1
|
Santiago C, Siegrist J, Africawala N, Handler A, Tasnim A, Anjum R, Turecek J, Lehnert BP, Renauld S, Choi J, Nolan-Tamariz M, Iskols M, Magee AR, Paradis S, Sharma N, Ginty DD. Activity-dependent development of the body's touch receptors. Neuron 2025:S0896-6273(25)00298-3. [PMID: 40381613 DOI: 10.1016/j.neuron.2025.04.015] [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: 09/22/2023] [Revised: 02/12/2025] [Accepted: 04/16/2025] [Indexed: 05/20/2025]
Abstract
We report a role for activity in the development of the primary sensory neurons that detect touch. Genetic deletion of Piezo2, the principal mechanosensitive ion channel in somatosensory neurons, caused profound changes in the formation of mechanosensory end-organ structures. Peripheral-nervous-system-specific deletion of the voltage-gated sodium channel Nav1.6 (Scn8a), which resulted in altered electrophysiological responses to mechanical stimuli, also disrupted somatosensory neuron morphologies, supporting a role for neuronal activity in end-organ formation. Single-cell RNA sequencing of Piezo2 mutants revealed changes in gene expression in sensory neurons activated by light mechanical forces, whereas other neuronal classes were minimally affected, and genetic deletion of Piezo2-dependent genes partially reproduced the defects in mechanosensory neuron structures observed in Piezo2 mutants. These findings indicate that mechanically evoked neuronal activity acts early in life to shape the maturation of mechanosensory end-organs that underlie our sense of gentle touch.
Collapse
Affiliation(s)
- Celine Santiago
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Julianna Siegrist
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Nusrat Africawala
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Annie Handler
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Aniqa Tasnim
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Rabia Anjum
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, MA 02453, USA
| | - Josef Turecek
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Brendan P Lehnert
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Sophia Renauld
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jinheon Choi
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Michael Nolan-Tamariz
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Michael Iskols
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Alexandra R Magee
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Suzanne Paradis
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, MA 02453, USA
| | - Nikhil Sharma
- Department of Biochemistry and Molecular Biophysics and Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University Medical Center, New York, NY 10032, USA
| | - David D Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
2
|
Li Y, Li N, Zhang J, Liu X, Xu Y, Kong L, Man R, Li J. FKBP51 protects hair cells of utricles from gentamicin-induced toxicity in vitro: possible relation to the activities of NF-κB signaling pathway. Food Chem Toxicol 2025; 202:115513. [PMID: 40324675 DOI: 10.1016/j.fct.2025.115513] [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: 02/09/2025] [Revised: 04/22/2025] [Accepted: 05/03/2025] [Indexed: 05/07/2025]
Abstract
FK506-binding protein 51 (FKBP51) belongs to the immunophilin family, which is related to regulation of cell growth and apoptosis. The present study was designed to explore the expression of FKBP51 in murine utricle hair cells (HCs) and the possible mechanisms underpinning the actions of FKBP51 relevant to gentamicin-mediated toxicity, with special attention given to nuclear factor-kappa B (NF-κB) activities in vitro. Here, we found, for the first time, that FKBP51 was widely expressed in both cytoplasm and cytomembrane. Moreover, the expression of FKBP51 in cultured HCs of utricle was downregulated after exposure to 1 mM gentamicin for 24 h. Then, the fkbp51 knockout mice were utilized to further investigate the role of FKBP51 in HCs in response to gentamicin. And the absence of FKBP51 led to severe HCs loss, accumulated ROS levels, and increased apoptotic factors, which could be alleviated by 2 mM N-acetyl-l-cysteine (NAC) to certain degree. In addition, mechanistic studies with 10 mM BAY 11-7082 showed that FKBP51 protected HCs against gentamicin-induced damage, at least in part, via blocking the NF-κB pathway activation. Taken together, our new findings suggest that FKBP51 might serve as a new target for the prevention of HCs of utricle from aminoglycoside-induced vestibular toxicity.
Collapse
Affiliation(s)
- Yanan Li
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Ning Li
- Department of Clinical Laboratory, Juye Hospital of Traditional Chinese Medicine, Juye, 274900, China
| | - Junhong Zhang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Xueyan Liu
- Shandong Institute of Scientific and Technical Information, Jinan, 250101, China
| | - Yue Xu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Ligang Kong
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
| | - Rongjun Man
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China.
| | - Jianfeng Li
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China.
| |
Collapse
|
3
|
Peineau T, Marcovich I, Rodriguez CVM, O'Malley S, Cui R, Ballesteros A, Holt JR. Mammalian TMC1 or 2 are necessary for scramblase activity in auditory hair cells. Hear Res 2025; 460:109229. [PMID: 40073458 PMCID: PMC12038766 DOI: 10.1016/j.heares.2025.109229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/18/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025]
Abstract
Sensory transduction in auditory hair cells gates mechanosensitive ion channels, converting sound information into electrical signals (Zheng and Holt, 2021). Previously, we found that Transmembrane channel (TMC) proteins 1 and 2 form the pore of hair cell transduction channels (Pan et al., 2013; 2018). The structure of C. elegans TMC proteins (Jeong et al., 2022; Clark et al., 2024) and predicted mammalian TMC structures (Hahn et al., 2009; Ballesteros et al., 2018; Pan et al., 2018) are reminiscent of TMEM16 proteins, which function as Ca2+-activated ion channels and lipid scramblases. Here, we investigated lipid scramblase activity in live auditory hair cells with pharmacologic or genetic disruption of TMC1, extending work reported by Ballesteros and Swartz (2022). We used annexin-V to label phosphatidylserine (PS) localized in the outer leaflet of hair cell stereocilia membranes. PS externalization was triggered by disruption of sensory transduction using the blocker, benzamil, or by genetic mutations that affect TMC1 permeation properties. We found that expression of either TMC1 or TMC2, was essential for PS externalization. Tmc1/Tmc2 knockout mice and Tmie mutant mice lacked PS externalization completely. We also determined that expression of exogenous human TMCs (hTMC1 or hTMC2) in Tmc1/Tmc2 knockout mice induced PS externalization. Lastly, we demonstrated that expression of a dominant mutation in Tmc1 evoked constitutive PS externalization, while a recessive mutation eliminated PS externalization. Our data suggest that disruption of sensory transduction may lead to dysregulation of membrane homeostasis in hair cells and thus may contribute to auditory dysfunction in mice and humans.
Collapse
Affiliation(s)
- Thibault Peineau
- Boston Children's Hospital and Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, USA
| | - Irina Marcovich
- Boston Children's Hospital and Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, USA
| | | | - Sydney O'Malley
- Boston Children's Hospital and Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, USA
| | - Runjia Cui
- National Institute on Deafness and other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Angela Ballesteros
- National Institute on Deafness and other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jeffrey R Holt
- Boston Children's Hospital and Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, USA.
| |
Collapse
|
4
|
Deng S, Yan Z. TMC1 and TMC2 function as the mechano-electrical transduction ion channel in hearing. Curr Opin Neurobiol 2025; 93:103026. [PMID: 40280017 DOI: 10.1016/j.conb.2025.103026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/29/2025]
Abstract
Mechanotransduction within the specialized cochlea hair cells is fundamental to perceiving sound waves. This intricate mechanism converts mechanical vibrations into electrical signals that the brain can interpret as sound. The molecular identity of the mechanoelectrical transducer continues to be a subject of intense debate. Transmembrane channel-like protein 1 (TMC1) was initially recognized as a deafness gene in humans, with subsequent studies revealing the hearing loss phenotype in Tmc1 mutant mice. Mechanotransduction currents were lost in the hair cells of Tmc1;Tmc2 double knockout mice, indicating the involvement of TMC1/2 in auditory mechanotransduction. Both TMC1/2 are expressed at the tip of stereocilia in hair cells, the subcellular site of auditory mechanotransduction. Notably, recent in vitro studies have overcome long-standing technical barriers that TMC1/2 are not localized to the cell membrane in heterologous expression and provided compelling evidence that TMC1/2 are mechanically gated ion channels, finally fulfilling both the essential and necessary criteria they must meet as sensory transducers. In hair cells, tip-links possibly relay force to TMC1/2 by tether gating or membrane-tension gating, while the molecular mechanisms underlying each gating mechanism require further investigation.
Collapse
Affiliation(s)
- Siqi Deng
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China; Department of Life Sciences, Hong Kong University of Science and Technology, Hong Kong, China
| | - Zhiqiang Yan
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China; Department of Neurobiology, School of Basic Medicine, Capital Medical University, Beijing, China; Institute for Medical Physiology, Chinese Institutes for Medical Research, Beijing, China.
| |
Collapse
|
5
|
Yao X, Zhang Y, Hong X, Xing Y, Xu Z. Esrp1 and Esrp2 regulate the stability of tmc1/ 2a mRNAs in zebrafish sensory hair cells. J Neurosci 2025; 45:e0837242025. [PMID: 40086870 PMCID: PMC12019119 DOI: 10.1523/jneurosci.0837-24.2025] [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: 05/04/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025] Open
Abstract
RNA-binding proteins (RBPs) are important for post-transcriptional RNA processing, including pre-mRNA alternative splicing, mRNA stability, and translation. Several RBPs have been shown to play pivotal roles in the inner ear, whose dysfunction leads to auditory and/or balance impairments. Epithelial splicing-regulatory protein 1 (ESRP1) regulates alternative splicing and mRNA stability, and mutations in ESRP1 gene have been associated with sensorineural hearing loss in humans. In Esrp1 knockout mouse embryos, alternative splicing of its target genes such as Fgfr2 is impaired, which eventually result in cochlear development deficits. However, Esrp1 knockout mice die soon after birth because of complications from cleft-lip and palate defects, impeding further investigations at later postnatal ages. In the present study, we explored the role of ESRP1 in hearing using zebrafish as a model. We showed that esrp1 and its paralog esrp2 are expressed in the inner ear and certain anterior lateral line (ALL) neuromasts. Furthermore, our data suggested that Esrp1 and Esrp2 are required for the mechano-electrical transduction (MET) function of hair cells. RNA sequencing results indicated a significant decrease in the levels of several mRNAs in esrp1/2 double knockout larvae. Among the dysregulated genes are tmc1 and tmc2a, which encode essential subunits of the MET complex. Further investigations demonstrated that Esrp1/2 could directly bind to tmc1 and tmc2a mRNAs and affect their stability. Taken together, we showed here that Esrp1 and Esrp2 regulate the MET function of zebrafish sensory hair cells by modulating the stability of tmc1 and tmc2a mRNAs.Significance statement ESRP1 is an important RNA-binding protein, whose malfunction has been associated with hearing loss in humans. Esrp1 knockout affects alternative splicing of its target mRNAs such as Fgfr2, eventually leading to cochlear development deficits in mice. However, Esrp1 knockout mice die soon after birth, precluding further investigations at later postnatal ages. In this study, we explored the role of ESRP1 in hearing using zebrafish as a model. Our results demonstrated that esrp1 and its paralog esrp2 are expressed in the zebrafish inner ear, and that esrp1/esrp2 double knockout compromised the mechano-electrical transduction (MET) function of hair cells. Additionally, we successfully identified tmc1 and tmc2a mRNAs as the targets of Esrp1/2, which encode essential subunits of the MET complex.
Collapse
Affiliation(s)
- Xuebo Yao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
| | - Yan Zhang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
- Department of Biochemistry and Department of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Xiaying Hong
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
| | - Yanyi Xing
- Women's Hospital, Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Zhigang Xu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
- Shandong Provincial Collaborative Innovation Center of Cell Biology, Shandong Normal University, Jinan, Shandong 250014, China
| |
Collapse
|
6
|
Chen Y, Li Y, Liu Y, Sun J, Feng W, Chen Y, Tian Y, Lei T, Huang P. Ectopic mouse TMC1 and TMC2 alone form mechanosensitive channels that are potently modulated by TMIE. Proc Natl Acad Sci U S A 2025; 122:e2403141122. [PMID: 39999170 PMCID: PMC11892609 DOI: 10.1073/pnas.2403141122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 01/13/2025] [Indexed: 02/27/2025] Open
Abstract
The mechanotransduction (MT) channel expressed in cochlear and vestibular hair cells converts the mechanical stimulation of sound and head movements into electrochemical signals. Recently, TMC1 and TMC2 (TMC1/2) have been recognized as the pore-forming subunit of the MT channel, but TMC1/2 functional expression in heterologous cells-which is critical for unequivocally identifying them as the bona fide pore-forming subunit of the MT channel-has not been achieved because ectopic TMC1/2 become trapped in the ER. Here, we report that adding a Fyn lipidation tag to mouse TMC1/2 (mTMC1/2) drove their cell-surface expression, and, importantly, full-length mTMC1/2 expressed alone functioned as mechanosensitive channels, underscoring the view that TMC1/2 constitute the sole pore-forming subunit of the MT channel. Moreover, mouse transmembrane inner ear (TMIE) (mTMIE) protein robustly stimulated TMC1/2 channel activity by modulating their gating. Intriguingly, the N-terminal 27 residues of mTMIE were dispensable for regulating TMC1/2 in our in vitro functional assay, whereas, in striking contrast, mutating mTMIE C76C77, the predicted palmitoylation sites, eliminated mTMIE stimulation of mTMC1/2, indicating a crucial role of the palmitoyl group in regulating TMC1/2 gating. mTMC1/2+mTMIE form 18 pS and 24 pS single channels, respectively. mTMC1/2+mTMIE single channels showed biophysical and pharmacological properties similar to those of the MT channel. Our findings provide insights into several fundamental and debated aspects of the function of TMC1/2 and TMIE, and our functional assay of TMC1/2 and TMIE in heterologous cells will facilitate further functional and structural characterization of these proteins and other MT-complex components.
Collapse
Affiliation(s)
- Yixuan Chen
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Hong Kong0000, China
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong0000, China
| | - Yulin Li
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong0000, China
| | - Yonghong Liu
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong0000, China
| | - Jiawen Sun
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Hong Kong0000, China
| | - Wanying Feng
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong0000, China
| | - Yanfei Chen
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong0000, China
| | - Ye Tian
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong0000, China
| | - Tianlun Lei
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong0000, China
| | - Pingbo Huang
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Hong Kong0000, China
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong0000, China
- State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong0000, China
| |
Collapse
|
7
|
Fu S, Pan X, Lu M, Dong J, Yan Z. Human TMC1 and TMC2 are mechanically gated ion channels. Neuron 2025; 113:411-425.e4. [PMID: 39674179 DOI: 10.1016/j.neuron.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 09/23/2024] [Accepted: 11/14/2024] [Indexed: 12/16/2024]
Abstract
Mammalian transmembrane channel-like proteins 1 and 2 (TMC1 and TMC2) have emerged as very promising candidate mechanotransduction channels in hair cells. However, controversy persists because the heterogeneously expressed TMC1/2 in cultured cells lack evidence of mechanical gating, primarily due to their absence from the plasma membrane. By employing domain swapping with OSCA1.1 and subsequent point mutations, we successfully identified membrane-localized mouse TMC1/2 mutants, demonstrating that they are mechanically gated in heterologous cells. Further, whole-genome CRISPRi screening enabled wild-type human TMC1/2 localization in the plasma membrane, where they responded robustly to poking stimuli. In addition, wild-type human TMC1/2 showed stretch-activated currents and clear single-channel current activities. Deafness-related TMC1 mutations altered the reversal potential of TMC1, indicating that TMC1/2 are pore-forming mechanotransduction channels. In summary, our study provides evidence that human TMC1/2 are pore-forming, mechanically activated ion channels, supporting their roles as mechanotransduction channels in hair cells.
Collapse
Affiliation(s)
- Songdi Fu
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China; Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China
| | - Xueqi Pan
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China; Department of Neurobiology, School of Basic Medicine, Capital Medical University, Beijing, China
| | - Mingshun Lu
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jianying Dong
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China; Department of Neurobiology, School of Basic Medicine, Capital Medical University, Beijing, China
| | - Zhiqiang Yan
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China; Department of Neurobiology, School of Basic Medicine, Capital Medical University, Beijing, China; Institute for Medical Physiology, Chinese Institutes for Medical Research, Beijing, China.
| |
Collapse
|
8
|
Wu S, Lin L, Hu Q, Yao X, Wang H, Liu S, Liu Q, Xi Y, Lin Y, Gong J, Hu R, Zhan W, Luo Y, He G, Liu Z, Xiong W, Wang Q, Xu Z, Bai F, Lu Q. Mechano-electrical transduction components TMC1-CIB2 undergo a Ca 2+-induced conformational change linked to hearing loss. Dev Cell 2025:S1534-5807(25)00004-8. [PMID: 39889697 DOI: 10.1016/j.devcel.2025.01.004] [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: 06/22/2023] [Revised: 08/20/2024] [Accepted: 01/09/2025] [Indexed: 02/03/2025]
Abstract
TMC1, a unique causative gene associated with deafness, exhibits variants with autosomal dominant and recessive inheritance patterns. TMC1 codes for the transmembrane channel-like protein 1 (TMC1), a key component of the mechano-electrical transduction (MET) machinery for hearing. However, the molecular mechanism of Ca2+ regulation in MET remains unclear. Calcium and integrin-binding protein 2 (CIB2), another MET component associated with deafness, can bind with Ca2+. Our study shows that TMC1-CIB2 complex undergoes a Ca2+-induced conformational change. We identified a vertebrate-specific binding site on TMC1 that interacts with apo CIB2, linked with hearing loss. Using an ex vivo mouse organotypic cochlea model, we demonstrated that disruption of the calcium-binding site of CIB2 perturbs the MET channel conductivity. After systematically analyzing the hearing loss variants, we observed dominant mutations of TMC1 cluster around the putative ion pore or at the binding interfaces with CIB2. These findings elucidate the molecular mechanisms underlying TMC1-linked hearing loss.
Collapse
Affiliation(s)
- Shaoxuan Wu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Lin Lin
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Qiaoyu Hu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; Innovation Center for AI and Drug Discovery, School of Pharmacy, East China Normal University, Shanghai 200062, China
| | - Xuebo Yao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
| | - Hongyang Wang
- Senior Department of Otolaryngology, Head and Neck Surgery, Department of Audiology and Vestibular Medicine, Chinese PLA Institute of Otolaryngology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China
| | - Shuang Liu
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Qingling Liu
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Yuehui Xi
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
| | - Yuzhe Lin
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Jianqiao Gong
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ruixing Hu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Wei Zhan
- National Facility for Protein Science in Shanghai, Shanghai Advanced Research Institute, Chinese Academy of Science, Zhangjiang Lab, Shanghai 201210, China
| | - Yi Luo
- Senior Department of Otolaryngology, Head and Neck Surgery, Department of Audiology and Vestibular Medicine, Chinese PLA Institute of Otolaryngology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China
| | - Guang He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Zhijun Liu
- National Facility for Protein Science in Shanghai, Shanghai Advanced Research Institute, Chinese Academy of Science, Zhangjiang Lab, Shanghai 201210, China
| | - Wei Xiong
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Qiuju Wang
- Senior Department of Otolaryngology, Head and Neck Surgery, Department of Audiology and Vestibular Medicine, Chinese PLA Institute of Otolaryngology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China.
| | - Zhigang Xu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China.
| | - Fang Bai
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; School of Information Science and Technology, ShanghaiTech University, Shanghai 201210, China; Shanghai Clinical Research and Trial Center, Shanghai 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| | - Qing Lu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China; Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China.
| |
Collapse
|
9
|
Giese APJ, Weng WH, Kindt KS, Chang HHV, Montgomery JS, Ratzan EM, Beirl AJ, Aponte Rivera R, Lotthammer JM, Walujkar S, Foster MP, Zobeiri OA, Holt JR, Riazuddin S, Cullen KE, Sotomayor M, Ahmed ZM. Complexes of vertebrate TMC1/2 and CIB2/3 proteins form hair-cell mechanotransduction cation channels. eLife 2025; 12:RP89719. [PMID: 39773557 PMCID: PMC11709434 DOI: 10.7554/elife.89719] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Abstract
Calcium and integrin-binding protein 2 (CIB2) and CIB3 bind to transmembrane channel-like 1 (TMC1) and TMC2, the pore-forming subunits of the inner-ear mechano-electrical transduction (MET) apparatus. These interactions have been proposed to be functionally relevant across mechanosensory organs and vertebrate species. Here, we show that both CIB2 and CIB3 can form heteromeric complexes with TMC1 and TMC2 and are integral for MET function in mouse cochlea and vestibular end organs as well as in zebrafish inner ear and lateral line. Our AlphaFold 2 models suggest that vertebrate CIB proteins can simultaneously interact with at least two cytoplasmic domains of TMC1 and TMC2 as validated using nuclear magnetic resonance spectroscopy of TMC1 fragments interacting with CIB2 and CIB3. Molecular dynamics simulations of TMC1/2 complexes with CIB2/3 predict that TMCs are structurally stabilized by CIB proteins to form cation channels. Overall, our work demonstrates that intact CIB2/3 and TMC1/2 complexes are integral to hair-cell MET function in vertebrate mechanosensory epithelia.
Collapse
Affiliation(s)
- Arnaud PJ Giese
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of MedicineBaltimoreUnited States
| | - Wei-Hsiang Weng
- Department of Chemistry and Biochemistry, The Ohio State UniversityColumbusUnited States
- Biophysics Graduate Program, The Ohio State UniversityColumbusUnited States
| | - Katie S Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | | | - Jonathan S Montgomery
- Department of Chemistry and Biochemistry, The Ohio State UniversityColumbusUnited States
- Ohio State Biochemistry Program, The Ohio State UniversityColumbusUnited States
| | - Evan M Ratzan
- Departments of Otolaryngology and Neurology, F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Alisha J Beirl
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Roberto Aponte Rivera
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Jeffrey M Lotthammer
- Department of Chemistry and Biochemistry, The Ohio State UniversityColumbusUnited States
| | - Sanket Walujkar
- Department of Chemistry and Biochemistry, The Ohio State UniversityColumbusUnited States
| | - Mark P Foster
- Department of Chemistry and Biochemistry, The Ohio State UniversityColumbusUnited States
- Biophysics Graduate Program, The Ohio State UniversityColumbusUnited States
- Ohio State Biochemistry Program, The Ohio State UniversityColumbusUnited States
| | - Omid A Zobeiri
- Department of Biomedical Engineering, McGill UniversityMontrealCanada
| | - Jeffrey R Holt
- Departments of Otolaryngology and Neurology, F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Saima Riazuddin
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of MedicineBaltimoreUnited States
| | - Kathleen E Cullen
- Departments of Biomedical Engineering, Neuroscience, and Otolaryngology and Head and Neck Surgery, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Marcos Sotomayor
- Department of Chemistry and Biochemistry, The Ohio State UniversityColumbusUnited States
- Biophysics Graduate Program, The Ohio State UniversityColumbusUnited States
- Ohio State Biochemistry Program, The Ohio State UniversityColumbusUnited States
| | - Zubair M Ahmed
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of MedicineBaltimoreUnited States
- Department of Biochemistry and Molecular Biology, University of Maryland School of MedicineBaltimoreUnited States
- Department of Ophthalmology and Visual Sciences, University of Maryland School of MedicineBaltimoreUnited States
| |
Collapse
|
10
|
Ma X, Chen X, Che Y, Zhu S, Wang X, Gao S, Wu J, Kong F, Cheng C, Wu Y, Guo J, Qi J, Chai R. The single-cell transcriptomic landscape of the topological differences in mammalian auditory receptors. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2398-2410. [PMID: 39083201 DOI: 10.1007/s11427-024-2672-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/01/2024] [Indexed: 10/22/2024]
Abstract
Mammalian hair cells (HCs) are arranged spirally along the cochlear axis and correspond to different frequency ranges. Serving as primary sound detectors, HCs spatially segregate component frequencies into a topographical map. HCs display significant diversity in anatomical and physiological characteristics, yet little is known about the organization of the cochleotopic map of HCs or the molecules involved in this process. Using single-cell RNA sequencing, we determined the distinct molecular profiles of inner hair cells and outer hair cells, and we identified numerous position-dependent genes that were expressed as gradients. Newly identified genes such as Ptn, Rxra, and Nfe2l2 were found to be associated with tonotopy. We employed the SCENIC algorithm to predict the transcription factors that potentially shape these tonotopic gradients. Furthermore, we confirmed that Nfe2l2, a tonotopy-related transcription factor, is critical in mice for sensing low-to-medium sound frequencies in vivo. the analysis of cell-cell communication revealed potential receptor-ligand networks linking inner hair cells to spiral ganglion neurons, including pathways such as BDNF-Ntrk and PTN-Scd4, which likely play essential roles in tonotopic maintenance. Overall, these findings suggest that molecular gradients serve as the organizing principle for maintaining the selection of sound frequencies by HCs.
Collapse
Affiliation(s)
- Xiangyu Ma
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Xin Chen
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Yuwei Che
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Siyao Zhu
- School of Engineering, Vanderbilt University, Nashville, 37240, USA
| | - Xinlin Wang
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Shan Gao
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Jiheng Wu
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Fanliang Kong
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Cheng Cheng
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing, 210096, China
- Research Institute of Otorhinolaryngology, Nanjing, 210096, China
| | - Yunhao Wu
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Jiamin Guo
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Jieyu Qi
- Department of Neurology, Aerospace Center Hospital, School of Life Sciences, Beijing Institute of Technology, Beijing, 100081, China.
- Advanced Technology Research Institute, Beijing Institute of Technology, Beijing, 100081, China.
| | - Renjie Chai
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
- Department of Neurology, Aerospace Center Hospital, School of Life Sciences, Beijing Institute of Technology, Beijing, 100081, China.
- Advanced Technology Research Institute, Beijing Institute of Technology, Beijing, 100081, China.
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Southeast University Shenzhen Research Institute, Shenzhen, 518063, China.
| |
Collapse
|
11
|
Hartig EI, Day M, Jarysta A, Tarchini B. Proteins required for stereocilia elongation during mammalian hair cell development ensure precise and steady heights during adult life. Proc Natl Acad Sci U S A 2024; 121:e2405455121. [PMID: 39320919 PMCID: PMC11459194 DOI: 10.1073/pnas.2405455121] [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: 03/15/2024] [Accepted: 08/05/2024] [Indexed: 09/26/2024] Open
Abstract
The hair bundle, or stereocilia bundle, is the mechanosensory compartment of hair cells (HCs) in the inner ear. To date, most mechanistic studies have focused on stereocilia bundle morphogenesis, and it remains unclear how this organelle critical for hearing preserves its precise dimensions during life in mammals. The GPSM2-GNAI complex occupies the distal tip of stereocilia in the tallest row and is required for their elongation during development. Here, we ablate GPSM2-GNAI in adult mouse HCs after normal stereocilia elongation is completed. We observe a progressive height reduction of the tallest row stereocilia totaling ~600 nm after 12 wk in Gpsm2 mutant inner HCs. To measure GPSM2 longevity at tips, we generated a HaloTag-Gpsm2 mouse strain and performed pulse-chase experiments in vivo. Estimates using pulse-chase or tracking loss of GPSM2 immunolabeling following Gpsm2 inactivation suggest that GPSM2 is relatively long-lived at stereocilia tips with a half-life of 9 to 10 d. Height reduction coincides with dampened auditory brainstem responses evoked by low-frequency stimuli in particular. Finally, GPSM2 is required for normal tip enrichment of elongation complex (EC) partners MYO15A, WHRN, and EPS8, mirroring their established codependence during development. Taken together, our results show that the EC is also essential in mature HCs to ensure precise and stable stereocilia height and for sensitive detection of a full range of sound frequencies.
Collapse
Affiliation(s)
- Elli I. Hartig
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, MA02111
- The Jackson Laboratory, Bar Harbor, ME04609
| | | | | | - Basile Tarchini
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, MA02111
- The Jackson Laboratory, Bar Harbor, ME04609
| |
Collapse
|
12
|
Zhu Z, Reid W, George SS, Ou V, Ó Maoiléidigh D. 3D morphology of an outer-hair-cell hair bundle increases its displacement and dynamic range. Biophys J 2024; 123:3433-3451. [PMID: 39161094 PMCID: PMC11480765 DOI: 10.1016/j.bpj.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 05/22/2024] [Accepted: 08/13/2024] [Indexed: 08/21/2024] Open
Abstract
In mammals, outer-hair-cell hair bundles (OHBs) transduce sound-induced forces into receptor currents and are required for the wide dynamic range and high sensitivity of hearing. OHBs differ conspicuously in morphology from other types of bundles. Here, we show that the 3D morphology of an OHB greatly impacts its mechanics and transduction. An OHB comprises rod-like stereocilia, which pivot on the surface of its sensory outer hair cell. Stereocilium pivot positions are arranged in columns and form a V shape. We measure the pivot positions and determine that OHB columns are far from parallel. To calculate the consequences of an OHB's V shape and far-from-parallel columns, we develop a mathematical model of an OHB that relates its pivot positions, 3D morphology, mechanics, and receptor current. We find that the 3D morphology of the OHB can halve its stiffness, can double its damping coefficient, and causes stereocilium displacements driven by stimulus forces to differ substantially across the OHB. Stereocilium displacements drive the opening and closing of ion channels through which the receptor current flows. Owing to the stereocilium-displacement differences, the currents passing through the ion channels can peak versus the stimulus frequency and vary considerably across the OHB. Consequently, the receptor current peaks versus the stimulus frequency. Ultimately, the OHB's 3D morphology can increase its receptor-current dynamic range more than twofold. Our findings imply that potential pivot-position changes owing to development, mutations, or location within the mammalian auditory organ might greatly alter OHB function.
Collapse
Affiliation(s)
- Zenghao Zhu
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, Stanford, California
| | - Wisam Reid
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, Stanford, California; Harvard Medical School, Boston, Massachusetts
| | - Shefin Sam George
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, Stanford, California
| | - Victoria Ou
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, Stanford, California
| | - Dáibhid Ó Maoiléidigh
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, Stanford, California.
| |
Collapse
|
13
|
Ebrahim S, Ballesteros A, Zheng WS, Mukherjee S, Hu G, Weng WH, Montgomery JS, Agyemang Y, Cui R, Sun W, Krystofiak E, Foster MP, Sotomayor M, Kachar B. Transmembrane channel-like 4 and 5 proteins at microvillar tips are potential ion channels and lipid scramblases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.22.609173. [PMID: 39229161 PMCID: PMC11370596 DOI: 10.1101/2024.08.22.609173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Microvilli-membrane bound actin protrusions on the surface of epithelial cells-are sites of critical processes including absorption, secretion, and adhesion. Increasing evidence suggests microvilli are mechanosensitive, but underlying molecules and mechanisms remain unknown. Here, we localize transmembrane channel-like proteins 4 and 5 (TMC4 and 5) and calcium and integrin binding protein 3 (CIB3) to microvillar tips in intestinal epithelial cells, near glycocalyx insertion sites. We find that TMC5 colocalizes with CIB3 in cultured cells and that a TMC5 fragment forms a complex with CIB3 in vitro. Homology and AlphaFold2 models reveal a putative ion permeation pathway in TMC4 and 5, and molecular dynamics simulations predict both proteins can conduct ions and perform lipid scrambling. These findings raise the possibility that TMC4 and 5 interact with CIB3 at microvillar tips to form a mechanosensitive complex, akin to TMC1 and 2, and CIB2 and 3, within the mechanotransduction channel complex at the tips of inner ear stereocilia.
Collapse
Affiliation(s)
- Seham Ebrahim
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA
| | - Angela Ballesteros
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
- Section on Sensory Physiology and Biophysics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - W. Sharon Zheng
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA
| | - Shounak Mukherjee
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Gaizun Hu
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA
| | - Wei-Hsiang Weng
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
- Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Jonathan S. Montgomery
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH 43210, USA
| | - Yaw Agyemang
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Runjia Cui
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Willy Sun
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Evan Krystofiak
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mark P. Foster
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
- Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH 43210, USA
| | - Marcos Sotomayor
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
- Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH 43210, USA
| | - Bechara Kachar
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
14
|
Hussain S, Sedlacek M, Cui R, Zhang-Hooks W, Bergles D, Bum-Shin J, Kindt KS, Kachar B. Spontaneous calcium transients in hair cell stereocilia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.12.607658. [PMID: 39185174 PMCID: PMC11343103 DOI: 10.1101/2024.08.12.607658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
The hair bundle of auditory and vestibular hair cells converts mechanical stimuli into electrical signals through mechanoelectrical transduction (MET). The MET apparatus is built around a tip link that connects neighboring stereocilia that are aligned in the direction of mechanosensitivity of the hair bundle. Upon stimulation, the MET channel complex responds to changes in tip-link tension and allows a cation influx into the cell. Ca2+ influx in stereocilia has been used as a signature of MET activity. Using genetically encoded Ca2+ sensors (GCaMP3, GCaMP6s) and high-performance fluorescence confocal microscopy, we detect spontaneous Ca2+ transients in individual stereocilia in developing and fully formed hair bundles. We demonstrate that this activity is abolished by MET channel blockers and thus likely originates from putative MET channels. We observe Ca2+ transients in the stereocilia of mice in tissue explants as well as in vivo in zebrafish hair cells, indicating this activity is functionally conserved. Within stereocilia, the origin of Ca2+ transients is not limited to the canonical MET site at the stereocilia tip but is also present along the stereocilia length. Remarkably, we also observe these Ca2+ transients in the microvilli-like structures on the hair cell surface in the early stages of bundle development, prior to the onset of MET. Ca2+ transients are also present in the tallest rows of stereocilia in auditory hair cells, structures not traditionally thought to contain MET channels. We hypothesize that this newly described activity may reflect stochastic and spontaneous MET channel opening. Localization of these transients to other regions of the stereocilia indicates the presence of a pool of channels or channel precursors. Our work provides insights into MET channel assembly, maturation, function, and turnover.
Collapse
Affiliation(s)
- Saman Hussain
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Miloslav Sedlacek
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Runjia Cui
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wendy Zhang-Hooks
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Dwight Bergles
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Otolaryngology-Head & Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jung Bum-Shin
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22908
| | - Katie S. Kindt
- Laboratory of Cellular Biology, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bechara Kachar
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
15
|
Giese APJ, Weng WH, Kindt KS, Chang HHV, Montgomery JS, Ratzan EM, Beirl AJ, Rivera RA, Lotthammer JM, Walujkar S, Foster MP, Zobeiri OA, Holt JR, Riazuddin S, Cullen KE, Sotomayor M, Ahmed ZM. Complexes of vertebrate TMC1/2 and CIB2/3 proteins form hair-cell mechanotransduction cation channels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.26.542533. [PMID: 37398045 PMCID: PMC10312449 DOI: 10.1101/2023.05.26.542533] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Calcium and integrin-binding protein 2 (CIB2) and CIB3 bind to transmembrane channel-like 1 (TMC1) and TMC2, the pore-forming subunits of the inner-ear mechano-electrical transduction (MET) apparatus. These interactions have been proposed to be functionally relevant across mechanosensory organs and vertebrate species. Here we show that both CIB2 and CIB3 can form heteromeric complexes with TMC1 and TMC2 and are integral for MET function in mouse cochlea and vestibular end organs as well as in zebrafish inner ear and lateral line. Our AlphaFold 2 models suggest that vertebrate CIB proteins can simultaneously interact with at least two cytoplasmic domains of TMC1 and TMC2 as validated using nuclear magnetic resonance spectroscopy of TMC1 fragments interacting with CIB2 and CIB3. Molecular dynamics simulations of TMC1/2 complexes with CIB2/3 predict that TMCs are structurally stabilized by CIB proteins to form cation channels. Overall, our work demonstrates that intact CIB2/3 and TMC1/2 complexes are integral to hair-cell MET function in vertebrate mechanosensory epithelia.
Collapse
Affiliation(s)
- Arnaud P J Giese
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Wei-Hsiang Weng
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
- Biophysics Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Katie S Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | | | - Jonathan S Montgomery
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH, USA
| | - Evan M Ratzan
- Departments of Otolaryngology and Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alisha J Beirl
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Roberto Aponte Rivera
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey M Lotthammer
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
| | - Sanket Walujkar
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
| | - Mark P Foster
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
- Biophysics Graduate Program, The Ohio State University, Columbus, OH, USA
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH, USA
| | - Omid A Zobeiri
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Jeffrey R Holt
- Departments of Otolaryngology and Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Saima Riazuddin
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kathleen E Cullen
- Departments of Biomedical Engineering, Neuroscience, and Otolaryngology and Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marcos Sotomayor
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
- Biophysics Graduate Program, The Ohio State University, Columbus, OH, USA
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH, USA
| | - Zubair M Ahmed
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
16
|
Slika E, Fuchs PA. Genetic tools for studying cochlear inhibition. Front Cell Neurosci 2024; 18:1372948. [PMID: 38560293 PMCID: PMC10978695 DOI: 10.3389/fncel.2024.1372948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 02/28/2024] [Indexed: 04/04/2024] Open
Abstract
Efferent feedback to the mammalian cochlea includes cholinergic medial olivocochlear neurons (MOCs) that release ACh to hyperpolarize and shunt the voltage change that drives electromotility of outer hair cells (OHCs). Via brainstem connectivity, MOCs are activated by sound in a frequency- and intensity-dependent manner, thereby reducing the amplification of cochlear vibration provided by OHC electromotility. Among other roles, this efferent feedback protects the cochlea from acoustic trauma. Lesion studies, as well as a variety of genetic mouse models, support the hypothesis of efferent protection from acoustic trauma. Genetic knockout and gain-of-function knockin of the unique α9α10-containing nicotinic acetylcholine receptor (nAChR) in hair cells show that acoustic protection correlates with the efficacy of cholinergic inhibition of OHCs. This protective effect was replicated by viral transduction of the gain-of-function α9L9'T nAChR into α9-knockout mice. Continued progress with "efferent gene therapy" will require a reliable method for visualizing nAChR expression in cochlear hair cells. To that end, mice expressing HA-tagged α9 or α10 nAChRs were generated using CRISPR technology. This progress will facilitate continued study of the hair cell nAChR as a therapeutic target to prevent hearing loss and potentially to ameliorate associated pathologies such as hyperacusis.
Collapse
Affiliation(s)
| | - Paul Albert Fuchs
- The Center for Hearing and Balance, Otolaryngology-Head and Neck Surgery, Johns Hopkins, University School of Medicine Baltimore, Baltimore, MD, United States
| |
Collapse
|
17
|
Lee JH, Perez-Flores MC, Park S, Kim HJ, Chen Y, Kang M, Kersigo J, Choi J, Thai PN, Woltz RL, Perez-Flores DC, Perkins G, Sihn CR, Trinh P, Zhang XD, Sirish P, Dong Y, Feng WW, Pessah IN, Dixon RE, Sokolowski B, Fritzsch B, Chiamvimonvat N, Yamoah EN. The Piezo channel is a mechano-sensitive complex component in the mammalian inner ear hair cell. Nat Commun 2024; 15:526. [PMID: 38228630 PMCID: PMC10791687 DOI: 10.1038/s41467-023-44230-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 12/04/2023] [Indexed: 01/18/2024] Open
Abstract
The inner ear is the hub where hair cells (HCs) transduce sound, gravity, and head acceleration stimuli to the brain. Hearing and balance rely on mechanosensation, the fastest sensory signals transmitted to the brain. The mechanoelectrical transducer (MET) channel is the entryway for the sound-balance-brain interface, but the channel-complex composition is not entirely known. Here, we report that the mouse utilizes Piezo1 (Pz1) and Piezo2 (Pz2) isoforms as MET-complex components. The Pz channels, expressed in HC stereocilia, and cell lines are co-localized and co-assembled with MET complex partners. Mice expressing non-functional Pz1 and Pz2 at the ROSA26 locus have impaired auditory and vestibular traits that can only be explained if the Pzs are integral to the MET complex. We suggest that Pz subunits constitute part of the MET complex and that interactions with other MET complex components yield functional MET units to generate HC MET currents.
Collapse
Affiliation(s)
- Jeong Han Lee
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Maria C Perez-Flores
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Seojin Park
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
- Prestige Biopharma, 11-12F, 44, Myongjigukje7-ro, Gangseo-gu, Busan, 67264, South Korea
| | - Hyo Jeong Kim
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Yingying Chen
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Mincheol Kang
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
- Prestige Biopharma, 11-12F, 44, Myongjigukje7-ro, Gangseo-gu, Busan, 67264, South Korea
| | | | - Jinsil Choi
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Phung N Thai
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA, 95616, USA
| | - Ryan L Woltz
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA, 95616, USA
| | | | - Guy Perkins
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA, 92093, USA
| | - Choong-Ryoul Sihn
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Pauline Trinh
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA, 95616, USA
| | - Xiao-Dong Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA, 95616, USA
| | - Padmini Sirish
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA, 95616, USA
| | - Yao Dong
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, 1089 VM3B, Davis, CA, 95616, USA
| | - Wayne Wei Feng
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, 1089 VM3B, Davis, CA, 95616, USA
| | - Isaac N Pessah
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, 1089 VM3B, Davis, CA, 95616, USA
| | - Rose E Dixon
- Department of Physiology & Membrane Biology, Tupper Hall, One Shields Avenue, Davis, CA, 95616, USA
| | - Bernd Sokolowski
- Department of Otolaryngology-Head and Neck Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, IA, USA
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA, 95616, USA
- VA Northern California Healthcare System, Sacramento, USA
| | - Ebenezer N Yamoah
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA.
| |
Collapse
|
18
|
Beurg M, Schwalbach ET, Fettiplace R. LHFPL5 is a key element in force transmission from the tip link to the hair cell mechanotransducer channel. Proc Natl Acad Sci U S A 2024; 121:e2318270121. [PMID: 38194445 PMCID: PMC10801851 DOI: 10.1073/pnas.2318270121] [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: 10/19/2023] [Accepted: 11/30/2023] [Indexed: 01/11/2024] Open
Abstract
During auditory transduction, sound-evoked vibrations of the hair cell stereociliary bundles open mechanotransducer (MET) ion channels via tip links extending from one stereocilium to its neighbor. How tension in the tip link is delivered to the channel is not fully understood. The MET channel comprises a pore-forming subunit, transmembrane channel-like protein (TMC1 or TMC2), aided by several accessory proteins, including LHFPL5 (lipoma HMGIC fusion partner-like 5). We investigated the role of LHFPL5 in transduction by comparing MET channel activation in outer hair cells of Lhfpl5-/- knockout mice with those in Lhfpl5+/- heterozygotes. The 10 to 90 percent working range of transduction in Tmc1+/+; Lhfpl5+/- was 52 nm, from which the single-channel gating force, Z, was evaluated as 0.34 pN. However, in Tmc1+/+; Lhfpl5-/- mice, the working range increased to 123 nm and Z more than halved to 0.13 pN, indicating reduced sensitivity. Tip link tension is thought to activate the channel via a gating spring, whose stiffness is inferred from the stiffness change on tip link destruction. The gating stiffness was ~40 percent of the total bundle stiffness in wild type but was virtually abolished in Lhfpl5-/-, implicating LHFPL5 as a principal component of the gating spring. The mutation Tmc1 p.D569N reduced the LHFPL5 immunolabeling in the stereocilia and like Lhfpl5-/- doubled the MET working range, but other deafness mutations had no effect on the dynamic range. We conclude that tip-link tension is transmitted to the channel primarily via LHFPL5; residual activation without LHFPL5 may occur by direct interaction between PCDH15 and TMC1.
Collapse
Affiliation(s)
- Maryline Beurg
- Department of Neuroscience, University of WisconsinSchool of Medicine and Public Health, Madison, WI53706
| | - Evan Travis Schwalbach
- Department of Neuroscience, University of WisconsinSchool of Medicine and Public Health, Madison, WI53706
| | - Robert Fettiplace
- Department of Neuroscience, University of WisconsinSchool of Medicine and Public Health, Madison, WI53706
| |
Collapse
|
19
|
Clark S, Mitra J, Elferich J, Goehring A, Ge J, Ha T, Gouaux E. Single molecule studies of the native hair cell mechanosensory transduction complex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.11.571162. [PMID: 38168376 PMCID: PMC10760052 DOI: 10.1101/2023.12.11.571162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Hearing and balance rely on the conversion of a mechanical stimulus into an electrical signal, a process known as mechanosensory transduction (MT). In vertebrates, this process is accomplished by an MT complex that is located in hair cells of the inner ear. While the past three decades of research have identified many subunits that are important for MT and revealed interactions between these subunits, the composition and organization of a functional complex remains unknown. The major challenge associated with studying the MT complex is its extremely low abundance in hair cells; current estimates of MT complex quantity range from 3-60 attomoles per cochlea or utricle, well below the detection limit of most biochemical assays that are used to characterize macromolecular complexes. Here we describe the optimization of two single molecule assays, single molecule pull-down (SiMPull) and single molecule array (SiMoA), to study the composition and quantity of native mouse MT complexes. We demonstrate that these assays are capable of detecting and quantifying low attomoles of the native MT subunits protocadherin-15 (PCDH15) and lipoma HMGIC fusion partner-like protein 5 (LHFPL5). Our results illuminate the stoichiometry of PCDH15- and LHFPL5-containing complexes and establish SiMPull and SiMoA as productive methods for probing the abundance, composition, and arrangement of subunits in the native MT complex.
Collapse
Affiliation(s)
- Sarah Clark
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
- Present address: Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Jaba Mitra
- Department of Materials Science and Engineering, University of Illinois Urbana-Champaign, IL 61801, USA
- Present address: Pacific Biosciences, Menlo Park, CA 94025, USA
| | - Johannes Elferich
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
- Present address: UMass Chan Medical School, Worcester, MA 01655, USA
| | - April Goehring
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Jingpeng Ge
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
- Present address: School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, 201210, China
| | - Taekjip Ha
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University, Baltimore, MD 21215, USA
- Present address: Howard Hughes Medical Institute, Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Present address: Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Eric Gouaux
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
- Howard Hughes Medical Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| |
Collapse
|
20
|
Cho SH, Yun Y, Lee DH, Cha JH, Lee SM, Lee J, Suh MH, Lee JH, Oh SH, Park MK, Lee SY. Novel autosomal dominant TMC1 variants linked to hearing loss: insight into protein-lipid interactions. BMC Med Genomics 2023; 16:320. [PMID: 38066485 PMCID: PMC10704677 DOI: 10.1186/s12920-023-01766-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 12/05/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND TMC1, which encodes transmembrane channel-like protein 1, forms the mechanoelectrical transduction (MET) channel in auditory hair cells, necessary for auditory function. TMC1 variants are known to cause autosomal dominant (DFNA36) and autosomal recessive (DFNB7/11) non-syndromic hearing loss, but only a handful of TMC1 variants underlying DFNA36 have been reported, hampering analysis of genotype-phenotype correlations. METHODS In this study, we retrospectively reviewed 338 probands in an in-house database of genetic hearing loss, evaluating the clinical phenotypes and genotypes of novel TMC1 variants associated with DFNA36. To analyze the structural impact of these variants, we generated two structural models of human TMC1, utilizing the Cryo-EM structure of C. elegans TMC1 as a template and AlphaFold protein structure database. Specifically, the lipid bilayer-embedded protein database was used to construct membrane-embedded models of TMC1. We then examined the effect of TMC1 variants on intramolecular interactions and predicted their potential pathogenicity. RESULTS We identified two novel TMC1 variants related to DFNA36 (c.1256T > C:p.Phe419Ser and c.1444T > C:p.Trp482Arg). The affected subjects had bilateral, moderate, late-onset, progressive sensorineural hearing loss with a down-sloping configuration. The Phe419 residue located in the transmembrane domain 4 of TMC1 faces outward towards the channel pore and is in close proximity to the hydrophobic tail of the lipid bilayer. The non-polar-to-polar variant (p.Phe419Ser) alters the hydrophobicity in the membrane, compromising protein-lipid interactions. On the other hand, the Trp482 residue located in the extracellular linker region between transmembrane domains 5 and 6 is anchored to the membrane interfaces via its aromatic rings, mediating several molecular interactions that stabilize the structure of TMC1. This type of aromatic ring-based anchoring is also observed in homologous transmembrane proteins such as OSCA1.2. Conversely, the substitution of Trp with Arg (Trp482Arg) disrupts the cation-π interaction with phospholipids located in the outer leaflet of the phospholipid bilayer, destabilizing protein-lipid interactions. Additionally, Trp482Arg collapses the CH-π interaction between Trp482 and Pro511, possibly reducing the overall stability of the protein. In parallel with the molecular modeling, the two mutants degraded significantly faster compared to the wild-type protein, compromising protein stability. CONCLUSIONS This results expand the genetic spectrum of disease-causing TMC1 variants related to DFNA36 and provide insight into TMC1 transmembrane protein-lipid interactions.
Collapse
Affiliation(s)
- Sung Ho Cho
- Seoul National University College of Medicine, Seoul, South Korea
| | - Yejin Yun
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Jongno-Gu, Daehak-Ro, 101, Seoul, South Korea
| | - Dae Hee Lee
- CTCELLS, Inc, 21, Yuseong-daero, 1205beon-gil, Yuseong-gu, Daejeon, Republic of Korea
| | - Joo Hyun Cha
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Jongno-Gu, Daehak-Ro, 101, Seoul, South Korea
| | - So Min Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Jongno-Gu, Daehak-Ro, 101, Seoul, South Korea
| | - Jehyun Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Jongno-Gu, Daehak-Ro, 101, Seoul, South Korea
| | - Myung Hwan Suh
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Jongno-Gu, Daehak-Ro, 101, Seoul, South Korea
| | - Jun Ho Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Jongno-Gu, Daehak-Ro, 101, Seoul, South Korea
| | - Seung-Ha Oh
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Jongno-Gu, Daehak-Ro, 101, Seoul, South Korea
| | - Moo Kyun Park
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Jongno-Gu, Daehak-Ro, 101, Seoul, South Korea.
- Sensory Organ Research Institute, Seoul National University Medical Research Center, Seoul, South Korea.
| | - Sang-Yeon Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Jongno-Gu, Daehak-Ro, 101, Seoul, South Korea.
- Sensory Organ Research Institute, Seoul National University Medical Research Center, Seoul, South Korea.
- Department of Genomic Medicine, Precision Medicine & Rare Disease Center, Seoul, South Korea.
| |
Collapse
|
21
|
Santiago C, Sharma N, Africawala N, Siegrist J, Handler A, Tasnim A, Anjum R, Turecek J, Lehnert BP, Renauld S, Nolan-Tamariz M, Iskols M, Magee AR, Paradis S, Ginty DD. Activity-dependent development of the body's touch receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.23.559109. [PMID: 37790437 PMCID: PMC10542488 DOI: 10.1101/2023.09.23.559109] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
We report a role for activity in the development of the primary sensory neurons that detect touch. Genetic deletion of Piezo2, the principal mechanosensitive ion channel in somatosensory neurons, caused profound changes in the formation of mechanosensory end organ structures and altered somatosensory neuron central targeting. Single cell RNA sequencing of Piezo2 conditional mutants revealed changes in gene expression in the sensory neurons activated by light mechanical forces, whereas other neuronal classes were less affected. To further test the role of activity in mechanosensory end organ development, we genetically deleted the voltage-gated sodium channel Nav1.6 (Scn8a) in somatosensory neurons throughout development and found that Scn8a mutants also have disrupted somatosensory neuron morphologies and altered electrophysiological responses to mechanical stimuli. Together, these findings indicate that mechanically evoked neuronal activity acts early in life to shape the maturation of the mechanosensory end organs that underlie our sense of gentle touch.
Collapse
Affiliation(s)
- Celine Santiago
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Nikhil Sharma
- Department of Molecular Pharmacology and Therapeutics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Nusrat Africawala
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Julianna Siegrist
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Annie Handler
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Aniqa Tasnim
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Rabia Anjum
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, MA, 02453, USA
| | - Josef Turecek
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Brendan P. Lehnert
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Sophia Renauld
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Michael Nolan-Tamariz
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Michael Iskols
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Alexandra R. Magee
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Suzanne Paradis
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, MA, 02453, USA
| | - David D. Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA
- Lead Contact
| |
Collapse
|
22
|
Park J, Bird JE. The actin cytoskeleton in hair bundle development and hearing loss. Hear Res 2023; 436:108817. [PMID: 37300948 PMCID: PMC10408727 DOI: 10.1016/j.heares.2023.108817] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/18/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023]
Abstract
Inner ear hair cells assemble mechanosensitive hair bundles on their apical surface that transduce sounds and accelerations. Each hair bundle is comprised of ∼ 100 individual stereocilia that are arranged into rows of increasing height and width; their specific and precise architecture being necessary for mechanoelectrical transduction (MET). The actin cytoskeleton is fundamental to establishing this architecture, not only by forming the structural scaffold shaping each stereocilium, but also by composing rootlets and the cuticular plate that together provide a stable foundation supporting each stereocilium. In concert with the actin cytoskeleton, a large assortment of actin-binding proteins (ABPs) function to cross-link actin filaments into specific topologies, as well as control actin filament growth, severing, and capping. These processes are individually critical for sensory transduction and are all disrupted in hereditary forms of human hearing loss. In this review, we provide an overview of actin-based structures in the hair bundle and the molecules contributing to their assembly and functional properties. We also highlight recent advances in mechanisms driving stereocilia elongation and how these processes are tuned by MET.
Collapse
Affiliation(s)
- Jinho Park
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, United States; Myology Institute, University of Florida, Gainesville, FL 32610, United States
| | - Jonathan E Bird
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, United States; Myology Institute, University of Florida, Gainesville, FL 32610, United States.
| |
Collapse
|
23
|
Aristizábal-Ramírez I, Dragich AK, Giese APJ, Sofia Zuluaga-Osorio K, Watkins J, Davies GK, Hadi SE, Riazuddin S, Vander Kooi CW, Ahmed ZM, Frolenkov GI. Calcium and Integrin-binding protein 2 (CIB2) controls force sensitivity of the mechanotransducer channels in cochlear outer hair cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.09.545606. [PMID: 37461484 PMCID: PMC10350036 DOI: 10.1101/2023.07.09.545606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Calcium and Integrin-Binding Protein 2 (CIB2) is an essential subunit of the mechano-electrical transduction (MET) complex in mammalian auditory hair cells. CIB2 binds to pore-forming subunits of the MET channel, TMC1/2 and is required for their transport and/or retention at the tips of mechanosensory stereocilia. Since genetic ablation of CIB2 results in complete loss of MET currents, the exact role of CIB2 in the MET complex remains elusive. Here, we generated a new mouse strain with deafness-causing p.R186W mutation in Cib2 and recorded small but still measurable MET currents in the cochlear outer hair cells. We found that R186W variant causes increase of the resting open probability of MET channels, steeper MET current dependence on hair bundle deflection (I-X curve), loss of fast adaptation, and increased leftward shifts of I-X curves upon hair cell depolarization. Combined with AlphaFold2 prediction that R186W disrupts one of the multiple interacting sites between CIB2 and TMC1/2, our data suggest that CIB2 mechanically constraints TMC1/2 conformations to ensure proper force sensitivity and dynamic range of the MET channels. Using a custom piezo-driven stiff probe deflecting the hair bundles in less than 10 µs, we also found that R186W variant slows down the activation of MET channels. This phenomenon, however, is unlikely to be due to direct effect on MET channels, since we also observed R186W-evoked disruption of the electron-dense material at the tips of mechanotransducing stereocilia and the loss of membrane-shaping BAIAP2L2 protein from the same location. We concluded that R186W variant of CIB2 disrupts force sensitivity of the MET channels and force transmission to these channels.
Collapse
|
24
|
Hakizimana P. The summating potential polarity encodes the ear health condition. Cell Mol Life Sci 2023; 80:163. [PMID: 37225973 DOI: 10.1007/s00018-023-04809-5] [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: 01/18/2023] [Revised: 05/10/2023] [Accepted: 05/14/2023] [Indexed: 05/26/2023]
Abstract
The summating potential (SP), the DC potential which, along with the AC response, is produced when the hair cells convert the vibrational mechanical energy of sound into electrical signals, is the most enigmatic of the cochlear potentials because its polarity and function have remained elusive for more than seven decades. Despite the tremendous socioeconomic consequences of noise-induced hearing loss and the profound physiological importance of understanding how loud noise exposure impairs the hair cell receptor activation, the relationship between the SP and noise-induced hearing impairment remains poorly characterized. Here, I show that in normally hearing ears, the SP polarity is positive and its amplitude relative to the AC response grows exponentially across frequencies, and becomes negative and decreases exponentially across frequencies following noise-induced hearing injury. Since the SP is thought to be generated by K+ outflow down the gradient through the hair cell basolateral K+ channels, the SP polarity switch to negative values is consistent with a noise-induced shift in the operating point of the hair cells.
Collapse
Affiliation(s)
- Pierre Hakizimana
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, 581 83, Linköping, Sweden.
| |
Collapse
|
25
|
Krey JF, Chatterjee P, Halford J, Cunningham CL, Perrin BJ, Barr-Gillespie PG. Control of stereocilia length during development of hair bundles. PLoS Biol 2023; 21:e3001964. [PMID: 37011103 PMCID: PMC10101650 DOI: 10.1371/journal.pbio.3001964] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 04/13/2023] [Accepted: 03/14/2023] [Indexed: 04/05/2023] Open
Abstract
Assembly of the hair bundle, the sensory organelle of the inner ear, depends on differential growth of actin-based stereocilia. Separate rows of stereocilia, labeled 1 through 3 from tallest to shortest, lengthen or shorten during discrete time intervals during development. We used lattice structured illumination microscopy and surface rendering to measure dimensions of stereocilia from mouse apical inner hair cells during early postnatal development; these measurements revealed a sharp transition at postnatal day 8 between stage III (row 1 and 2 widening; row 2 shortening) and stage IV (final row 1 lengthening and widening). Tip proteins that determine row 1 lengthening did not accumulate simultaneously during stages III and IV; while the actin-bundling protein EPS8 peaked at the end of stage III, GNAI3 peaked several days later-in early stage IV-and GPSM2 peaked near the end of stage IV. To establish the contributions of key macromolecular assemblies to bundle structure, we examined mouse mutants that eliminated tip links (Cdh23v2J or Pcdh15av3J), transduction channels (TmieKO), or the row 1 tip complex (Myo15ash2). Cdh23v2J/v2J and Pcdh15av3J/av3J bundles had adjacent stereocilia in the same row that were not matched in length, revealing that a major role of these cadherins is to synchronize lengths of side-by-side stereocilia. Use of the tip-link mutants also allowed us to distinguish the role of transduction from effects of transduction proteins themselves. While levels of GNAI3 and GPSM2, which stimulate stereocilia elongation, were greatly attenuated at the tips of TmieKO/KO row 1 stereocilia, they accumulated normally in Cdh23v2J/v2J and Pcdh15av3J/av3J stereocilia. These results reinforced the suggestion that the transduction proteins themselves facilitate localization of proteins in the row 1 complex. By contrast, EPS8 concentrates at tips of all TmieKO/KO, Cdh23v2J/v2J, and Pcdh15av3J/av3J stereocilia, correlating with the less polarized distribution of stereocilia lengths in these bundles. These latter results indicated that in wild-type hair cells, the transduction complex prevents accumulation of EPS8 at the tips of shorter stereocilia, causing them to shrink (rows 2 and 3) or disappear (row 4 and microvilli). Reduced rhodamine-actin labeling at row 2 stereocilia tips of tip-link and transduction mutants suggests that transduction's role is to destabilize actin filaments there. These results suggest that regulation of stereocilia length occurs through EPS8 and that CDH23 and PCDH15 regulate stereocilia lengthening beyond their role in gating mechanotransduction channels.
Collapse
Affiliation(s)
- Jocelyn F. Krey
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Paroma Chatterjee
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Julia Halford
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Christopher L. Cunningham
- Pittsburgh Hearing Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Benjamin J. Perrin
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| | - Peter G. Barr-Gillespie
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| |
Collapse
|
26
|
The tetraspan LHFPL5 is critical to establish maximal force sensitivity of the mechanotransduction channel of cochlear hair cells. Cell Rep 2023; 42:112245. [PMID: 36917610 DOI: 10.1016/j.celrep.2023.112245] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/28/2023] [Accepted: 02/27/2023] [Indexed: 03/14/2023] Open
Abstract
The mechanoelectrical transduction (MET) channel of cochlear hair cells is gated by the tip link, but the mechanisms that establish the exquisite force sensitivity of this MET channel are not known. Here, we show that the tetraspan lipoma HMGIC fusion partner-like 5 (LHFPL5) directly couples the tip link to the MET channel. Disruption of these interactions severely perturbs MET. Notably, the N-terminal cytoplasmic domain of LHFPL5 binds to an amphipathic helix in TMC1, a critical gating domain conserved between different MET channels. Mutations in the amphipathic helix of TMC1 or in the N-terminus of LHFPL5 that perturb interactions of LHFPL5 with the amphipathic helix affect channel responses to mechanical force. We conclude that LHFPL5 couples the tip link to the MET channel and that channel gating depends on a structural element in TMC1 that is evolutionarily conserved between MET channels. Overall, our findings support a tether model for transduction channel gating by the tip link.
Collapse
|
27
|
Qiu X, Müller U. Sensing sound: Cellular specializations and molecular force sensors. Neuron 2022; 110:3667-3687. [PMID: 36223766 PMCID: PMC9671866 DOI: 10.1016/j.neuron.2022.09.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/03/2022] [Accepted: 09/14/2022] [Indexed: 11/08/2022]
Abstract
Organisms of all phyla express mechanosensitive ion channels with a wide range of physiological functions. In recent years, several classes of mechanically gated ion channels have been identified. Some of these ion channels are intrinsically mechanosensitive. Others depend on accessory proteins to regulate their response to mechanical force. The mechanotransduction machinery of cochlear hair cells provides a particularly striking example of a complex force-sensing machine. This molecular ensemble is embedded into a specialized cellular compartment that is crucial for its function. Notably, mechanotransduction channels of cochlear hair cells are not only critical for auditory perception. They also shape their cellular environment and regulate the development of auditory circuitry. Here, we summarize recent discoveries that have shed light on the composition of the mechanotransduction machinery of cochlear hair cells and how this machinery contributes to the development and function of the auditory system.
Collapse
Affiliation(s)
- Xufeng Qiu
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ulrich Müller
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
28
|
The conductance and organization of the TMC1-containing mechanotransducer channel complex in auditory hair cells. Proc Natl Acad Sci U S A 2022; 119:e2210849119. [PMID: 36191207 PMCID: PMC9564823 DOI: 10.1073/pnas.2210849119] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We studied the role of TMC1 as the central component of the hair cell mechanotransducer (MET) channel by characterizing transduction in mice harboring mutations in the pore region. All Tmc1 mutations reduced the Ca2+ influx into the hair bundle. Two mutations (Tmc1 p.D528N or Tmc1 p.E520Q) also decreased channel conductance and two (Tmc1 p. D569N or Tmc1 p.W554L) lowered expression. These mutations endorse TMC1 as the pore of the MET channel. The MET channel also contains accessory subunits, LHFPL5 and TMIE. MET currents were small in Lhfpl5 or Tmie knockout mice. Nevertheless, MET channels could still be activated by hair bundle displacement; single-channel conductance was unaffected in Lhfpl5−/− but reduced in Tmie−/−, suggesting TMIE likely contributes to the pore. Transmembrane channel-like protein 1 (TMC1) is thought to form the ion-conducting pore of the mechanoelectrical transducer (MET) channel in auditory hair cells. Using single-channel analysis and ionic permeability measurements, we characterized six missense mutations in the purported pore region of mouse TMC1. All mutations reduced the Ca2+ permeability of the MET channel, triggering hair cell apoptosis and deafness. In addition, Tmc1 p.E520Q and Tmc1 p.D528N reduced channel conductance, whereas Tmc1 p.W554L and Tmc1 p.D569N lowered channel expression without affecting the conductance. Tmc1 p.M412K and Tmc1 p.T416K reduced only the Ca2+ permeability. The consequences of these mutations endorse TMC1 as the pore of the MET channel. The accessory subunits, LHFPL5 and TMIE, are thought to be involved in targeting TMC1 to the tips of the stereocilia. We found sufficient expression of TMC1 in outer hair cells of Lhfpl5 and Tmie knockout mice to determine the properties of the channels, which could still be gated by hair bundle displacement. Single-channel conductance was unaffected in Lhfpl5−/− but was reduced in Tmie−/−, implying TMIE very likely contributes to the pore. Both the working range and half-saturation point of the residual MET current in Lhfpl5−/− were substantially increased, suggesting that LHFPL5 is part of the mechanical coupling between the tip-link and the MET channel. Based on counts of numbers of stereocilia per bundle, we estimate that each PCDH15 and LHFPL5 monomer may contact two channels irrespective of location.
Collapse
|
29
|
Ballesteros A, Swartz KJ. Regulation of membrane homeostasis by TMC1 mechanoelectrical transduction channels is essential for hearing. SCIENCE ADVANCES 2022; 8:eabm5550. [PMID: 35921424 PMCID: PMC9348795 DOI: 10.1126/sciadv.abm5550] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
The mechanoelectrical transduction (MET) channel in auditory hair cells converts sound into electrical signals, enabling hearing. Transmembrane-like channel 1 and 2 (TMC1 and TMC2) are implicated in forming the pore of the MET channel. Here, we demonstrate that inhibition of MET channels, breakage of the tip links required for MET, or buffering of intracellular Ca... induces pronounced phosphatidylserine externalization, membrane blebbing, and ectosome release at the hair cell sensory organelle, culminating in the loss of TMC1. Membrane homeostasis triggered by MET channel inhibition requires Tmc1 but not Tmc2, and three deafness-causing mutations in Tmc1 cause constitutive phosphatidylserine externalization that correlates with deafness phenotype. Our results suggest that, in addition to forming the pore of the MET channel, TMC1 is a critical regulator of membrane homeostasis in hair cells, and that Tmc1-related hearing loss may involve alterations in membrane homeostasis.
Collapse
|
30
|
Caprara GA, Peng AW. Mechanotransduction in mammalian sensory hair cells. Mol Cell Neurosci 2022; 120:103706. [PMID: 35218890 PMCID: PMC9177625 DOI: 10.1016/j.mcn.2022.103706] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 02/14/2022] [Accepted: 02/18/2022] [Indexed: 11/23/2022] Open
Abstract
In the inner ear, the auditory and vestibular systems detect and translate sensory information regarding sound and balance. The sensory cells that transform mechanical input into an electrical signal in these systems are called hair cells. A specialized organelle on the apical surface of hair cells called the hair bundle detects mechanical signals. Displacement of the hair bundle causes mechanotransduction channels to open. The morphology and organization of the hair bundle, as well as the properties and characteristics of the mechanotransduction process, differ between the different hair cell types in the auditory and vestibular systems. These differences likely contribute to maximizing the transduction of specific signals in each system. This review will discuss the molecules essential for mechanotransduction and the properties of the mechanotransduction process, focusing our attention on recent data and differences between the auditory and vestibular systems.
Collapse
Affiliation(s)
- Giusy A Caprara
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - Anthony W Peng
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America.
| |
Collapse
|
31
|
Signatures of cochlear processing in neuronal coding of auditory information. Mol Cell Neurosci 2022; 120:103732. [PMID: 35489636 DOI: 10.1016/j.mcn.2022.103732] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 11/22/2022] Open
Abstract
The vertebrate ear is endowed with remarkable perceptual capabilities. The faintest sounds produce vibrations of magnitudes comparable to those generated by thermal noise and can nonetheless be detected through efficient amplification of small acoustic stimuli. Two mechanisms have been proposed to underlie such sound amplification in the mammalian cochlea: somatic electromotility and active hair-bundle motility. These biomechanical mechanisms may work in concert to tune auditory sensitivity. In addition to amplitude sensitivity, the hearing system shows exceptional frequency discrimination allowing mammals to distinguish complex sounds with great accuracy. For instance, although the wide hearing range of humans encompasses frequencies from 20 Hz to 20 kHz, our frequency resolution extends to one-thirtieth of the interval between successive keys on a piano. In this article, we review the different cochlear mechanisms underlying sound encoding in the auditory system, with a particular focus on the frequency decomposition of sounds. The relation between peak frequency of activation and location along the cochlea - known as tonotopy - arises from multiple gradients in biophysical properties of the sensory epithelium. Tonotopic mapping represents a major organizational principle both in the peripheral hearing system and in higher processing levels and permits the spectral decomposition of complex tones. The ribbon synapses connecting sensory hair cells to auditory afferents and the downstream spiral ganglion neurons are also tuned to process periodic stimuli according to their preferred frequency. Though sensory hair cells and neurons necessarily filter signals beyond a few kHz, many animals can hear well beyond this range. We finally describe how the cochlear structure shapes the neural code for further processing in order to send meaningful information to the brain. Both the phase-locked response of auditory nerve fibers and tonotopy are key to decode sound frequency information and place specific constraints on the downstream neuronal network.
Collapse
|
32
|
In vivo real-time imaging reveals megalin as the aminoglycoside gentamicin transporter into cochlea whose inhibition is otoprotective. Proc Natl Acad Sci U S A 2022; 119:2117946119. [PMID: 35197290 PMCID: PMC8892513 DOI: 10.1073/pnas.2117946119] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2022] [Indexed: 01/01/2023] Open
Abstract
Aminoglycosides (AGs) are commonly used antibiotics that cause deafness through the irreversible loss of cochlear sensory hair cells (HCs). How AGs enter the cochlea and then target HCs remains unresolved. Here, we performed time-lapse multicellular imaging of cochlea in live adult hearing mice via a chemo-mechanical cochleostomy. The in vivo tracking revealed that systemically administered Texas Red-labeled gentamicin (GTTR) enters the cochlea via the stria vascularis and then HCs selectively. GTTR uptake into HCs was completely abolished in transmembrane channel-like protein 1 (TMC1) knockout mice, indicating mechanotransducer channel-dependent AG uptake. Blockage of megalin, the candidate AG transporter in the stria vascularis, by binding competitor cilastatin prevented GTTR accumulation in HCs. Furthermore, cilastatin treatment markedly reduced AG-induced HC degeneration and hearing loss in vivo. Together, our in vivo real-time tracking of megalin-dependent AG transport across the blood-labyrinth barrier identifies new therapeutic targets for preventing AG-induced ototoxicity.
Collapse
|
33
|
Jeong H, Clark S, Goehring A, Dehghani-Ghahnaviyeh S, Rasouli A, Tajkhorshid E, Gouaux E. Structures of the TMC-1 complex illuminate mechanosensory transduction. Nature 2022; 610:796-803. [PMID: 36224384 PMCID: PMC9605866 DOI: 10.1038/s41586-022-05314-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/02/2022] [Indexed: 02/05/2023]
Abstract
The initial step in the sensory transduction pathway underpinning hearing and balance in mammals involves the conversion of force into the gating of a mechanosensory transduction channel1. Despite the profound socioeconomic impacts of hearing disorders and the fundamental biological significance of understanding mechanosensory transduction, the composition, structure and mechanism of the mechanosensory transduction complex have remained poorly characterized. Here we report the single-particle cryo-electron microscopy structure of the native transmembrane channel-like protein 1 (TMC-1) mechanosensory transduction complex isolated from Caenorhabditis elegans. The two-fold symmetric complex is composed of two copies each of the pore-forming TMC-1 subunit, the calcium-binding protein CALM-1 and the transmembrane inner ear protein TMIE. CALM-1 makes extensive contacts with the cytoplasmic face of the TMC-1 subunits, whereas the single-pass TMIE subunits reside on the periphery of the complex, poised like the handles of an accordion. A subset of complexes additionally includes a single arrestin-like protein, arrestin domain protein (ARRD-6), bound to a CALM-1 subunit. Single-particle reconstructions and molecular dynamics simulations show how the mechanosensory transduction complex deforms the membrane bilayer and suggest crucial roles for lipid-protein interactions in the mechanism by which mechanical force is transduced to ion channel gating.
Collapse
Affiliation(s)
- Hanbin Jeong
- grid.433851.80000 0004 0608 3919Vollum Institute, Oregon Health and Science University, Portland, OR USA
| | - Sarah Clark
- grid.433851.80000 0004 0608 3919Vollum Institute, Oregon Health and Science University, Portland, OR USA
| | - April Goehring
- grid.433851.80000 0004 0608 3919Vollum Institute, Oregon Health and Science University, Portland, OR USA ,grid.5288.70000 0000 9758 5690Howard Hughes Medical Institute, Oregon Health and Science University, Portland, OR USA
| | - Sepehr Dehghani-Ghahnaviyeh
- grid.35403.310000 0004 1936 9991Theoretical and Computational Biophysics Group, NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL USA ,grid.35403.310000 0004 1936 9991Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL USA ,grid.35403.310000 0004 1936 9991Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL USA
| | - Ali Rasouli
- grid.35403.310000 0004 1936 9991Theoretical and Computational Biophysics Group, NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL USA ,grid.35403.310000 0004 1936 9991Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL USA ,grid.35403.310000 0004 1936 9991Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL USA
| | - Emad Tajkhorshid
- grid.35403.310000 0004 1936 9991Theoretical and Computational Biophysics Group, NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL USA ,grid.35403.310000 0004 1936 9991Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL USA ,grid.35403.310000 0004 1936 9991Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL USA
| | - Eric Gouaux
- grid.433851.80000 0004 0608 3919Vollum Institute, Oregon Health and Science University, Portland, OR USA ,grid.5288.70000 0000 9758 5690Howard Hughes Medical Institute, Oregon Health and Science University, Portland, OR USA
| |
Collapse
|
34
|
Elferich J, Clark S, Ge J, Goehring A, Matsui A, Gouaux E. Molecular structures and conformations of protocadherin-15 and its complexes on stereocilia elucidated by cryo-electron tomography. eLife 2021; 10:74512. [PMID: 34964715 PMCID: PMC8776254 DOI: 10.7554/elife.74512] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/27/2021] [Indexed: 11/23/2022] Open
Abstract
Mechanosensory transduction (MT), the conversion of mechanical stimuli into electrical signals, underpins hearing and balance and is carried out within hair cells in the inner ear. Hair cells harbor actin-filled stereocilia, arranged in rows of descending heights, where the tips of stereocilia are connected to their taller neighbors by a filament composed of protocadherin 15 (PCDH15) and cadherin 23 (CDH23), deemed the ‘tip link.’ Tension exerted on the tip link opens an ion channel at the tip of the shorter stereocilia, thus converting mechanical force into an electrical signal. While biochemical and structural studies have provided insights into the molecular composition and structure of isolated portions of the tip link, the architecture, location, and conformational states of intact tip links, on stereocilia, remains unknown. Here, we report in situ cryo-electron microscopy imaging of the tip link in mouse stereocilia. We observe individual PCDH15 molecules at the tip and shaft of stereocilia and determine their stoichiometry, conformational heterogeneity, and their complexes with other filamentous proteins, perhaps including CDH23. The PCDH15 complexes occur in clusters, frequently with more than one copy of PCDH15 at the tip of stereocilia, suggesting that tip links might consist of more than one copy of PCDH15 complexes and, by extension, might include multiple MT complexes.
Collapse
Affiliation(s)
- Johannes Elferich
- Vollum Institute, Oregon Health and Science University, Portland, United States
| | - Sarah Clark
- Vollum Institute, Oregon Health and Science University, Portland, United States
| | - Jingpeng Ge
- Vollum Institute, Oregon Health and Science University, Portland, United States
| | - April Goehring
- Vollum Institute, Oregon Health and Science University, Portland, United States
| | - Aya Matsui
- Vollum Institute, Oregon Health and Science University, Portland, United States
| | - Eric Gouaux
- Vollum Institute, Oregon Health and Science University, Portland, United States
| |
Collapse
|
35
|
Miller KK, Atkinson P, Mendoza KR, Ó Maoiléidigh D, Grillet N. Dimensions of a Living Cochlear Hair Bundle. Front Cell Dev Biol 2021; 9:742529. [PMID: 34900993 PMCID: PMC8657763 DOI: 10.3389/fcell.2021.742529] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 10/13/2021] [Indexed: 11/23/2022] Open
Abstract
The hair bundle is the mechanosensory organelle of hair cells that detects mechanical stimuli caused by sounds, head motions, and fluid flows. Each hair bundle is an assembly of cellular-protrusions called stereocilia, which differ in height to form a staircase. Stereocilia have different heights, widths, and separations in different species, sensory organs, positions within an organ, hair-cell types, and even within a single hair bundle. The dimensions of the stereociliary assembly dictate how the hair bundle responds to stimuli. These hair-bundle properties have been measured previously only to a limited degree. In particular, mammalian data are either incomplete, lack control for age or position within an organ, or have artifacts owing to fixation or dehydration. Here, we provide a complete set of measurements for postnatal day (P) 11 C57BL/6J mouse apical inner hair cells (IHCs) obtained from living tissue, tissue mildly-fixed for fluorescent imaging, or tissue strongly fixed and dehydrated for scanning electronic microscopy (SEM). We found that hair bundles mildly-fixed for fluorescence had the same dimensions as living hair bundles, whereas SEM-prepared hair bundles shrank uniformly in stereociliary heights, widths, and separations. By determining the shrinkage factors, we imputed live dimensions from SEM that were too small to observe optically. Accordingly, we created the first complete blueprint of a living IHC hair bundle. We show that SEM-prepared measurements strongly affect calculations of a bundle’s mechanical properties – overestimating stereociliary deflection stiffness and underestimating the fluid coupling between stereocilia. The methods of measurement, the data, and the consequences we describe illustrate the high levels of accuracy and precision required to understand hair-bundle mechanotransduction.
Collapse
Affiliation(s)
- Katharine K Miller
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University, Stanford, CA, United States
| | - Patrick Atkinson
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University, Stanford, CA, United States
| | - Kyssia Ruth Mendoza
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University, Stanford, CA, United States
| | - Dáibhid Ó Maoiléidigh
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University, Stanford, CA, United States
| | - Nicolas Grillet
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|
36
|
Ramkumar V, Mukherjea D, Dhukhwa A, Rybak LP. Oxidative Stress and Inflammation Caused by Cisplatin Ototoxicity. Antioxidants (Basel) 2021; 10:antiox10121919. [PMID: 34943021 PMCID: PMC8750101 DOI: 10.3390/antiox10121919] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/22/2021] [Accepted: 10/28/2021] [Indexed: 02/06/2023] Open
Abstract
Hearing loss is a significant health problem that can result from a variety of exogenous insults that generate oxidative stress and inflammation. This can produce cellular damage and impairment of hearing. Radiation damage, ageing, damage produced by cochlear implantation, acoustic trauma and ototoxic drug exposure can all generate reactive oxygen species in the inner ear with loss of sensory cells and hearing loss. Cisplatin ototoxicity is one of the major causes of hearing loss in children and adults. This review will address cisplatin ototoxicity. It includes discussion of the mechanisms associated with cisplatin-induced hearing loss including uptake pathways for cisplatin entry, oxidative stress due to overpowering antioxidant defense mechanisms, and the recently described toxic pathways that are activated by cisplatin, including necroptosis and ferroptosis. The cochlea contains G-protein coupled receptors that can be activated to provide protection. These include adenosine A1 receptors, cannabinoid 2 receptors (CB2) and the Sphingosine 1-Phosphate Receptor 2 (S1PR2). A variety of heat shock proteins (HSPs) can be up-regulated in the cochlea. The use of exosomes offers a novel method of delivery of HSPs to provide protection. A reversible MET channel blocker that can be administered orally may block cisplatin uptake into the cochlear cells. Several protective agents in preclinical studies have been shown to not interfere with cisplatin efficacy. Statins have shown efficacy in reducing cisplatin ototoxicity without compromising patient response to treatment. Additional clinical trials could provide exciting findings in the prevention of cisplatin ototoxicity.
Collapse
Affiliation(s)
- Vickram Ramkumar
- Department of Pharmacology, School of Medicine, Southern Illinois University, 801 N. Rutledge Street, Springfield, IL 62702, USA; (V.R.); (A.D.)
| | - Debashree Mukherjea
- Department of Otolaryngology, School of Medicine, Southern Illinois University, 801 N. Rutledge Street, Springfield, IL 62702, USA;
| | - Asmita Dhukhwa
- Department of Pharmacology, School of Medicine, Southern Illinois University, 801 N. Rutledge Street, Springfield, IL 62702, USA; (V.R.); (A.D.)
| | - Leonard P. Rybak
- Department of Otolaryngology, School of Medicine, Southern Illinois University, 801 N. Rutledge Street, Springfield, IL 62702, USA;
- Correspondence: ; Fax: +1-217-545-6544
| |
Collapse
|
37
|
Beurg M, Nam JH, Fettiplace R. The speed of the hair cell mechanotransducer channel revealed by fluctuation analysis. J Gen Physiol 2021; 153:212584. [PMID: 34411238 PMCID: PMC8383808 DOI: 10.1085/jgp.202112959] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/01/2021] [Indexed: 11/22/2022] Open
Abstract
Although mechanoelectrical transducer (MET) channels have been extensively studied, uncertainty persists about their molecular architecture and single-channel conductance. We made electrical measurements from mouse cochlear outer hair cells (OHCs) to reexamine the MET channel conductance comparing two different methods. Analysis of fluctuations in the macroscopic currents showed that the channel conductance in apical OHCs determined from nonstationary noise analysis was about half that of single-channel events recorded after tip link destruction. We hypothesized that this difference reflects a bandwidth limitation in the noise analysis, which we tested by simulations of stochastic fluctuations in modeled channels. Modeling indicated that the unitary conductance depended on the relative values of the channel activation time constant and the applied low-pass filter frequency. The modeling enabled the activation time constant of the channel to be estimated for the first time, yielding a value of only a few microseconds. We found that the channel conductance, assayed with both noise and recording of single-channel events, was reduced by a third in a new deafness mutant, Tmc1 p.D528N. Our results indicate that noise analysis is likely to underestimate MET channel amplitude, which is better characterized from recordings of single-channel events.
Collapse
Affiliation(s)
- Maryline Beurg
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Jong-Hoon Nam
- Departments of Mechanical Engineering and Biomechanical Engineering, University of Rochester, Rochester, NY
| | - Robert Fettiplace
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, WI
| |
Collapse
|
38
|
Koo H, Hwang JY, Jung S, Park H, Bok J, Park JW. Position Specific Alternative Splicing and Gene Expression Profiles Along the Tonotopic Axis of Chick Cochlea. Front Mol Biosci 2021; 8:726976. [PMID: 34568429 PMCID: PMC8456117 DOI: 10.3389/fmolb.2021.726976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/25/2021] [Indexed: 11/17/2022] Open
Abstract
Alternative splicing (AS) refers to the production of multiple mRNA isoforms from a single gene due to alternative selection of exons or splice sites during pre-mRNA splicing. It is a primary mechanism of gene regulation in higher eukaryotes and significantly expands the functional complexity of eukaryotic organisms, contributing to animal development and disease. Recent studies have shown that AS also influences functional diversity by affecting the transcriptomic and proteomic profiles in a position-dependent manner in a single organ. The peripheral hearing organ, the cochlea, is organized to detect sounds at different frequencies depending on its location along the longitudinal axis. This unique functional configuration, the tonotopy, is known to be facilitated by differential gene expression along the cochlear duct. We profiled transcriptome-wide gene expression and AS changes that occur within the different positions of chick cochlea. These analyses revealed distinct gene expression profiles and AS, including a splicing program that is unique to tonotopy. Changes in the expression of splicing factors PTBP3, ESRP1, and ESRP2 were demonstrated to contribute to position-specific AS. RNA-binding motif enrichment analysis near alternatively spliced exons provided further insight into the combinatorial regulation of AS at different positions by different RNA-binding proteins. These data, along with gene ontology (GO) analysis, represent a comprehensive analysis of the dynamic regulation of AS at different positions in chick cochlea.
Collapse
Affiliation(s)
- Heiyeun Koo
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
- BK21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Jae Yeon Hwang
- Department of Computer Science and Engineering, University of Louisville, Louisville, KY, United States
| | - Sungbo Jung
- Department of Computer Science and Engineering, University of Louisville, Louisville, KY, United States
| | - Hyeyoung Park
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
| | - Jinwoong Bok
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
- BK21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, South Korea
| | - Juw Won Park
- Department of Computer Science and Engineering, University of Louisville, Louisville, KY, United States
- KY INBRE Bioinformatics Core, University of Louisville, Louisville, KY, United States
| |
Collapse
|
39
|
Chen Z, Zhu S, Kindig K, Wang S, Chou SW, Davis RW, Dercoli MR, Weaver H, Stepanyan R, McDermott BM. Tmc proteins are essential for zebrafish hearing where Tmc1 is not obligatory. Hum Mol Genet 2021; 29:2004-2021. [PMID: 32167554 DOI: 10.1093/hmg/ddaa045] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 02/13/2020] [Indexed: 12/19/2022] Open
Abstract
Perception of sound is initiated by mechanically gated ion channels at the tips of stereocilia. Mature mammalian auditory hair cells require transmembrane channel-like 1 (TMC1) for mechanotransduction, and mutations of the cognate genetic sequences result in dominant or recessive heritable deafness forms in humans and mice. In contrast, zebrafish lateral line hair cells, which detect water motion, require Tmc2a and Tmc2b. Here, we use standard and multiplex genome editing in conjunction with functional and behavioral assays to determine the reliance of zebrafish hearing and vestibular organs on Tmc proteins. Surprisingly, our approach using multiple mutant alleles demonstrates that hearing in zebrafish is not dependent on Tmc1, nor is it fully dependent on Tmc2a and Tmc2b. Hearing however is absent in triple-mutant zebrafish that lack Tmc1, Tmc2a and Tmc2b. These outcomes reveal a striking resemblance of Tmc protein reliance in the vestibular sensory epithelia of mammals to the maculae of zebrafish. Moreover, our findings disclose a logic of Tmc use where hearing depends on a complement of Tmc proteins beyond those employed to sense water motion.
Collapse
Affiliation(s)
- Zongwei Chen
- Department of Otolaryngology-Head and Neck Surgery, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.,Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Shaoyuan Zhu
- Department of Otolaryngology-Head and Neck Surgery, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.,Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Kayla Kindig
- Department of Otolaryngology-Head and Neck Surgery, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.,Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Shengxuan Wang
- Department of Otolaryngology-Head and Neck Surgery, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.,Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Shih-Wei Chou
- Department of Otolaryngology-Head and Neck Surgery, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.,Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Robin Woods Davis
- Department of Otolaryngology-Head and Neck Surgery, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.,Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Michael R Dercoli
- Department of Otolaryngology-Head and Neck Surgery, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.,Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Hannah Weaver
- Department of Otolaryngology-Head and Neck Surgery, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.,Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Ruben Stepanyan
- Department of Otolaryngology-Head and Neck Surgery, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.,Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Brian M McDermott
- Department of Otolaryngology-Head and Neck Surgery, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.,Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA.,Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.,Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
40
|
Zhao Q, Shen Y, Li X, Tian F, Yu X, Yobas L, Park H, Duan Y, Huang P. Analyzing protein-protein interactions in rare cells using microbead-based single-molecule pulldown assay. LAB ON A CHIP 2021; 21:3137-3149. [PMID: 34165117 DOI: 10.1039/d1lc00260k] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
For studying protein-protein interactions (PPIs) in general, a powerful and commonly used technique is conventional coimmunoprecipitation (co-IP/pulldown) followed by western blotting. However, the technique does not provide precise information regarding the kinetics and stoichiometry of PPIs. Another drawback is that the sensitivity of conventional co-IP is not suitable for examining PPIs in rare cells such as sensory hair cells, circulating tumor cells, embryonic stem cells, and subsets of immune cells. The current single-molecule pulldown (SiMPull) assay can potentially be used for studying PPIs in rare cells but its wide application is hindered by the high technical barrier and time consumption. We report an innovative, agarose microbead-based approach for SiMPull. We used commercially available, pre-surface-functionalized agarose microbeads to capture the protein of interest together with its binding partners specifically from cell extracts and observed these interactions under a microscope at the single-molecule level. Relative to the original method, microbead-based SiMPull is considerably faster, easier to use, and more reproducible and yet provides similar sensitivity and signal-to-background ratio; specifically, with the new method, sample-preparation time is substantially decreased (from ∼10 to ∼3 h). These crucial features should facilitate wide application of the powerful and versatile SiMPull method in common biological and clinical laboratories. Notably, by exploiting the simplicity and ultrahigh sensitivity of microbead-based SiMPull, we used the method in the study of rare auditory hair cells and γδ T cells for the first time.
Collapse
Affiliation(s)
- Qirui Zhao
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China.
| | - Yusheng Shen
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Hong Kong, China
| | - Xiaofen Li
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Hong Kong, China
| | - Fang Tian
- Department of Physics, Hong Kong University of Science and Technology, Hong Kong, China
| | - Xiaojie Yu
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China.
| | - Levent Yobas
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Hong Kong, China and Department of Electronic and Computer Engineering, Hong Kong University of Science and Technology, Hong Kong, China
| | - Hyokeun Park
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China. and Department of Physics, Hong Kong University of Science and Technology, Hong Kong, China
| | - Yuanyuan Duan
- Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, China
| | - Pingbo Huang
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China. and Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Hong Kong, China and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China and HKUST Shenzhen Research Institute, Hong Kong University of Science and Technology, Hong Kong, China and Hong Kong Branch of Guangdong Southern Marine Science and Engineering Laboratory (Guangzhou), Hong Kong University of Science and Technology, Hong Kong, China
| |
Collapse
|
41
|
Warren B, Nowotny M. Bridging the Gap Between Mammal and Insect Ears – A Comparative and Evolutionary View of Sound-Reception. Front Ecol Evol 2021. [DOI: 10.3389/fevo.2021.667218] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Insects must wonder why mammals have ears only in their head and why they evolved only one common principle of ear design—the cochlea. Ears independently evolved at least 19 times in different insect groups and therefore can be found in completely different body parts. The morphologies and functional characteristics of insect ears are as wildly diverse as the ecological niches they exploit. In both, insects and mammals, hearing organs are constrained by the same biophysical principles and their respective molecular processes for mechanotransduction are thought to share a common evolutionary origin. Due to this, comparative knowledge of hearing across animal phyla provides crucial insight into fundamental processes of auditory transduction, especially at the biomechanical and molecular level. This review will start by comparing hearing between insects and mammals in an evolutionary context. It will then discuss current findings about sound reception will help to bridge the gap between both research fields.
Collapse
|
42
|
Liu S, Wang S, Zou L, Xiong W. Mechanisms in cochlear hair cell mechano-electrical transduction for acquisition of sound frequency and intensity. Cell Mol Life Sci 2021; 78:5083-5094. [PMID: 33871677 PMCID: PMC11072359 DOI: 10.1007/s00018-021-03840-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 03/30/2021] [Accepted: 04/09/2021] [Indexed: 10/21/2022]
Abstract
Sound signals are acquired and digitized in the cochlea by the hair cells that further transmit the coded information to the central auditory pathways. Any defect in hair cell function may induce problems in the auditory system and hearing-based brain function. In the past 2 decades, our understanding of auditory transduction has been substantially deepened because of advances in molecular, structural, and functional studies. Results from these experiments can be perfectly embedded in the previously established profile from anatomical, histological, genetic, and biophysical research. This review aims to summarize the progress on the molecular and cellular mechanisms of the mechano-electrical transduction (MET) channel in the cochlear hair cells, which is involved in the acquisition of sound frequency and intensity-the two major parameters of an acoustic cue. We also discuss recent studies on TMC1, the molecule likely to form the MET channel pore.
Collapse
Affiliation(s)
- Shuang Liu
- School of Life Sciences, Tsinghua University, 1 Qinghuayuan, Beijing, 100084, China
- IDG/McGovern Institute for Brain Research at Tsinghua University, Tsinghua University, 1 Qinghuayuan, Beijing, 100084, China
| | - Shufeng Wang
- School of Life Sciences, Tsinghua University, 1 Qinghuayuan, Beijing, 100084, China
- IDG/McGovern Institute for Brain Research at Tsinghua University, Tsinghua University, 1 Qinghuayuan, Beijing, 100084, China
| | - Linzhi Zou
- School of Life Sciences, Tsinghua University, 1 Qinghuayuan, Beijing, 100084, China
- IDG/McGovern Institute for Brain Research at Tsinghua University, Tsinghua University, 1 Qinghuayuan, Beijing, 100084, China
| | - Wei Xiong
- School of Life Sciences, Tsinghua University, 1 Qinghuayuan, Beijing, 100084, China.
- IDG/McGovern Institute for Brain Research at Tsinghua University, Tsinghua University, 1 Qinghuayuan, Beijing, 100084, China.
| |
Collapse
|
43
|
Abstract
Sound-induced mechanical stimuli are detected by elaborate mechanosensory transduction (MT) machinery in highly specialized hair cells of the inner ear. Genetic studies of inherited deafness in the past decades have uncovered several molecular constituents of the MT complex, and intense debate has surrounded the molecular identity of the pore-forming subunits. How the MT components function in concert in response to physical stimulation is not fully understood. In this review, we summarize and discuss multiple lines of evidence supporting the hypothesis that transmembrane channel-like 1 is a long-sought MT channel subunit. We also review specific roles of other components of the MT complex, including protocadherin 15, cadherin 23, lipoma HMGIC fusion partner-like 5, transmembrane inner ear, calcium and integrin-binding family member 2, and ankyrins. Based on these recent advances, we propose a unifying theory of hair cell MT that may reconcile most of the functional discoveries obtained to date. Finally, we discuss key questions that need to be addressed for a comprehensive understanding of hair cell MT at molecular and atomic levels.
Collapse
Affiliation(s)
- Wang Zheng
- Departments of Otolaryngology and Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA;
| | - Jeffrey R Holt
- Departments of Otolaryngology and Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA;
| |
Collapse
|
44
|
Trouillet A, Miller KK, George SS, Wang P, Ali NES, Ricci A, Grillet N. Loxhd1 Mutations Cause Mechanotransduction Defects in Cochlear Hair Cells. J Neurosci 2021; 41:3331-3343. [PMID: 33707295 PMCID: PMC8051682 DOI: 10.1523/jneurosci.0975-20.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 02/22/2021] [Accepted: 03/02/2021] [Indexed: 12/27/2022] Open
Abstract
Sound detection happens in the inner ear via the mechanical deflection of the hair bundle of cochlear hair cells. The hair bundle is an apical specialization consisting of actin-filled membrane protrusions (called stereocilia) connected by tip links (TLs) that transfer the deflection force to gate the mechanotransduction channels. Here, we identified the hearing loss-associated Loxhd1/DFNB77 gene as being required for the mechanotransduction process. LOXHD1 consists of 15 polycystin lipoxygenase α-toxin (PLAT) repeats, which in other proteins can bind lipids and proteins. LOXHD1 was distributed along the length of the stereocilia. Two LOXHD1 mouse models with mutations in the 10th PLAT repeat exhibited mechanotransduction defects (in both sexes). While mechanotransduction currents in mutant inner hair cells (IHCs) were similar to wild-type levels in the first postnatal week, they were severely affected by postnatal day 11. The onset of the mechanotransduction phenotype was consistent with the temporal progression of postnatal LOXHD1 expression/localization in the hair bundle. The mechanotransduction defect observed in Loxhd1-mutant IHCs was not accompanied by a morphologic defect of the hair bundle or a reduction in TL number. Using immunolocalization, we found that two proteins of the upper and lower TL protein complexes (Harmonin and LHFPL5) were maintained in the mutants, suggesting that the mechanotransduction machinery was present but not activatable. This work identified a novel LOXHD1-dependent step in hair bundle development that is critical for mechanotransduction in mature hair cells as well as for normal hearing function in mice and humans.SIGNIFICANCE STATEMENT Hair cells detect sound-induced forces via the hair bundle, which consists of membrane protrusions connected by tip links. The mechanotransduction machinery forms protein complexes at the tip-link ends. The current study showed that LOXHD1, a multirepeat protein responsible for hearing loss in humans and mice when mutated, was required for hair-cell mechanotransduction, but only after the first postnatal week. Using immunochemistry, we demonstrated that this defect was not caused by the mislocalization of the tip-link complex proteins Harmonin or LHFPL5, suggesting that the mechanotransduction protein complexes were maintained. This work identified a new step in hair bundle development, which is critical for both hair-cell mechanotransduction and hearing.
Collapse
Affiliation(s)
- Alix Trouillet
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University, Stanford, California 94305
| | - Katharine K Miller
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University, Stanford, California 94305
| | - Shefin Sam George
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University, Stanford, California 94305
| | - Pei Wang
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University, Stanford, California 94305
| | - Noor-E-Seher Ali
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University, Stanford, California 94305
| | - Anthony Ricci
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University, Stanford, California 94305
- Department of Molecular and Cellular Physiology, School of Medicine, Stanford University, Stanford, California 94305
| | - Nicolas Grillet
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University, Stanford, California 94305
| |
Collapse
|
45
|
Tani T, Koike-Tani M, Tran MT, Shribak M, Levic S. Postnatal structural development of mammalian Basilar Membrane provides anatomical basis for the maturation of tonotopic maps and frequency tuning. Sci Rep 2021; 11:7581. [PMID: 33828185 PMCID: PMC8027603 DOI: 10.1038/s41598-021-87150-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/16/2021] [Indexed: 02/01/2023] Open
Abstract
The basilar membrane (BM) of the mammalian cochlea constitutes a spiraling acellular ribbon that is intimately attached to the organ of Corti. Its graded stiffness, increasing from apex to the base of the cochlea provides the mechanical basis for sound frequency analysis. Despite its central role in auditory signal transduction, virtually nothing is known about the BM's structural development. Using polarized light microscopy, the present study characterized the architectural transformations of freshly dissected BM at time points during postnatal development and maturation. The results indicate that the BM structural elements increase progressively in size, becoming radially aligned and more tightly packed with maturation and reach the adult structural signature by postnatal day 20 (P20). The findings provide insight into structural details and developmental changes of the mammalian BM, suggesting that BM is a dynamic structure that changes throughout the life of an animal.
Collapse
Affiliation(s)
- Tomomi Tani
- Marine Biological Laboratory, Eugene Bell Center, Woods Hole, MA, USA
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Ikeda, Osaka, Japan
| | - Maki Koike-Tani
- Marine Biological Laboratory, Eugene Bell Center, Woods Hole, MA, USA
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan
| | - Mai Thi Tran
- Marine Biological Laboratory, Eugene Bell Center, Woods Hole, MA, USA
- College of Engineering and Computer Science, VinUniversity, Gia Lam District, Hanoi, Vietnam
| | - Michael Shribak
- Marine Biological Laboratory, Eugene Bell Center, Woods Hole, MA, USA
| | - Snezana Levic
- Marine Biological Laboratory, Eugene Bell Center, Woods Hole, MA, USA.
- Sensory Neuroscience Research Group, School of Pharmacy and Biomolecular Sciences, University of Brighton, Huxley Building, Brighton, BN2 4GJ, UK.
- Brighton and Sussex Medical School, University of Sussex, Brighton, BN1 9PX, UK.
| |
Collapse
|
46
|
New Tmc1 Deafness Mutations Impact Mechanotransduction in Auditory Hair Cells. J Neurosci 2021; 41:4378-4391. [PMID: 33824189 PMCID: PMC8152607 DOI: 10.1523/jneurosci.2537-20.2021] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 03/19/2021] [Accepted: 03/27/2021] [Indexed: 12/05/2022] Open
Abstract
Transmembrane channel-like protein isoform 1 (TMC1) is a major component of the mechano-electrical transducer (MET) channel in cochlear hair cells and is subject to numerous mutations causing deafness. We report a new dominant human deafness mutation, TMC1 p.T422K, and have characterized the homologous mouse mutant, Tmc1 p.T416K, which caused deafness and outer hair cell (OHC) loss by the fourth postnatal week. MET channels showed decreased Ca2+ permeability and resting open probability, but no change in single-channel conductance or expression. Three adjacent deafness mutations are TMC1 p.L416R, p.G417R, and p.M418K, the last homologous to the mouse Beethoven that exhibits similar channel effects. All substitute a positive for a neutral residue, which could produce charge screening in the channel pore or influence binding of an accessory subunit. Channel properties were compared in mice of both sexes between dominant (Tmc1 p.T416K, Tmc1 p.D569N) and recessive (Tmc1 p.W554L, Tmc1 p.D528N) mutations of residues near the putative pore of the channel. Tmc1 p.W554L and p.D569N exhibit reduced maximum current with no effect on single-channel conductance, implying a smaller number of channels transported to the stereociliary tips; this may stem from impaired TMC1 binding to LHFPL5. Tmc1 p.D528N, located in the pore's narrowest region, uniquely caused large reductions in MET channel conductance and block by dihydrostreptomycin (DHS). For Tmc1 p.T416K and Tmc1 p.D528N, transduction loss occurred between P15 and P20. We propose two mechanisms linking channel mutations and deafness: decreased Ca2+ permeability, common to all mutants, and decreased resting open probability in low Ca2+, confined to dominant mutations. SIGNIFICANCE STATEMENT Transmembrane channel-like protein isoform 1 (TMC1) is thought to be a major component of the mechanotransducer channel in auditory hair cells, but the protein organization and channel structure are still uncertain. We made four mouse lines harboring Tmc1 point mutations that alter channel properties, causing hair cell degeneration and deafness. These include a mouse homolog of a new human deafness mutation pT416K that decreased channel Ca2+ permeability by introducing a positively-charged amino acid in the putative pore. All mutations are consistent with the channel structure predicted from modeling, but only one, p.D528N near the external face of the pore, substantially reduced channel conductance and Ca2+ permeability and virtually abolished block by dihydrostreptomycin (DHS), strongly endorsing its siting within the pore.
Collapse
|
47
|
McGrath J, Tung CY, Liao X, Belyantseva IA, Roy P, Chakraborty O, Li J, Berbari NF, Faaborg-Andersen CC, Barzik M, Bird JE, Zhao B, Balakrishnan L, Friedman TB, Perrin BJ. Actin at stereocilia tips is regulated by mechanotransduction and ADF/cofilin. Curr Biol 2021; 31:1141-1153.e7. [PMID: 33400922 PMCID: PMC8793668 DOI: 10.1016/j.cub.2020.12.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/21/2020] [Accepted: 12/07/2020] [Indexed: 11/30/2022]
Abstract
Stereocilia on auditory sensory cells are actin-based protrusions that mechanotransduce sound into an electrical signal. These stereocilia are arranged into a bundle with three rows of increasing length to form a staircase-like morphology that is required for hearing. Stereocilia in the shorter rows, but not the tallest row, are mechanotransducing because they have force-sensitive channels localized at their tips. The onset of mechanotransduction during mouse postnatal development refines stereocilia length and width. However, it is unclear how actin is differentially regulated between stereocilia in the tallest row of the bundle and the shorter, mechanotransducing rows. Here, we show actin turnover is increased at the tips of mechanotransducing stereocilia during bundle maturation. Correspondingly, from birth to postnatal day 6, these stereocilia had increasing amounts of available actin barbed ends, where monomers can be added or lost readily, as compared with the non-mechanotransducing stereocilia in the tallest row. The increase in available barbed ends depended on both mechanotransduction and MYO15 or EPS8, which are required for the normal specification and elongation of the tallest row of stereocilia. We also found that loss of the F-actin-severing proteins ADF and cofilin-1 decreased barbed end availability at stereocilia tips. These proteins enriched at mechanotransducing stereocilia tips, and their localization was perturbed by the loss of mechanotransduction, MYO15, or EPS8. Finally, stereocilia lengths and widths were dysregulated in Adf and Cfl1 mutants. Together, these data show that actin is remodeled, likely by a severing mechanism, in response to mechanotransduction.
Collapse
Affiliation(s)
- Jamis McGrath
- Department of Biology, Indiana University-Purdue University Indianapolis, 723 West Michigan Street, Indianapolis, IN 46202, USA
| | - Chun-Yu Tung
- Department of Biology, Indiana University-Purdue University Indianapolis, 723 West Michigan Street, Indianapolis, IN 46202, USA
| | - Xiayi Liao
- Department of Biology, Indiana University-Purdue University Indianapolis, 723 West Michigan Street, Indianapolis, IN 46202, USA
| | - Inna A Belyantseva
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, NIH, 35A Convent Drive, Bethesda, MD 20892, USA
| | - Pallabi Roy
- Department of Biology, Indiana University-Purdue University Indianapolis, 723 West Michigan Street, Indianapolis, IN 46202, USA
| | - Oisorjo Chakraborty
- Department of Biology, Indiana University-Purdue University Indianapolis, 723 West Michigan Street, Indianapolis, IN 46202, USA
| | - Jinan Li
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, 1160 West Michigan Street, Indianapolis, IN 46202, USA
| | - Nicolas F Berbari
- Department of Biology, Indiana University-Purdue University Indianapolis, 723 West Michigan Street, Indianapolis, IN 46202, USA
| | - Christian C Faaborg-Andersen
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, NIH, 35A Convent Drive, Bethesda, MD 20892, USA
| | - Melanie Barzik
- Section on Sensory Cell Biology, National Institute on Deafness and Other Communication Disorders, NIH, 35A Convent Drive, Bethesda, MD 20892, USA
| | - Jonathan E Bird
- Department of Pharmacology and Therapeutics, University of Florida, 1200 Newell Drive, Gainesville, FL 32610, USA
| | - Bo Zhao
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, 1160 West Michigan Street, Indianapolis, IN 46202, USA
| | - Lata Balakrishnan
- Department of Biology, Indiana University-Purdue University Indianapolis, 723 West Michigan Street, Indianapolis, IN 46202, USA
| | - Thomas B Friedman
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, NIH, 35A Convent Drive, Bethesda, MD 20892, USA
| | - Benjamin J Perrin
- Department of Biology, Indiana University-Purdue University Indianapolis, 723 West Michigan Street, Indianapolis, IN 46202, USA.
| |
Collapse
|
48
|
Bankoti K, Generotti C, Hwa T, Wang L, O'Malley BW, Li D. Advances and challenges in adeno-associated viral inner-ear gene therapy for sensorineural hearing loss. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:209-236. [PMID: 33850952 PMCID: PMC8010215 DOI: 10.1016/j.omtm.2021.03.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
There is growing attention and effort focused on treating the root cause of sensorineural hearing loss rather than managing associated secondary characteristic features. With recent substantial advances in understanding sensorineural hearing-loss mechanisms, gene delivery has emerged as a promising strategy for the biological treatment of hearing loss associated with genetic dysfunction. There are several successful and promising proof-of-principle examples of transgene deliveries in animal models; however, there remains substantial further progress to be made in these avenues before realizing their clinical application in humans. Herein, we review different aspects of development, ongoing preclinical studies, and challenges to the clinical transition of transgene delivery of the inner ear toward the restoration of lost auditory and vestibular function.
Collapse
Affiliation(s)
- Kamakshi Bankoti
- Department of Otorhinolaryngology, Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Charles Generotti
- Department of Otorhinolaryngology, Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tiffany Hwa
- Department of Otorhinolaryngology, Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lili Wang
- Department of Medicine, Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bert W O'Malley
- Department of Otorhinolaryngology, Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daqing Li
- Department of Otorhinolaryngology, Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
49
|
Effertz T, Moser T, Oliver D. Recent advances in cochlear hair cell nanophysiology: subcellular compartmentalization of electrical signaling in compact sensory cells. Fac Rev 2021; 9:24. [PMID: 33659956 PMCID: PMC7886071 DOI: 10.12703/r/9-24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In recent years, genetics, physiology, and structural biology have advanced into the molecular details of the sensory physiology of auditory hair cells. Inner hair cells (IHCs) and outer hair cells (OHCs) mediate two key functions: active amplification and non-linear compression of cochlear vibrations by OHCs and sound encoding by IHCs at their afferent synapses with the spiral ganglion neurons. OHCs and IHCs share some molecular physiology, e.g. mechanotransduction at the apical hair bundles, ribbon-type presynaptic active zones, and ionic conductances in the basolateral membrane. Unique features enabling their specific function include prestin-based electromotility of OHCs and indefatigable transmitter release at the highest known rates by ribbon-type IHC active zones. Despite their compact morphology, the molecular machineries that either generate electrical signals or are driven by these signals are essentially all segregated into local subcellular structures. This review provides a brief account on recent insights into the molecular physiology of cochlear hair cells with a specific focus on organization into membrane domains.
Collapse
Affiliation(s)
- Thomas Effertz
- InnerEarLab, Department of Otorhinolaryngology, University Medical Center Göttingen, 37099 Göttingen, Germany
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37099 Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany
- Synaptic Nanophysiology Group, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
- Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, 37075 Göttingen, Germany
| | - Dominik Oliver
- Institute for Physiology and Pathophysiology, Philipps University, Deutschhausstraße 2, 35037 Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Universities of Marburg and Giessen, Germany
- DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodelling, GRK 2213, Philipps University, Marburg, Germany
| |
Collapse
|
50
|
Janesick A, Scheibinger M, Benkafadar N, Kirti S, Ellwanger DC, Heller S. Cell-type identity of the avian cochlea. Cell Rep 2021; 34:108900. [PMID: 33761346 DOI: 10.1016/j.celrep.2021.108900] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 01/22/2021] [Accepted: 03/03/2021] [Indexed: 02/06/2023] Open
Abstract
In contrast to mammals, birds recover naturally from acquired hearing loss, which makes them an ideal model for inner ear regeneration research. Here, we present a validated single-cell RNA sequencing resource of the avian cochlea. We describe specific markers for three distinct types of sensory hair cells, including a previously unknown subgroup, which we call superior tall hair cells. We identify markers for the supporting cells associated with tall hair cells, which represent the facultative stem cells of the avian inner ear. Likewise, we present markers for supporting cells that are located below the short cochlear hair cells. We further infer spatial expression gradients of hair cell genes along the tonotopic axis of the cochlea. This resource advances neurobiology, comparative biology, and regenerative medicine by providing a basis for comparative studies with non-regenerating mammalian cochleae and for longitudinal studies of the regenerating avian cochlea.
Collapse
Affiliation(s)
- Amanda Janesick
- Department of Otolaryngology - Head & Neck Surgery, Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Palo Alto, CA 94305, USA.
| | - Mirko Scheibinger
- Department of Otolaryngology - Head & Neck Surgery, Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Nesrine Benkafadar
- Department of Otolaryngology - Head & Neck Surgery, Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Sakin Kirti
- Department of Otolaryngology - Head & Neck Surgery, Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Palo Alto, CA 94305, USA; Case Western Reserve University, Cleveland, OH 44106, USA
| | - Daniel C Ellwanger
- Department of Otolaryngology - Head & Neck Surgery, Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Palo Alto, CA 94305, USA; Genome Analysis Unit, Amgen Research, Amgen, Inc., South San Francisco, CA 94080, USA
| | - Stefan Heller
- Department of Otolaryngology - Head & Neck Surgery, Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Palo Alto, CA 94305, USA.
| |
Collapse
|