51
|
Ontogeny of cellular organization and LGR5 expression in porcine cochlea revealed using tissue clearing and 3D imaging. iScience 2022; 25:104695. [PMID: 35865132 PMCID: PMC9294204 DOI: 10.1016/j.isci.2022.104695] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/20/2022] [Accepted: 06/27/2022] [Indexed: 11/23/2022] Open
Abstract
Over 11% of the world's population experience hearing loss. Although there are promising studies to restore hearing in rodent models, the size, ontogeny, genetics, and frequency range of hearing of most rodents' cochlea do not match that of humans. The porcine cochlea can bridge this gap as it shares many anatomical, physiological, and genetic similarities with its human counterpart. Here, we provide a detailed methodology to process and image the porcine cochlea in 3D using tissue clearing and light-sheet microscopy. The resulting 3D images can be employed to compare cochleae across different ages and conditions, investigate the ontogeny of cochlear cytoarchitecture, and produce quantitative expression maps of LGR5, a marker of cochlear progenitors in mice. These data reveal that hair cell organization, inner ear morphology, cellular cartography in the organ of Corti, and spatiotemporal expression of LGR5 are dynamic over developmental stages in a pattern not previously documented.
Collapse
|
52
|
Kelley MW. Cochlear Development; New Tools and Approaches. Front Cell Dev Biol 2022; 10:884240. [PMID: 35813214 PMCID: PMC9260282 DOI: 10.3389/fcell.2022.884240] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/19/2022] [Indexed: 12/21/2022] Open
Abstract
The sensory epithelium of the mammalian cochlea, the organ of Corti, is comprised of at least seven unique cell types including two functionally distinct types of mechanosensory hair cells. All of the cell types within the organ of Corti are believed to develop from a population of precursor cells referred to as prosensory cells. Results from previous studies have begun to identify the developmental processes, lineage restrictions and signaling networks that mediate the specification of many of these cell types, however, the small size of the organ and the limited number of each cell type has hampered progress. Recent technical advances, in particular relating to the ability to capture and characterize gene expression at the single cell level, have opened new avenues for understanding cellular specification in the organ of Corti. This review will cover our current understanding of cellular specification in the cochlea, discuss the most commonly used methods for single cell RNA sequencing and describe how results from a recent study using single cell sequencing provided new insights regarding cellular specification.
Collapse
|
53
|
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
|
54
|
Laporte E, Hermans F, De Vriendt S, Vennekens A, Lambrechts D, Nys C, Cox B, Vankelecom H. Decoding the activated stem cell phenotype of the neonatally maturing pituitary. eLife 2022; 11:75742. [PMID: 35699412 PMCID: PMC9333987 DOI: 10.7554/elife.75742] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 06/13/2022] [Indexed: 12/02/2022] Open
Abstract
The pituitary represents the endocrine master regulator. In mouse, the gland undergoes active maturation immediately after birth. Here, we in detail portrayed the stem cell compartment of neonatal pituitary. Single-cell RNA-sequencing pictured an active gland, revealing proliferative stem as well as hormonal (progenitor) cell populations. The stem cell pool displayed a hybrid epithelial/mesenchymal phenotype, characteristic of development-involved tissue stem cells. Organoid culturing recapitulated the stem cells’ phenotype, interestingly also reproducing their paracrine activity. The pituitary stem cell-activating interleukin-6 advanced organoid growth, although the neonatal stem cell compartment was not visibly affected in Il6−/− mice, likely due to cytokine family redundancy. Further transcriptomic analysis exposed a pronounced WNT pathway in the neonatal gland, shown to be involved in stem cell activation and to overlap with the (fetal) human pituitary transcriptome. Following local damage, the neonatal gland efficiently regenerates, despite absence of additional stem cell proliferation, or upregulated IL-6 or WNT expression, all in line with the already high stem cell activation status, thereby exposing striking differences with adult pituitary. Together, our study decodes the stem cell compartment of neonatal pituitary, exposing an activated state in the maturing gland. Understanding stem cell activation is key to potential pituitary regenerative prospects. The pituitary gland is a pea-sized structure found just below the brain that produces hormones controlling everything from growth and stress to reproduction and immunity. To perform its role, the pituitary gland needs specialised hormone-producing cells, but it also contains stem cells. These stem cells can divide to produce more cells like themselves, or differentiate into cells of different types, including hormone-producing cells. In mice, the stem cells of the pituitary gland appear to be activated in the first few weeks after birth, and later become ‘quiescent’ (or lazy) in the adult pituitary gland. However, it remains unclear how the activated state found in the maturing gland is established and regulated. To answer this question, Laporte et al. used single-cell RNA sequencing, a technique that allows researchers to profile which genes are active in individual cells, which can provide vital information about the state and activity of a tissue. The researchers compared the cells of the maturing pituitary gland of newborn mice to the cells in the established gland of adult mice. This analysis revealed that the maturing pituitary gland is a dynamic tissue, with populations of cells that are actively dividing (including the stem cells), which the mature pituitary gland lacks. Additionally, Laporte et al. established the molecular basis for the activated state of the stem cells in the maturing pituitary gland, which relies on the activation of a cell signalling pathway called WNT. To confirm these findings, Laporte et al. used an organoid system that allowed them to recapitulate the stem cell compartment of the maturing pituitary gland in a dish. When Laporte et al. blocked WNT signalling in these organoids, the organoids failed to form or divide. Furthermore, blocking the pathway directly in newborn mice reduced the number of dividing stem cells in the pituitary gland. Both findings support the notion that WNT signalling is required to establish the activated state of the maturing pituitary gland in newborn mice. Laporte et al. also wanted to know whether the newborn pituitary gland responded to injury differently than the adult gland. It had already been established that the adult pituitary stem cells become activated upon injury, and that the gland has some regenerative capacity. However, when Laporte et al. injured the newborn pituitary gland, the gland was able to fully regenerate, despite the stem cells not becoming more activated. This is likely because these cells are already activated (or ‘primed’), and do not require further activation to divide and repair the gland with the help of other proliferating cells. With these results, Laporte et al. shed light on the activated state of the stem cells in the pituitary gland of newborn mice. This provides insight into the role of these stem cells, as well as unveiling possible routes towards regenerating pituitary tissue. This could eventually prove useful in medicine, in cases when the pituitary gland is damaged or removed.
Collapse
Affiliation(s)
- Emma Laporte
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Florian Hermans
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Silke De Vriendt
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | | | | | - Charlotte Nys
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Benoit Cox
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Hugo Vankelecom
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| |
Collapse
|
55
|
Ma X, Zhang S, Qin S, Guo J, Yuan J, Qiang R, Zhou S, Cao W, Yang J, Ma F, Chai R. Transcriptomic and epigenomic analyses explore the potential role of H3K4me3 in neomycin-induced cochlear Lgr5+ progenitor cell regeneration of hair cells. Hum Cell 2022; 35:1030-1044. [DOI: 10.1007/s13577-022-00727-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 05/17/2022] [Indexed: 12/14/2022]
|
56
|
Hinton AS, Yang-Hood A, Schrader AD, Loose C, Ohlemiller KK, McLean WJ. Approaches to Treat Sensorineural Hearing Loss by Hair-Cell Regeneration: The Current State of Therapeutic Developments and Their Potential Impact on Audiological Clinical Practice. J Am Acad Audiol 2022; 32:661-669. [PMID: 35609593 PMCID: PMC9129918 DOI: 10.1055/s-0042-1750281] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Sensorineural hearing loss (SNHL) is typically a permanent and often progressive condition that is commonly attributed to sensory cell loss. All vertebrates except mammals can regenerate lost sensory cells. Thus, SNHL is currently only treated with hearing aids or cochlear implants. There has been extensive research to understand how regeneration occurs in nonmammals, how hair cells form during development, and what limits regeneration in maturing mammals. These studies motivated efforts to identify therapeutic interventions to regenerate hair cells as a treatment for hearing loss, with a focus on targeting supporting cells to form new sensory hair cells. The approaches include gene therapy and small molecule delivery to the inner ear. At the time of this publication, early-stage clinical trials have been conducted to test targets that have shown evidence of regenerating sensory hair cells in preclinical models. As these potential treatments move closer to a clinical reality, it will be important to understand which therapeutic option is most appropriate for a given population. It is also important to consider which audiological tests should be administered to identify hearing improvement while considering the pharmacokinetics and mechanism of a given approach. Some impacts on audiological practice could include implementing less common audiological measures as standard procedure. As devices are not capable of repairing the damaged underlying biology, hair-cell regeneration treatments could allow patients to benefit more from their devices, move from a cochlear implant candidate to a hearing aid candidate, or move a subject to not needing an assistive device. Here, we describe the background, current state, and future implications of hair-cell regeneration research.
Collapse
Affiliation(s)
| | - Aizhen Yang-Hood
- Department of Otolaryngology, Central Institute for the Deaf, Fay and Carl Simons Center for Hearing and Deafness, Washington University School of Medicine, Saint Louis, Missouri
| | - Angela D Schrader
- Department of Otolaryngology, Central Institute for the Deaf, Fay and Carl Simons Center for Hearing and Deafness, Washington University School of Medicine, Saint Louis, Missouri
| | | | - Kevin K Ohlemiller
- Department of Otolaryngology, Central Institute for the Deaf, Fay and Carl Simons Center for Hearing and Deafness, Washington University School of Medicine, Saint Louis, Missouri
| | - Will J McLean
- Frequency Therapeutics, Lexington, Massachusetts.,Department of Surgery, University of Connecticut School of Medicine, Farmington, Connecticut
| |
Collapse
|
57
|
Lee MP, Waldhaus J. In vitro and in vivo models: What have we learnt about inner ear regeneration and treatment for hearing loss? Mol Cell Neurosci 2022; 120:103736. [PMID: 35577314 PMCID: PMC9551661 DOI: 10.1016/j.mcn.2022.103736] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 04/29/2022] [Accepted: 05/10/2022] [Indexed: 01/07/2023] Open
Abstract
The sensory cells of the inner ear, called hair cells, do not regenerate spontaneously and therefore, hair cell loss and subsequent hearing loss are permanent in humans. Conversely, functional hair cell regeneration can be observed in non-mammalian vertebrate species like birds and fish. Also, during postnatal development in mice, limited regenerative capacity and the potential to isolate stem cells were reported. Together, these findings spurred the interest of current research aiming to investigate the endogenous regenerative potential in mammals. In this review, we summarize current in vitro based approaches and briefly introduce different in vivo model organisms utilized to study hair cell regeneration. Furthermore, we present an overview of the findings that were made synergistically using both, the in vitro and in vivo based tools.
Collapse
Affiliation(s)
- Mary P Lee
- Department of Otolaryngology-Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Joerg Waldhaus
- Department of Otolaryngology-Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
58
|
García-Añoveros J, Clancy JC, Foo CZ, García-Gómez I, Zhou Y, Homma K, Cheatham MA, Duggan A. Tbx2 is a master regulator of inner versus outer hair cell differentiation. Nature 2022; 605:298-303. [PMID: 35508658 PMCID: PMC9803360 DOI: 10.1038/s41586-022-04668-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 03/21/2022] [Indexed: 01/03/2023]
Abstract
The cochlea uses two types of mechanosensory cell to detect sounds. A single row of inner hair cells (IHCs) synapse onto neurons to transmit sensory information to the brain, and three rows of outer hair cells (OHCs) selectively amplify auditory inputs1. So far, two transcription factors have been implicated in the specific differentiation of OHCs, whereas, to our knowledge, none has been identified in the differentiation of IHCs2-4. One such transcription factor for OHCs, INSM1, acts during a crucial embryonic period to consolidate the OHC fate, preventing OHCs from transdifferentiating into IHCs2. In the absence of INSM1, embryonic OHCs misexpress a core set of IHC-specific genes, which we predict are involved in IHC differentiation. Here we find that one of these genes, Tbx2, is a master regulator of IHC versus OHC differentiation in mice. Ablation of Tbx2 in embryonic IHCs results in their development as OHCs, expressing early OHC markers such as Insm1 and eventually becoming completely mature OHCs in the position of IHCs. Furthermore, Tbx2 is epistatic to Insm1: in the absence of both genes, cochleae generate only OHCs, which suggests that TBX2 is necessary for the abnormal transdifferentiation of INSM1-deficient OHCs into IHCs, as well as for normal IHC differentiation. Ablation of Tbx2 in postnatal, largely differentiated IHCs makes them transdifferentiate directly into OHCs, replacing IHC features with those of mature and not embryonic OHCs. Finally, ectopic expression of Tbx2 in OHCs results in their transdifferentiation into IHCs. Hence, Tbx2 is both necessary and sufficient to make IHCs distinct from OHCs and maintain this difference throughout development.
Collapse
Affiliation(s)
- Jaime García-Añoveros
- Department of Anesthesiology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,Department of Neuroscience, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,Hugh Knowles Center for Clinical and Basic Science in Hearing and its Disorders, Northwestern University, Chicago, IL, USA.,These authors jointly supervised this work: Jaime García-Añoveros, Anne Duggan.,Correspondence and requests for materials should be addressed to Jaime García-Añoveros or Anne Duggan. ;
| | - John C. Clancy
- Department of Anesthesiology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Chuan Zhi Foo
- Department of Anesthesiology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,Driskill Graduate Program in Life Sciences, Northwestern University, Chicago, IL, USA
| | - Ignacio García-Gómez
- Department of Anesthesiology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Yingjie Zhou
- Department of Anesthesiology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, USA
| | - Kazuaki Homma
- Hugh Knowles Center for Clinical and Basic Science in Hearing and its Disorders, Northwestern University, Chicago, IL, USA.,Department of Otolaryngology–Head and Neck Surgery, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Mary Ann Cheatham
- Hugh Knowles Center for Clinical and Basic Science in Hearing and its Disorders, Northwestern University, Chicago, IL, USA.,Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, USA
| | - Anne Duggan
- Department of Anesthesiology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.,These authors jointly supervised this work: Jaime García-Añoveros, Anne Duggan.,Correspondence and requests for materials should be addressed to Jaime García-Añoveros or Anne Duggan. ;
| |
Collapse
|
59
|
Waissbluth S, Maass JC, Sanchez HA, Martínez AD. Supporting Cells and Their Potential Roles in Cisplatin-Induced Ototoxicity. Front Neurosci 2022; 16:867034. [PMID: 35573297 PMCID: PMC9104564 DOI: 10.3389/fnins.2022.867034] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
Cisplatin is a known ototoxic chemotherapy drug, causing irreversible hearing loss. Evidence has shown that cisplatin causes inner ear damage as a result of adduct formation, a proinflammatory environment and the generation of reactive oxygen species within the inner ear. The main cochlear targets for cisplatin are commonly known to be the outer hair cells, the stria vascularis and the spiral ganglion neurons. Further evidence has shown that certain transporters can mediate cisplatin influx into the inner ear cells including organic cation transporter 2 (OCT2) and the copper transporter Ctr1. However, the expression profiles for these transporters within inner ear cells are not consistent in the literature, and expression of OCT2 and Ctr1 has also been observed in supporting cells. Organ of Corti supporting cells are essential for hair cell activity and survival. Special interest has been devoted to gap junction expression by these cells as certain mutations have been linked to hearing loss. Interestingly, cisplatin appears to affect connexin expression in the inner ear. While investigations regarding cisplatin-induced hearing loss have been focused mainly on the known targets previously mentioned, the role of supporting cells for cisplatin-induced ototoxicity has been overlooked. In this mini review, we discuss the implications of supporting cells expressing OCT2 and Ctr1 as well as the potential role of gap junctions in cisplatin-induced cytotoxicity.
Collapse
Affiliation(s)
- Sofia Waissbluth
- Department of Otolaryngology, Pontificia Universidad Católica de Chile, Santiago, Chile
- *Correspondence: Sofia Waissbluth, ;
| | - Juan Cristóbal Maass
- Department of Otolaryngology, Hospital Clínico de la Universidad de Chile, Santiago, Chile
| | - Helmuth A. Sanchez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Instituto de Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Agustín D. Martínez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Instituto de Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
60
|
Tao Y, Liu X, Yang L, Chu C, Tan F, Yu Z, Ke J, Li X, Zheng X, Zhao X, Qi J, Lin CP, Chai R, Zhong G, Wu H. AAV-ie-K558R mediated cochlear gene therapy and hair cell regeneration. Signal Transduct Target Ther 2022; 7:109. [PMID: 35449181 PMCID: PMC9023545 DOI: 10.1038/s41392-022-00938-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/29/2022] [Accepted: 02/14/2022] [Indexed: 12/19/2022] Open
Abstract
The cochlea consists of multiple types of cells, including hair cells, supporting cells and spiral ganglion neurons, and is responsible for converting mechanical forces into electric signals that enable hearing. Genetic and environmental factors can result in dysfunctions of cochlear and auditory systems. In recent years, gene therapy has emerged as a promising treatment in animal deafness models. One major challenge of the gene therapy for deafness is to effectively deliver genes to specific cells of cochleae. Here, we screened and identified an AAV-ie mutant, AAV-ie-K558R, that transduces hair cells and supporting cells in the cochleae of neonatal mice with high efficiency. AAV-ie-K558R is a safe vector with no obvious deficits in the hearing system. We found that AAV-ie-K558R can partially restore the hearing loss in Prestin KO mice and, importantly, deliver Atoh1 into cochlear supporting cells to generate hair cell-like cells. Our results demonstrate the clinical potential of AAV-ie-K558R for treating the hearing loss caused by hair cell death.
Collapse
Affiliation(s)
- Yong Tao
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200011, PR China
| | - Xiaoyi Liu
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, PR China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, PR China
| | - Liu Yang
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, PR China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, PR China
| | - Cenfeng Chu
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, PR China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, PR China
| | - Fangzhi Tan
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, PR China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, PR China
| | - Zehua Yu
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, PR China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, PR China
| | - Junzi Ke
- iHuman Institute, ShanghaiTech University, Shanghai, 201210, PR China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, PR China
| | - Xiang Li
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200011, PR China
| | - Xiaofei Zheng
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200011, PR China
| | - Xingle Zhao
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China.,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200011, PR China
| | - Jieyu Qi
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, PR China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, PR China
| | - Chao-Po Lin
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, PR China
| | - Renjie Chai
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, PR China. .,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, PR China. .,Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing, PR China. .,Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, 100069, Beijing, PR China.
| | - Guisheng Zhong
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China. .,iHuman Institute, ShanghaiTech University, Shanghai, 201210, PR China. .,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, PR China.
| | - Hao Wu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China. .,Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China. .,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, 200011, PR China.
| |
Collapse
|
61
|
Chen J, Gao D, Chen J, Hou S, He B, Li Y, Li S, Zhang F, Sun X, Jin Y, Sun L, Yang J. Pseudo-Temporal Analysis of Single-Cell RNA Sequencing Reveals Trans-Differentiation Potential of Greater Epithelial Ridge Cells Into Hair Cells During Postnatal Development of Cochlea in Rats. Front Mol Neurosci 2022; 15:832813. [PMID: 35370544 PMCID: PMC8966675 DOI: 10.3389/fnmol.2022.832813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/08/2022] [Indexed: 11/23/2022] Open
Abstract
The hair cells of the cochlea play a decisive role in the process of hearing damage and recovery, yet knowledge of their regeneration process is still limited. Greater epithelial ridge (GER) cells, a type of cell present during cochlear development that has the characteristics of a precursor sensory cell, disappear at the time of maturation of hearing development. Its development and evolution remain mysterious for many years. Here, we performed single-cell RNA sequencing to profile the gene expression landscapes of rats’ cochlear basal membrane from P1, P7, and P14 and identified eight major subtypes of GER cells. Furthermore, single-cell trajectory analysis for GER cells and hair cells indicated that among the different subtypes of GER, four subtypes had transient cell proliferation after birth and could transdifferentiate into inner and outer hair cells, and two of them mainly transdifferentiated into inner hair cells. The other two subtypes eventually transdifferentiate into outer hair cells. Our study lays the groundwork for elucidating the mechanisms of the key regulatory genes and signaling pathways in the trans-differentiation of GER cell subtypes into hair cells and provides potential clues to understand hair cell regeneration.
Collapse
Affiliation(s)
- Jianyong Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Ear Science, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Otolaryngology and Translational Medicine, Shanghai, China
| | - Dekun Gao
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Ear Science, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Otolaryngology and Translational Medicine, Shanghai, China
| | - Junmin Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Ear Science, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Otolaryngology and Translational Medicine, Shanghai, China
| | - Shule Hou
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Ear Science, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Otolaryngology and Translational Medicine, Shanghai, China
| | - Baihui He
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Ear Science, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Otolaryngology and Translational Medicine, Shanghai, China
| | - Yue Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Ear Science, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Otolaryngology and Translational Medicine, Shanghai, China
| | - Shuna Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Ear Science, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Otolaryngology and Translational Medicine, Shanghai, China
| | - Fan Zhang
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Ear Science, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Otolaryngology and Translational Medicine, Shanghai, China
| | - Xiayu Sun
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Ear Science, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Otolaryngology and Translational Medicine, Shanghai, China
| | - Yulian Jin
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Ear Science, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Otolaryngology and Translational Medicine, Shanghai, China
- *Correspondence: Yulian Jin,
| | - Lianhua Sun
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Ear Science, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Otolaryngology and Translational Medicine, Shanghai, China
- Lianhua Sun,
| | - Jun Yang
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Ear Science, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Otolaryngology and Translational Medicine, Shanghai, China
- Jun Yang,
| |
Collapse
|
62
|
Single-cell transcriptome analysis reveals three sequential phases of gene expression during zebrafish sensory hair cell regeneration. Dev Cell 2022; 57:799-819.e6. [PMID: 35316618 PMCID: PMC9188816 DOI: 10.1016/j.devcel.2022.03.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/19/2021] [Accepted: 02/28/2022] [Indexed: 12/20/2022]
Abstract
Loss of sensory hair cells (HCs) in the mammalian inner ear leads to permanent hearing and vestibular defects, whereas loss of HCs in zebrafish results in their regeneration. We used single-cell RNA sequencing (scRNA-seq) to characterize the transcriptional dynamics of HC regeneration in zebrafish at unprecedented spatiotemporal resolution. We uncovered three sequentially activated modules: first, an injury/inflammatory response and downregulation of progenitor cell maintenance genes within minutes after HC loss; second, the transient activation of regeneration-specific genes; and third, a robust re-activation of developmental gene programs, including HC specification, cell-cycle activation, ribosome biogenesis, and a metabolic switch to oxidative phosphorylation. The results are relevant not only for our understanding of HC regeneration and how we might be able to trigger it in mammals but also for regenerative processes in general. The data are searchable and publicly accessible via a web-based interface.
Collapse
|
63
|
Li XJ, Morgan C, Goff LA, Doetzlhofer A. Follistatin promotes LIN28B-mediated supporting cell reprogramming and hair cell regeneration in the murine cochlea. SCIENCE ADVANCES 2022; 8:eabj7651. [PMID: 35148175 PMCID: PMC8836811 DOI: 10.1126/sciadv.abj7651] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 12/18/2021] [Indexed: 05/27/2023]
Abstract
Hair cell (HC) loss within the inner ear cochlea is a leading cause for deafness in humans. Before the onset of hearing, immature supporting cells (SCs) in neonatal mice have some limited capacity for HC regeneration. Here, we show that in organoid culture, transient activation of the progenitor-specific RNA binding protein LIN28B and Activin antagonist follistatin (FST) enhances regenerative competence of maturing/mature cochlear SCs by reprogramming them into progenitor-like cells. Transcriptome profiling and mechanistic studies reveal that LIN28B drives SC reprogramming, while FST is required to counterbalance hyperactivation of transforming growth factor-β-type signaling by LIN28B. Last, we show that LIN28B and FST coactivation enhances spontaneous cochlear HC regeneration in neonatal mice and that LIN28B may be part of an endogenous repair mechanism that primes SCs for HC regeneration. These findings indicate that SC dedifferentiation is critical for HC regeneration and identify LIN28B and FST as main regulators.
Collapse
Affiliation(s)
- Xiao-Jun Li
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Charles Morgan
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Loyal A. Goff
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Angelika Doetzlhofer
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Otolaryngology and Center for Hearing and Balance, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
64
|
Rai V, Tu S, Frank JR, Zuo J. Molecular Pathways Modulating Sensory Hair Cell Regeneration in Adult Mammalian Cochleae: Progress and Perspectives. Int J Mol Sci 2021; 23:ijms23010066. [PMID: 35008497 PMCID: PMC8745006 DOI: 10.3390/ijms23010066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/20/2021] [Accepted: 12/20/2021] [Indexed: 12/30/2022] Open
Abstract
Noise-induced, drug-related, and age-related disabling hearing loss is a major public health problem and affect approximately 466 million people worldwide. In non-mammalian vertebrates, the death of sensory hair cells (HCs) induces the proliferation and transdifferentiation of adjacent supporting cells into new HCs; however, this capacity is lost in juvenile and adult mammalian cochleae leading to permanent hearing loss. At present, cochlear implants and hearing devices are the only available treatments and can help patients to a certain extent; however, no biological approach or FDA-approved drug is effective to treat disabling hearing loss and restore hearing. Recently, regeneration of mammalian cochlear HCs by modulating molecular pathways or transcription factors has offered some promising results, although the immaturity of the regenerated HCs remains the biggest concern. Furthermore, most of the research done is in neonates and not in adults. This review focuses on critically summarizing the studies done in adult mammalian cochleae and discusses various strategies to elucidate novel transcription factors for better therapeutics.
Collapse
Affiliation(s)
| | | | | | - Jian Zuo
- Correspondence: ; Tel.: +1-(402)-280-2916
| |
Collapse
|
65
|
Xu Z, Rai V, Zuo J. TUB and ZNF532 Promote the Atoh1-Mediated Hair Cell Regeneration in Mouse Cochleae. Front Cell Neurosci 2021; 15:759223. [PMID: 34819838 PMCID: PMC8606527 DOI: 10.3389/fncel.2021.759223] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/15/2021] [Indexed: 12/31/2022] Open
Abstract
Hair cell (HC) regeneration is a promising therapy for permanent sensorineural hearing loss caused by HC loss in mammals. Atoh1 has been shown to convert supporting cells (SCs) to HCs in neonatal cochleae; its combinations with other factors can improve the efficiency of HC regeneration. To identify additional transcription factors for efficient Atoh1-mediated HC regeneration, here we optimized the electroporation procedure for explant culture of neonatal mouse organs of Corti and tested multiple transcription factors, Six2, Ikzf2, Lbh, Arid3b, Hmg20 a, Tub, Sall1, and Znf532, for their potential to promote Atoh1-mediated conversion of SCs to HCs. These transcription factors are expressed highly in HCs but differentially compared to the converted HCs based on previous studies, and are also potential co-reprograming factors for Atoh1-mediated SC-to-HC conversion by literature review. P0.5 cochlear explants were electroporated with these transcription factors alone or jointly with Atoh1. We found that Sox2+ progenitors concentrated within the lateral greater epithelial ridge (GER) can be electroporated efficiently with minimal HC damage. Atoh1 ectopic expression promoted HC regeneration in Sox2+ lateral GER cells. Transcription factors Tub and Znf532, but not the other six tested, promoted the HC regeneration mediated by Atoh1, consistent with previous studies that Isl1 promotes Atoh1-mediated HC conversionex vivo and in vivo and that both Tub and Znf532 are downstream targets of Isl1. Thus, our studies revealed an optimized electroporation method that can transfect the Sox2+ lateral GER cells efficiently with minimal damage to the endogenous HCs. Our results also demonstrate the importance of the Isl1/Tub/Znf532 pathway in promoting Atoh1-mediated HC regeneration.
Collapse
Affiliation(s)
- Zhenhang Xu
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States.,Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, China
| | - Vikrant Rai
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| | - Jian Zuo
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| |
Collapse
|
66
|
Udagawa T, Atkinson PJ, Milon B, Abitbol JM, Song Y, Sperber M, Huarcaya Najarro E, Scheibinger M, Elkon R, Hertzano R, Cheng AG. Lineage-tracing and translatomic analysis of damage-inducible mitotic cochlear progenitors identifies candidate genes regulating regeneration. PLoS Biol 2021; 19:e3001445. [PMID: 34758021 PMCID: PMC8608324 DOI: 10.1371/journal.pbio.3001445] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 11/22/2021] [Accepted: 10/18/2021] [Indexed: 12/21/2022] Open
Abstract
Cochlear supporting cells (SCs) are glia-like cells critical for hearing function. In the neonatal cochlea, the greater epithelial ridge (GER) is a mitotically quiescent and transient organ, which has been shown to nonmitotically regenerate SCs. Here, we ablated Lgr5+ SCs using Lgr5-DTR mice and found mitotic regeneration of SCs by GER cells in vivo. With lineage tracing, we show that the GER houses progenitor cells that robustly divide and migrate into the organ of Corti to replenish ablated SCs. Regenerated SCs display coordinated calcium transients, markers of the SC subtype inner phalangeal cells, and survive in the mature cochlea. Via RiboTag, RNA-sequencing, and gene clustering algorithms, we reveal 11 distinct gene clusters comprising markers of the quiescent and damaged GER, and damage-responsive genes driving cell migration and mitotic regeneration. Together, our study characterizes GER cells as mitotic progenitors with regenerative potential and unveils their quiescent and damaged translatomes.
Collapse
Affiliation(s)
- Tomokatsu Udagawa
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Otorhinolaryngology, The Jikei University School of Medicine, Tokyo, Japan
| | - Patrick J. Atkinson
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Beatrice Milon
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Julia M. Abitbol
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Yang Song
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Michal Sperber
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Elvis Huarcaya Najarro
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Mirko Scheibinger
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Ran Elkon
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ronna Hertzano
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Alan G. Cheng
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
67
|
He Z, Ding Y, Mu Y, Xu X, Kong W, Chai R, Chen X. Stem Cell-Based Therapies in Hearing Loss. Front Cell Dev Biol 2021; 9:730042. [PMID: 34746126 PMCID: PMC8567027 DOI: 10.3389/fcell.2021.730042] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 10/04/2021] [Indexed: 12/19/2022] Open
Abstract
In recent years, neural stem cell transplantation has received widespread attention as a new treatment method for supplementing specific cells damaged by disease, such as neurodegenerative diseases. A number of studies have proved that the transplantation of neural stem cells in multiple organs has an important therapeutic effect on activation and regeneration of cells, and restore damaged neurons. This article describes the methods for inducing the differentiation of endogenous and exogenous stem cells, the implantation operation and regulation of exogenous stem cells after implanted into the inner ear, and it elaborates the relevant signal pathways of stem cells in the inner ear, as well as the clinical application of various new materials. At present, stem cell therapy still has limitations, but the role of this technology in the treatment of hearing diseases has been widely recognized. With the development of related research, stem cell therapy will play a greater role in the treatment of diseases related to the inner ear.
Collapse
Affiliation(s)
- Zuhong He
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yanyan Ding
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yurong Mu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxiang Xu
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Weijia Kong
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Renjie Chai
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, School of Life Sciences and Technology, Southeast University, Nanjing, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China.,Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Xiong Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
68
|
Engerer P, Petridou E, Williams PR, Suzuki SC, Yoshimatsu T, Portugues R, Misgeld T, Godinho L. Notch-mediated re-specification of neuronal identity during central nervous system development. Curr Biol 2021; 31:4870-4878.e5. [PMID: 34534440 DOI: 10.1016/j.cub.2021.08.049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 06/27/2021] [Accepted: 08/18/2021] [Indexed: 11/27/2022]
Abstract
Neuronal identity has long been thought of as immutable, so that once a cell acquires a specific fate, it is maintained for life.1 Studies using the overexpression of potent transcription factors to experimentally reprogram neuronal fate in the mouse neocortex2,3 and retina4,5 have challenged this notion by revealing that post-mitotic neurons can switch their identity. Whether fate reprogramming is part of normal development in the central nervous system (CNS) is unclear. While there are some reports of physiological cell fate reprogramming in invertebrates,6,7 and in the vertebrate peripheral nervous system,8 endogenous fate reprogramming in the vertebrate CNS has not been documented. Here, we demonstrate spontaneous fate re-specification in an interneuron lineage in the zebrafish retina. We show that the visual system homeobox 1 (vsx1)-expressing lineage, which has been associated exclusively with excitatory bipolar cell (BC) interneurons,9-12 also generates inhibitory amacrine cells (ACs). We identify a role for Notch signaling in conferring plasticity to nascent vsx1 BCs, allowing suitable transcription factor programs to re-specify them to an AC fate. Overstimulating Notch signaling enhances this physiological phenotype so that both daughters of a vsx1 progenitor differentiate into ACs and partially differentiated vsx1 BCs can be converted into ACs. Furthermore, this physiological re-specification can be mimicked to allow experimental induction of an entirely distinct fate, that of retinal projection neurons, from the vsx1 lineage. Our observations reveal unanticipated plasticity of cell fate during retinal development.
Collapse
Affiliation(s)
- Peter Engerer
- Institute of Neuronal Cell Biology, Technische Universität München, Biedersteiner Strasse 29, 80802 Munich, Germany
| | - Eleni Petridou
- Institute of Neuronal Cell Biology, Technische Universität München, Biedersteiner Strasse 29, 80802 Munich, Germany; Graduate School of Systemic Neurosciences (GSN), Ludwig-Maximilian University of Munich, Großhaderner Strasse 2, 82152 Planegg-Martinsried, Germany
| | - Philip R Williams
- Institute of Neuronal Cell Biology, Technische Universität München, Biedersteiner Strasse 29, 80802 Munich, Germany
| | - Sachihiro C Suzuki
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | - Takeshi Yoshimatsu
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | - Ruben Portugues
- Institute of Neuroscience, Technische Universität München, Biedersteiner Strasse 29, 80802 Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Thomas Misgeld
- Institute of Neuronal Cell Biology, Technische Universität München, Biedersteiner Strasse 29, 80802 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377 Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Leanne Godinho
- Institute of Neuronal Cell Biology, Technische Universität München, Biedersteiner Strasse 29, 80802 Munich, Germany.
| |
Collapse
|
69
|
Guo L, Cao W, Niu Y, He S, Chai R, Yang J. Autophagy Regulates the Survival of Hair Cells and Spiral Ganglion Neurons in Cases of Noise, Ototoxic Drug, and Age-Induced Sensorineural Hearing Loss. Front Cell Neurosci 2021; 15:760422. [PMID: 34720884 PMCID: PMC8548757 DOI: 10.3389/fncel.2021.760422] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/15/2021] [Indexed: 12/24/2022] Open
Abstract
Inner ear hair cells (HCs) and spiral ganglion neurons (SGNs) are the core components of the auditory system. However, they are vulnerable to genetic defects, noise exposure, ototoxic drugs and aging, and loss or damage of HCs and SGNs results in permanent hearing loss due to their limited capacity for spontaneous regeneration in mammals. Many efforts have been made to combat hearing loss including cochlear implants, HC regeneration, gene therapy, and antioxidant drugs. Here we review the role of autophagy in sensorineural hearing loss and the potential targets related to autophagy for the treatment of hearing loss.
Collapse
Affiliation(s)
- Lingna Guo
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China.,Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wei Cao
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuguang Niu
- Department of Ambulatory Medicine, The First Medical Center of PLA General Hospital, Beijing, China
| | - Shuangba He
- Department of Otolaryngology Head and Neck Surgery, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Renjie Chai
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Jianming Yang
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
70
|
Riley BB. Comparative assessment of Fgf's diverse roles in inner ear development: A zebrafish perspective. Dev Dyn 2021; 250:1524-1551. [PMID: 33830554 DOI: 10.1002/dvdy.343] [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: 02/09/2021] [Revised: 03/26/2021] [Accepted: 03/26/2021] [Indexed: 01/21/2023] Open
Abstract
Progress in understanding mechanisms of inner ear development has been remarkably rapid in recent years. The research community has benefited from the availability of several diverse model organisms, including zebrafish, chick, and mouse. The complexity of the inner ear has proven to be a challenge, and the complexity of the mammalian cochlea in particular has been the subject of intense scrutiny. Zebrafish lack a cochlea and exhibit a number of other differences from amniote species, hence they are sometimes seen as less relevant for inner ear studies. However, accumulating evidence shows that underlying cellular and molecular mechanisms are often highly conserved. As a case in point, consideration of the diverse functions of Fgf and its downstream effectors reveals many similarities between vertebrate species, allowing meaningful comparisons the can benefit the entire research community. In this review, I will discuss mechanisms by which Fgf controls key events in early otic development in zebrafish and provide direct comparisons with chick and mouse.
Collapse
Affiliation(s)
- Bruce B Riley
- Biology Department, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
71
|
MECOM promotes supporting cell proliferation and differentiation in cochlea. J Otol 2021; 17:59-66. [PMID: 35949554 PMCID: PMC9349018 DOI: 10.1016/j.joto.2021.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 12/12/2022] Open
Abstract
Permanent damage to hair cells (HCs) is the leading cause of sensory deafness. Supporting cells (SCs) are essential in the restoration of hearing in mammals because they can proliferate and differentiate to HCs. MDS1 and EVI1 complex locus (MECOM) is vital in early development and cell differentiation and regulates the TGF-β signaling pathway to adapt to pathophysiological events, such as hematopoietic proliferation, differentiation and cells death. In addition, MECOM plays an essential role in neurogenesis and craniofacial development. However, the role of MECOM in the development of cochlea and its way to regulate related signaling are not fully understood. To address this problem, this study examined the expression of MECOM during the development of cochlea and observed a significant increase of MECOM at the key point of auditory epithelial morphogenesis, indicating that MECOM may have a vital function in the formation of cochlea and regeneration of HCs. Meanwhile, we tried to explore the possible effect and potential mechanism of MECOM in SC proliferation and HC regeneration. Findings from this study indicate that overexpression of MECOM markedly increases the proliferation of SCs in the inner ear, and the expression of Smad3 and Cdkn2b related to TGF signaling is significantly down-regulated, corresponding to the overexpression of MECOM. Collectively, these data may provide an explanation of the vital function of MECOM in SC proliferation and trans-differentiation into HCs, as well as its regulation. The interaction between MECOM, Wnt, Notch and the TGF-β signaling may provide a feasible approach to induce the regeneration of HCs.
Collapse
|
72
|
Chen Y, Qiang R, Zhang Y, Cao W, Wu L, Jiang P, Ai J, Ma X, Dong Y, Gao X, Li H, Lu L, Zhang S, Chai R. The Expression and Roles of the Super Elongation Complex in Mouse Cochlear Lgr5+ Progenitor Cells. Front Cell Neurosci 2021; 15:735723. [PMID: 34658793 PMCID: PMC8519586 DOI: 10.3389/fncel.2021.735723] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 09/14/2021] [Indexed: 12/04/2022] Open
Abstract
The super elongation complex (SEC) has been reported to play a key role in the proliferation and differentiation of mouse embryonic stem cells. However, the expression pattern and function of the SEC in the inner ear has not been investigated. Here, we studied the inner ear expression pattern of three key SEC components, AFF1, AFF4, and ELL3, and found that these three proteins are all expressed in both cochlear hair cells (HCs)and supporting cells (SCs). We also cultured Lgr5+ inner ear progenitors in vitro for sphere-forming assays and differentiation assays in the presence of the SEC inhibitor flavopiridol. We found that flavopiridol treatment decreased the proliferation ability of Lgr5+ progenitors, while the differentiation ability of Lgr5+ progenitors was not affected. Our results suggest that the SEC might play important roles in regulating inner ear progenitors and thus regulating HC regeneration. Therefore, it will be very meaningful to further investigate the detailed roles of the SEC signaling pathway in the inner ear in vivo in order to develop effective treatments for sensorineural hearing loss.
Collapse
Affiliation(s)
- Yin Chen
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Ruiying Qiang
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, School of Life Sciences and Technology, Southeast University, Nanjing, China
| | - Yuan Zhang
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, School of Life Sciences and Technology, Southeast University, Nanjing, China
| | - Wei Cao
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Hospital of Anhui Medical University, Hefei, China
| | - Leilei Wu
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, School of Life Sciences and Technology, Southeast University, Nanjing, China
| | - Pei Jiang
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, School of Life Sciences and Technology, Southeast University, Nanjing, China
| | - Jingru Ai
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, School of Life Sciences and Technology, Southeast University, Nanjing, China
| | - Xiangyu Ma
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, School of Life Sciences and Technology, Southeast University, Nanjing, China
| | - Ying Dong
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, School of Life Sciences and Technology, Southeast University, Nanjing, China
| | - Xia Gao
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - He Li
- Department of Otolaryngology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ling Lu
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Shasha Zhang
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, School of Life Sciences and Technology, Southeast University, Nanjing, China
| | - Renjie Chai
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, School of Life Sciences and Technology, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| |
Collapse
|
73
|
Kwan KY, White PM. Understanding the differentiation and epigenetics of cochlear sensory progenitors in pursuit of regeneration. Curr Opin Otolaryngol Head Neck Surg 2021; 29:366-372. [PMID: 34374667 PMCID: PMC8452321 DOI: 10.1097/moo.0000000000000741] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PURPOSE OF REVIEW Sensory hair cells (HCs) of the inner ear are responsible for our ability to hear and balance. Loss of these cells results in hearing loss. Stem cell replacement and in situ regeneration have the potential to replace lost HCs. Newly discovered contributions of transcription factor regulatory networks and epigenetic mechanisms in regulating HC differentiation and regeneration are placed into context of the literature. RECENT FINDINGS A wealth of new data has helped to define cochlear sensory progenitors in their developmental trajectories. This includes transcription factor networks, epigenetic manipulations, and cochlear HC subtype specification. SUMMARY Understanding how sensory progenitors differ and how HC subtypes arise will substantially inform efforts in hearing restoration.
Collapse
Affiliation(s)
- Kelvin Y. Kwan
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway, New Jersey
| | - Patricia M. White
- Department of Neuroscience, Ernest J. Del Monte Institute of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
74
|
Xu S, Yang N. Research Progress on the Mechanism of Cochlear Hair Cell Regeneration. Front Cell Neurosci 2021; 15:732507. [PMID: 34489646 PMCID: PMC8417573 DOI: 10.3389/fncel.2021.732507] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 07/28/2021] [Indexed: 12/26/2022] Open
Abstract
Mammalian inner ear hair cells do not have the ability to spontaneously regenerate, so their irreversible damage is the main cause of sensorineural hearing loss. The damage and loss of hair cells are mainly caused by factors such as aging, infection, genetic factors, hypoxia, autoimmune diseases, ototoxic drugs, or noise exposure. In recent years, research on the regeneration and functional recovery of mammalian auditory hair cells has attracted more and more attention in the field of auditory research. How to regenerate and protect hair cells or auditory neurons through biological methods and rebuild auditory circuits and functions are key scientific issues that need to be resolved in this field. This review mainly summarizes and discusses the recent research progress in gene therapy and molecular mechanisms related to hair cell regeneration in the field of sensorineural hearing loss.
Collapse
Affiliation(s)
- Shan Xu
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, China
| | - Ning Yang
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
75
|
Erni ST, Gill JC, Palaferri C, Fernandes G, Buri M, Lazarides K, Grandgirard D, Edge ASB, Leib SL, Roccio M. Hair Cell Generation in Cochlear Culture Models Mediated by Novel γ-Secretase Inhibitors. Front Cell Dev Biol 2021; 9:710159. [PMID: 34485296 PMCID: PMC8414802 DOI: 10.3389/fcell.2021.710159] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 07/26/2021] [Indexed: 12/30/2022] Open
Abstract
Sensorineural hearing loss is prevalent within society affecting the quality of life of 460 million worldwide. In the majority of cases, this is due to insult or degeneration of mechanosensory hair cells in the cochlea. In adult mammals, hair cell loss is irreversible as sensory cells are not replaced spontaneously. Genetic inhibition of Notch signaling had been shown to induce hair cell formation by transdifferentiation of supporting cells in young postnatal rodents and provided an impetus for targeting Notch pathway with small molecule inhibitors for hearing restoration. Here, the oto-regenerative potential of different γ-secretase inhibitors (GSIs) was evaluated in complementary assay models, including cell lines, organotypic cultures of the organ of Corti and cochlear organoids to characterize two novel GSIs (CPD3 and CPD8). GSI-treatment induced hair cell gene expression in all these models and was effective in increasing hair cell numbers, in particular outer hair cells, both in baseline conditions and in response to ototoxic damage. Hair cells were generated from transdifferentiation of supporting cells. Similar findings were obtained in cochlear organoid cultures, used for the first time to probe regeneration following sisomicin-induced damage. Finally, effective absorption of a novel GSI through the round window membrane and hair cell induction was attained in a whole cochlea culture model and in vivo pharmacokinetic comparisons of transtympanic delivery of GSIs and different vehicle formulations were successfully conducted in guinea pigs. This preclinical evaluation of targeting Notch signaling with novel GSIs illustrates methods of characterization for hearing restoration molecules, enabling translation to more complex animal studies and clinical research.
Collapse
Affiliation(s)
- Silvia T Erni
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Laboratory of Inner Ear Research, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - John C Gill
- Audion Therapeutics B.V., Amsterdam, Netherlands
| | - Carlotta Palaferri
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Laboratory of Inner Ear Research, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Gabriella Fernandes
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Laboratory of Inner Ear Research, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Michelle Buri
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Laboratory of Inner Ear Research, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | | | - Denis Grandgirard
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Albert S B Edge
- Massachusetts Eye and Ear, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States.,Harvard Stem Cell Institute, Cambridge, MA, United States
| | - Stephen L Leib
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Marta Roccio
- Cluster for Regenerative Neuroscience, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Laboratory of Inner Ear Research, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich, Zurich, Switzerland.,Department of Otorhinolaryngology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
76
|
Sun S, Li S, Luo Z, Ren M, He S, Wang G, Liu Z. Dual expression of Atoh1 and Ikzf2 promotes transformation of adult cochlear supporting cells into outer hair cells. eLife 2021; 10:66547. [PMID: 34477109 PMCID: PMC8439656 DOI: 10.7554/elife.66547] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 09/02/2021] [Indexed: 12/24/2022] Open
Abstract
Mammalian cochlear outer hair cells (OHCs) are essential for hearing. Severe hearing impairment follows OHC degeneration. Previous attempts at regenerating new OHCs from cochlear supporting cells (SCs) have been unsuccessful, notably lacking expression of the key OHC motor protein, Prestin. Thus, regeneration of Prestin+ OHCs represents a barrier to restore auditory function in vivo. Here, we reported the successful in vivo conversion of adult mouse cochlear SCs into Prestin+ OHC-like cells through the concurrent induction of two key transcriptional factors known to be necessary for OHC development: Atoh1 and Ikzf2. Single-cell RNA sequencing revealed the upregulation of 729 OHC genes and downregulation of 331 SC genes in OHC-like cells. The resulting differentiation status of these OHC-like cells was much more advanced than previously achieved. This study thus established an efficient approach to induce the regeneration of Prestin+ OHCs, paving the way for in vivo cochlear repair via SC transdifferentiation.
Collapse
Affiliation(s)
- Suhong Sun
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Shuting Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhengnan Luo
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Minhui Ren
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Shunji He
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Guangqin Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhiyong Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| |
Collapse
|
77
|
Hickman TT, Hashimoto K, Liberman LD, Liberman MC. Cochlear Synaptic Degeneration and Regeneration After Noise: Effects of Age and Neuronal Subgroup. Front Cell Neurosci 2021; 15:684706. [PMID: 34434091 PMCID: PMC8380781 DOI: 10.3389/fncel.2021.684706] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/30/2021] [Indexed: 01/24/2023] Open
Abstract
In CBA/CaJ mice, confocal analysis has shown that acoustic overexposure can immediately destroy synapses between auditory-nerve fibers (ANFs) and their peripheral targets, the inner hair cells (IHCs), and that years later, a corresponding number of ANF cell bodies degenerate. In guinea pig, post-exposure disappearance of pre-synaptic ribbons can be equally dramatic, however, post-exposure recovery to near-baseline counts has been reported. Since confocal counts are confounded by thresholding issues, the fall and rise of synaptic ribbon counts could represent “regeneration,” i.e., terminal retraction, re-extension and synaptogenesis, or “recovery,” i.e., down- and subsequent up-regulation of synaptic markers. To clarify, we counted pre-synaptic ribbons, assessed their juxtaposition with post-synaptic receptors, measured the extension of ANF terminals, and quantified the spatial organization and size gradients of these synaptic elements around the hair cell. Present results in guinea pigs exposed as adults (14 months), along with prior results in juveniles (1 month), suggest there is post-exposure neural regeneration in the guinea pig, but not the CBA/CaJ mouse, and that this regenerative capacity extends into adulthood. The results also show, for the first time, that the acute synaptic loss is concentrated on the modiolar side of IHCs, consistent with a selective loss of the high-threshold ANFs with low spontaneous rates. The morphological similarities between the post-exposure neurite extension and synaptogenesis, seen spontaneously in the guinea pig, and in CBA/CaJ only with forced overexpression of neurotrophins, suggest that the key difference may be in the degree of sustained or injury-induced expression of these signaling molecules in the cochlea.
Collapse
Affiliation(s)
- Tyler T Hickman
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA, United States.,Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA, United States
| | - Ken Hashimoto
- Department of Otolaryngology-Head and Neck Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Leslie D Liberman
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA, United States
| | - M Charles Liberman
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA, United States.,Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
78
|
Xu J, Yu D, Dong X, Xie X, Xu M, Guo L, Huang L, Tang Q, Gan L. GATA3 maintains the quiescent state of cochlear supporting cells by regulating p27 kip1. Sci Rep 2021; 11:15779. [PMID: 34349220 PMCID: PMC8338922 DOI: 10.1038/s41598-021-95427-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 07/26/2021] [Indexed: 01/22/2023] Open
Abstract
Haplo-insufficiency of the GATA3 gene causes hypoparathyroidism, sensorineural hearing loss, and renal disease (HDR) syndrome. Previous studies have shown that Gata3 is required for the development of the prosensory domain and spiral ganglion neurons (SGNs) of the mouse cochlea during embryogenesis. However, its role in supporting cells (SCs) after cell fate specification is largely unknown. In this study, we used tamoxifen-inducible Sox2CreERT2 mice to delete Gata3 in SCs of the neonatal mouse cochlea and showed that loss of Gata3 resulted in the proliferation of SCs, including the inner pillar cells (IPCs), inner border cells (IBCs), and lateral greater epithelium ridge (GER). In addition, loss of Gata3 resulted in the down-regulation of p27kip1, a cell cycle inhibitor, in the SCs of Gata3-CKO neonatal cochleae. Chromatin immunoprecipitation analysis revealed that GATA3 directly binds to p27kip1 promoter and could maintain the quiescent state of cochlear SCs by regulating p27kip1 expression. Furthermore, RNA-seq analysis revealed that loss of Gata3 function resulted in the change in the expression of genes essential for the development and function of cochlear SCs, including Tectb, Cyp26b1, Slitrk6, Ano1, and Aqp4.
Collapse
Affiliation(s)
- Jiadong Xu
- College of Life Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, NY, 14642, USA
| | - Dongliang Yu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, Zhejiang, China
| | - Xuhui Dong
- Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, NY, 14642, USA
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Xiaoling Xie
- Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, NY, 14642, USA
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Mei Xu
- College of Life Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, NY, 14642, USA
| | - Luming Guo
- College of Life Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, NY, 14642, USA
| | - Liang Huang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Qi Tang
- Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, NY, 14642, USA
- Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Gan
- Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, NY, 14642, USA.
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
79
|
Alesci A, Pergolizzi S, Lo Cascio P, Fumia A, Lauriano ER. Neuronal regeneration: Vertebrates comparative overview and new perspectives for neurodegenerative diseases. ACTA ZOOL-STOCKHOLM 2021. [DOI: 10.1111/azo.12397] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Alessio Alesci
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences University of Messina Messina Italy
| | - Simona Pergolizzi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences University of Messina Messina Italy
| | - Patrizia Lo Cascio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences University of Messina Messina Italy
| | - Angelo Fumia
- Department of Clinical and Experimental Medicine University of Messina Messina Italy
| | - Eugenia Rita Lauriano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences University of Messina Messina Italy
| |
Collapse
|
80
|
Abstract
Hearing loss is often caused by death of sensory hair cells (HCs) in the inner ear. HCs are vulnerable to some ototoxic drugs, such as aminoglycosides(AGs) and the cisplatin.The most predominant form of drug-induced cell death is apoptosis. Many efforts have been made to protect HCs from cell death after ototoxic drug exposure. These mechanisms and potential targets of HCs protection will be discussed in this review.And we also propose further investigation in the field of HCs necrosis and regeneration, as well as future clinical utilization.
Collapse
|
81
|
Mao H, Chen Y. Noise-Induced Hearing Loss: Updates on Molecular Targets and Potential Interventions. Neural Plast 2021; 2021:4784385. [PMID: 34306060 PMCID: PMC8279877 DOI: 10.1155/2021/4784385] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/12/2021] [Indexed: 12/18/2022] Open
Abstract
Noise overexposure leads to hair cell loss, synaptic ribbon reduction, and auditory nerve deterioration, resulting in transient or permanent hearing loss depending on the exposure severity. Oxidative stress, inflammation, calcium overload, glutamate excitotoxicity, and energy metabolism disturbance are the main contributors to noise-induced hearing loss (NIHL) up to now. Gene variations are also identified as NIHL related. Glucocorticoid is the only approved medication for NIHL treatment. New pharmaceuticals targeting oxidative stress, inflammation, or noise-induced neuropathy are emerging, highlighted by the nanoparticle-based drug delivery system. Given the complexity of the pathogenesis behind NIHL, deeper and more comprehensive studies still need to be fulfilled.
Collapse
Affiliation(s)
- Huanyu Mao
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai 200031, China
| | - Yan Chen
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai 200031, China
| |
Collapse
|
82
|
Sun F, Zhou K, Tian KY, Zhang XY, Liu W, Wang J, Zhong CP, Qiu JH, Zha DJ. Atrial Natriuretic Peptide Promotes Neurite Outgrowth and Survival of Cochlear Spiral Ganglion Neurons in vitro Through NPR-A/cGMP/PKG Signaling. Front Cell Dev Biol 2021; 9:681421. [PMID: 34268307 PMCID: PMC8276373 DOI: 10.3389/fcell.2021.681421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/10/2021] [Indexed: 01/22/2023] Open
Abstract
Sensorineural hearing loss (SNHL) is a dominant public health issue affecting millions of people around the globe, which is correlated with the irreversible deterioration of the hair cells and spiral ganglion neurons (SGNs) within the cochlea. Strategies using bioactive molecules that regulate neurite regeneration and neuronal survival to reestablish connections between auditory epithelium or implanted electrodes and SGN neurites would become attractive therapeutic candidates for SNHL. As an intracellular second messenger, cyclic guanosine-3’,5’-monophosphate (cGMP) can be synthesized through activation of particulate guanylate cyclase-coupled natriuretic peptide receptors (NPRs) by natriuretic peptides, which in turn modulates multiple aspects of neuronal functions including neuronal development and neuronal survival. As a cardiac-derived hormone, atrial natriuretic peptide (ANP), and its specific receptors (NPR-A and NPR-C) are broadly expressed in the nervous system where they might be involved in the maintenance of diverse neural functions. Despite former literatures and our reports indicating the existence of ANP and its receptors within the inner ear, particularly in the spiral ganglion, their potential regulatory mechanisms underlying functional properties of auditory neurons are still incompletely understood. Our recently published investigation revealed that ANP could promote the neurite outgrowth of SGNs by activating NPR-A/cGMP/PKG cascade in a dose-dependent manner. In the present research, the influence of ANP and its receptor-mediated downstream signaling pathways on neurite outgrowth, neurite attraction, and neuronal survival of SGNs in vitro was evaluated by employing cultures of organotypic explant and dissociated neuron from postnatal rats. Our data indicated that ANP could support and attract neurite outgrowth of SGNs and possess a high capacity to improve neuronal survival of SGNs against glutamate-induced excitotoxicity by triggering the NPR-A/cGMP/PKG pathway. The neuroregenerative and neuroprotective effects of ANP/NPRA/cGMP/PKG-dependent signaling on SGNs would represent an attractive therapeutic candidate for hearing impairment.
Collapse
Affiliation(s)
- Fei Sun
- Department of Otolaryngology-Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ke Zhou
- Department of Laboratory Medicine, Institute of Clinical Laboratory Medicine of PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ke-Yong Tian
- Department of Otolaryngology-Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xin-Yu Zhang
- Department of Otolaryngology-Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wei Liu
- Department of Otolaryngology-Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jie Wang
- Department of Otolaryngology-Head and Neck Surgery, The Affiliated Children Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Cui-Ping Zhong
- Department of Otolaryngology-Head and Neck Surgery, The 940th Hospital of Joint Logistics Support Force of PLA, Lanzhou, China
| | - Jian-Hua Qiu
- Department of Otolaryngology-Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ding-Jun Zha
- Department of Otolaryngology-Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
83
|
Deletion of Clusterin Protects Cochlear Hair Cells against Hair Cell Aging and Ototoxicity. Neural Plast 2021; 2021:9979157. [PMID: 34194490 PMCID: PMC8181089 DOI: 10.1155/2021/9979157] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/15/2021] [Accepted: 04/28/2021] [Indexed: 01/06/2023] Open
Abstract
Hearing loss is a debilitating disease that affects 10% of adults worldwide. Most sensorineural hearing loss is caused by the loss of mechanosensitive hair cells in the cochlea, often due to aging, noise, and ototoxic drugs. The identification of genes that can be targeted to slow aging and reduce the vulnerability of hair cells to insults is critical for the prevention of sensorineural hearing loss. Our previous cell-specific transcriptome analysis of adult cochlear hair cells and supporting cells showed that Clu, encoding a secreted chaperone that is involved in several basic biological events, such as cell death, tumor progression, and neurodegenerative disorders, is expressed in hair cells and supporting cells. We generated Clu-null mice (C57BL/6) to investigate its role in the organ of Corti, the sensory epithelium responsible for hearing in the mammalian cochlea. We showed that the deletion of Clu did not affect the development of hair cells and supporting cells; hair cells and supporting cells appeared normal at 1 month of age. Auditory function tests showed that Clu-null mice had hearing thresholds comparable to those of wild-type littermates before 3 months of age. Interestingly, Clu-null mice displayed less hair cell and hearing loss compared to their wildtype littermates after 3 months. Furthermore, the deletion of Clu is protected against aminoglycoside-induced hair cell loss in both in vivo and in vitro models. Our findings suggested that the inhibition of Clu expression could represent a potential therapeutic strategy for the alleviation of age-related and ototoxic drug-induced hearing loss.
Collapse
|
84
|
Kastan N, Gnedeva K, Alisch T, Petelski AA, Huggins DJ, Chiaravalli J, Aharanov A, Shakked A, Tzahor E, Nagiel A, Segil N, Hudspeth AJ. Small-molecule inhibition of Lats kinases may promote Yap-dependent proliferation in postmitotic mammalian tissues. Nat Commun 2021; 12:3100. [PMID: 34035288 PMCID: PMC8149661 DOI: 10.1038/s41467-021-23395-3] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 04/20/2021] [Indexed: 02/04/2023] Open
Abstract
Hippo signaling is an evolutionarily conserved pathway that restricts growth and regeneration predominantly by suppressing the activity of the transcriptional coactivator Yap. Using a high-throughput phenotypic screen, we identified a potent and non-toxic activator of Yap. In vitro kinase assays show that the compound acts as an ATP-competitive inhibitor of Lats kinases-the core enzymes in Hippo signaling. The substance prevents Yap phosphorylation and induces proliferation of supporting cells in the murine inner ear, murine cardiomyocytes, and human Müller glia in retinal organoids. RNA sequencing indicates that the inhibitor reversibly activates the expression of transcriptional Yap targets: upon withdrawal, a subset of supporting-cell progeny exits the cell cycle and upregulates genes characteristic of sensory hair cells. Our results suggest that the pharmacological inhibition of Lats kinases may promote initial stages of the proliferative regeneration of hair cells, a process thought to be permanently suppressed in the adult mammalian inner ear.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Cell Line
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cell Proliferation/genetics
- Ependymoglial Cells/cytology
- Ependymoglial Cells/drug effects
- Ependymoglial Cells/metabolism
- HEK293 Cells
- Hair Cells, Auditory, Inner/cytology
- Hair Cells, Auditory, Inner/drug effects
- Hair Cells, Auditory, Inner/metabolism
- Humans
- Mice, Knockout
- Mice, Transgenic
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Small Molecule Libraries/pharmacology
- Tumor Suppressor Proteins/antagonists & inhibitors
- Tumor Suppressor Proteins/metabolism
- YAP-Signaling Proteins
- Mice
Collapse
Affiliation(s)
- Nathaniel Kastan
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
- Laboratory of Sensory Neuroscience, The Rockefeller University, New York, NY, USA
| | - Ksenia Gnedeva
- Tina and Rick Caruso Department of Otolaryngology-Head and Neck Surgery, University of Southern California, Los Angles, CA, USA.
| | - Theresa Alisch
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
- Laboratory of Sensory Neuroscience, The Rockefeller University, New York, NY, USA
| | - Aleksandra A Petelski
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
- Laboratory of Sensory Neuroscience, The Rockefeller University, New York, NY, USA
- Department of Bioengineering and Barnett Institute, Northeastern University, Boston, MA, USA
| | - David J Huggins
- Tri-Institutional Therapeutics Discovery Institute, New York, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Jeanne Chiaravalli
- High-Throughput Screening Resource Center, The Rockefeller University, New York, NY, USA
- Institut Pasteur, Paris, France
| | - Alla Aharanov
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Avraham Shakked
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Eldad Tzahor
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Aaron Nagiel
- Department of Surgery Children's Hospital Los Angeles, Vision Center, Los Angeles, CA, USA
- Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
- USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Neil Segil
- Tina and Rick Caruso Department of Otolaryngology-Head and Neck Surgery, University of Southern California, Los Angles, CA, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angles, CA, USA
| | - A J Hudspeth
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
- Laboratory of Sensory Neuroscience, The Rockefeller University, New York, NY, USA
| |
Collapse
|
85
|
Selective ablation of inner hair cells and subsequent in-situ hair cell regeneration in the neonatal mouse cochlea. Hear Res 2021; 407:108275. [PMID: 34089989 DOI: 10.1016/j.heares.2021.108275] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 04/24/2021] [Accepted: 05/10/2021] [Indexed: 11/24/2022]
Abstract
Loss of hair cells (HCs) accounts for most sensorineural hearing loss, and regeneration of cochlear HCs is considered as the ultimate strategy for restoring hearing. Several lines of evidence have shown that Lgr5+ progenitor cells can spontaneously regenerate new HCs after HC loss at the neonatal stage, and most of which are immature. IHCs are resistant to ototoxic drugs and noise and cannot be ablated efficiently in order to precisely investigate IHC regeneration in existing hearing injury models, and thus we generated a new transgenic mouse model by inserting diphtheria toxin receptor (DTR) under the control of the Vglut3 promoter. In this model, IHCs were selectively ablated in a dose-dependent manner after the injection of diphtheria toxin (DT) at the neonatal stage, while OHCs remained intact with normal hair bundle structures until adulthood. With this IHC-specific injury model, we observed HC regeneration from Lgr5+ progenitors after IHC ablation at the neonatal stage. Some of the newly generated HCs replaced the lost IHCs in-situ and re-build the structure of the organ of Corti through the asymmetrical mitosis of progenitor cells. While, the majority of the regenerated HCs did not survive until adulthood, and the loss of spiral ganglion neurons was observed after the IHC ablation, which led to profound hearing loss after DT injection in Vglut3DTR+ mice at the neonatal stage. The model presented here shows promise for investigating the mechanisms behind IHC loss and subsequent regeneration.
Collapse
|
86
|
Canonical Wnt Signaling Pathway on Polarity Formation of Utricle Hair Cells. Neural Plast 2021; 2021:9950533. [PMID: 34122536 PMCID: PMC8166501 DOI: 10.1155/2021/9950533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/26/2021] [Accepted: 05/11/2021] [Indexed: 02/05/2023] Open
Abstract
As part of the inner ear, the vestibular system is responsible for sense of balance, which consists of three semicircular canals, the utricle, and the saccule. Increasing evidence has indicated that the noncanonical Wnt/PCP signaling pathway plays a significant role in the development of the polarity of the inner ear. However, the role of canonical Wnt signaling in the polarity of the vestibule is still not completely clear. In this study, we found that canonical Wnt pathway-related genes are expressed in the early stage of development of the utricle and change dynamically. We conditionally knocked out β-catenin, a canonical Wnt signaling core protein, and found that the cilia orientation of hair cells was disordered with reduced number of hair cells in the utricle. Moreover, regulating the canonical Wnt pathway (Licl and IWP2) in vitro also affected hair cell polarity and indicated that Axin2 may be important in this process. In conclusion, our results not only confirm that the regulation of canonical Wnt signaling affects the number of hair cells in the utricle but also provide evidence for its role in polarity development.
Collapse
|
87
|
Abdul-Aziz D, Hathiramani N, Phung L, Sykopetrites V, Edge ASB. HIC1 Represses Atoh1 Transcription and Hair Cell Differentiation in the Cochlea. Stem Cell Reports 2021; 16:797-809. [PMID: 33770497 PMCID: PMC8072069 DOI: 10.1016/j.stemcr.2021.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 02/26/2021] [Accepted: 02/27/2021] [Indexed: 11/23/2022] Open
Abstract
Across species, expression of the basic helix-loop-helix transcription factor ATOH1 promotes differentiation of cochlear supporting cells to sensory hair cells required for hearing. In mammals, this process is limited to development, whereas nonmammalian vertebrates can also regenerate hair cells after injury. The mechanistic basis for this difference is not fully understood. Hypermethylated in cancer 1 (HIC1) is a transcriptional repressor known to inhibit Atoh1 in the cerebellum. We therefore investigated its potential role in cochlear hair cell differentiation. We find that Hic1 is expressed throughout the postnatal murine cochlear sensory epithelium. In cochlear organoids, Hic1 knockdown induces Atoh1 expression and promotes hair cell differentiation, while Hic1 overexpression hinders differentiation. Wild-type HIC1, but not the DNA-binding mutant C521S, suppresses activity of the Atoh1 autoregulatory enhancer and blocks its responsiveness to β-catenin activation. Our findings reveal the importance of HIC1 repression of Atoh1 in the cochlea, which may be targeted to promote hair cell regeneration.
Collapse
Affiliation(s)
- Dunia Abdul-Aziz
- Department of Otolaryngology, Harvard Medical School, Boston, MA, USA; Eaton Peabody Laboratory, Massachusetts Eye and Ear, Boston, MA, USA
| | | | - Lauren Phung
- Eaton Peabody Laboratory, Massachusetts Eye and Ear, Boston, MA, USA
| | - Vittoria Sykopetrites
- Eaton Peabody Laboratory, Massachusetts Eye and Ear, Boston, MA, USA; Università degli Studi di Milano, Milan, Italy
| | - Albert S B Edge
- Department of Otolaryngology, Harvard Medical School, Boston, MA, USA; Eaton Peabody Laboratory, Massachusetts Eye and Ear, Boston, MA, USA; Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
88
|
Gao J, Fan L, Zhao L, Su Y. The interaction of Notch and Wnt signaling pathways in vertebrate regeneration. CELL REGENERATION (LONDON, ENGLAND) 2021; 10:11. [PMID: 33791915 PMCID: PMC8012441 DOI: 10.1186/s13619-020-00072-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 12/14/2020] [Indexed: 12/21/2022]
Abstract
Regeneration is an evolutionarily conserved process in animal kingdoms, however, the regenerative capacities differ from species and organ/tissues. Mammals possess very limited regenerative potential to replace damaged organs, whereas non-mammalian species usually have impressive abilities to regenerate organs. The regeneration process requires proper spatiotemporal regulation from key signaling pathways. The canonical Notch and Wnt signaling pathways, two fundamental signals guiding animal development, have been demonstrated to play significant roles in the regeneration of vertebrates. In recent years, increasing evidence has implicated the cross-talking between Notch and Wnt signals during organ regeneration. In this review, we summarize the roles of Notch signaling and Wnt signaling during several representative organ regenerative events, emphasizing the functions and molecular bases of their interplay in these processes, shedding light on utilizing these two signaling pathways to enhance regeneration in mammals and design legitimate therapeutic strategies.
Collapse
Affiliation(s)
- Junying Gao
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, Shandong, China.,College of Fisheries, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Lixia Fan
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, Shandong, China.,College of Fisheries, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Long Zhao
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, Shandong, China. .,College of Fisheries, Ocean University of China, Qingdao, 266003, Shandong, China.
| | - Ying Su
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, Shandong, China. .,College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, Shandong, China.
| |
Collapse
|
89
|
Chen Y, Gu Y, Li Y, Li GL, Chai R, Li W, Li H. Generation of mature and functional hair cells by co-expression of Gfi1, Pou4f3, and Atoh1 in the postnatal mouse cochlea. Cell Rep 2021; 35:109016. [PMID: 33882317 DOI: 10.1016/j.celrep.2021.109016] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 12/15/2020] [Accepted: 03/30/2021] [Indexed: 12/12/2022] Open
Abstract
The mammalian cochlea cannot regenerate functional hair cells (HCs) spontaneously. Atoh1 overexpression as well as other strategies are unable to generate functional HCs. Here, we simultaneously upregulated the expression of Gfi1, Pou4f3, and Atoh1 in postnatal cochlear supporting cells (SCs) in vivo, which efficiently converted SCs into HCs. The newly regenerated HCs expressed HC markers Myo7a, Calbindin, Parvalbumin, and Ctbp2 and were innervated by neurites. Importantly, many new HCs expressed the mature and terminal marker Prestin or vesicular glutamate transporter 3 (vGlut3), depending on the subtypes of the source SCs. Finally, our patch-clamp analysis showed that the new HCs in the medial region acquired a large K+ current, fired spikes transiently, and exhibited signature refinement of ribbon synapse functions, in close resemblance to native wild-type inner HCs. We demonstrated that co-upregulating Gfi1, Pou4f3, and Atoh1 enhances the efficiency of HC generation and promotes the functional maturation of new HCs.
Collapse
Affiliation(s)
- Yan Chen
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai 200031, China
| | - Yuyan Gu
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai 200031, China
| | - Yige Li
- MOE Key Laboratory for Developmental Genes and Human Disease, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing 210096, China
| | - Geng-Lin Li
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai 200031, China
| | - Renjie Chai
- MOE Key Laboratory for Developmental Genes and Human Disease, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing 210096, China; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing, China.
| | - Wenyan Li
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai 200031, China.
| | - Huawei Li
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China; Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai 200031, China; The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
90
|
Iyer AA, Groves AK. Transcription Factor Reprogramming in the Inner Ear: Turning on Cell Fate Switches to Regenerate Sensory Hair Cells. Front Cell Neurosci 2021; 15:660748. [PMID: 33854418 PMCID: PMC8039129 DOI: 10.3389/fncel.2021.660748] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/08/2021] [Indexed: 12/15/2022] Open
Abstract
Non-mammalian vertebrates can restore their auditory and vestibular hair cells naturally by triggering the regeneration of adjacent supporting cells. The transcription factor ATOH1 is a key regulator of hair cell development and regeneration in the inner ear. Following the death of hair cells, supporting cells upregulate ATOH1 and give rise to new hair cells. However, in the mature mammalian cochlea, such natural regeneration of hair cells is largely absent. Transcription factor reprogramming has been used in many tissues to convert one cell type into another, with the long-term hope of achieving tissue regeneration. Reprogramming transcription factors work by altering the transcriptomic and epigenetic landscapes in a target cell, resulting in a fate change to the desired cell type. Several studies have shown that ATOH1 is capable of reprogramming cochlear non-sensory tissue into cells resembling hair cells in young animals. However, the reprogramming ability of ATOH1 is lost with age, implying that the potency of individual hair cell-specific transcription factors may be reduced or lost over time by mechanisms that are still not clear. To circumvent this, combinations of key hair cell transcription factors have been used to promote hair cell regeneration in older animals. In this review, we summarize recent findings that have identified and studied these reprogramming factor combinations for hair cell regeneration. Finally, we discuss the important questions that emerge from these findings, particularly the feasibility of therapeutic strategies using reprogramming factors to restore human hearing in the future.
Collapse
Affiliation(s)
- Amrita A. Iyer
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Program in Genetics & Genomics, Houston, TX, United States
| | - Andrew K. Groves
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Program in Genetics & Genomics, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
91
|
Fernández-Hernández I, Marsh EB, Bonaguidi MA. Mechanosensory neuron regeneration in adult Drosophila. Development 2021; 148:dev.187534. [PMID: 33597190 DOI: 10.1242/dev.187534] [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: 12/17/2019] [Accepted: 02/09/2021] [Indexed: 01/01/2023]
Abstract
Auditory and vestibular mechanosensory hair cells do not regenerate following injury or aging in the adult mammalian inner ear, inducing irreversible hearing loss and balance disorders for millions of people. Research on model systems showing replacement of mechanosensory cells can provide mechanistic insights into developing new regenerative therapies. Here, we developed lineage tracing systems to reveal the generation of mechanosensory neurons in the Johnston's organ (JO) of intact adult Drosophila, which are the functional counterparts to hair cells in vertebrates. New JO neurons develop cilia and target central brain circuitry. Unexpectedly, mitotic recombination clones point to JO neuron self-replication as a likely source of neuronal plasticity. This mechanism is further enhanced upon treatment with experimental and ototoxic compounds. Our findings introduce a new platform to expedite research on mechanisms and compounds mediating mechanosensory cell regeneration, with nascent implications for hearing and balance restoration.
Collapse
Affiliation(s)
- Ismael Fernández-Hernández
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Evan B Marsh
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Michael A Bonaguidi
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA .,Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.,Department of Gerontology, Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.,Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033 USA
| |
Collapse
|
92
|
Chen M, Huang J. Retinoic acid induces differentiation of cochlear neural progenitor cells into hair cells. Braz J Otorhinolaryngol 2021; 88:962-967. [PMID: 33707121 PMCID: PMC9615533 DOI: 10.1016/j.bjorl.2021.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 01/05/2021] [Accepted: 01/30/2021] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION Inner ear progenitor cells have the potential for multi-directional differentiation. Retinoic acid is an important requirement for the development of the inner ear. Blocking the Curtyr's retinoic acid signaling pathway can significantly reduce the number of hair cells. Therefore, we believe that retinoic acid may induce the regeneration of inner ear hair cells. OBJECTIVE To investigate whether the cochlear neural progenitor cells maintain the characteristics of stem cells during recovery and subculture, whether retinoic acid can induce cochlear neural progenitor cells into hair cells in vitro, and whether retinoic acid promotes or inhibits the proliferation of cochlear neural progenitor cells during differentiation. METHODS Cochlear neural progenitor cells were cultured and induced in DMEM/F12+RA (10-6M) and then detected the expressions of hair cell markers (Math1 and MyosinVIIa) by immunofluorescence cytochemistry and realtime-polymerase chain reaction, and the proliferation of cochlear neural progenitor cells was detected by Brdu. RESULTS The nestin of cochlear neural progenitor cells was positively expressed. The ratios of Math1-positive cells in the control group and experimental group were 1.5% and 63%, respectively; the ratios of MyosinVIIa-positive cells in the control group and experimental group were 0.96% and 56%, respectively (p<0.05). The ratios of Brdu+-labeled cells in retinoic acid group, group PBS, and group FBS were 20.6%, 29.9%, and 54.3%, respectively; however, the proliferation rate in the experimental group decreased. CONCLUSION Retinoic acid can promote cochlear neural progenitor cells to differentiate into the hair cells.
Collapse
Affiliation(s)
- Minyun Chen
- The Second Affiliated Hospital of Fujian Medical University, Department of Otolaryngology, Fujian, China
| | - Jianmin Huang
- Fujian Medical University Union Hospital, Department of Otolaryngology, Fujian, China.
| |
Collapse
|
93
|
Kubota M, Scheibinger M, Jan TA, Heller S. Greater epithelial ridge cells are the principal organoid-forming progenitors of the mouse cochlea. Cell Rep 2021; 34:108646. [PMID: 33472062 PMCID: PMC7847202 DOI: 10.1016/j.celrep.2020.108646] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 11/01/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023] Open
Abstract
In mammals, hearing loss is irreversible due to the lack of regenerative potential of non-sensory cochlear cells. Neonatal cochlear cells, however, can grow into organoids that harbor sensory epithelial cells, including hair cells and supporting cells. Here, we purify different cochlear cell types from neonatal mice, validate the composition of the different groups with single-cell RNA sequencing (RNA-seq), and assess the various groups' potential to grow into inner ear organoids. We find that the greater epithelial ridge (GER), a transient cell population that disappears during post-natal cochlear maturation, harbors the most potent organoid-forming cells. We identified three distinct GER cell groups that correlate with a specific spatial distribution of marker genes. Organoid formation was synergistically enhanced when the cells were cultured at increasing density. This effect is not due to diffusible signals but requires direct cell-to-cell contact. Our findings improve the development of cell-based assays to study culture-generated inner ear cell types.
Collapse
Affiliation(s)
- Marie Kubota
- Department of Otolaryngology - Head & Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Mirko Scheibinger
- Department of Otolaryngology - Head & Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Taha A Jan
- Department of Otolaryngology - Head & Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Otolaryngology - Head & Neck Surgery, University of California San Francisco, San Francisco, CA 94115, USA
| | - Stefan Heller
- Department of Otolaryngology - Head & Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
94
|
Kothapalli A, Staecker H, Mellott AJ. Supervised machine learning for automated classification of human Wharton's Jelly cells and mechanosensory hair cells. PLoS One 2021; 16:e0245234. [PMID: 33417611 PMCID: PMC7793269 DOI: 10.1371/journal.pone.0245234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/23/2020] [Indexed: 11/23/2022] Open
Abstract
Tissue engineering and gene therapy strategies offer new ways to repair permanent damage to mechanosensory hair cells (MHCs) by differentiating human Wharton's Jelly cells (HWJCs). Conventionally, these strategies require the classification of each cell as differentiated or undifferentiated. Automated classification tools, however, may serve as a novel method to rapidly classify these cells. In this paper, images from previous work, where HWJCs were differentiated into MHC-like cells, were examined. Various cell features were extracted from these images, and those which were pertinent to classification were identified. Different machine learning models were then developed, some using all extracted data and some using only certain features. To evaluate model performance, the area under the curve (AUC) of the receiver operating characteristic curve was primarily used. This paper found that limiting algorithms to certain features consistently improved performance. The top performing model, a voting classifier model consisting of two logistic regressions, a support vector machine, and a random forest classifier, obtained an AUC of 0.9638. Ultimately, this paper illustrates the viability of a novel machine learning pipeline to automate the classification of undifferentiated and differentiated cells. In the future, this research could aid in automated strategies that determine the viability of MHC-like cells after differentiation.
Collapse
Affiliation(s)
- Abihith Kothapalli
- Blue Valley West High School, Overland Park, KS, United States of America
| | - Hinrich Staecker
- Department of Otolaryngology, Head and Neck Surgery, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Adam J. Mellott
- Department of Plastic Surgery, University of Kansas Medical Center, Kansas City, KS, United States of America
| |
Collapse
|
95
|
Zhao T, Zheng T, Yu H, Hu BH, Hu B, Ma P, Yang Y, Yang N, Hu J, Cao T, Chen G, Yan B, Peshoff M, Hatzoglou M, Geng R, Li B, Zheng QY. Autophagy impairment as a key feature for acetaminophen-induced ototoxicity. Cell Death Dis 2021; 12:3. [PMID: 33414397 PMCID: PMC7791066 DOI: 10.1038/s41419-020-03328-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023]
Abstract
Macroautophagy/autophagy is a highly conserved self-digestion pathway that plays an important role in cytoprotection under stress conditions. Autophagy is involved in hepatotoxicity induced by acetaminophen (APAP) in experimental animals and in humans. APAP also causes ototoxicity. However, the role of autophagy in APAP-induced auditory hair cell damage is unclear. In the present study, we investigated autophagy mechanisms during APAP-induced cell death in a mouse auditory cell line (HEI-OC1) and mouse cochlear explant culture. We found that the expression of LC3-II protein and autophagic structures was increased in APAP-treated HEI-OC1 cells; however, the degradation of SQSTM1/p62 protein, the yellow puncta of mRFP-GFP-LC3 fluorescence, and the activity of lysosomal enzymes decreased in APAP-treated HEI-OC1 cells. The degradation of p62 protein and the expression of lysosomal enzymes also decreased in APAP-treated mouse cochlear explants. These data indicate that APAP treatment compromises autophagic degradation and causes lysosomal dysfunction. We suggest that lysosomal dysfunction may be directly responsible for APAP-induced autophagy impairment. Treatment with antioxidant N-acetylcysteine (NAC) partially alleviated APAP-induced autophagy impairment and apoptotic cell death, suggesting the involvement of oxidative stress in APAP-induced autophagy impairment. Inhibition of autophagy by knocking down of Atg5 and Atg7 aggravated APAP-induced ER and oxidative stress and increased apoptotic cell death. This study provides a better understanding of the mechanism responsible for APAP ototoxicity, which is important for future exploration of treatment strategies for the prevention of hearing loss caused by ototoxic medications.
Collapse
Affiliation(s)
- Tong Zhao
- Hearing and Speech Rehabilitation Institute, College of Special Education, Binzhou Medical University, Yantai, China
| | - Tihua Zheng
- Hearing and Speech Rehabilitation Institute, College of Special Education, Binzhou Medical University, Yantai, China
| | - Huining Yu
- Hearing and Speech Rehabilitation Institute, College of Special Education, Binzhou Medical University, Yantai, China
| | - Bo Hua Hu
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, USA
| | - Bing Hu
- Department of Otolaryngology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Peng Ma
- Department of Genetics, School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Ying Yang
- Hearing and Speech Rehabilitation Institute, College of Special Education, Binzhou Medical University, Yantai, China
| | - Naidi Yang
- Key Laboratory of Flexible Electronics & Institute of Advanced Materials, Nanjing Tech University, Nanjing, China
| | - Juan Hu
- Department of Otolaryngology-Head & Neck Surgery, Second Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Tongtao Cao
- Hearing and Speech Rehabilitation Institute, College of Special Education, Binzhou Medical University, Yantai, China
| | - Gang Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Bin Yan
- Hearing and Speech Rehabilitation Institute, College of Special Education, Binzhou Medical University, Yantai, China
| | - Melina Peshoff
- Department of Otolaryngology, Case Western Reserve University, Cleveland, OH, USA
| | - Maria Hatzoglou
- Department of Genetics, Case Western Reserve University, Cleveland, OH, USA
| | - Ruishuang Geng
- Hearing and Speech Rehabilitation Institute, College of Special Education, Binzhou Medical University, Yantai, China.
| | - Bo Li
- Hearing and Speech Rehabilitation Institute, College of Special Education, Binzhou Medical University, Yantai, China.
| | - Qing Yin Zheng
- Department of Otolaryngology, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
96
|
Bai H, Yang S, Xi C, Wang X, Xu J, Weng M, Zhao R, Jiang L, Gao X, Bing J, Zhang M, Zhang X, Han Z, Zeng S. Signaling pathways (Notch, Wnt, Bmp and Fgf) have additive effects on hair cell regeneration in the chick basilar papilla after streptomycin injury in vitro: Additive effects of signaling pathways on hair cell regeneration. Hear Res 2020; 401:108161. [PMID: 33422722 DOI: 10.1016/j.heares.2020.108161] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 11/12/2020] [Accepted: 12/27/2020] [Indexed: 01/07/2023]
Abstract
Hair cells can be regenerated after damage by transdifferentiation in which a supporting cell directly differentiates into a hair cell without mitosis. However, such regeneration is at the cost of exhausting the support cells in the mammalian mature cochlea. Thus, more effective methods should be found to promote mitotic regeneration but partially preserve support cells after damage. To address the issue, we first injured hair cells in the chick basilar papillae (BP) by treatment with streptomycin in vitro. We then compared the mitotic regeneration on the neural side in the middle part of BP after treatment with a pharmacological inhibitor or agonist of the Notch (DAPT), Wnt (LiCl), Bmp (Noggin) or Fgf (SU5402) signaling pathway, with that after treatment with combinations of two or three inhibitors or agonist of these pathways. Our results indicate that treatments with a single inhibitor or agonist of the Notch, Wnt, Bmp or Fgf signaling pathway could significantly increase mitotic regeneration as well as direct transdifferentiation. The results also show that hair cells (Myosin 7a+), support cells (Sox2+) and mitotically regenerated hair cells (Myosin 7a+/Sox2+/BrdU+) increased significantly on the neural side in the middle part of BP after two or three combinations of the inhibition of Notch, Bmp or Fgf signaling pathway or the activation of Wnt signaling pathway, besides the reported coregulatory effects of Notch and Wnt signaling. The study of the effects of systematic combinations of pathway modulators provided more insight into hair cell regeneration from mitosis.
Collapse
Affiliation(s)
- Huanju Bai
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, 100875 China
| | - Siyuan Yang
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, College of Life Sciences, Hainan Normal University, Haikou, 571158 China; Hainan Instistute of Science and Technology, Haikou, 571126 China
| | - Chao Xi
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, 100875 China
| | - Xi Wang
- Department of Otorhinolaryngolgoy, The General Hospital of the PLA Rocket Force, Beijing, 100088 China
| | - Jincao Xu
- Department of Otorhinolaryngolgoy, The General Hospital of the PLA Rocket Force, Beijing, 100088 China
| | - Menglu Weng
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, 100875 China
| | - Ruxia Zhao
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, 100875 China
| | - Lingling Jiang
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, 100875 China
| | - Xue Gao
- Department of Otorhinolaryngolgoy, The General Hospital of the PLA Rocket Force, Beijing, 100088 China
| | - Jie Bing
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, 100875 China
| | - Meiguang Zhang
- Department of Otorhinolaryngolgoy, The General Hospital of the PLA Rocket Force, Beijing, 100088 China
| | - Xinwen Zhang
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, College of Life Sciences, Hainan Normal University, Haikou, 571158 China
| | - Zhongming Han
- Department of Otorhinolaryngolgoy, The General Hospital of the PLA Rocket Force, Beijing, 100088 China; Department of Otorhinolaryngolgoy, He Bei YanDa Hospital, Hebei Medical University, Hebei, China 065201.
| | - Shaoju Zeng
- Beijing Key Laboratory of Gene Resource and Molecular Development, Beijing Normal University, 100875 China.
| |
Collapse
|
97
|
Roccio M. Directed differentiation and direct reprogramming: Applying stem cell technologies to hearing research. Stem Cells 2020; 39:375-388. [PMID: 33378797 DOI: 10.1002/stem.3315] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/24/2020] [Accepted: 12/01/2020] [Indexed: 12/26/2022]
Abstract
Hearing loss is the most widely spread sensory disorder in our society. In the majority of cases, it is caused by the loss or malfunctioning of cells in the cochlea: the mechanosensory hair cells, which act as primary sound receptors, and the connecting auditory neurons of the spiral ganglion, which relay the signal to upper brain centers. In contrast to other vertebrates, where damage to the hearing organ can be repaired through the activity of resident cells, acting as tissue progenitors, in mammals, sensory cell damage or loss is irreversible. The understanding of gene and cellular functions, through analysis of different animal models, has helped to identify causes of disease and possible targets for hearing restoration. Translation of these findings to novel therapeutics is, however, hindered by the lack of cellular assays, based on human sensory cells, to evaluate the conservation of molecular pathways across species and the efficacy of novel therapeutic strategies. In the last decade, stem cell technologies enabled to generate human sensory cell types in vitro, providing novel tools to study human inner ear biology, model disease, and validate therapeutics. This review focuses specifically on two technologies: directed differentiation of pluripotent stem cells and direct reprogramming of somatic cell types to sensory hair cells and neurons. Recent development in the field are discussed as well as how these tools could be implemented to become routinely adopted experimental models for hearing research.
Collapse
Affiliation(s)
- Marta Roccio
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich (USZ), and University of Zurich (UZH), Zurich, Switzerland
| |
Collapse
|
98
|
Matsunaga M, Kita T, Yamamoto R, Yamamoto N, Okano T, Omori K, Sakamoto S, Nakagawa T. Initiation of Supporting Cell Activation for Hair Cell Regeneration in the Avian Auditory Epithelium: An Explant Culture Model. Front Cell Neurosci 2020; 14:583994. [PMID: 33281558 PMCID: PMC7688741 DOI: 10.3389/fncel.2020.583994] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/14/2020] [Indexed: 01/08/2023] Open
Abstract
Sensorineural hearing loss is a common disability often caused by the loss of sensory hair cells in the cochlea. Hair cell (HCs) regeneration has long been the main target for the development of novel therapeutics for sensorineural hearing loss. In the mammalian cochlea, hair cell regeneration is limited, but the auditory epithelia of non-mammalian organisms retain the capacity for hair cell regeneration. In the avian basilar papilla (BP), supporting cells (SCs), which give rise to regenerated hair cells, are usually quiescent. Hair cell loss induces both direct transdifferentiation and mitotic division of supporting cells. Here, we established an explant culture model for hair cell regeneration in chick basilar papillae and validated it for investigating the initial phase of hair cell regeneration. The histological assessment demonstrated hair cell regeneration via direct transdifferentiation of supporting cells. Labeling with 5-ethynyl-2′-deoxyuridine (EdU) revealed the occurrence of mitotic division in the supporting cells at specific locations in the basilar papillae, while no EdU labeling was observed in newly generated hair cells. RNA sequencing indicated alterations in known signaling pathways associated with hair cell regeneration, consistent with previous findings. Also, unbiased analyses of RNA sequencing data revealed novel genes and signaling pathways that may be related to the induction of supporting cell activation in the chick basilar papillae. These results indicate the advantages of our explant culture model of the chick basilar papillae for exploring the molecular mechanisms of hair cell regeneration.
Collapse
Affiliation(s)
- Mami Matsunaga
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoko Kita
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryosuke Yamamoto
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Norio Yamamoto
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takayuki Okano
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koichi Omori
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Takayuki Nakagawa
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
99
|
Developmental and Functional Hair Cell-Like Cells Induced by Atoh1 Overexpression in the Adult Mammalian Cochlea In Vitro. Neural Plast 2020; 2020:8885813. [PMID: 33204251 PMCID: PMC7661126 DOI: 10.1155/2020/8885813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 09/17/2020] [Accepted: 10/21/2020] [Indexed: 12/22/2022] Open
Abstract
Hair cells (HCs) in the mammalian cochleae cannot spontaneously regenerate once damaged, resulting in permanent hearing loss. It has been shown that Atoh1 overexpression induces hair cell-like cells (HCLCs) in the cochlea of newborn rodents, but this is hard to achieve in adult mammals. In this study, we used a three-dimensional cochlear culture system and an adenoviral-mediated delivery vector to overexpress Atoh1 in adult mouse cochleae. HCLCs were successfully induced from 3 days after virus infection (3 DVI) in vitro, and the number increased with time. HCLCs were myosin7a positive and distinguishable from remnant HCs in a culture environment. Meanwhile, patch-clamp results showed that noninactive outward potassium currents (sustained outward potassium currents) could be recorded in HCLCs and that their magnitude increased with time, similar to normal HCs. Furthermore, transient HCN currents were recorded in some HCLCs, indicating that the HCLCs experienced a developmental stage similar to normal HCs. We also compared the electrophysiological features of HCLCs from adult mice with native HCs and found the HCLCs gradually matured, similar to the normal HCs. Meanwhile, HCLCs from adult mice possessed the same bundles as developmental HCs. However, these HCLCs did not express prestin, which is a special marker for outer hair cells (OHCs), even at 13 DVI. These results demonstrate that Atoh1 overexpression induces HCLC formation in the adult mammalian cochlea and that these HCLCs were functional and experienced a developmental process similar to that of normal HCs.
Collapse
|
100
|
White PM. Perspectives on Human Hearing Loss, Cochlear Regeneration, and the Potential for Hearing Restoration Therapies. Brain Sci 2020; 10:E756. [PMID: 33092183 PMCID: PMC7589617 DOI: 10.3390/brainsci10100756] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/13/2020] [Accepted: 10/16/2020] [Indexed: 12/23/2022] Open
Abstract
Most adults who acquire hearing loss find it to be a disability that is poorly corrected by current prosthetics. This gap drives current research in cochlear mechanosensory hair cell regeneration and in hearing restoration. Birds and fish can spontaneously regenerate lost hair cells through a process that has become better defined in the last few years. Findings from these studies have informed new research on hair cell regeneration in the mammalian cochlea. Hair cell regeneration is one part of the greater problem of hearing restoration, as hearing loss can stem from a myriad of causes. This review discusses these issues and recent findings, and places them in the greater social context of need and community.
Collapse
Affiliation(s)
- Patricia M White
- Department of Neuroscience, Ernest J. Del Monte Institute for Neuroscience, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY 14642, USA
| |
Collapse
|