1
|
Qin T, Bronner ME. Multifaceted roles of sonic hedgehog signaling in mammalian inner ear development. Dev Biol 2025; 524:97-104. [PMID: 40349906 DOI: 10.1016/j.ydbio.2025.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
The inner ear is an intricate structure that houses six sensory organs responsible for both hearing and balance. The development of the inner ear begins with the formation of the otic placode, a transient ectodermal thickening that emerges early during embryonic development. The otic placode undergoes a series of morphological changes, from thickening to invagination and then pinching off from the ectoderm to form the otic vesicle, which further differentiates into the specialized structures of the inner ear. These developmental processes require a coordinated interplay between intrinsic transcription factors and extrinsic signaling molecules, which regulate the patterning, proliferation, and differentiation of the inner ear components. In this review, we focus on the role of Sonic hedgehog (Shh) signaling in the development of the mammalian inner ear. We explore how Shh signaling is involved at multiple stages of inner ear development, including the patterning of the otic vesicle and the differentiation of specific cell types within the cochlea. Additionally, we discuss the mechanisms by which Shh signaling integrates with other signaling pathways and transcription factors to ensure the proper development and function of the inner ear. Understanding the molecular basis of these processes provides valuable insights into inner ear development and its disorders.
Collapse
Affiliation(s)
- Tianli Qin
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.
| |
Collapse
|
2
|
Jahanshir E, Llamas J, Kim Y, Biju K, Oak S, Gnedeva K. The Hippo pathway and p27 Kip1 cooperate to suppress mitotic regeneration in the organ of Corti and the retina. Proc Natl Acad Sci U S A 2025; 122:e2411313122. [PMID: 40178894 PMCID: PMC12002246 DOI: 10.1073/pnas.2411313122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 02/19/2025] [Indexed: 04/05/2025] Open
Abstract
The mature mammalian auditory sensory organ, the organ of Corti (OC), lacks the capacity for regenerating hair cells, leading to permanent hearing impairment. In contrast, the vestibular system has a limited capacity for hair cell regeneration, which we have shown to be further enhanced by inhibiting the Hippo pathway. Here, we demonstrate that, despite similar transcriptional responses, only vestibular and not auditory supporting cells proliferate as a result of Yap activation following Hippo inhibition. Mechanistically, we identify p27Kip1, a cell cycle kinase inhibitor encoded by Cdkn1b, as an additional barrier preventing cell cycle reentry specifically in the OC. We show that while in both systems Yap stimulates p27Kip1 degradation through activation of its direct target gene Skp2, this protein-level control is antagonized by an unusually high level of Cdkn1b transcription in the cochlea. Consequently, p27Kip1 activity is maintained in the OC even in the presence of constitutively active Yap5SA, counteracting its mitogenic effects. Supporting this model, inactivation of the Hippo pathway in the Cdkn1b-deficient background is sufficient to induce adult auditory supporting cell proliferation in vivo. Furthermore, we show that the synergistic interaction between Hippo and p27Kip1 is conserved in the retina where inhibition of both pathways potently induces Müller glia proliferation and initiates neuronal regeneration. Our work uncovers the molecular mechanism preventing quiescent adult sensory progenitor cells, supporting cells in the ear and Müller glia in the eye, from reentering the cell cycle after damage-the key step toward sensory receptor regeneration blocked in mammals.
Collapse
Affiliation(s)
- Eva Jahanshir
- University of Southern California Caruso Department of Otolaryngology—Head and Neck Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
| | - Juan Llamas
- University of Southern California Caruso Department of Otolaryngology—Head and Neck Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
| | - Yeeun Kim
- University of Southern California Caruso Department of Otolaryngology—Head and Neck Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
| | - Kevin Biju
- University of Southern California Caruso Department of Otolaryngology—Head and Neck Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
| | - Sanyukta Oak
- University of Southern California Caruso Department of Otolaryngology—Head and Neck Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
| | - Ksenia Gnedeva
- University of Southern California Caruso Department of Otolaryngology—Head and Neck Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
| |
Collapse
|
3
|
Rainey RN, Houman SD, Menendez L, Chang R, Tao L, Bugacov H, McMahon AP, Kalluri R, Oghalai JS, Groves AK, Segil N. Inducible, virus-free direct lineage reprogramming enhances scalable generation of human inner ear hair cell-like cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.20.639352. [PMID: 40060658 PMCID: PMC11888184 DOI: 10.1101/2025.02.20.639352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
Mammalian inner ear sensory hair cells are highly sensitive to environmental stress and do not regenerate, making hearing loss progressive and permanent. The paucity and extreme inaccessibility of these cells hinder the development of regenerative and otoprotective strategies, Direct lineage reprogramming to generate large quantities of hair cell-like cells in vitro offers a promising approach to overcome these experimental bottlenecks. Previously, we identified four transcription factors-Six1, Atoh1, Pou4f3, and Gfi1 (SAPG)-capable of converting mouse embryonic fibroblasts, adult tail tip fibroblasts, and postnatal mouse supporting cells into induced hair cell-like cells through retroviral or lentiviral transduction (Menendez et al., 2020). Here, we developed a virus-free, inducible system using a stable human induced pluripotent stem (iPS) cell line carrying doxycycline-inducible SAPG. Our inducible system significantly increases reprogramming efficiency compared to retroviral methods, achieving a ~19-fold greater conversion to a hair cell fate in half the time. Immunostaining, Western blot, and single-nucleus RNA-seq analyses confirm the expression of hair cell-specific markers and activation of hair cell gene networks in reprogrammed cells. The reprogrammed hair cells closely resemble developing fetal hair cells, as evidenced by comparison with a human fetal inner ear dataset. Electrophysiological analysis reveals that the induced hair cell-like cells exhibit diverse voltage-dependent ion currents, including robust, quick-activating, slowly inactivating currents characteristic of primary hair cells. This virus-free approach improves scalability, reproducibility, and the modeling of hair cell differentiation, offering significant potential for hair cell regenerative strategies and preclinical drug discovery targeting ototoxicity and otoprotection.
Collapse
Affiliation(s)
- Robert N Rainey
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of University of Southern California, Los Angeles, California, United States
| | - Sam D Houman
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of University of Southern California, Los Angeles, California, United States
- Present address: Touro University of California, College of Osteopathic Medicine, Vallejo, California, United States
| | - Louise Menendez
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of University of Southern California, Los Angeles, California, United States
| | - Ryan Chang
- Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California, United States
| | - Litao Tao
- Biomedical Sciences Department, School of Medicine, Creighton University, Omaha, Nebraska, United States
| | - Helena Bugacov
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of University of Southern California, Los Angeles, California, United States
- Present address: Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of University of Southern California, Los Angeles, California, United States
- Present address: Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States
| | - Radha Kalluri
- Zilkha Neurogenetic Institute, Keck School of Medicine of University of Southern California, Los Angeles, California, United States
- USC Caruso Department of Otolaryngology - Head and Neck Surgery, Keck School of Medicine of University of Southern California, Los Angeles, California, United States
| | - John S Oghalai
- USC Caruso Department of Otolaryngology - Head and Neck Surgery, Keck School of Medicine of University of Southern California, Los Angeles, California, United States
| | - Andrew K Groves
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Neil Segil
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of University of Southern California, Los Angeles, California, United States
- Zilkha Neurogenetic Institute, Keck School of Medicine of University of Southern California, Los Angeles, California, United States
| |
Collapse
|
4
|
Song Y, Dou Z, Liu W, Zhang A, Gao X, Shi H, Zhang Z, Gao J, Jin Y. The chromatin remodeler Brg1 is essential for cochlear sensory epithelium differentiation and patterning. Hum Mol Genet 2025:ddaf019. [PMID: 39927735 DOI: 10.1093/hmg/ddaf019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/14/2025] [Accepted: 01/30/2025] [Indexed: 02/11/2025] Open
Abstract
Human genome analyses have revealed that abnormal BAF (BRG1/BRM-associated factor) complex is highly associated with hearing loss. However, the underlying pathogenesis remains largely unknown. Disrupted structure and function of the organ of Corti is the most prevalent cause of sensorineural hearing loss in mammals. Here, we investigated the role of Brg1-based BAF complex during the differentiation and development of the auditory sensory epithelium, a crucial period for the formation of the organ of Corti. Our findings indicate that deletion of Brg1 leads to premature hair cell (HC) differentiation by inactivating Sonic hedgehog (Shh) signaling. Despite the formation of HCs, subsequent differentiation of inner hair cells (IHCs) and outer hair cells (OHCs) was impaired. Additionally, we observed that the mosaic-like arrangement of HCs and supporting cells (SCs) was disrupted resulting in abnormal sensory epithelium patterning. Furthermore, we found the planar cell polarity of the Brg1-deficient cochlea was abnormal. Our study demonstrates the pivotal role of Brg1 in the differentiation and patterning of the organ of Corti.
Collapse
Affiliation(s)
- Yuning Song
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, 27 Shanda Nanlu, Jinan 250100, China
| | - Zhilin Dou
- Key Laboratory for Experimental Teratology of the Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xilu, Jinan 250012, China
| | - Wenwen Liu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, 4 Duanxing Xilu, Jinan 250022, China
| | - Aizhen Zhang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, 4 Duanxing Xilu, Jinan 250022, China
| | - Xiaotong Gao
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, 27 Shanda Nanlu, Jinan 250100, China
| | - Hongbiao Shi
- Key Laboratory for Experimental Teratology of the Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xilu, Jinan 250012, China
| | - Zhixiong Zhang
- Key Laboratory for Experimental Teratology of the Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xilu, Jinan 250012, China
| | - Jiangang Gao
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, 27 Shanda Nanlu, Jinan 250100, China
| | - Yecheng Jin
- Key Laboratory for Experimental Teratology of the Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xilu, Jinan 250012, China
| |
Collapse
|
5
|
Wang S, Chakraborty S, Fu Y, Lee MP, Liu J, Waldhaus J. 3D reconstruction of the mouse cochlea from scRNA-seq data suggests morphogen-based principles in apex-to-base specification. Dev Cell 2024; 59:1538-1552.e6. [PMID: 38593801 PMCID: PMC11187690 DOI: 10.1016/j.devcel.2024.03.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 04/03/2023] [Accepted: 03/13/2024] [Indexed: 04/11/2024]
Abstract
In the mammalian auditory system, frequency discrimination depends on numerous morphological and physiological properties of the organ of Corti, which gradually change along the apex-to-base (tonotopic) axis of the organ. For example, the basilar membrane stiffness changes tonotopically, thus affecting the tuning properties of individual hair cells. At the molecular level, those frequency-specific characteristics are mirrored by gene expression gradients; however, the molecular mechanisms controlling tonotopic gene expression in the mouse cochlea remain elusive. Through analyzing single-cell RNA sequencing (scRNA-seq) data from E12.5 and E14.5 time points, we predicted that morphogens, rather than a cell division-associated mechanism, confer spatial identity in the extending cochlea. Subsequently, we reconstructed the developing cochlea in 3D space from scRNA-seq data to investigate the molecular pathways mediating positional information. The retinoic acid (RA) and hedgehog pathways were found to form opposing apex-to-base gradients, and functional interrogation using mouse cochlear explants suggested that both pathways jointly specify the longitudinal axis.
Collapse
Affiliation(s)
- Shuze Wang
- Department of Otolaryngology-Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Saikat Chakraborty
- Department of Otolaryngology-Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yujuan Fu
- Biomedical Informatics and Medical Education, University of Washington, Seattle, WA 98195, USA
| | - Mary P Lee
- Department of Otolaryngology-Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jie Liu
- Department of Computational Medicine and Bioinformatics, 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
|
6
|
McGovern MM, Hosamani IV, Niu Y, Nguyen KY, Zong C, Groves AK. Expression of Atoh1, Gfi1, and Pou4f3 in the mature cochlea reprograms nonsensory cells into hair cells. Proc Natl Acad Sci U S A 2024; 121:e2304680121. [PMID: 38266052 PMCID: PMC10835112 DOI: 10.1073/pnas.2304680121] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 12/08/2023] [Indexed: 01/26/2024] Open
Abstract
Mechanosensory hair cells of the mature mammalian organ of Corti do not regenerate; consequently, loss of hair cells leads to permanent hearing loss. Although nonmammalian vertebrates can regenerate hair cells from neighboring supporting cells, many humans with severe hearing loss lack both hair cells and supporting cells, with the organ of Corti being replaced by a flat epithelium of nonsensory cells. To determine whether the mature cochlea can produce hair cells in vivo, we reprogrammed nonsensory cells adjacent to the organ of Corti with three hair cell transcription factors: Gfi1, Atoh1, and Pou4f3. We generated numerous hair cell-like cells in nonsensory regions of the cochlea and new hair cells continued to be added over a period of 9 wk. Significantly, cells adjacent to reprogrammed hair cells expressed markers of supporting cells, suggesting that transcription factor reprogramming of nonsensory cochlear cells in adult animals can generate mosaics of sensory cells like those seen in the organ of Corti. Generating such sensory mosaics by reprogramming may represent a potential strategy for hearing restoration in humans.
Collapse
Affiliation(s)
| | - Ishwar V. Hosamani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX77030
| | - Yichi Niu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX77030
| | - Ken Y. Nguyen
- Department of Neuroscience, Baylor College of Medicine, Houston, TX77030
| | - Chenghang Zong
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX77030
| | - Andrew K. Groves
- Department of Neuroscience, Baylor College of Medicine, Houston, TX77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX77030
| |
Collapse
|
7
|
Xia M, Wu M, Li Y, Liu Y, Jia G, Lou Y, Ma J, Gao Q, Xie M, Chen Y, He Y, Li H, Li W. Varying mechanical forces drive sensory epithelium formation. SCIENCE ADVANCES 2023; 9:eadf2664. [PMID: 37922362 PMCID: PMC10624343 DOI: 10.1126/sciadv.adf2664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 10/05/2023] [Indexed: 11/05/2023]
Abstract
The mechanical cues of the external microenvironment have been recognized as essential clues driving cell behavior. Although intracellular signals modulating cell fate during sensory epithelium development is well understood, the driving force of sensory epithelium formation remains elusive. Here, we manufactured a hybrid hydrogel with tunable mechanical properties for the cochlear organoids culture and revealed that the extracellular matrix (ECM) drives sensory epithelium formation through shifting stiffness in a stage-dependent pattern. As the driving force, moderate ECM stiffness activated the expansion of cochlear progenitor cell (CPC)-derived epithelial organoids by modulating the integrin α3 (ITGA3)/F-actin cytoskeleton/YAP signaling. Higher stiffness induced the transition of CPCs into sensory hair cells (HCs) through increasing the intracellular Ca2+ signaling mediated by PIEZO2 and then activating KLF2 to accomplish the cell specification . Our results identify the molecular mechanism of sensory epithelium formation guided by ECM mechanical force and contribute to developing therapeutic approaches for HC regeneration.
Collapse
Affiliation(s)
- Mingyu Xia
- ENT institute and Otorhinolaryngology Department of 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
| | - Mingxuan Wu
- ENT institute and Otorhinolaryngology Department of 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
| | - Yuanrong Li
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yaoqian Liu
- ENT institute and Otorhinolaryngology Department of 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
| | - Gaogan Jia
- ENT institute and Otorhinolaryngology Department of 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
| | - Yiyun Lou
- ENT institute and Otorhinolaryngology Department of 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
| | - Jiaoyao Ma
- ENT institute and Otorhinolaryngology Department of 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
| | - Qing Gao
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
| | - Mingjun Xie
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Plastic and Reconstructive Surgery Center, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Yuewei Chen
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Huawei Li
- ENT institute and Otorhinolaryngology Department of 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
- Shanghai Engineering Research Centre of Cochlear Implant, Shanghai 200031, China
| | - Wenyan Li
- ENT institute and Otorhinolaryngology Department of 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
|
8
|
Moysan L, Fazekas F, Fekete A, Köles L, Zelles T, Berekméri E. Ca 2+ Dynamics of Gap Junction Coupled and Uncoupled Deiters' Cells in the Organ of Corti in Hearing BALB/c Mice. Int J Mol Sci 2023; 24:11095. [PMID: 37446272 DOI: 10.3390/ijms241311095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/19/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
ATP, as a paracrine signalling molecule, induces intracellular Ca2+ elevation via the activation of purinergic receptors on the surface of glia-like cochlear supporting cells. These cells, including the Deiters' cells (DCs), are also coupled by gap junctions that allow the propagation of intercellular Ca2+ waves via diffusion of Ca2+ mobilising second messenger IP3 between neighbouring cells. We have compared the ATP-evoked Ca2+ transients and the effect of two different gap junction (GJ) blockers (octanol and carbenoxolone, CBX) on the Ca2+ transients in DCs located in the apical and middle turns of the hemicochlea preparation of BALB/c mice (P14-19). Octanol had no effect on Ca2+ signalling, while CBX inhibited the ATP response, more prominently in the middle turn. Based on astrocyte models and using our experimental results, we successfully simulated the Ca2+ dynamics in DCs in different cochlear regions. The mathematical model reliably described the Ca2+ transients in the DCs and suggested that the tonotopical differences could originate from differences in purinoceptor and Ca2+ pump expressions and in IP3-Ca2+ release mechanisms. The cochlear turn-dependent effect of CBX might be the result of the differing connexin isoform composition of GJs along the tonotopic axis. The contribution of IP3-mediated Ca2+ signalling inhibition by CBX cannot be excluded.
Collapse
Affiliation(s)
- Louise Moysan
- Department of Zoology, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary
| | - Fruzsina Fazekas
- Department of Zoology, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary
| | - Adam Fekete
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - László Köles
- Department of Oral Biology, Semmelweis University, H-1089 Budapest, Hungary
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| | - Tibor Zelles
- Department of Oral Biology, Semmelweis University, H-1089 Budapest, Hungary
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, H-1083 Budapest, Hungary
| | - Eszter Berekméri
- Department of Zoology, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| |
Collapse
|
9
|
Thompson MJ, Young CA, Munnamalai V, Umulis DM. Early radial positional information in the cochlea is optimized by a precise linear BMP gradient and enhanced by SOX2. Sci Rep 2023; 13:8567. [PMID: 37237002 PMCID: PMC10219982 DOI: 10.1038/s41598-023-34725-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Positional information encoded in signaling molecules is essential for early patterning in the prosensory domain of the developing cochlea. The sensory epithelium, the organ of Corti, contains an exquisite repeating pattern of hair cells and supporting cells. This requires precision in the morphogen signals that set the initial radial compartment boundaries, but this has not been investigated. To measure gradient formation and morphogenetic precision in developing cochlea, we developed a quantitative image analysis procedure measuring SOX2 and pSMAD1/5/9 profiles in mouse embryos at embryonic day (E)12.5, E13.5, and E14.5. Intriguingly, we found that the pSMAD1/5/9 profile forms a linear gradient up to the medial ~ 75% of the PSD from the pSMAD1/5/9 peak in the lateral edge during E12.5 and E13.5. This is a surprising activity readout for a diffusive BMP4 ligand secreted from a tightly constrained lateral region since morphogens typically form exponential or power-law gradient shapes. This is meaningful for gradient interpretation because while linear profiles offer the theoretically highest information content and distributed precision for patterning, a linear morphogen gradient has not yet been observed. Furthermore, this is unique to the cochlear epithelium as the pSMAD1/5/9 gradient is exponential in the surrounding mesenchyme. In addition to the information-optimized linear profile, we found that while pSMAD1/5/9 is stable during this timeframe, an accompanying gradient of SOX2 shifts dynamically. Last, through joint decoding maps of pSMAD1/5/9 and SOX2, we see that there is a high-fidelity mapping between signaling activity and position in the regions that will become Kölliker's organ and the organ of Corti. Mapping is ambiguous in the prosensory domain precursory to the outer sulcus. Altogether, this research provides new insights into the precision of early morphogenetic patterning cues in the radial cochlea prosensory domain.
Collapse
Affiliation(s)
- Matthew J Thompson
- Weldon School of Biomedical Engineering, Purdue University, 206 S Martin Jischke Dr, West Lafayette, IN, 47907, USA
| | - Caryl A Young
- University of Maine, 168 College Ave, Orono, ME, 04469, USA
| | - Vidhya Munnamalai
- University of Maine, 168 College Ave, Orono, ME, 04469, USA.
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA.
| | - David M Umulis
- Weldon School of Biomedical Engineering, Purdue University, 206 S Martin Jischke Dr, West Lafayette, IN, 47907, USA.
| |
Collapse
|
10
|
Li S, He S, Lu Y, Jia S, Liu Z. Epistatic genetic interactions between Insm1 and Ikzf2 during cochlear outer hair cell development. Cell Rep 2023; 42:112504. [PMID: 37171961 DOI: 10.1016/j.celrep.2023.112504] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/17/2023] [Accepted: 04/27/2023] [Indexed: 05/14/2023] Open
Abstract
The cochlea harbors two types of sound receptors, outer hair cells (OHCs) and inner hair cells (IHCs). OHCs transdifferentiate into IHCs in Insm1 mutants, and OHCs in Ikzf2-deficient mice are dysfunctional and maintain partial IHC gene expression. Insm1 potentially acts as a positive but indirect regulator of Ikzf2, considering that Insm1 is expressed earlier than Ikzf2 and primarily functions as a transcriptional repressor. However, direct evidence of this possibility is lacking. Here, we report the following results: first, Insm1 overexpression in IHCs leads to ectopic Ikzf2 expression. Second, Ikzf2 expression is repressed in Insm1-deficient OHCs, and forced expression of Ikzf2 mitigates the OHC abnormality in Insm1 mutants. Last, dual ablation of Insm1 and Ikzf2 generates a similar OHC phenotype as does Insm1 ablation alone. Collectively, our findings reveal the transcriptional cascade from Insm1 to Ikzf2, which should facilitate future investigation of the molecular mechanisms underlying OHC development and regeneration.
Collapse
Affiliation(s)
- Shuting Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shunji He
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ying Lu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shiqi Jia
- The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Zhiyong Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China.
| |
Collapse
|
11
|
Generation of p27icreER transgenic mice: A tool for inducible gene expression in supporting cells in the cochlea. Hear Res 2023; 431:108727. [PMID: 36905855 DOI: 10.1016/j.heares.2023.108727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/21/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023]
Abstract
The loss of cochlear hair cells (HCs) is an important cause of sensorineural hearing loss, and finding ways to regenerate HCs would be the ideal way forward for restoring hearing. In this research field, tamoxifen-inducible Cre recombinase (iCreER) transgenic mice and the Cre-loxp system are widely used to manipulate gene expression in supporting cells (SCs), which lie beneath the sensory HCs and are a natural source for HC regeneration. However, many iCreER transgenic lines are of limited utility because they cannot target all subtypes of SCs or they cannot be used in the adult stage. In this study, a new line of iCreER transgenic mice, the p27-P2A-iCreERT2 knock-in mouse strain, was generated by inserting the P2A-iCreERT2 cassette immediately in front of the stop codon of p27, which kept the endogenous expression and function of p27 intact. Using a reporter mouse line with tdTomato fluorescence, we showed that the p27iCreER transgenic line can target all subtypes of cochlear SCs, including Claudius cells. p27-CreER activity in SCs was observed in both the postnatal and the adult stage, suggesting that this mouse strain can be useful for research work in adult cochlear HC regeneration. We then overexpressed Gfi1, Pou4f3, and Atoh1 in p27+ SCs of P6/7 mice using this strain and successfully induced many new Myo7a/tdTomato double-positive cells, further confirming that the p27-P2A-iCreERT2 mouse strain is a new and reliable tool for cochlear HC regeneration and hearing restoration.
Collapse
|
12
|
IGF-1 Controls Metabolic Homeostasis and Survival in HEI-OC1 Auditory Cells through AKT and mTOR Signaling. Antioxidants (Basel) 2023; 12:antiox12020233. [PMID: 36829792 PMCID: PMC9952701 DOI: 10.3390/antiox12020233] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Insulin-like growth factor 1 (IGF-1) is a trophic factor for the nervous system where it exerts pleiotropic effects, including the regulation of metabolic homeostasis. IGF-1 deficiency induces morphological alterations in the cochlea, apoptosis and hearing loss. While multiple studies have addressed the role of IGF-1 in hearing protection, its potential function in the modulation of otic metabolism remains unclear. Here, we report that "House Ear Institute-organ of Corti 1" (HEI-OC1) auditory cells express IGF-system genes that are regulated during their differentiation. Upon binding to its high-affinity receptor IGF1R, IGF-1 activates AKT and mTOR signaling to stimulate anabolism and, concomitantly, to reduce autophagic catabolism in HEI-OC1 progenitor cells. Notably, IGF-1 stimulation during HEI-OC1 differentiation to mature otic cells sustained both constructive metabolism and autophagic flux, possibly to favor cell remodeling. IGF1R engagement and downstream AKT signaling promoted HEI-OC1 cell survival by maintaining redox balance, even when cells were challenged with the ototoxic agent cisplatin. Our findings establish that IGF-1 not only serves an important function in otic metabolic homeostasis but also activates antioxidant defense mechanisms to promote hair cell survival during the stress response to insults.
Collapse
|
13
|
Sufu- and Spop-mediated regulation of Gli2 is essential for the control of mammalian cochlear hair cell differentiation. Proc Natl Acad Sci U S A 2022; 119:e2206571119. [PMID: 36252002 PMCID: PMC9618052 DOI: 10.1073/pnas.2206571119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Development of mammalian auditory epithelium, the organ of Corti, requires precise control of both cell cycle withdrawal and differentiation. Sensory progenitors (prosensory cells) in the cochlear apex exit the cell cycle first but differentiate last. Sonic hedgehog (Shh) signaling is required for the spatiotemporal regulation of prosensory cell differentiation, but the underlying mechanisms remain unclear. Here, we show that suppressor of fused (Sufu), a negative regulator of Shh signaling, is essential for controlling the timing and progression of hair cell (HC) differentiation. Removal of Sufu leads to abnormal Atoh1 expression and a severe delay of HC differentiation due to elevated Gli2 mRNA expression. Later in development, HC differentiation defects are restored in the Sufu mutant by the action of speckle-type PDZ protein (Spop), which promotes Gli2 protein degradation. Deletion of both Sufu and Spop results in robust Gli2 activation, exacerbating HC differentiation defects. We further demonstrate that Gli2 inhibits HC differentiation through maintaining the progenitor state of Sox2+ prosensory cells. Along the basal-apical axis of the developing cochlea, the Sox2 expression level is higher in the progenitor cells than in differentiating cells and is down-regulated from base to apex as differentiation proceeds. The dynamic spatiotemporal change of Sox2 expression levels is controlled by Shh signaling through Gli2. Together, our results reveal key functions of Gli2 in sustaining the progenitor state, thereby preventing HC differentiation and in turn governing the basal-apical progression of HC differentiation in the cochlea.
Collapse
|
14
|
Liu L, Zou L, Li K, Hou H, Hu Q, Liu S, Li J, Song C, Chen J, Wang S, Wang Y, Li C, Du H, Li JL, Chen F, Xu Z, Sun W, Sun Q, Xiong W. Template-independent genome editing in the Pcdh15 av-3j mouse, a model of human DFNB23 nonsyndromic deafness. Cell Rep 2022; 40:111061. [PMID: 35830793 DOI: 10.1016/j.celrep.2022.111061] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 04/15/2022] [Accepted: 06/14/2022] [Indexed: 11/03/2022] Open
Abstract
Although frameshift mutations lead to 22% of inherited Mendelian disorders in humans, there is no efficient in vivo gene therapy strategy available to date, particularly in nondividing cells. Here, we show that nonhomologous end-joining (NHEJ)-mediated nonrandom editing profiles compensate the frameshift mutation in the Pcdh15 gene and restore the lost mechanotransduction function in postmitotic hair cells of Pcdh15av-3J mice, an animal model of human nonsyndromic deafness DFNB23. Identified by an ex vivo evaluation system in cultured cochlear explants, the selected guide RNA restores reading frame in approximately 50% of indel products and recovers mechanotransduction in more than 70% of targeted hair cells. In vivo treatment shows that half of the animals gain improvements in auditory responses, and balance function is restored in the majority of injected mutant mice. These results demonstrate that NHEJ-mediated reading-frame restoration is a simple and efficient strategy in postmitotic systems.
Collapse
Affiliation(s)
- Lian Liu
- School of Life Sciences, Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research at Tsinghua University, Tsinghua University, Beijing 100084, China
| | - Linzhi Zou
- School of Life Sciences, Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research at Tsinghua University, Tsinghua University, Beijing 100084, China
| | - Kuan Li
- School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Hanqing Hou
- School of Life Sciences, Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research at Tsinghua University, Tsinghua University, Beijing 100084, China
| | - Qun Hu
- School of Life Sciences, Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research at Tsinghua University, Tsinghua University, Beijing 100084, China
| | - Shuang Liu
- School of Life Sciences, Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research at Tsinghua University, Tsinghua University, Beijing 100084, China
| | - Jie Li
- School of Life Sciences, Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research at Tsinghua University, Tsinghua University, Beijing 100084, China
| | - Chenmeng Song
- School of Life Sciences, Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research at Tsinghua University, Tsinghua University, Beijing 100084, China
| | - Jiaofeng Chen
- School of Life Sciences, Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research at Tsinghua University, Tsinghua University, Beijing 100084, China
| | - Shufeng Wang
- School of Life Sciences, Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research at Tsinghua University, Tsinghua University, Beijing 100084, China
| | - Yangzhen Wang
- School of Life Sciences, Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research at Tsinghua University, Tsinghua University, Beijing 100084, China
| | - Changri Li
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Haibo Du
- School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
| | - Jun-Liszt Li
- Chinese Institute for Brain Research, Beijing 102206, China; Academy for Advanced Interdisciplinary Studies (AAIS), Peking University, Beijing 100871, China
| | - Fangyi Chen
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Zhigang Xu
- School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
| | - Wenzhi Sun
- Chinese Institute for Brain Research, Beijing 102206, China; School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Qianwen Sun
- School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Wei Xiong
- School of Life Sciences, Tsinghua University, Beijing 100084, China; IDG/McGovern Institute for Brain Research at Tsinghua University, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
15
|
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
|
16
|
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
|
17
|
Single-cell transcriptomic landscapes of the otic neuronal lineage at multiple early embryonic ages. Cell Rep 2022; 38:110542. [PMID: 35320729 DOI: 10.1016/j.celrep.2022.110542] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/03/2021] [Accepted: 02/25/2022] [Indexed: 11/20/2022] Open
Abstract
Inner ear vestibular and spiral ganglion neurons (VGNs and SGNs) are known to play pivotal roles in balance control and sound detection. However, the molecular mechanisms underlying otic neurogenesis at early embryonic ages have remained unclear. Here, we use single-cell RNA sequencing to reveal the transcriptomes of mouse otic tissues at three embryonic ages, embryonic day 9.5 (E9.5), E11.5, and E13.5, covering proliferating and undifferentiated otic neuroblasts and differentiating VGNs and SGNs. We validate the high quality of our studies by using multiple assays, including genetic fate mapping analysis, and we uncover several genes upregulated in neuroblasts or differentiating VGNs and SGNs, such as Shox2, Myt1, Casz1, and Sall3. Notably, our findings suggest a general cascaded differentiation trajectory during early otic neurogenesis. The comprehensive understanding of early otic neurogenesis provided by our study holds critical implications for both basic and translational research.
Collapse
|
18
|
Li S, Fan T, Li C, Wang Y, Li J, Liu Z. Fate-mapping analysis of cochlear cells expressing Atoh1 mRNA via a new Atoh1 3*HA-P2A-Cre knockin mouse strain. Dev Dyn 2022; 251:1156-1174. [PMID: 35038200 DOI: 10.1002/dvdy.453] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Atoh1 is recognized to be essential for cochlear hair cell (HC) development. However, Atoh1 temporal and spatial expression patterns remain widely debated. Here, we aimed to obtain evidence to resolve the controversies regarding Atoh1 expression by generating a new knockin mouse strain: Atoh13*HA-P2A-Cre . RESULTS Fate-mapping analysis of Atoh13*HA-P2A-Cre/+ ; Rosa26-CAG-LSL-tdTomato (Ai9)/+ mice enabled us to concurrently characterize the temporal expression of Atoh1 protein (through HA-tag immunostaining) and visualize the cells expressing Atoh1 mRNA (as tdTomato+ cells). Our findings show that whereas Atoh1 mRNA expression is rapidly turned on in early cochlear progenitors, Atoh1 protein is only detected in differentiating HCs or progenitors just committed to the HC fate. Cre activity is also stronger in Atoh13*HA-P2A-Cre/+ than in previous mouse models, because almost all cochlear HCs and nearby supporting cells here are tdTomato+. Furthermore, tdTomato, but not HA, is expressed in middle and apical spiral ganglion neurons. CONCLUSION Collectively, our findings indicate that Atoh13*HA-P2A-Cre can serve as a powerful genetic model in the developmental biology field. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Shuting Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Ting Fan
- ENT Institute and Otorhinolaryngology Department, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine, National Health and Family Planning Commission (NHFPC), Shanghai, China
| | - Chao 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
| | - Yunfeng Wang
- ENT Institute and Otorhinolaryngology Department, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine, National Health and Family Planning Commission (NHFPC), Shanghai, China
| | - Jian Li
- Clinical Laboratory Center, Children's Hospital of Fudan University, Shanghai, China
| | - Zhiyong Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| |
Collapse
|
19
|
Luo Z, Zhang J, Qiao L, Lu F, Liu Z. Mapping Genome-wide Binding Sites of Prox1 in Mouse Cochlea Using the CUT&RUN Approach. Neurosci Bull 2021; 37:1703-1707. [PMID: 34351548 DOI: 10.1007/s12264-021-00757-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/25/2021] [Indexed: 11/28/2022] Open
Affiliation(s)
- 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, 200031, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jixiang Zhang
- University of Chinese Academy of Sciences, Beijing, 100049, China.,State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lianyong Qiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Falong Lu
- University of Chinese Academy of Sciences, Beijing, 100049, China. .,State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Zhiyong Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China. .,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 201210, China.
| |
Collapse
|
20
|
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
|
21
|
McLean WJ, Hinton AS, Herby JT, Salt AN, Hartsock JJ, Wilson S, Lucchino DL, Lenarz T, Warnecke A, Prenzler N, Schmitt H, King S, Jackson LE, Rosenbloom J, Atiee G, Bear M, Runge CL, Gifford RH, Rauch SD, Lee DJ, Langer R, Karp JM, Loose C, LeBel C. Improved Speech Intelligibility in Subjects With Stable Sensorineural Hearing Loss Following Intratympanic Dosing of FX-322 in a Phase 1b Study. Otol Neurotol 2021; 42:e849-e857. [PMID: 33617194 PMCID: PMC8279894 DOI: 10.1097/mao.0000000000003120] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
OBJECTIVES There are no approved pharmacologic therapies for chronic sensorineural hearing loss (SNHL). The combination of CHIR99021+valproic acid (CV, FX-322) has been shown to regenerate mammalian cochlear hair cells ex vivo. The objectives were to characterize the cochlear pharmacokinetic profile of CV in guinea pigs, then measure FX-322 in human perilymph samples, and finally assess safety and audiometric effects of FX-322 in humans with chronic SNHL. STUDY DESIGNS Middle ear residence, cochlear distribution, and elimination profiles of FX-322 were assessed in guinea pigs. Human perilymph sampling following intratympanic FX-322 dosing was performed in an open-label study in cochlear implant subjects. Unilateral intratympanic FX-322 was assessed in a Phase 1b prospective, randomized, double-blinded, placebo-controlled clinical trial. SETTING Three private otolaryngology practices in the US. PATIENTS Individuals diagnosed with mild to moderately severe chronic SNHL (≤70 dB standard pure-tone average) in one or both ears that was stable for ≥6 months, medical histories consistent with noise-induced or idiopathic sudden SNHL, and no significant vestibular symptoms. INTERVENTIONS Intratympanic FX-322. MAIN OUTCOME MEASURES Pharmacokinetics of FX-322 in perilymph and safety and audiometric effects. RESULTS After intratympanic delivery in guinea pigs and humans, FX-322 levels in the cochlear extended high-frequency region were observed and projected to be pharmacologically active in humans. A single dose of FX-322 in SNHL subjects was well tolerated with mild, transient treatment-related adverse events (n = 15 FX-322 vs 8 placebo). Of the six patients treated with FX-322 who had baseline word recognition in quiet scores below 90%, four showed clinically meaningful improvements (absolute word recognition improved 18-42%, exceeding the 95% confidence interval determined by previously published criteria). No significant changes in placebo-injected ears were observed. At the group level, FX-322 subjects outperformed placebo group in word recognition in quiet when averaged across all time points, with a mean improvement from baseline of 18.9% (p = 0.029). For words in noise, the treated group showed a mean 1.3 dB signal-to-noise ratio improvement (p = 0.012) relative to their baseline scores while placebo-treated subjects did not (-0.21 dB, p = 0.71). CONCLUSIONS Delivery of FX-322 to the extended high-frequency region of the cochlea is well tolerated and enhances speech recognition performance in multiple subjects with stable chronic hearing loss.
Collapse
Affiliation(s)
- Will J. McLean
- Frequency Therapeutics, Woburn, MA & Farmington, CT
- Department of Surgery, University of Connecticut School of Medicine, Farmington, CT
| | | | | | - Alec N. Salt
- Department of Otolaryngology, Central Institute for the Deaf, Fay and Carl Simons Center for Hearing and Deafness, Washington University School of Medicine, Saint Louis, MO
| | - Jared J. Hartsock
- Department of Otolaryngology, Central Institute for the Deaf, Fay and Carl Simons Center for Hearing and Deafness, Washington University School of Medicine, Saint Louis, MO
| | - Sam Wilson
- Frequency Therapeutics, Woburn, MA & Farmington, CT
| | | | - Thomas Lenarz
- Department of Otolaryngology and Cluster of Excellence of the German Research Foundation “Hearing4all”, Hannover Medical School, Hannover, Germany
| | - Athanasia Warnecke
- Department of Otolaryngology and Cluster of Excellence of the German Research Foundation “Hearing4all”, Hannover Medical School, Hannover, Germany
| | - Nils Prenzler
- Department of Otolaryngology and Cluster of Excellence of the German Research Foundation “Hearing4all”, Hannover Medical School, Hannover, Germany
| | - Heike Schmitt
- Department of Otolaryngology and Cluster of Excellence of the German Research Foundation “Hearing4all”, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | - Christina L. Runge
- Department of Otolaryngology and Communication Sciences, Medical College of Wisconsin, Milwaukee, WI
| | - René H. Gifford
- Department of Hearing and Speech Sciences, Vanderbilt University Medical Center, Nashville, TN
| | - Steven D. Rauch
- Department of Otolaryngology, Harvard Medical School and Massachusetts Eye and Ear, Boston
| | - Daniel J. Lee
- Department of Otolaryngology, Harvard Medical School and Massachusetts Eye and Ear, Boston
| | - Robert Langer
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Jeffrey M. Karp
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School Boston MA
- Harvard-MIT Division of Health Science and Technology
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA
| | | | - Carl LeBel
- Frequency Therapeutics, Woburn, MA & Farmington, CT
| |
Collapse
|
22
|
Enhancer decommissioning imposes an epigenetic barrier to sensory hair cell regeneration. Dev Cell 2021; 56:2471-2485.e5. [PMID: 34331868 DOI: 10.1016/j.devcel.2021.07.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/24/2021] [Accepted: 07/08/2021] [Indexed: 01/02/2023]
Abstract
Adult mammalian tissues such as heart, brain, retina, and the sensory structures of the inner ear do not effectively regenerate, although a latent capacity for regeneration exists at embryonic and perinatal times. We explored the epigenetic basis for this latent regenerative potential in the mouse inner ear and its rapid loss during maturation. In perinatal supporting cells, whose fate is maintained by Notch-mediated lateral inhibition, the hair cell enhancer network is epigenetically primed (H3K4me1) but silenced (active H3K27 de-acetylation and trimethylation). Blocking Notch signaling during the perinatal period of plasticity rapidly eliminates epigenetic silencing and allows supporting cells to transdifferentiate into hair cells. Importantly, H3K4me1 priming of the hair cell enhancers in supporting cells is removed during the first post-natal week, coinciding with the loss of transdifferentiation potential. We hypothesize that enhancer decommissioning during cochlear maturation contributes to the failure of hair cell regeneration in the mature organ of Corti.
Collapse
|
23
|
Yu HV, Tao L, Llamas J, Wang X, Nguyen JD, Trecek T, Segil N. POU4F3 pioneer activity enables ATOH1 to drive diverse mechanoreceptor differentiation through a feed-forward epigenetic mechanism. Proc Natl Acad Sci U S A 2021; 118:e2105137118. [PMID: 34266958 PMCID: PMC8307294 DOI: 10.1073/pnas.2105137118] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
During embryonic development, hierarchical cascades of transcription factors interact with lineage-specific chromatin structures to control the sequential steps in the differentiation of specialized cell types. While examples of transcription factor cascades have been well documented, the mechanisms underlying developmental changes in accessibility of cell type-specific enhancers remain poorly understood. Here, we show that the transcriptional "master regulator" ATOH1-which is necessary for the differentiation of two distinct mechanoreceptor cell types, hair cells in the inner ear and Merkel cells of the epidermis-is unable to access much of its target enhancer network in the progenitor populations of either cell type when it first appears, imposing a block to further differentiation. This block is overcome by a feed-forward mechanism in which ATOH1 first stimulates expression of POU4F3, which subsequently acts as a pioneer factor to provide access to closed ATOH1 enhancers, allowing hair cell and Merkel cell differentiation to proceed. Our analysis also indicates the presence of both shared and divergent ATOH1/POU4F3-dependent enhancer networks in hair cells and Merkel cells. These cells share a deep developmental lineage relationship, deriving from their common epidermal origin, and suggesting that this feed-forward mechanism preceded the evolutionary divergence of these very different mechanoreceptive cell types.
Collapse
Affiliation(s)
- Haoze V Yu
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology at University of Southern California, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033
| | - Litao Tao
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology at University of Southern California, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033
| | - Juan Llamas
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology at University of Southern California, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033
| | - Xizi Wang
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology at University of Southern California, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033
| | - John D Nguyen
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology at University of Southern California, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033
| | - Talon Trecek
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology at University of Southern California, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033
| | - Neil Segil
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology at University of Southern California, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033;
- Caruso Department of Otolaryngology - Head and Neck Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033
| |
Collapse
|
24
|
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
|
25
|
Elliott KL, Pavlinkova G, Chizhikov VV, Yamoah EN, Fritzsch B. Neurog1, Neurod1, and Atoh1 are essential for spiral ganglia, cochlear nuclei, and cochlear hair cell development. Fac Rev 2021; 10:47. [PMID: 34131657 PMCID: PMC8170689 DOI: 10.12703/r/10-47] [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] [Indexed: 01/11/2023] Open
Abstract
We review the molecular basis of three related basic helix–loop–helix (bHLH) genes (Neurog1, Neurod1, and Atoh1) and upstream regulators Eya1/Six1, Sox2, Pax2, Gata3, Fgfr2b, Foxg1, and Lmx1a/b during the development of spiral ganglia, cochlear nuclei, and cochlear hair cells. Neuronal development requires early expression of Neurog1, followed by its downstream target Neurod1, which downregulates Atoh1 expression. In contrast, hair cells and cochlear nuclei critically depend on Atoh1 and require Neurod1 and Neurog1 expression for various aspects of development. Several experiments show a partial uncoupling of Atoh1/Neurod1 (spiral ganglia and cochlea) and Atoh1/Neurog1/Neurod1 (cochlear nuclei). In this review, we integrate the cellular and molecular mechanisms that regulate the development of auditory system and provide novel insights into the restoration of hearing loss, beyond the limited generation of lost sensory neurons and hair cells.
Collapse
Affiliation(s)
- Karen L Elliott
- Department of Biology, University of Iowa, Iowa City, IA, USA
| | - Gabriela Pavlinkova
- Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czechia
| | - Victor V Chizhikov
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Ebenezer N Yamoah
- Department of Physiology and Cell Biology, University of Nevada, Reno, NV, USA
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
26
|
Elliott KL, Pavlínková G, Chizhikov VV, Yamoah EN, Fritzsch B. Development in the Mammalian Auditory System Depends on Transcription Factors. Int J Mol Sci 2021; 22:ijms22084189. [PMID: 33919542 PMCID: PMC8074135 DOI: 10.3390/ijms22084189] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/16/2022] Open
Abstract
We review the molecular basis of several transcription factors (Eya1, Sox2), including the three related genes coding basic helix–loop–helix (bHLH; see abbreviations) proteins (Neurog1, Neurod1, Atoh1) during the development of spiral ganglia, cochlear nuclei, and cochlear hair cells. Neuronal development requires Neurog1, followed by its downstream target Neurod1, to cross-regulate Atoh1 expression. In contrast, hair cells and cochlear nuclei critically depend on Atoh1 and require Neurod1 expression for interactions with Atoh1. Upregulation of Atoh1 following Neurod1 loss changes some vestibular neurons’ fate into “hair cells”, highlighting the significant interplay between the bHLH genes. Further work showed that replacing Atoh1 by Neurog1 rescues some hair cells from complete absence observed in Atoh1 null mutants, suggesting that bHLH genes can partially replace one another. The inhibition of Atoh1 by Neurod1 is essential for proper neuronal cell fate, and in the absence of Neurod1, Atoh1 is upregulated, resulting in the formation of “intraganglionic” HCs. Additional genes, such as Eya1/Six1, Sox2, Pax2, Gata3, Fgfr2b, Foxg1, and Lmx1a/b, play a role in the auditory system. Finally, both Lmx1a and Lmx1b genes are essential for the cochlear organ of Corti, spiral ganglion neuron, and cochlear nuclei formation. We integrate the mammalian auditory system development to provide comprehensive insights beyond the limited perception driven by singular investigations of cochlear neurons, cochlear hair cells, and cochlear nuclei. A detailed analysis of gene expression is needed to understand better how upstream regulators facilitate gene interactions and mammalian auditory system development.
Collapse
Affiliation(s)
- Karen L. Elliott
- Department of Biology, University of Iowa, Iowa City, IA 52242, USA;
| | - Gabriela Pavlínková
- Institute of Biotechnology of the Czech Academy of Sciences, 25250 Vestec, Czechia;
| | - Victor V. Chizhikov
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Ebenezer N. Yamoah
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV 89557, USA;
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, IA 52242, USA;
- Correspondence:
| |
Collapse
|
27
|
Wang S, Lee MP, Jones S, Liu J, Waldhaus J. Mapping the regulatory landscape of auditory hair cells from single-cell multi-omics data. Genome Res 2021; 31:1885-1899. [PMID: 33837132 DOI: 10.1101/gr.271080.120] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 03/23/2021] [Indexed: 11/25/2022]
Abstract
Auditory hair cells transduce sound to the brain and in mammals these cells reside together with supporting cells in the sensory epithelium of the cochlea, called the organ of Corti. To establish the organ's delicate function during development and differentiation, spatiotemporal gene expression is strictly controlled by chromatin accessibility and cell type-specific transcription factors, jointly representing the regulatory landscape. Bulk-sequencing technology and cellular heterogeneity obscured investigations on the interplay between transcription factors and chromatin accessibility in inner ear development. To study the formation of the regulatory landscape in hair cells, we collected single-cell chromatin accessibility profiles accompanied by single-cell RNA data from genetically labeled murine hair cells and supporting cells after birth. Using an integrative approach, we predicted cell type-specific activating and repressing functions of developmental transcription factors. Furthermore, by integrating gene expression and chromatin accessibility datasets, we reconstructed gene regulatory networks. Then, using a comparative approach, 20 hair cell-specific activators and repressors, including putative downstream target genes, were identified. Clustering of target genes resolved groups of related transcription factors and was utilized to infer their developmental functions. Finally, the heterogeneity in the single-cell data allowed us to spatially reconstruct transcriptional as well as chromatin accessibility trajectories, indicating that gradual changes in the chromatin accessibility landscape were lagging behind the transcriptional identity of hair cells along the organ's longitudinal axis. Overall, this study provides a strategy to spatially reconstruct the formation of a lineage specific regulatory landscape using a single-cell multi-omics approach.
Collapse
Affiliation(s)
- Shuze Wang
- University of Michigan, Kresge Hearing Research Institute
| | - Mary P Lee
- University of Michigan, Kresge Hearing Research Institute
| | - Scott Jones
- University of Michigan, Kresge Hearing Research Institute
| | | | - Joerg Waldhaus
- University of Michigan, Kresge Hearing Research Institute;
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
Su Y, Yang LM, Ornitz DM. FGF20-FGFR1 signaling through MAPK and PI3K controls sensory progenitor differentiation in the organ of Corti. Dev Dyn 2021; 250:134-144. [PMID: 32735383 PMCID: PMC8415122 DOI: 10.1002/dvdy.231] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Fibroblast Growth Factor 20 (FGF20)-FGF receptor 1 (FGFR1) signaling is essential for cochlear hair cell (HC) and supporting cell (SC) differentiation. In other organ systems, FGFR1 signals through several intracellular pathways including MAPK (ERK), PI3K, phospholipase C ɣ (PLCɣ), and p38. Previous studies implicated MAPK and PI3K pathways in HC and SC development. We hypothesized that one or both would be important downstream mediators of FGF20-FGFR1 signaling for HC differentiation. RESULTS By inhibiting pathways downstream of FGFR1 in cochlea explant cultures, we established that both MAPK and PI3K pathways are required for HC differentiation while PLCɣ and p38 pathways are not. Examining the canonical PI3K pathway, we found that while AKT is necessary for HC differentiation, it is not sufficient to rescue the Fgf20-/- phenotype. To determine whether PI3K functions downstream of FGF20, we inhibited Phosphatase and Tensin Homolog (PTEN) in Fgf20-/- explants. Overactivation of PI3K resulted in a partial rescue of the Fgf20-/- phenotype, demonstrating a requirement for PI3K downstream of FGF20. Consistent with a requirement for the MAPK pathway for FGF20-regulated HC differentiation, we show that treating Fgf20-/- explants with FGF9 increased levels of dpERK. CONCLUSIONS Together, these data provide evidence that both MAPK and PI3K are important downstream mediators of FGF20-FGFR1 signaling during HC and SC differentiation.
Collapse
Affiliation(s)
- Yutao Su
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Lu M Yang
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
31
|
Webber JL, Clancy JC, Zhou Y, Yraola N, Homma K, García-Añoveros J. Axodendritic versus axosomatic cochlear efferent termination is determined by afferent type in a hierarchical logic of circuit formation. SCIENCE ADVANCES 2021; 7:7/4/eabd8637. [PMID: 33523928 PMCID: PMC7817091 DOI: 10.1126/sciadv.abd8637] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/03/2020] [Indexed: 05/09/2023]
Abstract
Hearing involves a stereotyped neural network communicating cochlea and brain. How this sensorineural circuit assembles is largely unknown. The cochlea houses two types of mechanosensory hair cells differing in function (sound transmission versus amplification) and location (inner versus outer compartments). Inner (IHCs) and outer hair cells (OHCs) are each innervated by a distinct pair of afferent and efferent neurons: IHCs are contacted by type I afferents receiving axodendritic efferent contacts; OHCs are contacted by type II afferents and axosomatically terminating efferents. Using an Insm1 mouse mutant with IHCs in the position of OHCs, we discover a hierarchical sequence of instructions in which first IHCs attract, and OHCs repel, type I afferents; second, type II afferents innervate hair cells not contacted by type I afferents; and last, afferent fiber type determines if and how efferents innervate, whether axodendritically on the afferent, axosomatically on the hair cell, or not at all.
Collapse
Affiliation(s)
- Jemma L Webber
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - John C Clancy
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Yingjie Zhou
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Natalia Yraola
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Kazuaki Homma
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- The Hugh Knowles Center for Clinical and Basic Science in Hearing and its Disorders, Northwestern University, Chicago, IL 60611, USA
| | - Jaime García-Añoveros
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
- The Hugh Knowles Center for Clinical and Basic Science in Hearing and its Disorders, Northwestern University, Chicago, IL 60611, USA
- Departments of Neurology and Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
32
|
Moon KH, Ma JH, Min H, Koo H, Kim H, Ko HW, Bok J. Dysregulation of sonic hedgehog signaling causes hearing loss in ciliopathy mouse models. eLife 2020; 9:56551. [PMID: 33382037 PMCID: PMC7806262 DOI: 10.7554/elife.56551] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 12/31/2020] [Indexed: 12/11/2022] Open
Abstract
Defective primary cilia cause a range of diseases known as ciliopathies, including hearing loss. The etiology of hearing loss in ciliopathies, however, remains unclear. We analyzed cochleae from three ciliopathy mouse models exhibiting different ciliogenesis defects: Intraflagellar transport 88 (Ift88), Tbc1d32 (a.k.a. bromi), and Cilk1 (a.k.a. Ick) mutants. These mutants showed multiple developmental defects including shortened cochlear duct and abnormal apical patterning of the organ of Corti. Although ciliogenic defects in cochlear hair cells such as misalignment of the kinocilium are often associated with the planar cell polarity pathway, our results showed that inner ear defects in these mutants are primarily due to loss of sonic hedgehog signaling. Furthermore, an inner ear-specific deletion of Cilk1 elicits low-frequency hearing loss attributable to cellular changes in apical cochlear identity that is dedicated to low-frequency sound detection. This type of hearing loss may account for hearing deficits in some patients with ciliopathies.
Collapse
Affiliation(s)
- Kyeong-Hye Moon
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea.,BK21 PLUS project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ji-Hyun Ma
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyehyun Min
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Heiyeun Koo
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea.,BK21 PLUS project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - HongKyung Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyuk Wan Ko
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jinwoong Bok
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea.,BK21 PLUS project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea.,Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
33
|
Mittal R, Bencie N, Liu G, Eshraghi N, Nisenbaum E, Blanton SH, Yan D, Mittal J, Dinh CT, Young JI, Gong F, Liu XZ. Recent advancements in understanding the role of epigenetics in the auditory system. Gene 2020; 761:144996. [PMID: 32738421 PMCID: PMC8168289 DOI: 10.1016/j.gene.2020.144996] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/21/2020] [Indexed: 11/19/2022]
Abstract
Sensorineural deafness in mammals is most commonly caused by damage to inner ear sensory epithelia, or hair cells, and can be attributed to genetic and environmental causes. After undergoing trauma, many non-mammalian organisms, including reptiles, birds, and zebrafish, are capable of regenerating damaged hair cells. Mammals, however, are not capable of regenerating damaged inner ear sensory epithelia, so that hair cell damage is permanent and can lead to hearing loss. The field of epigenetics, which is the study of various phenotypic changes caused by modification of genetic expression rather than alteration of DNA sequence, has seen numerous developments in uncovering biological mechanisms of gene expression and creating various medical treatments. However, there is a lack of information on the precise contribution of epigenetic modifications in the auditory system, specifically regarding their correlation with development of inner ear (cochlea) and consequent hearing impairment. Current studies have suggested that epigenetic modifications influence differentiation, development, and protection of auditory hair cells in cochlea, and can lead to hair cell degeneration. The objective of this article is to review the existing literature and discuss the advancements made in understanding epigenetic modifications of inner ear sensory epithelial cells. The analysis of the emerging epigenetic mechanisms related to inner ear sensory epithelial cells development, differentiation, protection, and regeneration will pave the way to develop novel therapeutic strategies for hearing loss.
Collapse
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Nicole Bencie
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - George Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Nicolas Eshraghi
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Eric Nisenbaum
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Susan H Blanton
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA; Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Denise Yan
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jeenu Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Christine T Dinh
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Juan I Young
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Feng Gong
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Xue Zhong Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA; Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
34
|
Recent advancements in understanding the role of epigenetics in the auditory system. Gene 2020. [DOI: 10.1016/j.gene.2020.144996
expr 848609818 + 898508594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
|
35
|
Cohen R, Amir-Zilberstein L, Hersch M, Woland S, Loza O, Taiber S, Matsuzaki F, Bergmann S, Avraham KB, Sprinzak D. Mechanical forces drive ordered patterning of hair cells in the mammalian inner ear. Nat Commun 2020; 11:5137. [PMID: 33046691 PMCID: PMC7550578 DOI: 10.1038/s41467-020-18894-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 09/15/2020] [Indexed: 01/03/2023] Open
Abstract
Periodic organization of cells is required for the function of many organs and tissues. The development of such periodic patterns is typically associated with mechanisms based on intercellular signaling such as lateral inhibition and Turing patterning. Here we show that the transition from disordered to ordered checkerboard-like pattern of hair cells and supporting cells in the mammalian hearing organ, the organ of Corti, is likely based on mechanical forces rather than signaling events. Using time-lapse imaging of mouse cochlear explants, we show that hair cells rearrange gradually into a checkerboard-like pattern through a tissue-wide shear motion that coordinates intercalation and delamination events. Using mechanical models of the tissue, we show that global shear and local repulsion forces on hair cells are sufficient to drive the transition from disordered to ordered cellular pattern. Our findings suggest that mechanical forces drive ordered hair cell patterning in a process strikingly analogous to the process of shear-induced crystallization in polymer and granular physics.
Collapse
Affiliation(s)
- Roie Cohen
- George S. Wise Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, 6997801, Tel Aviv, Israel.,The Center for Physics and Chemistry of Living Systems, Tel Aviv University, 6997801, Tel Aviv, Israel.,Faculty of Exact Sciences, Raymond and Beverly Sackler School of Physics and Astronomy, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Liat Amir-Zilberstein
- George S. Wise Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Micha Hersch
- Department of Computational Biology, University of Lausanne, 1015, Lausanne, Switzerland.,Swiss Institute of Bioinformatics, 1015, Lausanne, Switzerland
| | - Shiran Woland
- George S. Wise Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Olga Loza
- George S. Wise Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Shahar Taiber
- George S. Wise Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, 6997801, Tel Aviv, Israel.,Sackler Faculty of Medicine and Sagol School of Neuroscience, Department of Human Molecular Genetics and Biochemistry, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Fumio Matsuzaki
- Laboratory of Cell Asymmetry, RIKEN Center for Biosystems Dynamics Research, Kobe, 650-0047, Japan
| | - Sven Bergmann
- Department of Computational Biology, University of Lausanne, 1015, Lausanne, Switzerland.,Swiss Institute of Bioinformatics, 1015, Lausanne, Switzerland.,Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Karen B Avraham
- Sackler Faculty of Medicine and Sagol School of Neuroscience, Department of Human Molecular Genetics and Biochemistry, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - David Sprinzak
- George S. Wise Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, 6997801, Tel Aviv, Israel. .,The Center for Physics and Chemistry of Living Systems, Tel Aviv University, 6997801, Tel Aviv, Israel.
| |
Collapse
|
36
|
Menendez L, Trecek T, Gopalakrishnan S, Tao L, Markowitz AL, Yu HV, Wang X, Llamas J, Huang C, Lee J, Kalluri R, Ichida J, Segil N. Generation of inner ear hair cells by direct lineage conversion of primary somatic cells. eLife 2020; 9:e55249. [PMID: 32602462 PMCID: PMC7326493 DOI: 10.7554/elife.55249] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023] Open
Abstract
The mechanoreceptive sensory hair cells in the inner ear are selectively vulnerable to numerous genetic and environmental insults. In mammals, hair cells lack regenerative capacity, and their death leads to permanent hearing loss and vestibular dysfunction. Their paucity and inaccessibility has limited the search for otoprotective and regenerative strategies. Growing hair cells in vitro would provide a route to overcome this experimental bottleneck. We report a combination of four transcription factors (Six1, Atoh1, Pou4f3, and Gfi1) that can convert mouse embryonic fibroblasts, adult tail-tip fibroblasts and postnatal supporting cells into induced hair cell-like cells (iHCs). iHCs exhibit hair cell-like morphology, transcriptomic and epigenetic profiles, electrophysiological properties, mechanosensory channel expression, and vulnerability to ototoxin in a high-content phenotypic screening system. Thus, direct reprogramming provides a platform to identify causes and treatments for hair cell loss, and may help identify future gene therapy approaches for restoring hearing.
Collapse
Affiliation(s)
- Louise Menendez
- Department of Stem Cell and Regenerative Medicine, University of Southern CaliforniaLos AngelesUnited States
- Eli and Edythe Broad Center, University of Southern CaliforniaLos AngelesUnited States
- Zilkha Neurogenetic Institute, University of Southern CaliforniaLos AngelesUnited States
| | - Talon Trecek
- Department of Stem Cell and Regenerative Medicine, University of Southern CaliforniaLos AngelesUnited States
- Eli and Edythe Broad Center, University of Southern CaliforniaLos AngelesUnited States
| | - Suhasni Gopalakrishnan
- Department of Stem Cell and Regenerative Medicine, University of Southern CaliforniaLos AngelesUnited States
- Eli and Edythe Broad Center, University of Southern CaliforniaLos AngelesUnited States
- Zilkha Neurogenetic Institute, University of Southern CaliforniaLos AngelesUnited States
| | - Litao Tao
- Department of Stem Cell and Regenerative Medicine, University of Southern CaliforniaLos AngelesUnited States
- Eli and Edythe Broad Center, University of Southern CaliforniaLos AngelesUnited States
| | - Alexander L Markowitz
- Zilkha Neurogenetic Institute, University of Southern CaliforniaLos AngelesUnited States
- USC Caruso Department of Otolaryngology – Head and Neck Surgery, University of Southern CaliforniaLos AngelesUnited States
| | - Haoze V Yu
- Department of Stem Cell and Regenerative Medicine, University of Southern CaliforniaLos AngelesUnited States
- Eli and Edythe Broad Center, University of Southern CaliforniaLos AngelesUnited States
| | - Xizi Wang
- Department of Stem Cell and Regenerative Medicine, University of Southern CaliforniaLos AngelesUnited States
- Eli and Edythe Broad Center, University of Southern CaliforniaLos AngelesUnited States
| | - Juan Llamas
- Department of Stem Cell and Regenerative Medicine, University of Southern CaliforniaLos AngelesUnited States
- Eli and Edythe Broad Center, University of Southern CaliforniaLos AngelesUnited States
| | | | - James Lee
- DRVision TechnologiesBellevueUnited States
| | - Radha Kalluri
- Zilkha Neurogenetic Institute, University of Southern CaliforniaLos AngelesUnited States
- USC Caruso Department of Otolaryngology – Head and Neck Surgery, University of Southern CaliforniaLos AngelesUnited States
| | - Justin Ichida
- Department of Stem Cell and Regenerative Medicine, University of Southern CaliforniaLos AngelesUnited States
- Eli and Edythe Broad Center, University of Southern CaliforniaLos AngelesUnited States
- Zilkha Neurogenetic Institute, University of Southern CaliforniaLos AngelesUnited States
| | - Neil Segil
- Department of Stem Cell and Regenerative Medicine, University of Southern CaliforniaLos AngelesUnited States
- Eli and Edythe Broad Center, University of Southern CaliforniaLos AngelesUnited States
- USC Caruso Department of Otolaryngology – Head and Neck Surgery, University of Southern CaliforniaLos AngelesUnited States
| |
Collapse
|
37
|
Abstract
The cochlea, a coiled structure located in the ventral region of the inner ear, acts as the primary structure for the perception of sound. Along the length of the cochlear spiral is the organ of Corti, a highly derived and rigorously patterned sensory epithelium that acts to convert auditory stimuli into neural impulses. The development of the organ of Corti requires a series of inductive events that specify unique cellular characteristics and axial identities along its three major axes. Here, we review recent studies of the cellular and molecular processes regulating several aspects of cochlear development, such as axial patterning, cochlear outgrowth and cellular differentiation. We highlight how the precise coordination of multiple signaling pathways is required for the successful formation of a complete organ of Corti.
Collapse
Affiliation(s)
- Elizabeth Carroll Driver
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthew W Kelley
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
38
|
Yang LM, Stout L, Rauchman M, Ornitz DM. Analysis of FGF20-regulated genes in organ of Corti progenitors by translating ribosome affinity purification. Dev Dyn 2020; 249:1217-1242. [PMID: 32492250 DOI: 10.1002/dvdy.211] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Understanding the mechanisms that regulate hair cell (HC) differentiation in the organ of Corti (OC) is essential to designing genetic therapies for hearing loss due to HC loss or damage. We have previously identified Fibroblast Growth Factor 20 (FGF20) as having a key role in HC and supporting cell differentiation in the mouse OC. To investigate the genetic landscape regulated by FGF20 signaling in OC progenitors, we employ Translating Ribosome Affinity Purification combined with Next Generation RNA Sequencing (TRAPseq) in the Fgf20 lineage. RESULTS We show that TRAPseq targeting OC progenitors effectively enriched for RNA from this rare cell population. TRAPseq identified differentially expressed genes (DEGs) downstream of FGF20, including Etv4, Etv5, Etv1, Dusp6, Hey1, Hey2, Heyl, Tectb, Fat3, Cpxm2, Sall1, Sall3, and cell cycle regulators such as Cdc20. Analysis of Cdc20 conditional-null mice identified decreased cochlea length, while analysis of Sall1-null and Sall1-ΔZn2-10 mice, which harbor a mutation that causes Townes-Brocks syndrome, identified a decrease in outer hair cell number. CONCLUSIONS We present two datasets: genes with enriched expression in OC progenitors, and DEGs downstream of FGF20 in the embryonic day 14.5 cochlea. We validate select DEGs via in situ hybridization and in vivo functional studies in mice.
Collapse
Affiliation(s)
- Lu M Yang
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Lisa Stout
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael Rauchman
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
39
|
Organ of Corti size is governed by Yap/Tead-mediated progenitor self-renewal. Proc Natl Acad Sci U S A 2020; 117:13552-13561. [PMID: 32482884 DOI: 10.1073/pnas.2000175117] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Precise control of organ growth and patterning is executed through a balanced regulation of progenitor self-renewal and differentiation. In the auditory sensory epithelium-the organ of Corti-progenitor cells exit the cell cycle in a coordinated wave between E12.5 and E14.5 before the initiation of sensory receptor cell differentiation, making it a unique system for studying the molecular mechanisms controlling the switch between proliferation and differentiation. Here we identify the Yap/Tead complex as a key regulator of the self-renewal gene network in organ of Corti progenitor cells. We show that Tead transcription factors bind directly to the putative regulatory elements of many stemness- and cell cycle-related genes. We also show that the Tead coactivator protein, Yap, is degraded specifically in the Sox2-positive domain of the cochlear duct, resulting in down-regulation of Tead gene targets. Further, conditional loss of the Yap gene in the inner ear results in the formation of significantly smaller auditory and vestibular sensory epithelia, while conditional overexpression of a constitutively active version of Yap, Yap5SA, is sufficient to prevent cell cycle exit and to prolong sensory tissue growth. We also show that viral gene delivery of Yap5SA in the postnatal inner ear sensory epithelia in vivo drives cell cycle reentry after hair cell loss. Taken together, these data highlight the key role of the Yap/Tead transcription factor complex in maintaining inner ear progenitors during development, and suggest new strategies to induce sensory cell regeneration.
Collapse
|
40
|
Wang L, Kempton JB, Jiang H, Jodelka FM, Brigande AM, Dumont RA, Rigo F, Lentz JJ, Hastings ML, Brigande JV. Fetal antisense oligonucleotide therapy for congenital deafness and vestibular dysfunction. Nucleic Acids Res 2020; 48:5065-5080. [PMID: 32249312 PMCID: PMC7229850 DOI: 10.1093/nar/gkaa194] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/13/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
Disabling hearing loss impacts ∼466 million individuals worldwide with 34 million children affected. Gene and pharmacotherapeutic strategies to rescue auditory function in mouse models of human deafness are most effective when administered before hearing onset, after which therapeutic efficacy is significantly diminished or lost. We hypothesize that preemptive correction of a mutation in the fetal inner ear prior to maturation of the sensory epithelium will optimally restore sensory function. We previously demonstrated that transuterine microinjection of a splice-switching antisense oligonucleotide (ASO) into the amniotic cavity immediately surrounding the embryo on embryonic day 13-13.5 (E13-13.5) corrected pre-mRNA splicing in the juvenile Usher syndrome type 1c (Ush1c) mouse mutant. Here, we show that this strategy only marginally rescues hearing and partially rescues vestibular function. To improve therapeutic outcomes, we microinjected ASO directly into the E12.5 inner ear. A single intra-otic dose of ASO corrects harmonin RNA splicing, restores harmonin protein expression in sensory hair cell bundles, prevents hair cell loss, improves hearing sensitivity, and ameliorates vestibular dysfunction. Improvements in auditory and vestibular function were sustained well into adulthood. Our results demonstrate that an ASO pharmacotherapeutic administered to a developing organ system in utero preemptively corrects pre-mRNA splicing to abrogate the disease phenotype.
Collapse
Affiliation(s)
- Lingyan Wang
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - J Beth Kempton
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - Han Jiang
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - Francine M Jodelka
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Alev M Brigande
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - Rachel A Dumont
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, CA 92010 USA
| | - Jennifer J Lentz
- Department of Otorhinolaryngology, Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Michelle L Hastings
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - John V Brigande
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
41
|
Spatiotemporally controlled overexpression of cyclin D1 triggers generation of supernumerary cells in the postnatal mouse inner ear. Hear Res 2020; 390:107951. [PMID: 32244147 DOI: 10.1016/j.heares.2020.107951] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 02/04/2020] [Accepted: 03/10/2020] [Indexed: 02/06/2023]
Abstract
The retinoblastoma family of pocket proteins (pRBs), composed of Rb1, p107, and p130 are negative regulators of cell-cycle progression. The deletion of any individual pRB in the auditory system triggers hair cells' (HCs) and supporting cells' (SCs) proliferation to different extents. Nevertheless, accessing their combined role in the inner ear through conditional or complete knockout methods is limited by the early mortality of the triple knockout. In quiescent cells, hyperphosphorylation and inactivation of the pRBs are maintained through the activity of the Cyclin-D1-cdk4/6 complex. Cyclin D1 (CycD1) is expressed in the embryonic and neonatal inner ear. In the mature organ of Corti (OC), CycD1 expression is significantly downregulated, paralleling the OC mitotic quiescence. Earlier studies showed that CycD1 overexpression leads to cell-cycle reactivation in cultures of inner ear explants. Here, we characterize a Cre-activated, Doxycycline (Dox)-controlled, conditional CycD1 overexpression model, which when bred to a tetracycline-controlled transcriptional activator and the Atoh1-cre mouse lines, allow for transient CycD1 overexpression and pRBs' downregulation in the inner ear in a reversible fashion. Analyses of postnatal mice's inner ears at various time points revealed the presence of supernumerary cells throughout the length of the cochlea and in the vestibular end-organs. Notably, most supernumerary cells were observed in the inner hair cells' (IHCs) region, expressed myosin VIIa (M7a), and showed no signs of apoptosis at any of the time points analyzed. Auditory and vestibular phenotypes were similar between the different genotypes and treatment groups. The fact that no significant differences were observed in auditory and vestibular function supports the notion that the supernumerary cells detected in the adult mice cochlea and macular end-organs may not impair auditory functions.
Collapse
|
42
|
Brown R, Groves AK. Hear, Hear for Notch: Control of Cell Fates in the Inner Ear by Notch Signaling. Biomolecules 2020; 10:biom10030370. [PMID: 32121147 PMCID: PMC7175228 DOI: 10.3390/biom10030370] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/24/2020] [Accepted: 02/26/2020] [Indexed: 02/08/2023] Open
Abstract
The vertebrate inner ear is responsible for detecting sound, gravity, and head motion. These mechanical forces are detected by mechanosensitive hair cells, arranged in a series of sensory patches in the vestibular and cochlear regions of the ear. Hair cells form synapses with neurons of the VIIIth cranial ganglion, which convey sound and balance information to the brain. They are surrounded by supporting cells, which nourish and protect the hair cells, and which can serve as a source of stem cells to regenerate hair cells after damage in non-mammalian vertebrates. The Notch signaling pathway plays many roles in the development of the inner ear, from the earliest formation of future inner ear ectoderm on the side of the embryonic head, to regulating the production of supporting cells, hair cells, and the neurons that innervate them. Notch signaling is re-deployed in non-mammalian vertebrates during hair cell regeneration, and attempts have been made to manipulate the Notch pathway to promote hair cell regeneration in mammals. In this review, we summarize the different modes of Notch signaling in inner ear development and regeneration, and describe how they interact with other signaling pathways to orchestrate the fine-grained cellular patterns of the ear.
Collapse
Affiliation(s)
- Rogers Brown
- Program in Developmental Biology; Baylor College of Medicine, Houston, TX 77030, USA;
| | - Andrew K. Groves
- Program in Developmental Biology; Baylor College of Medicine, Houston, TX 77030, USA;
- Department of Neuroscience; Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Correspondence: ; Tel.: +1-713-798-8743
| |
Collapse
|
43
|
Montcouquiol M, Kelley MW. Development and Patterning of the Cochlea: From Convergent Extension to Planar Polarity. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a033266. [PMID: 30617059 DOI: 10.1101/cshperspect.a033266] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Within the mammalian cochlea, sensory hair cells and supporting cells are aligned in curvilinear rows that extend along the length of the tonotopic axis. In addition, all of the cells within the epithelium are uniformly polarized across the orthogonal neural-abneural axis. Finally, each hair cell is intrinsically polarized as revealed by the presence of an asymmetrically shaped and apically localized stereociliary bundle. It has been known for some time that many of the developmental processes that regulate these patterning events are mediated, to some extent, by the core planar cell polarity (PCP) pathway. This article will review more recent work demonstrating how components of the PCP pathway interact with cytoskeletal motor proteins to regulate cochlear outgrowth. Finally, a signaling pathway originally identified for its role in asymmetric cell divisions has recently been shown to mediate several aspects of intrinsic hair cell polarity, including kinocilia migration, bundle shape, and elongation.
Collapse
Affiliation(s)
- Mireille Montcouquiol
- INSERM, Neurocentre Magendie, U1215, F-33077 Bordeaux, France.,University of Bordeaux, Neurocentre Magendie, U1215, F-33077 Bordeaux, France
| | - Matthew W Kelley
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
44
|
Novel insights into inner ear development and regeneration for targeted hearing loss therapies. Hear Res 2019; 397:107859. [PMID: 31810596 DOI: 10.1016/j.heares.2019.107859] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/06/2019] [Accepted: 11/25/2019] [Indexed: 02/06/2023]
Abstract
Sensorineural hearing loss is the most common sensory deficit in humans. Despite the global scale of the problem, only limited treatment options are available today. The mammalian inner ear is a highly specialized postmitotic organ, which lacks proliferative or regenerative capacity. Since the discovery of hair cell regeneration in non-mammalian species however, much attention has been placed on identifying possible strategies to reactivate similar responses in humans. The development of successful regenerative approaches for hearing loss strongly depends on a detailed understanding of the mechanisms that control human inner ear cellular specification, differentiation and function, as well as on the development of robust in vitro cellular assays, based on human inner ear cells, to study these processes and optimize therapeutic interventions. We summarize here some aspects of inner ear development and strategies to induce regeneration that have been investigated in rodents. Moreover, we discuss recent findings in human inner ear development and compare the results with findings from animal models. Finally, we provide an overview of strategies for in vitro generation of human sensory cells from pluripotent and somatic progenitors that may provide a platform for drug development and validation of therapeutic strategies in vitro.
Collapse
|
45
|
Munnamalai V, Fekete DM. The acquisition of positional information across the radial axis of the cochlea. Dev Dyn 2019; 249:281-297. [PMID: 31566832 DOI: 10.1002/dvdy.118] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 09/10/2019] [Accepted: 09/10/2019] [Indexed: 12/11/2022] Open
Affiliation(s)
- Vidhya Munnamalai
- The Jackson Laboratory Bar Harbor Maine
- Graduate Program of Biomedical Sciences and EngineeringUniversity of Maine Orono Maine
- The Neuroscience ProgramSackler School of Biomedical Sciences, Tufts University Boston Massachusetts
| | - Donna M. Fekete
- Department of Biological SciencesPurdue University West Lafayette Indiana
- Purdue Institute for Integrative Neuroscience West Lafayette Indiana
- Purdue Center for Cancer Research West Lafayette Indiana
| |
Collapse
|
46
|
Unidirectional and stage-dependent roles of Notch1 in Wnt-responsive Lgr5 + cells during mouse inner ear development. Front Med 2019; 13:705-712. [PMID: 31598881 DOI: 10.1007/s11684-019-0703-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 06/03/2019] [Indexed: 10/25/2022]
Abstract
Wnt and Notch signaling play crucial roles in the determination of the prosensory domain and in the differentiation of hair cells (HCs) and supporting cells during mouse inner ear development; however, the relationship between the two signaling pathways in the mouse cochlea remains largely unknown. Here, we investigated the interactions between Notch and Wnt signaling on the basis of the bidirectional regulation of Notch1 specifically in Wnt-responsive Lgr5+ progenitors during different cochlear development stages. We found that the downregulation of Notch1 in Lgr5+ cells from embryonic day (E) 14.5 to E18.5 can drive the quiescent Lgr5+ cells to re-enter the cell cycle and differentiate into extra HCs, whereas the upregulation of Notch1 expression did not affect the proliferation or differentiation of otic progenitor cells. No effect was observed on the upregulation or downregulation of Notch1 in Lgr5+ cells from E10.5 to E14.5. We concluded that the roles of Notch1 in Wnt-responsive Lgr5+ cells are unidirectional and stage dependent and Notch1 serves as a negative regulator for Lgr5+ progenitor activation during cochlear differentiation. Our findings improved the understanding of the interactions between Notch and Wnt signaling in cochlear development.
Collapse
|
47
|
Li C, Wang Y, Wang G, Lu Y, He S, Sun Y, Liu Z. Fate-mapping analysis using Rorb-IRES-Cre reveals apical-to-basal gradient of Rorb expression in mouse cochlea. Dev Dyn 2019; 249:173-186. [PMID: 31487081 DOI: 10.1002/dvdy.111] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 08/12/2019] [Accepted: 08/27/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Conditional loss-of-function studies are widely conducted using the Cre/Loxp system because this helps circumvent embryonic or neonatal lethality problems. However, Cre strains specific to the inner ear are lacking, and thus lethality frequently occurs even in conditional knockout studies. RESULTS Here, we report a Rorb-IRES-Cre knockin mouse strain in which the Cre recapitulates the expression pattern of endogenous Rorb (RAR-related orphan receptor beta). Analysis of Rorb-IRES-Cre/+; Rosa26-CAG-LSL-tdTomato/+ cochlear samples revealed that tdTomato was expressed at the apical turn only by E12.5. TdTomato was observed in the apical and middle turns but was minimally expressed in the basal turn at E15.5, E18.5, and P5. However, most of the auditory hair cells (HCs) and supporting cells (SCs) in all three turns were tdTomato+ at P15 and P30. Intriguingly, no tdTomato+ vestibular cells were detected until P5 and a few cells were present at P15 and P30. Finally, we also confirmed Rorb mRNA and protein expression in cochlear HCs and SCs at P30. CONCLUSIONS We reveal that Rorb expression exhibits an apical-to-basal gradient in cochleae. The cochlear-specific and apical-to-basal-gradient Rorb Cre activity should enable discrimination of gene functions in cochlear vs vestibular regions as well as low-frequency vs high-frequency regions in the cochlea.
Collapse
Affiliation(s)
- Chao Li
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yunfeng Wang
- ENT Institute and Otorhinolaryngology Department, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine, National Health and Family Planning Commission (NHFPC), Shanghai, China
| | - Guangqin Wang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ying Lu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shunji He
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuwei Sun
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhiyong Liu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
48
|
Roccio M, Edge ASB. Inner ear organoids: new tools to understand neurosensory cell development, degeneration and regeneration. Development 2019; 146:146/17/dev177188. [PMID: 31477580 DOI: 10.1242/dev.177188] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The development of therapeutic interventions for hearing loss requires fundamental knowledge about the signaling pathways controlling tissue development as well as the establishment of human cell-based assays to validate therapeutic strategies ex vivo Recent advances in the field of stem cell biology and organoid culture systems allow the expansion and differentiation of tissue-specific progenitors and pluripotent stem cells in vitro into functional hair cells and otic-like neurons. We discuss how inner ear organoids have been developed and how they offer for the first time the opportunity to validate drug-based therapies, gene-targeting approaches and cell replacement strategies.
Collapse
Affiliation(s)
- Marta Roccio
- Inner Ear Research Laboratory, Department of Biomedical Research (DBMR), University of Bern, Bern 3008, Switzerland .,Department of Otorhinolaryngology, Head & Neck Surgery, Inselspital, Bern University Hospital, University of Bern, Bern 3010, Switzerland
| | - Albert S B Edge
- Department of Otolaryngology, Harvard Medical School, Boston, MA 02115, USA.,Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA.,Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
49
|
Yang LM, Cheah KSE, Huh SH, Ornitz DM. Sox2 and FGF20 interact to regulate organ of Corti hair cell and supporting cell development in a spatially-graded manner. PLoS Genet 2019; 15:e1008254. [PMID: 31276493 PMCID: PMC6636783 DOI: 10.1371/journal.pgen.1008254] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 07/17/2019] [Accepted: 06/18/2019] [Indexed: 01/24/2023] Open
Abstract
The mouse organ of Corti, housed inside the cochlea, contains hair cells and supporting cells that transduce sound into electrical signals. These cells develop in two main steps: progenitor specification followed by differentiation. Fibroblast Growth Factor (FGF) signaling is important in this developmental pathway, as deletion of FGF receptor 1 (Fgfr1) or its ligand, Fgf20, leads to the loss of hair cells and supporting cells from the organ of Corti. However, whether FGF20-FGFR1 signaling is required during specification or differentiation, and how it interacts with the transcription factor Sox2, also important for hair cell and supporting cell development, has been a topic of debate. Here, we show that while FGF20-FGFR1 signaling functions during progenitor differentiation, FGFR1 has an FGF20-independent, Sox2-dependent role in specification. We also show that a combination of reduction in Sox2 expression and Fgf20 deletion recapitulates the Fgfr1-deletion phenotype. Furthermore, we uncovered a strong genetic interaction between Sox2 and Fgf20, especially in regulating the development of hair cells and supporting cells towards the basal end and the outer compartment of the cochlea. To explain this genetic interaction and its effects on the basal end of the cochlea, we provide evidence that decreased Sox2 expression delays specification, which begins at the apex of the cochlea and progresses towards the base, while Fgf20-deletion results in premature onset of differentiation, which begins near the base of the cochlea and progresses towards the apex. Thereby, Sox2 and Fgf20 interact to ensure that specification occurs before differentiation towards the cochlear base. These findings reveal an intricate developmental program regulating organ of Corti development along the basal-apical axis of the cochlea.
Collapse
Affiliation(s)
- Lu M. Yang
- Department of Developmental Biology; Washington University School of Medicine; St. Louis, Missouri, United States of America
| | - Kathryn S. E. Cheah
- School of Biomedical Sciences; The University of Hong Kong; Pokfulam, Hong Kong, China
| | - Sung-Ho Huh
- Department of Developmental Biology; Washington University School of Medicine; St. Louis, Missouri, United States of America
- Holland Regenerative Medicine Program, and the Department of Neurological Sciences; University of Nebraska Medical Center; Omaha, Nebraska, United States of America
- * E-mail: (DMO); (SH)
| | - David M. Ornitz
- Department of Developmental Biology; Washington University School of Medicine; St. Louis, Missouri, United States of America
- * E-mail: (DMO); (SH)
| |
Collapse
|
50
|
Bardhan T, Jeng J, Waldmann M, Ceriani F, Johnson SL, Olt J, Rüttiger L, Marcotti W, Holley MC. Gata3 is required for the functional maturation of inner hair cells and their innervation in the mouse cochlea. J Physiol 2019; 597:3389-3406. [PMID: 31069810 PMCID: PMC6636704 DOI: 10.1113/jp277997] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/07/2019] [Indexed: 01/14/2023] Open
Abstract
KEY POINTS The physiological maturation of auditory hair cells and their innervation requires precise temporal and spatial control of cell differentiation. The transcription factor gata3 is essential for the earliest stages of auditory system development and for survival and synaptogenesis in auditory sensory afferent neurons. We show that during postnatal development in the mouse inner ear gata3 is required for the biophysical maturation, growth and innervation of inner hair cells; in contrast, it is required only for the survival of outer hair cells. Loss of gata3 in inner hair cells causes progressive hearing loss and accounts for at least some of the deafness associated with the human hypoparathyroidism, deafness and renal anomaly (HDR) syndrome. The results show that gata3 is critical for later stages of mammalian auditory system development where it plays distinct, complementary roles in the coordinated maturation of sensory hair cells and their innervation. ABSTRACT The zinc finger transcription factor gata3 regulates inner ear development from the formation of the embryonic otic placode. Throughout development, gata3 is expressed dynamically in all the major cochlear cell types. Its role in afferent formation is well established but its possible involvement in hair cell maturation remains unknown. Here, we find that in heterozygous gata3 null mice (gata3+/- ) outer hair cells (OHCs) differentiate normally but their numbers are significantly lower. In contrast, inner hair cells (IHCs) survive normally but they fail to acquire adult basolateral membrane currents, retain pre-hearing current and efferent innervation profiles and have fewer ribbon synapses. Targeted deletion of gata3 driven by otoferlin-cre recombinase (gata3fl/fl otof-cre+/- ) in IHCs does not affect OHCs or the number of IHC afferent synapses but it leads to a failure in IHC maturation comparable to that observed in gata3+/- mice. Auditory brainstem responses in gata3fl/fl otof-cre+/- mice reveal progressive hearing loss that becomes profound by 6-7 months, whilst distortion product otoacoustic emissions are no different to control animals up to this age. Our results, alongside existing data, indicate that gata3 has specific, complementary functions in different cell types during inner ear development and that its continued expression in the sensory epithelium orchestrates critical aspects of physiological development and neural connectivity. Furthermore, our work indicates that hearing loss in human hypoparathyroidism, deafness and renal anomaly (HDR) syndrome arises from functional deficits in IHCs as well as loss of function from OHCs and both afferent and efferent neurons.
Collapse
MESH Headings
- Animals
- Cell Differentiation/physiology
- Cochlea/metabolism
- Cochlea/physiology
- GATA3 Transcription Factor/metabolism
- Hair Cells, Auditory, Inner/metabolism
- Hair Cells, Auditory, Inner/physiology
- Hair Cells, Auditory, Outer/metabolism
- Hair Cells, Auditory, Outer/physiology
- Hair Cells, Vestibular/metabolism
- Hair Cells, Vestibular/physiology
- Hearing/physiology
- Hearing Loss/metabolism
- Hearing Loss/physiopathology
- Membrane Proteins/metabolism
- Mice, Knockout
- Mice, Transgenic
- Sensory Receptor Cells/metabolism
- Sensory Receptor Cells/physiology
- Synapses/metabolism
Collapse
Affiliation(s)
- Tanaya Bardhan
- Department of Biomedical ScienceUniversity of SheffieldSheffieldUK
| | - Jing‐Yi Jeng
- Department of Biomedical ScienceUniversity of SheffieldSheffieldUK
| | - Marco Waldmann
- Department of OtolaryngologyTübingen Hearing Research CenterSection of Physiological Acoustics and CommunicationUniversity of Tübingen72076TübingenGermany
| | - Federico Ceriani
- Department of Biomedical ScienceUniversity of SheffieldSheffieldUK
| | | | - Jennifer Olt
- Department of Biomedical ScienceUniversity of SheffieldSheffieldUK
| | - Lukas Rüttiger
- Department of OtolaryngologyTübingen Hearing Research CenterSection of Physiological Acoustics and CommunicationUniversity of Tübingen72076TübingenGermany
| | - Walter Marcotti
- Department of Biomedical ScienceUniversity of SheffieldSheffieldUK
| | | |
Collapse
|