-catenin, and YAP in the striola, and stimulated robust proliferation of mature, normally quiescent striolar SCs. The findings suggest that E-cadherin-rich junctions, which are not present in the SCs of the fish, amphibians, and birds which readily regenerate hair cells, are responsible in part for the mammalian ear's vulnerability to permanent balance and hearing deficits.SIGNIFICANCE STATEMENT Millions of people are affected by hearing and balance deficits that arise when loud sounds, ototoxic drugs, infections, and aging cause hair cell losses. Such deficits are permanent for humans and other mammals, but nonmammals can recover hearing and balance after supporting cells regenerate replacement hair cells. Mammalian supporting cells lose the capacity to proliferate around the time they develop unique, exceptionally reinforced, E-cadherin-rich intercellular junctions. Here, we report the discovery of a pharmacological treatment that thins F-actin bands, depletes E-cadherin, and stimulates proliferation in long-quiescent supporting cells within a balance epithelium from adult mice. The findings suggest that high E-cadherin in those supporting cell junctions may be responsible, in part, for the permanence of hair cell loss in mammals.
Collapse
|
18
|
Notch Signalling: The Multitask Manager of Inner Ear Development and Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1218:129-157. [DOI: 10.1007/978-3-030-34436-8_8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
19
|
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: 21] [Impact Index Per Article: 4.2] [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
|
20
|
Deletion of Brg1 causes stereocilia bundle fusion and cuticular plate loss in vestibular hair cells. Hear Res 2019; 377:247-259. [PMID: 31003036 DOI: 10.1016/j.heares.2019.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/16/2019] [Accepted: 04/03/2019] [Indexed: 11/22/2022]
Abstract
Brg1 is an ATPase subunit of the SWI/SNF chromatin-remodeling complex, and it is indispensable for the development and homeostasis of various organs. Conditional deletion of Brg1 in cochlea hair cells (HCs) leads to multiple structural defects and profound deafness. However, the premature death of Brg1-deficient cochlea HCs hindered further study of the role of Brg1. In contrast to cochlea HCs, Brg1-deficient vestibular HCs survived for a long time. Therefore, HC apical structure and vestibular function were examined in inner HC-specific conditional Brg1 knockout mice. Vestibular HCs exhibited fused and elongated stereocilia bundles after deletion of Brg1, and the cuticular plate was absent in most HCs with fused stereocilia bundles. HC loss was observed in conditional Brg1 knockout mice at the age of 12 months. Morphological defects and HC loss were primarily restricted in the striolar region of the utricle and saccule and in the central region of ampulla. The behavioral tests revealed that Brg1 deletion in HCs caused vestibular dysfunction in older adult mice. These results suggest that Brg1 may play specific roles in the maintenance of the HC stereocilia bundle and the cuticular plate.
Collapse
|
21
|
Molecular therapy for genetic and degenerative vestibular disorders. Curr Opin Otolaryngol Head Neck Surg 2018; 26:307-311. [PMID: 30045104 DOI: 10.1097/moo.0000000000000477] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW The primary purpose of this review is to summarize current literature in the field of vestibular regeneration with a focus on recent developments in molecular and gene therapies. RECENT FINDINGS Since the discovery of limited vestibular hair cell regeneration in mammals in the 1990s, many elegant studies have improved our knowledge of mechanisms of development and regeneration of the vestibular system. A better understanding of the developmental pathways of the vestibular organs has fueled various biological strategies to enhance regeneration, including novel techniques in deriving vestibular hair cells from embryonic and induced pluripotent stem cells. In addition, the identification of specific genetic mutations responsible for vestibular disorders has opened various opportunities for gene replacement therapy. SUMMARY Vestibular dysfunction is a significant clinical problem with limited therapeutic options, warranting research on biological strategies to repair/regenerate the vestibular organs to restore function. The use of gene therapy appears promising in animal models of vestibular dysfunction.
Collapse
|
22
|
Validation of Housekeeping Genes as Reference for Reverse-Transcription-qPCR Analysis in Busulfan-Injured Microvascular Endothelial Cells. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4953806. [PMID: 30386793 PMCID: PMC6189687 DOI: 10.1155/2018/4953806] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 09/10/2018] [Accepted: 09/17/2018] [Indexed: 12/17/2022]
Abstract
Endothelial cells (ECs) could express some important cytokines and signal molecules which play a key role in normal hematopoiesis and repopulation. Busulfan-induced vascular endothelial injury is an important feature after hematopoietic stem cell transplantation (HSCT). But the molecular mechanism of how the injured ECs affect hematopoietic reconstruction is still unknown. It is possibly through modulation of the change of some gene expression. RT-qPCR is one of the most popular methods used to accurately determine gene expression levels, based on stable reference gene (RG) selection from housekeeping genes. So our aim is to select stable RGs for more accurate measures of mRNA levels during Busulfan-induced vascular endothelial injury. In this study, 14 RGs were selected to investigate their expression stability in ECs during 72 hours of EC injury treated with Busulfan. Our results revealed extreme variation in RG stability compared by five statistical algorithms. ywhaz and alas1 were recognized as the two idlest RGs on account of the final ranking, while the two most usually used RGs (gapdh and actb) were not the most stable RGs. Next, these data were verified by testing signalling pathway genes ctnnb1, robo4, and notch1 based on the above four genes ywha, alas1, gapdh, and actb. It shows that the normalization of mRNA expression data using unstable RGs greatly affects gene fold change, which means the reliability of the biological conclusions is questionable. Based on the best RGs used, we also found that robo4 is significantly overexpressed in Busulfan-impaired ECs. In conclusion, our data reaffirms the importance of RGs selection for the valid analysis of gene expression in Busulfan-impaired ECs. And it also provides very useful guidance and basis for more accurate differential expression gene screening and future expanding biomolecule study of different drugs such as cyclophosphamide and fludarabine-injured ECs.
Collapse
|
23
|
Zhang J, Sun H, Salvi R, Ding D. Paraquat initially damages cochlear support cells leading to anoikis-like hair cell death. Hear Res 2018; 364:129-141. [PMID: 29563067 PMCID: PMC5984146 DOI: 10.1016/j.heares.2018.03.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/20/2018] [Accepted: 03/09/2018] [Indexed: 12/11/2022]
Abstract
Paraquat (PQ), one of the most widely used herbicides, is extremely dangerous because it generates the highly toxic superoxide radical. When paraquat was applied to cochlear organotypic cultures, it not only damaged the outer hair cells (OHCs) and inner hair cells (IHCs), but also caused dislocation of the hair cell rows. We hypothesized that the dislocation arose from damage to the support cells (SCs) that anchors hair cells within the epithelium. To test this hypothesis, rat postnatal cochlear cultures were treated with PQ. Shortly after PQ treatment, the rows of OHCs separated from one another and migrated radially away from IHCs suggesting loss of cell-cell adhesion that hold the hair cells in proper alignment. Hair cells dislocation was associated with extensive loss of SCs in the organ of Corti, loss of tympanic border cells (TBCs) beneath the basilar membrane, the early appearance of superoxide staining and caspase-8 labeling in SCs below the OHCs and disintegration of E-cadherin and β-catenin in the organ of Corti. Damage to the TBCs and SCs occurred prior to loss of OHC or IHC loss suggesting a form of detachment-induced apoptosis referred to as anoikis.
Collapse
Affiliation(s)
- Jianhui Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, China; Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, 14214, USA
| | - Hong Sun
- Department of Otorhinolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, China; Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, 14214, USA
| | - Richard Salvi
- Department of Otorhinolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, China; Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, 14214, USA; Department of Audiology and Speech-Language Pathology, Asia University, Taichung, Taiwan, ROC
| | - Dalian Ding
- Department of Otorhinolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, China; Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, 14214, USA.
| |
Collapse
|
24
|
Luo WW, Wang XW, Ma R, Chi FL, Chen P, Cong N, Gu YY, Ren DD, Yang JM. Junctional E-cadherin/p120-catenin Is Correlated with the Absence of Supporting Cells to Hair Cells Conversion in Postnatal Mice Cochleae. Front Mol Neurosci 2018. [PMID: 29515364 PMCID: PMC5826362 DOI: 10.3389/fnmol.2018.00020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Notch inhibition is known to generate supernumerary hair cells (HCs) at the expense of supporting cells (SCs) in the mammalian inner ear. However, inhibition of Notch activity becomes progressively less effective at inducing SC-to-HC conversion in the postnatal cochlea and balance organs as the animal ages. It has been suggested that the SC-to-HC conversion capacity is inversely correlated with E-cadherin accumulation in postnatal mammalian utricles. However, whether E-cadherin localization is linked to the SC-to-HC conversion capacity in the mammalian inner ear is poorly understood. In the present study, we treated cochleae from postnatal day 0 (P0) with the Notch signaling inhibitor DAPT and observed apparent SC-to-HC conversion along with E-cadherin/p120ctn disruption in the sensory region. In addition, the SC-to-HC conversion capacity and E-cadherin/p120ctn disorganization were robust in the apex but decreased toward the base. We further demonstrated that the ability to regenerate HCs and the disruption of E-cadherin/p120ctn concomitantly decreased with age and ceased at P7, even after extended DAPT treatments. This timing is consistent with E-cadherin/p120ctn accumulation in the postnatal cochleae. These results suggest that the decreasing capacity of SCs to transdifferentiate into HCs correlates with E-cadherin/p120ctn localization in the postnatal cochleae, which might account for the absence of SC-to-HC conversion in the mammalian cochlea.
Collapse
Affiliation(s)
- Wen-Wei Luo
- Department of Otology and Skull Base Surgery, Eye & ENT Hospital of Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine, Ministry of Health, Shanghai, China
| | - Xin-Wei Wang
- Department of Otology and Skull Base Surgery, Eye & ENT Hospital of Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine, Ministry of Health, Shanghai, China
| | - Rui Ma
- Department of Otology and Skull Base Surgery, Eye & ENT Hospital of Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine, Ministry of Health, Shanghai, China
| | - Fang-Lu Chi
- Department of Otology and Skull Base Surgery, Eye & ENT Hospital of Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine, Ministry of Health, Shanghai, China
| | - Ping Chen
- Department of Cell Biology, Emory University, Atlanta, GA, United States
| | - Ning Cong
- Department of Otology and Skull Base Surgery, Eye & ENT Hospital of Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine, Ministry of Health, Shanghai, China
| | - Yu-Yan Gu
- Department of Otology and Skull Base Surgery, Eye & ENT Hospital of Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine, Ministry of Health, Shanghai, China
| | - Dong-Dong Ren
- Department of Otology and Skull Base Surgery, Eye & ENT Hospital of Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine, Ministry of Health, Shanghai, China
| | - Juan-Mei Yang
- Department of Otology and Skull Base Surgery, Eye & ENT Hospital of Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine, Ministry of Health, Shanghai, China
| |
Collapse
|
25
|
Oncomodulin Expression Reveals New Insights into the Cellular Organization of the Murine Utricle Striola. J Assoc Res Otolaryngol 2018; 19:33-51. [PMID: 29318409 DOI: 10.1007/s10162-017-0652-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 12/18/2017] [Indexed: 01/19/2023] Open
Abstract
Oncomodulin (OCM, aka β-parvalbumin) is an EF-hand calcium binding protein that is expressed in a restricted set of hair cells in the peristriolar region of the mammalian utricle. In the present study, we determined the topologic distribution of OCM among hair cell phenotypes to advance our understanding of the cellular organization of the striola and the relationship of these phenotypes with characteristics of tissue polarity. The distributions of OCM-positive (OCM+) hair cells were quantified in utricles of mature C57Bl/6 mice. Immunohistochemistry was conducted using antibodies to OCM, calretinin, and β3-tubulin. Fluorophore-conjugated phalloidin was used to label hair cell stereocilia, which provided the basis for determining hair cell counts and morphologic polarizations. We found OCM expression in striolar types I and II hair cells, though the distributions were dissimilar to the native striolar type I and II distributions, favoring type I hair cells. The distribution of OCM immunoreactivity among striolar type I hair cells also reflected nonrandom distribution among type Ic and Id phenotypes (i.e., those receiving calretinin-positive and calretinin-negative calyces, respectively). However, many OCM+ hair cells were found lateral to the striola, and within the epithelial region encompassing OCM+ hair cells, the distributions of OCM+ types Ic and Id hair cells were similar to the native distributions of Ic and Id in this region. Summarily, these data provide a quantitative perspective supporting the existence of different underlying factors driving the topologic expression of OCM in hair cells than those responsible for tissue polarity characteristics associated within the utricular striola, including calretinin expression in afferent calyces.
Collapse
|
26
|
Cytoskeletal Stability in the Auditory Organ In Vivo: RhoA Is Dispensable for Wound Healing but Essential for Hair Cell Development. eNeuro 2017; 4:eN-NWR-0149-17. [PMID: 28929130 PMCID: PMC5602105 DOI: 10.1523/eneuro.0149-17.2017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/24/2017] [Accepted: 08/28/2017] [Indexed: 01/03/2023] Open
Abstract
Wound healing in the inner ear sensory epithelia is performed by the apical domains of supporting cells (SCs). Junctional F-actin belts of SCs are thin during development but become exceptionally thick during maturation. The functional significance of the thick belts is not fully understood. We have studied the role of F-actin belts during wound healing in the developing and adult cochlea of mice in vivo. We show that the thick belts serve as intracellular scaffolds that preserve the positions of surviving cells in the cochlear sensory epithelium. Junctions associated with the thick F-actin belts did not readily disassemble during wound healing. To compensate for this, basolateral membranes of SCs participated in the closure of surface breach. Because not only neighboring but also distant SCs contributed to wound healing by basolateral protrusions, this event appears to be triggered by contact-independent diffusible signals. In the search for regulators of wound healing, we inactivated RhoA in SCs, which, however, did not limit wound healing. RhoA inactivation in developing outer hair cells (OHCs) caused myosin II delocalization from the perijunctional domain and apical cell-surface enlargement. These abnormalities led to the extrusion of OHCs from the epithelium. These results demonstrate the importance of stability of the apical domain, both in wound repair by SCs and in development of OHCs, and that only this latter function is regulated by RhoA. Because the correct cytoarchitecture of the cochlear sensory epithelium is required for normal hearing, the stability of cell apices should be maintained in regenerative and protective interventions.
Collapse
|
27
|
Diphtheria Toxin-Induced Cell Death Triggers Wnt-Dependent Hair Cell Regeneration in Neonatal Mice. J Neurosci 2017; 36:9479-89. [PMID: 27605621 DOI: 10.1523/jneurosci.2447-15.2016] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 07/20/2016] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED Cochlear hair cells (HCs), the sensory cells that respond to sound, do not regenerate after damage in adult mammals, and their loss is a major cause of deafness. Here we show that HC regeneration in newborn mouse ears occurred spontaneously when the original cells were ablated by treatment with diphtheria toxin (DT) in ears that had been engineered to overexpress the DT receptor, but was not detectable when HCs were ablated in vivo by the aminoglycoside antibiotic neomycin. A variety of Wnts (Wnt1, Wnt2, Wnt2b, Wnt4, Wnt5a, Wnt7b, Wnt9a, Wnt9b, and Wnt11) and Wnt pathway component Krm2 were upregulated after DT damage. Nuclear β-catenin was upregulated in HCs and supporting cells of the DT-damaged cochlea. Pharmacological inhibition of Wnt decreased spontaneous regeneration, confirming a role of Wnt signaling in HC regeneration. Inhibition of Notch signaling further potentiated supporting cell proliferation and HC differentiation that occurred spontaneously. The absence of new HCs in the neomycin ears was correlated to less robust Wnt pathway activation, but the ears subjected to neomycin treatment nonetheless showed increased cell division and HC differentiation after subsequent forced upregulation of β-catenin. These studies suggest, first, that Wnt signaling plays a key role in regeneration, and, second, that the outcome of a regenerative response to damage in the newborn cochlea is determined by reaching a threshold level of Wnt signaling rather than its complete absence or presence. SIGNIFICANCE STATEMENT Sensory HCs of the inner ear do not regenerate in the adult, and their loss is a major cause of deafness. We found that HCs regenerated spontaneously in the newborn mouse after diphtheria toxin (DT)-induced, but not neomycin-induced, HC death. Regeneration depended on activation of Wnt signaling, and regeneration in DT-treated ears correlated to a higher level of Wnt activation than occurred in nonregenerating neomycin-treated ears. This is significant because insufficient regeneration caused by a failure to reach a threshold level of signaling, if true in the adult, has the potential to be exploited for development of clinical approaches for the treatment of deafness caused by HC loss.
Collapse
|
28
|
Gnedeva K, Jacobo A, Salvi JD, Petelski AA, Hudspeth AJ. Elastic force restricts growth of the murine utricle. eLife 2017; 6. [PMID: 28742024 PMCID: PMC5550282 DOI: 10.7554/elife.25681] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 07/21/2017] [Indexed: 12/30/2022] Open
Abstract
Dysfunctions of hearing and balance are often irreversible in mammals owing to the inability of cells in the inner ear to proliferate and replace lost sensory receptors. To determine the molecular basis of this deficiency we have investigated the dynamics of growth and cellular proliferation in a murine vestibular organ, the utricle. Based on this analysis, we have created a theoretical model that captures the key features of the organ’s morphogenesis. Our experimental data and model demonstrate that an elastic force opposes growth of the utricular sensory epithelium during development, confines cellular proliferation to the organ’s periphery, and eventually arrests its growth. We find that an increase in cellular density and the subsequent degradation of the transcriptional cofactor Yap underlie this process. A reduction in mechanical constraints results in accumulation and nuclear translocation of Yap, which triggers proliferation and restores the utricle’s growth; interfering with Yap’s activity reverses this effect. DOI:http://dx.doi.org/10.7554/eLife.25681.001
Collapse
Affiliation(s)
- Ksenia Gnedeva
- Howard Hughes Medical Institute and Laboratory of Sensory Neuroscience, The Rockefeller University, New York, United States.,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, United States
| | - Adrian Jacobo
- Howard Hughes Medical Institute and Laboratory of Sensory Neuroscience, The Rockefeller University, New York, United States
| | - Joshua D Salvi
- Howard Hughes Medical Institute and Laboratory of Sensory Neuroscience, The Rockefeller University, New York, United States
| | - Aleksandra A Petelski
- Howard Hughes Medical Institute and Laboratory of Sensory Neuroscience, The Rockefeller University, New York, United States
| | - A J Hudspeth
- Howard Hughes Medical Institute and Laboratory of Sensory Neuroscience, The Rockefeller University, New York, United States
| |
Collapse
|
29
|
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: 46] [Impact Index Per Article: 6.6] [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
|
30
|
Kelley MW, Stone JS. Development and Regeneration of Sensory Hair Cells. AUDITORY DEVELOPMENT AND PLASTICITY 2017. [DOI: 10.1007/978-3-319-21530-3_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
31
|
Li W, You D, Chen Y, Chai R, Li H. Regeneration of hair cells in the mammalian vestibular system. Front Med 2016; 10:143-51. [DOI: 10.1007/s11684-016-0451-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 04/11/2016] [Indexed: 11/25/2022]
|
32
|
A central to peripheral progression of cell cycle exit and hair cell differentiation in the developing mouse cristae. Dev Biol 2016; 411:1-14. [PMID: 26826497 DOI: 10.1016/j.ydbio.2016.01.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 01/26/2016] [Indexed: 01/15/2023]
Abstract
The inner ear contains six distinct sensory organs that each maintains some ability to regenerate hair cells into adulthood. In the postnatal cochlea, there appears to be a relationship between the developmental maturity of a region and its ability to regenerate as postnatal regeneration largely occurs in the apical turn, which is the last region to differentiate and mature during development. In the mature cristae there are also regional differences in regenerative ability, which led us to hypothesize that there may be a general relationship between the relative maturity of a region and the regenerative competence of that region in all of the inner ear sensory organs. By analyzing adult mouse cristae labeled embryonically with BrdU, we found that hair cell birth starts in the central region and progresses to the periphery with age. Since the peripheral region of the adult cristae also maintains active Notch signaling and some regenerative competence, these results are consistent with the hypothesis that the last regions to develop retain some of their regenerative ability into adulthood. Further, by analyzing embryonic day 14.5 inner ears we provide evidence for a wave of hair cell birth along the longitudinal axis of the cristae from the central regions to the outer edges. Together with the data from the adult inner ears labeled with BrdU as embryos, these results suggest that hair cell differentiation closely follows cell cycle exit in the cristae, unlike in the cochlea where they are uncoupled.
Collapse
|
33
|
Montgomery SC, Cox BC. Whole Mount Dissection and Immunofluorescence of the Adult Mouse Cochlea. J Vis Exp 2016. [PMID: 26779585 DOI: 10.3791/53561] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The organ of Corti, housed in the cochlea of the inner ear, contains mechanosensory hair cells and surrounding supporting cells which are organized in a spiral shape and have a tonotopic gradient for sound detection. The mouse cochlea is approximately 6 mm long and often divided into three turns (apex, middle, and base) for analysis. To investigate cell loss, cell division, or mosaic gene expression, the whole mount or surface preparation of the cochlea is useful. This dissection method allows visualization of all cells within the organ of Corti when combined with immunostaining and confocal microscopy to image cells at different planes in the z-axis. Multiple optical cross-sections can also be obtained from these z-stack images. In addition, the whole mount dissection method can be used for scanning electron microscopy, although a different fixation method is needed. Here, we present a method to isolate the organ of Corti as three intact cochlear turns (apex, middle, and base). This method can be used for mice ranging from one week of age through adulthood and differs from the technique used for neonatal samples where calcification of the cochlea is incomplete. A slightly modified version can be used for dissection of the rat cochlea. We also demonstrate a procedure for immunostaining with fluorescently tagged antibodies.
Collapse
Affiliation(s)
- Scott C Montgomery
- Department of Surgery, Division of Otolaryngology, Southern Illinois University, School of Medicine
| | - Brandon C Cox
- Department of Surgery, Division of Otolaryngology, Southern Illinois University, School of Medicine; Department of Pharmacology, Southern Illinois University, School of Medicine;
| |
Collapse
|
34
|
Single-cell RNA-Seq resolves cellular complexity in sensory organs from the neonatal inner ear. Nat Commun 2015; 6:8557. [PMID: 26469390 PMCID: PMC4634134 DOI: 10.1038/ncomms9557] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 09/03/2015] [Indexed: 12/22/2022] Open
Abstract
In the inner ear, cochlear and vestibular sensory epithelia utilize grossly similar cell types to transduce different stimuli: sound and acceleration. Each individual sensory epithelium is composed of highly heterogeneous populations of cells based on physiological and anatomical criteria. However, limited numbers of each cell type have impeded transcriptional characterization. Here we generated transcriptomes for 301 single cells from the utricular and cochlear sensory epithelia of newborn mice to circumvent this challenge. Cluster analysis indicates distinct profiles for each of the major sensory epithelial cell types, as well as less-distinct sub-populations. Asynchrony within utricles allows reconstruction of the temporal progression of cell-type-specific differentiation and suggests possible plasticity among cells at the sensory–nonsensory boundary. Comparisons of cell types from utricles and cochleae demonstrate divergence between auditory and vestibular cells, despite a common origin. These results provide significant insights into the developmental processes that form unique inner ear cell types. Heterogeneous sensory epithelia of the inner ear are difficult to study owing to the few cells that can be isolated. Here the authors provide insight into the developmental processes underlying the formation of these cells by single-cell RNA-Seq.
Collapse
|
35
|
Xia Y, Cao X, Xue X, Feng Z, Fan Q, Zheng Y, Feng C, Xu H, Xia C, Cheng Y. Development of hair cells in inner ear is associated with expression and promoter methylation of Notch-1 in postnatal mice. Int J Clin Exp Med 2015; 8:15542-15548. [PMID: 26629046 PMCID: PMC4658935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 08/27/2015] [Indexed: 06/05/2023]
Abstract
The present study was designed to investigate the correlation among the number of hair cells in inner ear, Notch-1 gene expression levels and its methylation status of the promoter region in the postnatal mice. The hair cells in inner ear were collected from postnatal mice at day 0, 4, 8 and 16 and counted by immunofluorescence. Notch-1 mRNA expression were measured by real-time quantitative polymerize chain reaction (PCR). Methylation levels of CpG islands in Notch-1 promoters were analyzed by matrix-assisted laser desorption/ionization-time of flight mass spectrometry. The results showed that the number of hair cells in the inner ear increased gradually after birth, which were positively correlated to Notch-1 mRNA expression. However, analysis on methylation of CpG sites in Notch-1 promoter showed that the methylation rates increased gradually after births, which were correlated with the decreased expression of Notch-1. Drug lesion induced the loss of hair cells, and stimulated the expression of Notch-1 mRNA expression, but didn't influence the methylation rates of Notch-1 promoter. We concluded that the Notch-1 mRNA expression level in inner ear tissues is correlated with the development of hair cells. CpG islands in Notch-1 promoter region manifest hypermethylation status when hair cells in inner ear are mature.
Collapse
Affiliation(s)
- Yanghui Xia
- Kunming Sanatorium of Chengdu Military RegionFengshan Door, Wenquan, Anning 650300, Yunnan, China
| | - Xianbao Cao
- Department of Head-Neck Otolaryngology of General Hospital of Chengdu Military Region212 Grand View Road, Kunming 650000, Yunnan, China
| | - Xijun Xue
- Department of Head-Neck Otolaryngology of General Hospital of Chengdu Military Region212 Grand View Road, Kunming 650000, Yunnan, China
| | - Ziliang Feng
- Disease prevention and Control Center of Chengdu Military Region168 Grand View Road, Kunming 650000, Yunnan, China
| | - Quanshui Fan
- Disease prevention and Control Center of Chengdu Military Region168 Grand View Road, Kunming 650000, Yunnan, China
| | - Ying Zheng
- Disease prevention and Control Center of Chengdu Military Region168 Grand View Road, Kunming 650000, Yunnan, China
| | - Chun Feng
- Department of Head-Neck Otolaryngology of General Hospital of Chengdu Military Region212 Grand View Road, Kunming 650000, Yunnan, China
| | - Hongmei Xu
- Department of Head-Neck Otolaryngology of General Hospital of Chengdu Military Region212 Grand View Road, Kunming 650000, Yunnan, China
| | - Chengqiong Xia
- Department of Head-Neck Otolaryngology of General Hospital of Chengdu Military Region212 Grand View Road, Kunming 650000, Yunnan, China
| | - Yingkun Cheng
- Department of Head-Neck Otolaryngology of General Hospital of Chengdu Military Region212 Grand View Road, Kunming 650000, Yunnan, China
| |
Collapse
|
36
|
Atkinson PJ, Huarcaya Najarro E, Sayyid ZN, Cheng AG. Sensory hair cell development and regeneration: similarities and differences. Development 2015; 142:1561-71. [PMID: 25922522 DOI: 10.1242/dev.114926] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Sensory hair cells are mechanoreceptors of the auditory and vestibular systems and are crucial for hearing and balance. In adult mammals, auditory hair cells are unable to regenerate, and damage to these cells results in permanent hearing loss. By contrast, hair cells in the chick cochlea and the zebrafish lateral line are able to regenerate, prompting studies into the signaling pathways, morphogen gradients and transcription factors that regulate hair cell development and regeneration in various species. Here, we review these findings and discuss how various signaling pathways and factors function to modulate sensory hair cell development and regeneration. By comparing and contrasting development and regeneration, we also highlight the utility and limitations of using defined developmental cues to drive mammalian hair cell regeneration.
Collapse
Affiliation(s)
- Patrick J Atkinson
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Elvis Huarcaya Najarro
- 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
| | - Alan G Cheng
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
37
|
Spatial and Age-Dependent Hair Cell Generation in the Postnatal Mammalian Utricle. Mol Neurobiol 2015; 53:1601-1612. [PMID: 25666161 DOI: 10.1007/s12035-015-9119-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 01/28/2015] [Indexed: 10/24/2022]
Abstract
Loss of vestibular hair cells is a common cause of balance disorders. Current treatment options for bilateral vestibular dysfunction are limited. During development, atonal homolog 1 (Atoh1) is sufficient and necessary for the formation of hair cells and provides a promising gene target to induce hair cell generation in the mammals. In this study, we used a transgenic mouse line to test the age and cell type specificity of hair cell induction in the postnatal utricle in mice. We found that forced Atoh1 expression in vivo can induce hair cell formation in the utricle from postnatal days 1 to 21, while the efficacy of hair cell induction is progressively reduced as the animals become older. In the utricle, the induction of hair cells occurs both within the sensory region and in cells in the transitional epithelium next to the sensory region. Within the sensory epithelium, the central region, known as the striola, is most subjective to the induction of hair cell formation. Furthermore, forced Atoh1 expression can promote proliferation in an age-dependent manner that mirrors the progressively reduced efficacy of hair cell induction in the postnatal utricle. These results suggest that targeting both cell proliferation and Atoh1 in the utricle striolar region may be explored to induce hair cell regeneration in mammals. The study also demonstrates the usefulness of the animal model that provides an in vivo Atoh1 induction model for vestibular regeneration studies.
Collapse
|
38
|
Steiner AB, Kim T, Cabot V, Hudspeth AJ. Dynamic gene expression by putative hair-cell progenitors during regeneration in the zebrafish lateral line. Proc Natl Acad Sci U S A 2014; 111:E1393-401. [PMID: 24706895 PMCID: PMC3986164 DOI: 10.1073/pnas.1318692111] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Hearing loss is most commonly caused by the destruction of mechanosensory hair cells in the ear. This condition is usually permanent: Despite the presence of putative hair-cell progenitors in the cochlea, hair cells are not naturally replenished in adult mammals. Unlike those of the mammalian ear, the progenitor cells of nonmammalian vertebrates can regenerate hair cells throughout life. The basis of this difference remains largely unexplored but may lie in molecular dissimilarities that affect how progenitors respond to hair-cell death. To approach this issue, we analyzed gene expression in hair-cell progenitors of the lateral-line system. We developed a transgenic line of zebrafish that expresses a red fluorescent protein in the presumptive hair-cell progenitors known as mantle cells. Fluorescence-activated cell sorting from the skins of transgenic larvae, followed by microarray-based expression analysis, revealed a constellation of transcripts that are specifically enriched in these cells. Gene expression analysis after hair-cell ablation uncovered a cohort of genes that are differentially regulated early in regeneration, suggesting possible roles in the response of progenitors to hair-cell death. These results provide a resource for studying hair-cell regeneration and the biology of sensory progenitor cells.
Collapse
Affiliation(s)
- Aaron B Steiner
- Howard Hughes Medical Institute and Laboratory of Sensory Neuroscience, The Rockefeller University, New York, NY 10065
| | | | | | | |
Collapse
|
39
|
Responses to cell loss become restricted as the supporting cells in mammalian vestibular organs grow thick junctional actin bands that develop high stability. J Neurosci 2014; 34:1998-2011. [PMID: 24478379 DOI: 10.1523/jneurosci.4355-13.2014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Sensory hair cell (HC) loss is a major cause of permanent hearing and balance impairments for humans and other mammals. Yet, fish, amphibians, reptiles, and birds readily replace HCs and recover from such sensory deficits. It is unknown what prevents replacement in mammals, but cell replacement capacity declines contemporaneously with massive postnatal thickening of F-actin bands at the junctions between vestibular supporting cells (SCs). In non-mammals, SCs can give rise to regenerated HCs, and the bands remain thin even in adults. Here we investigated the stability of the F-actin bands between SCs in ears from chickens and mice and Madin-Darby canine kidney cells. Pharmacological experiments and fluorescence recovery after photobleaching (FRAP) of SC junctions in utricles from mice that express a γ-actin-GFP fusion protein showed that the thickening F-actin bands develop increased resistance to depolymerization and exceptional stability that parallels a sharp decline in the cell replacement capacity of the maturing mammalian ear. The FRAP recovery rate and the mobile fraction of γ-actin-GFP both decreased as the bands thickened with age and became highly stabilized. In utricles from neonatal mice, time-lapse recordings in the vicinity of dying HCs showed that numerous SCs change shape and organize multicellular actin purse strings that reseal the epithelium. In contrast, adult SCs appeared resistant to deformation, with resealing responses limited to just a few neighboring SCs that did not form purse strings. The exceptional stability of the uniquely thick F-actin bands at the junctions of mature SCs may play an important role in restricting dynamic repair responses in mammalian vestibular epithelia.
Collapse
|
40
|
Gene-expression analysis of hair cell regeneration in the zebrafish lateral line. Proc Natl Acad Sci U S A 2014; 111:E1383-92. [PMID: 24706903 DOI: 10.1073/pnas.1402898111] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Deafness caused by the terminal loss of inner ear hair cells is one of the most common sensory diseases. However, nonmammalian animals (e.g., birds, amphibians, and fish) regenerate damaged hair cells. To understand better the reasons underpinning such disparities in regeneration among vertebrates, we set out to define at high resolution the changes in gene expression associated with the regeneration of hair cells in the zebrafish lateral line. We performed RNA-Seq analyses on regenerating support cells purified by FACS. The resulting expression data were subjected to pathway enrichment analyses, and the differentially expressed genes were validated in vivo via whole-mount in situ hybridizations. We discovered that cell cycle regulators are expressed hours before the activation of Wnt/β-catenin signaling following hair cell death. We propose that Wnt/β-catenin signaling is not involved in regulating the onset of proliferation but governs proliferation at later stages of regeneration. In addition, and in marked contrast to mammals, our data clearly indicate that the Notch pathway is significantly down-regulated shortly after injury, thus uncovering a key difference between the zebrafish and mammalian responses to hair cell injury. Taken together, our findings lay the foundation for identifying differences in signaling pathway regulation that could be exploited as potential therapeutic targets to promote either sensory epithelium or hair cell regeneration in mammals.
Collapse
|
41
|
Sensational placodes: neurogenesis in the otic and olfactory systems. Dev Biol 2014; 389:50-67. [PMID: 24508480 PMCID: PMC3988839 DOI: 10.1016/j.ydbio.2014.01.023] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 01/27/2014] [Accepted: 01/28/2014] [Indexed: 11/22/2022]
Abstract
For both the intricate morphogenetic layout of the sensory cells in the ear and the elegantly radial arrangement of the sensory neurons in the nose, numerous signaling molecules and genetic determinants are required in concert to generate these specialized neuronal populations that help connect us to our environment. In this review, we outline many of the proteins and pathways that play essential roles in the differentiation of otic and olfactory neurons and their integration into their non-neuronal support structures. In both cases, well-known signaling pathways together with region-specific factors transform thickened ectodermal placodes into complex sense organs containing numerous, diverse neuronal subtypes. Olfactory and otic placodes, in combination with migratory neural crest stem cells, generate highly specialized subtypes of neuronal cells that sense sound, position and movement in space, odors and pheromones throughout our lives.
Collapse
|
42
|
Burns JC, Collado MS, Oliver ER, Corwin JT. Specializations of intercellular junctions are associated with the presence and absence of hair cell regeneration in ears from six vertebrate classes. J Comp Neurol 2013; 521:1430-48. [PMID: 23124808 DOI: 10.1002/cne.23250] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 10/10/2012] [Accepted: 10/25/2012] [Indexed: 01/12/2023]
Abstract
Sensory hair cell losses lead to hearing and balance deficits that are permanent for mammals, but temporary for nonmammals because supporting cells in their ears give rise to replacement hair cells. In mice and humans, vestibular supporting cells grow exceptionally large circumferential F-actin belts and their junctions express E-cadherin in patterns that strongly correlate with postnatal declines in regeneration capacity. In contrast, chicken supporting cells retain thin F-actin belts throughout life and express little E-cadherin. To determine whether the junctions in chicken ears might be representative of other ears that also regenerate hair cells, we investigated inner ears from dogfish sharks, zebrafish, bullfrogs, Xenopus, turtles, and the lizard, Anolis. As in chickens, the supporting cells in adult zebrafish, Xenopus, and turtle ears retained thin circumferential F-actin belts and expressed little E-cadherin. Supporting cells in adult sharks and bullfrogs also retained thin belts, but were not tested for E-cadherin. Supporting cells in adult Anolis exhibited wide, but porous webs of F-actin and strong E-cadherin expression. Anolis supporting cells also showed some cell cycle reentry when cultured. The results reveal that the association between thin F-actin belts and low E-cadherin is shared by supporting cells in anamniotes, turtles, and birds, which all can regenerate hair cells. Divergent junctional specializations in supporting cells appear to have arisen independently in Anolis and mammals. The presence of webs of F-actin at the junctions in Anolis appears compatible with supporting cell proliferation, but the solid reinforcement of the F-actin belts in mammals is associated with its absence.
Collapse
Affiliation(s)
- Joseph C Burns
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA
| | | | | | | |
Collapse
|
43
|
Postnatal refinement of auditory hair cell planar polarity deficits occurs in the absence of Vangl2. J Neurosci 2013; 33:14001-16. [PMID: 23986237 DOI: 10.1523/jneurosci.1307-13.2013] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The distinctive planar polarity of auditory hair cells is evident in the polarized organization of the stereociliary bundle. Mutations in the core planar cell polarity gene Van Gogh-like 2 (Vangl2) result in hair cells that fail to properly orient their stereociliary bundles along the mediolateral axis of the cochlea. The severity of this phenotype is graded along the length of the cochlea, similar to the hair cell differentiation gradient, suggesting that an active refinement process corrects planar polarity phenotypes in Vangl2 knock-out (KO) mice. Because Vangl2 gene deletions are lethal, Vangl2 conditional knock-outs (CKOs) were generated to test this hypothesis. When crossed with Pax2-Cre, Vangl2 is deleted from the inner ear, yielding planar polarity phenotypes similar to Vangl2 KOs at late embryonic stages except that Vangl2 CKO mice are viable and do not have craniorachischisis like Vangl2 KOs. Quantification of planar polarity deficits through postnatal development demonstrates the activity of a Vangl2-independent refinement process that rescues the planar polarity phenotype within 10 d of birth. In contrast, the Pax2-Cre;Vangl2 CKO has profound changes in the shape and distribution of outer pillar cell and Deiters' cell phalangeal processes that are not corrected during the period of planar polarity refinement. Auditory brainstem response analyses of adult mice show a 10-15 dB shift in auditory threshold, and distortion product otoacoustic emission measurements indicate that this mild hearing deficit is of cochlear origin. Together, these data demonstrate a Vangl2-independent refinement mechanism that actively reorients auditory stereociliary bundles and reveals an unexpected role of Vangl2 during supporting cell morphogenesis.
Collapse
|
44
|
Hair cell generation by notch inhibition in the adult mammalian cristae. J Assoc Res Otolaryngol 2013; 14:813-28. [PMID: 23989618 DOI: 10.1007/s10162-013-0414-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 08/12/2013] [Indexed: 10/26/2022] Open
Abstract
Balance disorders caused by hair cell loss in the sensory organs of the vestibular system pose a significant health problem worldwide, particularly in the elderly. Currently, this hair cell loss is permanent as there is no effective treatment. This is in stark contrast to nonmammalian vertebrates who robustly regenerate hair cells after damage. This disparity in regenerative potential highlights the need for further manipulation in order to stimulate more robust hair cell regeneration in mammals. In the utricle, Notch signaling is required for maintaining the striolar support cell phenotype into the second postnatal week. Notch signaling has further been implicated in hair cell regeneration after damage in the mature utricle. Here, we investigate the role of Notch signaling in the mature mammalian cristae in order to characterize the Notch-mediated regenerative potential of these sensory organs. For these studies, we used the γ-secretase inhibitor, N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT), in conjunction with a method we developed to culture cristae in vitro. In postnatal and adult cristae, we found that 5 days of DAPT treatment resulted in a downregulation of the Notch effectors Hes1 and Hes5 and also an increase in the total number of Gfi1(+) hair cells. Hes5, as reported by Hes5-GFP, was downregulated specifically in peripheral support cells. Using lineage tracing with proteolipid protein (PLP)/CreER;mTmG mice, we found that these hair cells arose through transdifferentiation of support cells in cristae explanted from mice up to 10 weeks of age. These transdifferentiated cells arose without proliferation and were capable of taking on a hair cell morphology, migrating to the correct cell layer, and assembling what appears to be a stereocilia bundle with a long kinocilium. Overall, these data show that Notch signaling is active in the mature cristae and suggest that it may be important in maintaining the support cell fate in a subset of peripheral support cells.
Collapse
|
45
|
Du X, Li W, Gao X, West MB, Saltzman WM, Cheng CJ, Stewart C, Zheng J, Cheng W, Kopke RD. Regeneration of mammalian cochlear and vestibular hair cells through Hes1/Hes5 modulation with siRNA. Hear Res 2013; 304:91-110. [PMID: 23850665 DOI: 10.1016/j.heares.2013.06.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 05/16/2013] [Accepted: 06/27/2013] [Indexed: 12/31/2022]
Abstract
The Notch pathway is a cell signaling pathway determining initial specification and subsequent cell fate in the inner ear. Previous studies have suggested that new hair cells (HCs) can be regenerated in the inner ear by manipulating the Notch pathway. In the present study, delivery of siRNA to Hes1 and Hes5 using a transfection reagent or siRNA to Hes1 encapsulated within poly(lactide-co-glycolide acid) (PLGA) nanoparticles increased HC numbers in non-toxin treated organotypic cultures of cochleae and maculae of postnatal day 3 mouse pups. An increase in HCs was also observed in cultured cochleae and maculae of mouse pups pre-conditioned with a HC toxin (4-hydroxy-2-nonenal or neomycin) and then treated with the various siRNA formulations. Treating cochleae with siRNA to Hes1 associated with a transfection reagent or siRNA to Hes1 delivered by PLGA nanoparticles decreased Hes1 mRNA and up-regulated Atoh1 mRNA expression allowing supporting cells (SCs) to acquire a HC fate. Experiments using cochleae and maculae of p27(kip1)/-GFP transgenic mouse pups demonstrated that newly generated HCs trans-differentiated from SCs. Furthermore, PLGA nanoparticles are non-toxic to inner ear tissue, readily taken up by cells within the tissue of interest, and present a synthetic delivery system that is a safe alternative to viral vectors. These results indicate that when delivered using a suitable vehicle, Hes siRNAs are potential therapeutic molecules that may have the capacity to regenerate new HCs in the inner ear and possibly restore human hearing and balance function.
Collapse
Affiliation(s)
- Xiaoping Du
- Hough Ear Institute, P.O. Box 23206, Oklahoma City, OK 73112, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Sensory hair cells are exquisitely sensitive vertebrate mechanoreceptors that mediate the senses of hearing and balance. Understanding the factors that regulate the development of these cells is important, not only to increase our understanding of ear development and its functional physiology but also to shed light on how these cells may be replaced therapeutically. In this review, we describe the signals and molecular mechanisms that initiate hair cell development in vertebrates, with particular emphasis on the transcription factor Atoh1, which is both necessary and sufficient for hair cell development. We then discuss recent findings on how microRNAs may modulate the formation and maturation of hair cells. Last, we review recent work on how hair cells are regenerated in many vertebrate groups and the factors that conspire to prevent this regeneration in mammals.
Collapse
Affiliation(s)
- Andrew K Groves
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA.
| | | | | |
Collapse
|
47
|
Coupling the cell cycle to development and regeneration of the inner ear. Semin Cell Dev Biol 2013; 24:507-13. [PMID: 23665151 DOI: 10.1016/j.semcdb.2013.04.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 04/23/2013] [Indexed: 12/19/2022]
Abstract
Cell cycle exit and acquirement of a postmitotic state is essential for the proper development of organs. In the present review, we examine the role of the cell cycle control in the sensory epithelia of the mammalian inner ear. We describe the roles of the core cell cycle regulators in the proliferation of prosensory cells and in the initiation and maintenance of terminal mitosis of the sensory epithelia. We also discuss how other intracellular signalling may influence the cell cycle. Finally, we address the question of whether manipulations of the cell cycle may have the potential to create replacement cells for the damaged inner sensory epithelia.
Collapse
|
48
|
Burns JC, Corwin JT. A historical to present-day account of efforts to answer the question: "what puts the brakes on mammalian hair cell regeneration?". Hear Res 2013; 297:52-67. [PMID: 23333259 PMCID: PMC3594491 DOI: 10.1016/j.heares.2013.01.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2012] [Revised: 12/20/2012] [Accepted: 01/07/2013] [Indexed: 12/17/2022]
Abstract
Hearing and balance deficits often affect humans and other mammals permanently, because their ears stop producing hair cells within a few days after birth. But production occurs throughout life in the ears of sharks, bony fish, amphibians, reptiles, and birds allowing them to replace lost hair cells and quickly recover after temporarily experiencing the kinds of sensory deficits that are irreversible for mammals. Since the mid 1970s, researchers have been asking what puts the brakes on hair cell regeneration in mammals. Here we evaluate the headway that has been made and assess current evidence for alternative mechanistic hypotheses that have been proposed to account for the limits to hair cell regeneration in mammals.
Collapse
Affiliation(s)
- Joseph C Burns
- Department of Neuroscience, University of Virginia, School of Medicine, Charlottesville, VA 22908, USA.
| | | |
Collapse
|
49
|
Rubel EW, Furrer SA, Stone JS. A brief history of hair cell regeneration research and speculations on the future. Hear Res 2013; 297:42-51. [PMID: 23321648 DOI: 10.1016/j.heares.2012.12.014] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 12/18/2012] [Accepted: 12/19/2012] [Indexed: 12/24/2022]
Abstract
Millions of people worldwide suffer from hearing and balance disorders caused by loss of the sensory hair cells that convert sound vibrations and head movements into electrical signals that are conveyed to the brain. In mammals, the great majority of hair cells are produced during embryogenesis. Hair cells that are lost after birth are virtually irreplaceable, leading to permanent disability. Other vertebrates, such as fish and amphibians, produce hair cells throughout life. However, hair cell replacement after damage to the mature inner ear was either not investigated or assumed to be impossible until studies in the late 1980s proved this to be false. Adult birds were shown to regenerate lost hair cells in the auditory sensory epithelium after noise- and ototoxic drug-induced damage. Since then, the field of hair cell regeneration has continued to investigate the capacity of the auditory and vestibular epithelia in vertebrates (fishes, birds, reptiles, and mammals) to regenerate hair cells and to recover function, the molecular mechanisms governing these regenerative capabilities, and the prospect of designing biologically-based treatments for hearing loss and balance disorders. Here, we review the major findings of the field during the past 25 years and speculate how future inner ear repair may one day be achieved.
Collapse
Affiliation(s)
- Edwin W Rubel
- Virginia Merrill Bloedel Hearing Research Center and Department of Otolaryngology and Head & Neck Surgery, University of Washington, Seattle, WA 98195, USA.
| | | | | |
Collapse
|
50
|
|