51
|
Calcium signaling and genetic rare diseases: An auditory perspective. Cell Calcium 2023; 110:102702. [PMID: 36791536 DOI: 10.1016/j.ceca.2023.102702] [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: 12/14/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/07/2023]
Abstract
Deafness is a highly heterogeneous disorder which stems, for 50%, from genetic origins. Sensory transduction relies mainly on sensory hair cells of the cochlea, in the inner ear. Calcium is key for the function of these cells and acts as a fundamental signal transduction. Its homeostasis depends on three factors: the calcium influx, through the mechanotransduction channel at the apical pole of the hair cell as well as the voltage-gated calcium channel at the base of the cells; the calcium buffering via Ca2+-binding proteins in the cytoplasm, but also in organelles such as mitochondria and the reticulum endoplasmic mitochondria-associated membranes with specialized proteins; and the calcium extrusion through the Ca-ATPase pump, located all over the plasma membrane. In addition, the synaptic transmission to the central nervous system is also controlled by calcium. Genetic studies of inherited deafness have tremendously helped understand the underlying molecular pathways of calcium signaling. In this review, we discuss these different factors in light of the associated genetic diseases (syndromic and non-syndromic deafness) and the causative genes.
Collapse
|
52
|
Functional consequences of Genetics variant in TMC1 and TMC2 within a United Arab Emirates family with Pre-lingual hearing loss. Saudi J Biol Sci 2023; 30:103520. [PMID: 36568409 PMCID: PMC9772550 DOI: 10.1016/j.sjbs.2022.103520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/30/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
Hearing loss (HL) is the most prevalent sensory disorder whose etiology comes from environmental and/or genetic factors. Approximately 60 % of HL cases are due to mutations in genes responsible for maintaining a normal hearing function. Despite the monogenic inheritance of hereditary hearing loss (HHL), its diagnosis is challenging as both clinical and genetic heterogeneity characterizes it. Through the development of next-generation sequencing (NGS) techniques, the number of identified mutations responsible for HHL has increased exponentially during the last decade. Mutations in the TMC1 have been reported in several patients with nonsyndromic hereditary hearing loss (NSHHL), more precisely in cases with an autosomal recessive inheritance pattern. In this study, we conducted whole-exome sequencing (WES) analysis of a United Arabs Emirates (UAE) family with autosomal recessive nonsyndromic hearing loss (ARNSHL). This analysis revealed segregation of the TMC1 missense mutation c.596A > T (p.Asn199Ile) with the disease. Bioinformatics analysis supported the pathogenic effect of this mutation and predicted its impact at the proteomics level. Molecular docking analysis of TMC2WT, TMC2R123K, TMC2Q205R, and TMC2R123K + Q205R. Finally, protein docking results suggest a role for TMC2 variants in the phenotypic variability observed within the investigated family.
Collapse
Key Words
- ARNSHL, Autosomal Recessive Nonsyndromic Hearing Loss
- GATK, Genome Analysis Toolkit
- GnomAD, Genome Aggregation Database
- HHL, Hereditary Hearing Loss
- HL, Hearing Loss
- NGS, Next Generation Sequencing
- NSHHL, Nonsyndromic Hereditary Hearing Loss
- Non-syndromic Hearing Loss
- PCR, Polymerase Chain Reaction
- Phenotypic variability
- Protein docking
- TMC1
- UAE, United Arabs Emirates
- WES, Whole-Exome Sequencing
- Whole-exome sequencing
- c.596A > T mutation
Collapse
|
53
|
Li J. Liquid-liquid phase separation in hair cell stereocilia development and maintenance. Comput Struct Biotechnol J 2023; 21:1738-1745. [PMID: 36890881 PMCID: PMC9986246 DOI: 10.1016/j.csbj.2023.02.040] [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: 10/28/2022] [Revised: 02/22/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
As an emerging concept, liquid-liquid phase separation (LLPS) in biological systems has shed light on the formation mechanisms of membrane-less compartments in cells. The process is driven by multivalent interactions of biomolecules such as proteins and/or nucleic acids, allowing them to form condensed structures. In the inner ear hair cells, LLPS-based biomolecular condensate assembly plays a vital role in the development and maintenance of stereocilia, the mechanosensing organelles located at the apical surface of hair cells. This review aims to summarize recent findings on the molecular basis governing the LLPS of Usher syndrome-related gene-encoding proteins and their binding partners, which may ultimately result in the formation of upper tip-link density and tip complex density in hair cell stereocilia, offering a better understanding of this severe inherited disease that causes deaf-blindness.
Collapse
Affiliation(s)
- Jianchao Li
- Department of Otorhinolaryngology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China.,Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
54
|
Wang X, Zhou Y, Wang D, Wang Y, Zhou Z, Ma X, Liu X, Dong Y. Cisplatin-induced ototoxicity: From signaling network to therapeutic targets. Biomed Pharmacother 2023; 157:114045. [PMID: 36455457 DOI: 10.1016/j.biopha.2022.114045] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/15/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022] Open
Abstract
Administration of cisplatin, a common chemotherapeutic drug, has an inevitable side effect of sensorineural hearing loss. The main etiologies are stria vascularis injury, spiral ganglion degeneration, and hair cell death. Over several decades, the research scope of cisplatin-induced ototoxicity has expanded with the discovery of the molecular mechanism mediating inner ear cell death, highlighting the roles of reactive oxygen species and transport channels for cisplatin uptake into inner ear cells. Upon entering hair cells, cisplatin disrupts organelle metabolism, induces oxidative stress, and targets DNA to cause intracellular damage. Recent studies have also reported the role of inflammation in cisplatin-induced ototoxicity. In this article, we preform a narrative review of the latest reported molecular mechanisms of cisplatin-induced ototoxicity, from extracellular to intracellular. We build up a signaling network starting with cisplatin entering into the inner ear through the blood labyrinth barrier, disrupting cochlear endolymph homeostasis, and activating inflammatory responses of the outer hair cells. After entering the hair cells, cisplatin causes hair cell death via DNA damage, redox system imbalance, and mitochondrial and endoplasmic reticulum dysfunction, culminating in programmed cell death including apoptosis, necroptosis, autophagic death, pyroptosis, and ferroptosis. Based on the mentioned mechanisms, prominent therapeutic targets, such as channel-blocking drugs of cisplatin transporter, construction of cisplatin structural analogues, anti-inflammatory drugs, antioxidants, cell death inhibitors, and others, were collated. Considering the recent research efforts, we have analyzed the feasibility of the aforementioned therapeutic strategies and proposed our otoprotective approaches to overcome cisplatin-induced ototoxicity.
Collapse
Affiliation(s)
- Xilu Wang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yingying Zhou
- Department of Obstetrics & gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Dali Wang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yi Wang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhaoyu Zhou
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiulan Ma
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaofang Liu
- Department of Surgical Oncology, the First Affiliated Hospital of China Medical University, Shenyang, China.
| | - Yaodong Dong
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
55
|
Chen YS, Cabrera E, Tucker BJ, Shin TJ, Moawad JV, Totten DJ, Booth KT, Nelson RF. TMPRSS3 expression is limited in spiral ganglion neurons: implication for successful cochlear implantation. J Med Genet 2022; 59:1219-1226. [PMID: 35961784 DOI: 10.1136/jmg-2022-108654] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/15/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND It is well established that biallelic mutations in transmembrane protease, serine 3 (TMPRSS3) cause hearing loss. Currently, there is controversy regarding the audiological outcomes after cochlear implantation (CI) for TMPRSS3-associated hearing loss. This controversy creates confusion among healthcare providers regarding the best treatment options for individuals with TMPRSS3-related hearing loss. METHODS A literature review was performed to identify all published cases of patients with TMPRSS3-associated hearing loss who received a CI. CI outcomes of this cohort were compared with published adult CI cohorts using postoperative consonant-nucleus-consonant (CNC) word performance. TMPRSS3 expression in mouse cochlea and human auditory nerves (HAN) was determined by using hybridisation chain reaction and single-cell RNA-sequencing analysis. RESULTS In aggregate, 27 patients (30 total CI ears) with TMPRSS3-associated hearing loss treated with CI, and 85% of patients reported favourable outcomes. Postoperative CNC word scores in patients with TMPRSS3-associated hearing loss were not significantly different than those seen in adult CI cohorts (8 studies). Robust Tmprss3 expression occurs throughout the mouse organ of Corti, the spindle and root cells of the lateral wall and faint staining within <5% of the HAN, representing type II spiral ganglion neurons. Adult HAN express negligible levels of TMPRSS3. CONCLUSION The clinical features after CI and physiological expression of TMPRSS3 suggest against a major role of TMPRSS3 in auditory neurons.
Collapse
Affiliation(s)
- Yuan-Siao Chen
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ernesto Cabrera
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Brady J Tucker
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Timothy J Shin
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jasmine V Moawad
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Douglas J Totten
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kevin T Booth
- Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
- Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Rick F Nelson
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
56
|
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
|
57
|
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
|
58
|
Scheibinger M, Janesick A, Benkafadar N, Ellwanger DC, Jan TA, Heller S. Cell-type identity of the avian utricle. Cell Rep 2022; 40:111432. [PMID: 36170825 PMCID: PMC9588199 DOI: 10.1016/j.celrep.2022.111432] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/18/2022] [Accepted: 09/08/2022] [Indexed: 11/30/2022] Open
Abstract
The avian utricle, a vestibular organ of the inner ear, displays turnover of sensory hair cells throughout life. This is in sharp contrast to the mammalian utricle, which shows limited regenerative capacity. Here, we use single-cell RNA sequencing to identify distinct marker genes for the different sensory hair cell subtypes of the chicken utricle, which we validated in situ. We provide markers for spatially distinct supporting cell populations and identify two transitional cell populations of dedifferentiating supporting cells and developing hair cells. Trajectory reconstruction resulted in an inventory of gene expression dynamics of natural hair cell generation in the avian utricle. Scheibinger et al. provide a single-cell transcriptomic atlas of the chicken utricle, a vestibular organ. Hair cell and supporting cell subtypes are defined by marker genes, and trajectories of gene expression dynamics during hair cell turnover are shown. This resource provides a baseline to study inner ear damage and regeneration.
Collapse
Affiliation(s)
- Mirko Scheibinger
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Amanda Janesick
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nesrine Benkafadar
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Daniel C Ellwanger
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Taha A Jan
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Stefan Heller
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
59
|
cAMP and voltage modulate rat auditory mechanotransduction by decreasing the stiffness of gating springs. Proc Natl Acad Sci U S A 2022; 119:e2107567119. [PMID: 35858439 PMCID: PMC9335186 DOI: 10.1073/pnas.2107567119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Regulation of auditory sensitivity contributes to the precision, dynamic range, and protection of the auditory system. Regulation of the hair cell mechanotransduction channel is a major contributor to controlling the sensitivity of the auditory transduction process. The gating spring is a critical piece of the mechanotransduction machinery because it opens and closes the mechanotransduction channel, and its stiffness regulates the sensitivity of the mechanotransduction process. In the present work, we characterize the effect of the second-messenger signaling molecule cyclic adenosine monophosphate (cAMP) and identify that it reduces gating spring stiffness likely through an exchange protein directly activated by cAMP (EPAC)-mediated pathway. This is a unique physiologic mechanism to regulate gating spring stiffness. Hair cells of the auditory and vestibular systems transform mechanical input into electrical potentials through the mechanoelectrical transduction process (MET). Deflection of the mechanosensory hair bundle increases tension in the gating springs that open MET channels. Regulation of MET channel sensitivity contributes to the auditory system’s precision, wide dynamic range and, potentially, protection from overexcitation. Modulating the stiffness of the gating spring modulates the sensitivity of the MET process. Here, we investigated the role of cyclic adenosine monophosphate (cAMP) in rat outer hair cell MET and found that cAMP up-regulation lowers the sensitivity of the channel in a manner consistent with decreasing gating spring stiffness. Direct measurements of the mechanical properties of the hair bundle confirmed a decrease in gating spring stiffness with cAMP up-regulation. In parallel, we found that prolonged depolarization mirrored the effects of cAMP. Finally, a limited number of experiments implicate that cAMP activates the exchange protein directly activated by cAMP to mediate the changes in MET sensitivity. These results reveal that cAMP signaling modulates gating spring stiffness to affect auditory sensitivity.
Collapse
|
60
|
Akyuz N, Karavitaki KD, Pan B, Tamvakologos PI, Brock KP, Li Y, Marks DS, Corey DP. Mechanical gating of the auditory transduction channel TMC1 involves the fourth and sixth transmembrane helices. SCIENCE ADVANCES 2022; 8:eabo1126. [PMID: 35857511 PMCID: PMC9278870 DOI: 10.1126/sciadv.abo1126] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 05/27/2022] [Indexed: 05/27/2023]
Abstract
The transmembrane (TM) channel-like 1 (TMC1) and TMC2 proteins play a central role in auditory transduction, forming ion channels that convert sound into electrical signals. However, the molecular mechanism of their gating remains unknown. Here, using predicted structural models as a guide, we probed the effects of 12 mutations on the mechanical gating of the transduction currents in native hair cells of Tmc1/2-null mice expressing virally introduced TMC1 variants. Whole-cell electrophysiological recordings revealed that mutations within the pore-lining TM4 and TM6 helices modified gating, reducing the force sensitivity or shifting the open probability of the channels, or both. For some of the mutants, these changes were accompanied by a change in single-channel conductance. Our observations are in line with a model wherein conformational changes in the TM4 and TM6 helices are involved in the mechanical gating of the transduction channel.
Collapse
Affiliation(s)
- Nurunisa Akyuz
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Bifeng Pan
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Kelly P. Brock
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Yaqiao Li
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Debora S. Marks
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - David P. Corey
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
61
|
Qian F, Wei G, Gao Y, Wang X, Gong J, Guo C, Wang X, Zhang X, Zhao J, Wang C, Xu M, Hu Y, Yin G, Kang J, Chai R, Xie G, Liu D. Single-cell RNA-sequencing of zebrafish hair cells reveals novel genes potentially involved in hearing loss. Cell Mol Life Sci 2022; 79:385. [PMID: 35753015 PMCID: PMC11072488 DOI: 10.1007/s00018-022-04410-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 05/18/2022] [Accepted: 06/01/2022] [Indexed: 01/22/2023]
Abstract
Hair cells play key roles in hearing and balance, and hair cell loss would result in hearing loss or vestibular dysfunction. Cellular and molecular research in hair cell biology provides us a better understanding of hearing and deafness. Zebrafish, owing to their hair cell-enriched organs, have been widely applied in hair cell-related research worldwide. Similar to mammals, zebrafish have inner ear hair cells. In addition, they also have lateral line neuromast hair cells. These different types of hair cells vary in morphology and function. However, systematic analysis of their molecular characteristics remains lacking. In this study, we analyzed the GFP+ cells isolated from Tg(Brn3c:mGFP) larvae with GFP expression in all hair cells using single-cell RNA-sequencing (scRNA-seq). Three subtypes of hair cells, namely macula hair cell (MHC), crista hair cell (CHC), and neuromast hair cell (NHC), were characterized and validated by whole-mount in situ hybridization analysis of marker genes. The hair cell scRNA-seq data revealed hair cell-specific genes, including hearing loss genes that have been identified in humans and novel genes potentially involved in hair cell formation and function. Two novel genes were discovered to specifically function in NHCs and MHCs, corresponding to their specific expression in NHCs and MHCs. This study allows us to understand the specific genes in hair cell subpopulations of zebrafish, which will shed light on the genetics of both human vestibular and cochlear hair cell function.
Collapse
Affiliation(s)
- Fuping Qian
- School of Life Sciences, Nantong Laboratory of Development and Diseases, Nantong University, Nantong, 226019, China
| | - Guanyun Wei
- School of Life Sciences, Nantong Laboratory of Development and Diseases, Nantong University, Nantong, 226019, China
| | - Yajing Gao
- Co-Innovation Center of Neuroregeneration, School of Life SciencesKey Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong, 226019, China
| | - Xin Wang
- Co-Innovation Center of Neuroregeneration, School of Life SciencesKey Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong, 226019, China
| | - Jie Gong
- School of Life Sciences, Nantong Laboratory of Development and Diseases, Nantong University, Nantong, 226019, China
| | - Chao Guo
- School of Life Sciences, Nantong Laboratory of Development and Diseases, Nantong University, Nantong, 226019, China
| | - Xiaoning Wang
- Co-Innovation Center of Neuroregeneration, School of Life SciencesKey Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong, 226019, China
| | - Xu Zhang
- Co-Innovation Center of Neuroregeneration, School of Life SciencesKey Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong, 226019, China
| | - Jinxiang Zhao
- Co-Innovation Center of Neuroregeneration, School of Life SciencesKey Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong, 226019, China
| | - Cheng Wang
- School of Life Sciences, Nantong Laboratory of Development and Diseases, Nantong University, Nantong, 226019, China
| | - Mengting Xu
- Co-Innovation Center of Neuroregeneration, School of Life SciencesKey Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong, 226019, China
| | - Yuebo Hu
- Co-Innovation Center of Neuroregeneration, School of Life SciencesKey Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong, 226019, China
| | - Guoli Yin
- Co-Innovation Center of Neuroregeneration, School of Life SciencesKey Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong, 226019, China
| | - Jiahui Kang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, 226001, China
| | - Renjie Chai
- Co-Innovation Center of Neuroregeneration, School of Life SciencesKey Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong, 226019, China.
- State Key Laboratory of Bioelectronics, Co-Innovation Center of Neuroregeneration, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, School of Life Science and Technology, Southeast University, Nanjing, 210096, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing, 100864, China.
- Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China.
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Gangcai Xie
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, 226001, China.
| | - Dong Liu
- School of Life Sciences, Nantong Laboratory of Development and Diseases, Nantong University, Nantong, 226019, China.
- Co-Innovation Center of Neuroregeneration, School of Life SciencesKey Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong, 226019, China.
| |
Collapse
|
62
|
Abstract
High-resolution immunofluorescence imaging of cochlear hair bundles faces many challenges due to the hair bundle’s small dimensions, fragile nature, and complex organization. Here, we describe an optimized protocol for hair-bundle protein immunostaining and localization. We detail the steps and solutions for extracting and fixing the mouse inner ear and for dissecting the organ of Corti. We further emphasize the optimal permeabilization, blocking, staining, and mounting conditions as well as the parameters for high-resolution microscopy imaging. For complete details on the use and execution of this protocol, please refer to Trouillet et al. (2021). Techniques for dissecting the mouse cochlea and the organ of Corti Dissection, permeabilization, blocking parameters to detect hair bundle proteins Mounting method to localize protein in the hair bundles
Publisher’s note: Undertaking any experimental protocol requires adherence to local institutional guidelines for laboratory safety and ethics.
Collapse
Affiliation(s)
- Katharine K Miller
- Stanford University, Department of Otolaryngology - Head & Neck Surgery, Stanford, CA, USA
| | - Pei Wang
- Stanford University, Department of Otolaryngology - Head & Neck Surgery, Stanford, CA, USA
| | - Nicolas Grillet
- Stanford University, Department of Otolaryngology - Head & Neck Surgery, Stanford, CA, USA
| |
Collapse
|
63
|
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
|
64
|
Maudoux A, Vitry S, El-Amraoui A. Vestibular Deficits in Deafness: Clinical Presentation, Animal Modeling, and Treatment Solutions. Front Neurol 2022; 13:816534. [PMID: 35444606 PMCID: PMC9013928 DOI: 10.3389/fneur.2022.816534] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
The inner ear is responsible for both hearing and balance. These functions are dependent on the correct functioning of mechanosensitive hair cells, which convert sound- and motion-induced stimuli into electrical signals conveyed to the brain. During evolution of the inner ear, the major changes occurred in the hearing organ, whereas the structure of the vestibular organs remained constant in all vertebrates over the same period. Vestibular deficits are highly prevalent in humans, due to multiple intersecting causes: genetics, environmental factors, ototoxic drugs, infections and aging. Studies of deafness genes associated with balance deficits and their corresponding animal models have shed light on the development and function of these two sensory systems. Bilateral vestibular deficits often impair individual postural control, gaze stabilization, locomotion and spatial orientation. The resulting dizziness, vertigo, and/or falls (frequent in elderly populations) greatly affect patient quality of life. In the absence of treatment, prosthetic devices, such as vestibular implants, providing information about the direction, amplitude and velocity of body movements, are being developed and have given promising results in animal models and humans. Novel methods and techniques have led to major progress in gene therapies targeting the inner ear (gene supplementation and gene editing), 3D inner ear organoids and reprograming protocols for generating hair cell-like cells. These rapid advances in multiscale approaches covering basic research, clinical diagnostics and therapies are fostering interdisciplinary research to develop personalized treatments for vestibular disorders.
Collapse
Affiliation(s)
- Audrey Maudoux
- Unit Progressive Sensory Disorders, Pathophysiology and Therapy, Institut Pasteur, Institut de l'Audition, Université de Paris, INSERM-UMRS1120, Paris, France
- Center for Balance Evaluation in Children (EFEE), Otolaryngology Department, Assistance Publique des Hôpitaux de Paris, Robert-Debré University Hospital, Paris, France
| | - Sandrine Vitry
- Unit Progressive Sensory Disorders, Pathophysiology and Therapy, Institut Pasteur, Institut de l'Audition, Université de Paris, INSERM-UMRS1120, Paris, France
| | - Aziz El-Amraoui
- Unit Progressive Sensory Disorders, Pathophysiology and Therapy, Institut Pasteur, Institut de l'Audition, Université de Paris, INSERM-UMRS1120, Paris, France
| |
Collapse
|
65
|
The genetic and phenotypic landscapes of Usher syndrome: from disease mechanisms to a new classification. Hum Genet 2022; 141:709-735. [PMID: 35353227 PMCID: PMC9034986 DOI: 10.1007/s00439-022-02448-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/04/2022] [Indexed: 12/16/2022]
Abstract
Usher syndrome (USH) is the most common cause of deaf–blindness in humans, with a prevalence of about 1/10,000 (~ 400,000 people worldwide). Cochlear implants are currently used to reduce the burden of hearing loss in severe-to-profoundly deaf patients, but many promising treatments including gene, cell, and drug therapies to restore the native function of the inner ear and retinal sensory cells are under investigation. The traditional clinical classification of Usher syndrome defines three major subtypes—USH1, 2 and 3—according to hearing loss severity and onset, the presence or absence of vestibular dysfunction, and age at onset of retinitis pigmentosa. Pathogenic variants of nine USH genes have been initially reported: MYO7A, USH1C, PCDH15, CDH23, and USH1G for USH1, USH2A, ADGRV1, and WHRN for USH2, and CLRN1 for USH3. Based on the co-occurrence of hearing and vision deficits, the list of USH genes has been extended to few other genes, but with limited supporting information. A consensus on combined criteria for Usher syndrome is crucial for the development of accurate diagnosis and to improve patient management. In recent years, a wealth of information has been obtained concerning the properties of the Usher proteins, related molecular networks, potential genotype–phenotype correlations, and the pathogenic mechanisms underlying the impairment or loss of hearing, balance and vision. The advent of precision medicine calls for a clear and more precise diagnosis of Usher syndrome, exploiting all the existing data to develop a combined clinical/genetic/network/functional classification for Usher syndrome.
Collapse
|
66
|
Dal Cortivo G, Dell’Orco D. Calcium- and Integrin-Binding Protein 2 (CIB2) in Physiology and Disease: Bright and Dark Sides. Int J Mol Sci 2022; 23:ijms23073552. [PMID: 35408910 PMCID: PMC8999013 DOI: 10.3390/ijms23073552] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 03/19/2022] [Accepted: 03/22/2022] [Indexed: 12/04/2022] Open
Abstract
Calcium- and integrin-binding protein 2 (CIB2) is a small EF-hand protein capable of binding Mg2+ and Ca2+ ions. While its biological function remains largely unclear, an increasing number of studies have shown that CIB2 is an essential component of the mechano-transduction machinery that operates in cochlear hair cells. Mutations in the gene encoding CIB2 have been associated with non-syndromic deafness. In addition to playing an important role in the physiology of hearing, CIB2 has been implicated in a multitude of very different processes, ranging from integrin signaling in platelets and skeletal muscle to autophagy, suggesting extensive functional plasticity. In this review, we summarize the current understanding of biochemical and biophysical properties of CIB2 and the biological roles that have been proposed for the protein in a variety of processes. We also highlight the many molecular aspects that remain unclarified and deserve further investigation.
Collapse
|
67
|
Perez-Flores MC, Verschooten E, Lee JH, Kim HJ, Joris PX, Yamoah EN. Intrinsic mechanical sensitivity of mammalian auditory neurons as a contributor to sound-driven neural activity. eLife 2022; 11:74948. [PMID: 35266451 PMCID: PMC8942473 DOI: 10.7554/elife.74948] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 03/09/2022] [Indexed: 11/18/2022] Open
Abstract
Mechanosensation – by which mechanical stimuli are converted into a neuronal signal – is the basis for the sensory systems of hearing, balance, and touch. Mechanosensation is unmatched in speed and its diverse range of sensitivities, reaching its highest temporal limits with the sense of hearing; however, hair cells (HCs) and the auditory nerve (AN) serve as obligatory bottlenecks for sounds to engage the brain. Like other sensory neurons, auditory neurons use the canonical pathway for neurotransmission and millisecond-duration action potentials (APs). How the auditory system utilizes the relatively slow transmission mechanisms to achieve ultrafast speed, and high audio-frequency hearing remains an enigma. Here, we address this paradox and report that the mouse, and chinchilla, AN are mechanically sensitive, and minute mechanical displacement profoundly affects its response properties. Sound-mimicking sinusoidal mechanical and electrical current stimuli affect phase-locked responses. In a phase-dependent manner, the two stimuli can also evoke suppressive responses. We propose that mechanical sensitivity interacts with synaptic responses to shape responses in the AN, including frequency tuning and temporal phase locking. Combining neurotransmission and mechanical sensation to control spike patterns gives the mammalian AN a secondary receptor role, an emerging theme in primary neuronal functions.
Collapse
Affiliation(s)
| | - Eric Verschooten
- Laboratory of Auditory Neurophysiology, University of Leuven, Leuven, Belgium
| | | | | | - Philip X Joris
- Laboratory of Auditory Neurophysiology, University of Leuven, Leuven, Belgium
| | | |
Collapse
|
68
|
Li J, Liu C, Kaefer S, Youssef M, Zhao B. The Mechanotransduction Channel and Organic Cation Transporter Are Critical for Cisplatin Ototoxicity in Murine Hair Cells. Front Mol Neurosci 2022; 15:835448. [PMID: 35221917 PMCID: PMC8866953 DOI: 10.3389/fnmol.2022.835448] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/10/2022] [Indexed: 11/17/2022] Open
Abstract
Cisplatin is one of the most widely used chemotherapeutic drugs across the world. However, the serious ototoxic effects, leading to permanent hair cell death and hearing loss, significantly limit the utility of cisplatin. In zebrafish, the functional mechanotransduction channel is required for cisplatin ototoxicity. However, it is still unclear the extent to which the mechanotransduction channel is involved in cisplatin uptake and ototoxicity in mammalian hair cells. Herein, we show that genetically disrupting mechanotransduction in mouse partially protects hair cells from cisplatin-induced hair cell death. Using a fluorescent-dye conjugated cisplatin, we monitored cisplatin uptake in cochlear explants and found that functional mechanotransduction is required for the uptake of cisplatin in murine hair cells. In addition, cimetidine, an inhibitor of the organic cation transporter, also partially protects hair cells from cisplatin ototoxicity. Notably, the otoprotective effects of cimetidine do not require mechanotransduction. These findings suggest that both the mechanotransduction channel and the organic cation transporter are critical for cisplatin ototoxicity in murine hair cells.
Collapse
|
69
|
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
|
70
|
Holt JR, Tobin M, Elferich J, Gouaux E, Ballesteros A, Yan Z, Ahmed ZM, Nicolson T. Putting the Pieces Together: the Hair Cell Transduction Complex. J Assoc Res Otolaryngol 2021; 22:601-608. [PMID: 34617206 PMCID: PMC8599550 DOI: 10.1007/s10162-021-00808-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/16/2021] [Indexed: 11/30/2022] Open
Abstract
Identification of the components of the mechanosensory transduction complex in hair cells has been a major research interest for many auditory and vestibular scientists and has attracted attention from outside the field. The past two decades have witnessed a number of significant advances with emergence of compelling evidence implicating at least a dozen distinct molecular components of the transduction machinery. Yet, how the pieces of this ensemble fit together and function in harmony to enable the senses of hearing and balance has not been clarified. The goal of this review is to summarize a 2021 symposium presented at the annual mid-winter meeting of the Association for Research in Otolaryngology. The symposium brought together the latest insights from within and beyond the field to examine individual components of the transduction complex and how these elements interact at molecular, structural, and biophysical levels to gate mechanosensitive channels and initiate sensory transduction in the inner ear. The review includes a brief historical background to set the stage for topics to follow that focus on structure, properties, and interactions of proteins such as CDH23, PCDH15, LHFPL5, TMIE, TMC1/2, and CIB2/3. We aim to present the diversity of ideas in this field and highlight emerging theories and concepts. This review will not only provide readers with a deeper appreciation of the components of the transduction apparatus and how they function together, but also bring to light areas of broad agreement, areas of scientific controversy, and opportunities for future scientific discovery.
Collapse
Affiliation(s)
- Jeffrey R Holt
- Departments of Otolaryngology & Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| | - Mélanie Tobin
- Laboratoire Physico-Chimie Curie, Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Paris, 75005, France
- Departments of Otorhinolaryngology, Neuroscience and Neurology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Johannes Elferich
- Vollum Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Eric Gouaux
- Vollum Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
- Howard Hughes Medical Institute, Vollum Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Angela Ballesteros
- Molecular Physiology and Biophysics Section, Disorders and Stroke, National Institute of Neurological, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Zhiqiang Yan
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Zubair M Ahmed
- Department of Otolaryngology Head & Neck Surgery, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA
| | - Teresa Nicolson
- Department of Otolaryngology-Head & Neck Surgery, Stanford University, Stanford, CA, USA
| |
Collapse
|
71
|
Sun S, Li S, Luo Z, Ren M, He S, Wang G, Liu Z. Dual expression of Atoh1 and Ikzf2 promotes transformation of adult cochlear supporting cells into outer hair cells. eLife 2021; 10:66547. [PMID: 34477109 PMCID: PMC8439656 DOI: 10.7554/elife.66547] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 09/02/2021] [Indexed: 12/24/2022] Open
Abstract
Mammalian cochlear outer hair cells (OHCs) are essential for hearing. Severe hearing impairment follows OHC degeneration. Previous attempts at regenerating new OHCs from cochlear supporting cells (SCs) have been unsuccessful, notably lacking expression of the key OHC motor protein, Prestin. Thus, regeneration of Prestin+ OHCs represents a barrier to restore auditory function in vivo. Here, we reported the successful in vivo conversion of adult mouse cochlear SCs into Prestin+ OHC-like cells through the concurrent induction of two key transcriptional factors known to be necessary for OHC development: Atoh1 and Ikzf2. Single-cell RNA sequencing revealed the upregulation of 729 OHC genes and downregulation of 331 SC genes in OHC-like cells. The resulting differentiation status of these OHC-like cells was much more advanced than previously achieved. This study thus established an efficient approach to induce the regeneration of Prestin+ OHCs, paving the way for in vivo cochlear repair via SC transdifferentiation.
Collapse
Affiliation(s)
- Suhong Sun
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Shuting Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhengnan Luo
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Minhui Ren
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Shunji He
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Guangqin Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhiyong Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| |
Collapse
|
72
|
Yan K, Zong W, Du H, Zhai X, Ren R, Liu S, Xiong W, Wang Y, Xu Z. BAIAP2L2 is required for the maintenance of mechanotransducing stereocilia of cochlear hair cells. J Cell Physiol 2021; 237:774-788. [PMID: 34346063 DOI: 10.1002/jcp.30545] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/21/2021] [Accepted: 07/24/2021] [Indexed: 01/05/2023]
Abstract
Stereocilia are actin-based cell protrusions of inner ear hair cells that play an essential role in mechano-electrical transduction (MET). Stereocilia are organized into several rows of increasing heights with the MET protein complex localized at the tips of shorter row stereocilia. At the tips of shorter row mechanotransducing stereocilia also resides a so-called "row 2 protein complex" whose dysfunction causes degeneration of the mechanotransducing stereocilia. In the present work, we show that BAIAP2L2 is localized at the tips of shorter row stereocilia in neonatal and adult mouse cochlear hair cells. Baiap2l2 inactivation causes degeneration of the mechanotransducing stereocilia, which eventually leads to profound hearing loss in mice of either sex. Consistently, electrophysiology and FM 1-43FX dye uptake results confirm that MET currents are compromised in Baiap2l2 knockout mice. Moreover, BAIAP2L2 binds to known row 2 complex components EPS8L2, TWF2, and CAPZB2, and the stereociliary tip localization of CAPZB2 is dependent on functional BAIAP2L2. Interestingly, BAIAP2L2 also binds to CIB2, a known MET complex component, and the stereociliary tip localization of BAIAP2L2 is abolished in Cib2 knockout mice. In conclusion, our present data suggest that BAIAP2L2 is a row 2 complex component, and is required for the maintenance of mechanotransducing stereocilia. Meanwhile, specific MET components such as CIB2 might play a direct role in stereocilia maintenance through binding to BAIAP2L2.
Collapse
Affiliation(s)
- Keji Yan
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, Shandong, China
| | - Wen Zong
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong, China
| | - Haibo Du
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, Shandong, China
| | - Xiaoyan Zhai
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, Shandong, China
| | - Rui Ren
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, Shandong, China
| | - Shuang Liu
- School of Life Sciences, IDG/McGovern Institute for Brain Research at Tsinghua, Tsinghua University, Beijing, China
| | - Wei Xiong
- School of Life Sciences, IDG/McGovern Institute for Brain Research at Tsinghua, Tsinghua University, Beijing, China
| | - Yanfei Wang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, Shandong, China
| | - Zhigang Xu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, Shandong, China.,Shandong Provincial Collaborative Innovation Center of Cell Biology, Shandong Normal University, Jinan, Shandong, China
| |
Collapse
|
73
|
Liang X, Qiu X, Dionne G, Cunningham CL, Pucak ML, Peng G, Kim YH, Lauer A, Shapiro L, Müller U. CIB2 and CIB3 are auxiliary subunits of the mechanotransduction channel of hair cells. Neuron 2021; 109:2131-2149.e15. [PMID: 34089643 PMCID: PMC8374959 DOI: 10.1016/j.neuron.2021.05.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 01/17/2021] [Accepted: 05/07/2021] [Indexed: 12/25/2022]
Abstract
CIB2 is a Ca2+- and Mg2+-binding protein essential for mechanoelectrical transduction (MET) by cochlear hair cells, but not by vestibular hair cells that co-express CIB2 and CIB3. Here, we show that in cochlear hair cells, CIB3 can functionally substitute for CIB2. Using X-ray crystallography, we demonstrate that CIB2 and CIB3 are structurally similar to KChIP proteins, auxiliary subunits of voltage-gated Kv4 channels. CIB2 and CIB3 bind to TMC1/2 through a domain in TMC1/2 flanked by transmembrane domains 2 and 3. The co-crystal structure of the CIB-binding domain in TMC1 with CIB3 reveals that interactions are mediated through a conserved CIB hydrophobic groove, similar to KChIP1 binding of Kv4. Functional studies in mice show that CIB2 regulates TMC1/2 localization and function in hair cells, processes that are affected by deafness-causing CIB2 mutations. We conclude that CIB2 and CIB3 are MET channel auxiliary subunits with striking similarity to Kv4 channel auxiliary subunits.
Collapse
Affiliation(s)
- Xiaoping Liang
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Xufeng Qiu
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Gilman Dionne
- Department of Biochemistry and Molecular Biophysics, Zuckerman Mind Brain, Department of Systems Biology, Columbia University, New York, NY 10032, USA
| | - Christopher L Cunningham
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michele L Pucak
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Guihong Peng
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ye-Hyun Kim
- Department of Otolaryngology-HNS, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Amanda Lauer
- Department of Otolaryngology-HNS, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lawrence Shapiro
- Department of Biochemistry and Molecular Biophysics, Zuckerman Mind Brain, Department of Systems Biology, Columbia University, New York, NY 10032, USA.
| | - Ulrich Müller
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
74
|
Fuster-García C, García-Bohórquez B, Rodríguez-Muñoz A, Aller E, Jaijo T, Millán JM, García-García G. Usher Syndrome: Genetics of a Human Ciliopathy. Int J Mol Sci 2021; 22:6723. [PMID: 34201633 PMCID: PMC8268283 DOI: 10.3390/ijms22136723] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/21/2022] Open
Abstract
Usher syndrome (USH) is an autosomal recessive syndromic ciliopathy characterized by sensorineural hearing loss, retinitis pigmentosa and, sometimes, vestibular dysfunction. There are three clinical types depending on the severity and age of onset of the symptoms; in addition, ten genes are reported to be causative of USH, and six more related to the disease. These genes encode proteins of a diverse nature, which interact and form a dynamic protein network called the "Usher interactome". In the organ of Corti, the USH proteins are essential for the correct development and maintenance of the structure and cohesion of the stereocilia. In the retina, the USH protein network is principally located in the periciliary region of the photoreceptors, and plays an important role in the maintenance of the periciliary structure and the trafficking of molecules between the inner and the outer segments of photoreceptors. Even though some genes are clearly involved in the syndrome, others are controversial. Moreover, expression of some USH genes has been detected in other tissues, which could explain their involvement in additional mild comorbidities. In this paper, we review the genetics of Usher syndrome and the spectrum of mutations in USH genes. The aim is to identify possible mutation associations with the disease and provide an updated genotype-phenotype correlation.
Collapse
Affiliation(s)
- Carla Fuster-García
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
- Biomedical Research Network for Rare Diseases, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - Belén García-Bohórquez
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
| | - Ana Rodríguez-Muñoz
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
| | - Elena Aller
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
- Biomedical Research Network for Rare Diseases, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
- Genetics Unit, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - Teresa Jaijo
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
- Biomedical Research Network for Rare Diseases, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
- Genetics Unit, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - José M. Millán
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
- Biomedical Research Network for Rare Diseases, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - Gema García-García
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
- Biomedical Research Network for Rare Diseases, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| |
Collapse
|
75
|
Sethna S, Scott PA, Giese APJ, Duncan T, Jian X, Riazuddin S, Randazzo PA, Redmond TM, Bernstein SL, Riazuddin S, Ahmed ZM. CIB2 regulates mTORC1 signaling and is essential for autophagy and visual function. Nat Commun 2021; 12:3906. [PMID: 34162842 PMCID: PMC8222345 DOI: 10.1038/s41467-021-24056-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 05/26/2021] [Indexed: 02/06/2023] Open
Abstract
Age-related macular degeneration (AMD) is a multifactorial neurodegenerative disorder. Although molecular mechanisms remain elusive, deficits in autophagy have been associated with AMD. Here we show that deficiency of calcium and integrin binding protein 2 (CIB2) in mice, leads to age-related pathologies, including sub-retinal pigment epithelium (RPE) deposits, marked accumulation of drusen markers APOE, C3, Aβ, and esterified cholesterol, and impaired visual function, which can be rescued using exogenous retinoids. Cib2 mutant mice exhibit reduced lysosomal capacity and autophagic clearance, and increased mTORC1 signaling-a negative regulator of autophagy. We observe concordant molecular deficits in dry-AMD RPE/choroid post-mortem human tissues. Mechanistically, CIB2 negatively regulates mTORC1 by preferentially binding to 'nucleotide empty' or inactive GDP-loaded Rheb. Upregulated mTORC1 signaling has been implicated in lymphangioleiomyomatosis (LAM) cancer. Over-expressing CIB2 in LAM patient-derived fibroblasts downregulates hyperactive mTORC1 signaling. Thus, our findings have significant implications for treatment of AMD and other mTORC1 hyperactivity-associated disorders.
Collapse
Affiliation(s)
- Saumil Sethna
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Patrick A Scott
- Department of Ophthalmology & Visual Sciences, University of Louisville, Louisville, KY, USA
| | - Arnaud P J Giese
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Todd Duncan
- Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xiaoying Jian
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sheikh Riazuddin
- Allama Iqbal Medical College, University of Health Sciences, Lahore, Pakistan
| | - Paul A Randazzo
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - T Michael Redmond
- Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Steven L Bernstein
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Saima Riazuddin
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Zubair M Ahmed
- Department of Otorhinolaryngology - Head & Neck Surgery, 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
|
76
|
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
|
77
|
Farhadi M, Razmara E, Balali M, Hajabbas Farshchi Y, Falah M. How Transmembrane Inner Ear (TMIE) plays role in the auditory system: A mystery to us. J Cell Mol Med 2021; 25:5869-5883. [PMID: 33987950 PMCID: PMC8256367 DOI: 10.1111/jcmm.16610] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 04/26/2021] [Indexed: 01/19/2023] Open
Abstract
Different cellular mechanisms contribute to the hearing sense, so it is obvious that any disruption in such processes leads to hearing impairment that greatly influences the global economy and quality of life of the patients and their relatives. In the past two decades, transmembrane inner ear (TMIE) protein has received a great deal of research interest because its impairments cause hereditary deafness in humans. This evolutionarily conserved membrane protein contributes to a fundamental complex that plays role in the maintenance and function of the sensory hair cells. Although the critical roles of the TMIE in mechanoelectrical transduction or hearing procedures have been discussed, there are little to no review papers summarizing the roles of the TMIE in the auditory system. In order to fill this gap, herein, we discuss the important roles of this protein in the auditory system including its role in mechanotransduction, olivocochlear synapse, morphology and different signalling pathways; we also review the genotype-phenotype correlation that can per se show the possible roles of this protein in the auditory system.
Collapse
Affiliation(s)
- Mohammad Farhadi
- ENT and Head and Neck Research Center and DepartmentThe Five Senses Health InstituteHazrat Rasoul Akram HospitalIran University of Medical SciencesTehranIran
| | - Ehsan Razmara
- Australian Regenerative Medicine InstituteMonash UniversityClaytonVICAustralia
| | - Maryam Balali
- ENT and Head and Neck Research Center and DepartmentThe Five Senses Health InstituteHazrat Rasoul Akram HospitalIran University of Medical SciencesTehranIran
| | - Yeganeh Hajabbas Farshchi
- Department of Cellular and Molecular BiologyTehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Masoumeh Falah
- ENT and Head and Neck Research Center and DepartmentThe Five Senses Health InstituteHazrat Rasoul Akram HospitalIran University of Medical SciencesTehranIran
| |
Collapse
|
78
|
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
|
79
|
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
|
80
|
de Joya EM, Colbert BM, Tang PC, Lam BL, Yang J, Blanton SH, Dykxhoorn DM, Liu X. Usher Syndrome in the Inner Ear: Etiologies and Advances in Gene Therapy. Int J Mol Sci 2021; 22:3910. [PMID: 33920085 PMCID: PMC8068832 DOI: 10.3390/ijms22083910] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/02/2021] [Accepted: 04/05/2021] [Indexed: 02/06/2023] Open
Abstract
Hearing loss is the most common sensory disorder with ~466 million people worldwide affected, representing about 5% of the population. A substantial portion of hearing loss is genetic. Hearing loss can either be non-syndromic, if hearing loss is the only clinical manifestation, or syndromic, if the hearing loss is accompanied by a collage of other clinical manifestations. Usher syndrome is a syndromic form of genetic hearing loss that is accompanied by impaired vision associated with retinitis pigmentosa and, in many cases, vestibular dysfunction. It is the most common cause of deaf-blindness. Currently cochlear implantation or hearing aids are the only treatments for Usher-related hearing loss. However, gene therapy has shown promise in treating Usher-related retinitis pigmentosa. Here we review how the etiologies of Usher-related hearing loss make it a good candidate for gene therapy and discuss how various forms of gene therapy could be applied to Usher-related hearing loss.
Collapse
Affiliation(s)
- Evan M. de Joya
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.M.J.); (B.M.C.); (P.-C.T.); (S.H.B.)
- Dr. John T. Macdonald Foundation Department of Human Genetics, John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Brett M. Colbert
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.M.J.); (B.M.C.); (P.-C.T.); (S.H.B.)
- Dr. John T. Macdonald Foundation Department of Human Genetics, John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- Medical Scientist Training Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Pei-Ciao Tang
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.M.J.); (B.M.C.); (P.-C.T.); (S.H.B.)
| | - Byron L. Lam
- Bascom Palmer Eye Institute, University of Miami School of Medicine, Miami, FL 33136, USA;
| | - Jun Yang
- John A. Moran Eye Center, Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT 84132, USA;
| | - Susan H. Blanton
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.M.J.); (B.M.C.); (P.-C.T.); (S.H.B.)
- Dr. John T. Macdonald Foundation Department of Human Genetics, John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Derek M. Dykxhoorn
- Dr. John T. Macdonald Foundation Department of Human Genetics, John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Xuezhong Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (E.M.J.); (B.M.C.); (P.-C.T.); (S.H.B.)
- Dr. John T. Macdonald Foundation Department of Human Genetics, John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
81
|
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
|
82
|
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
|
83
|
Abstract
Mechanosensing is a key feature through which organisms can receive inputs from the environment and convert them into specific functional and behavioral outputs. Mechanosensation occurs in many cells and tissues, regulating a plethora of molecular processes based on the distribution of forces and stresses both at the cell membrane and at the intracellular organelles levels, through complex interactions between cells’ microstructures, cytoskeleton, and extracellular matrix. Although several primary and secondary mechanisms have been shown to contribute to mechanosensation, a fundamental pathway in simple organisms and mammals involves the presence of specialized sensory neurons and the presence of different types of mechanosensitive ion channels on the neuronal cell membrane. In this contribution, we present a review of the main ion channels which have been proven to be significantly involved in mechanotransduction in neurons. Further, we discuss recent studies focused on the biological mechanisms and modeling of mechanosensitive ion channels’ gating, and on mechanotransduction modeling at different scales and levels of details.
Collapse
|
84
|
Zhu S, Chen Z, Wang H, McDermott BM. Tmc Reliance Is Biased by the Hair Cell Subtype and Position Within the Ear. Front Cell Dev Biol 2021; 8:570486. [PMID: 33490059 PMCID: PMC7817542 DOI: 10.3389/fcell.2020.570486] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 10/21/2020] [Indexed: 12/04/2022] Open
Abstract
Hair cells are heterogenous, enabling varied roles in sensory systems. An emerging hypothesis is that the transmembrane channel-like (Tmc) proteins of the hair cell’s mechanotransduction apparatus vary within and between organs to permit encoding of different mechanical stimuli. Five anatomical variables that may coincide with different Tmc use by a hair cell within the ear are the containing organ, cell morphology, cell position within an organ, axis of best sensitivity for the cell, and the hair bundle’s orientation within this axis. Here, we test this hypothesis in the organs of the zebrafish ear using a suite of genetic mutations. Transgenesis and quantitative measurements demonstrate two morphologically distinct hair cell types in the central thickness of a vestibular organ, the lateral crista: short and tall. In contrast to what has been observed, we find that tall hair cells that lack Tmc1 generally have substantial reductions in mechanosensitivity. In short hair cells that lack Tmc2 isoforms, mechanotransduction is largely abated. However, hair cell Tmc dependencies are not absolute, and an exceptional class of short hair cell that depends on Tmc1 is present, termed a short hair cell erratic. To further test anatomical variables that may influence Tmc use, we map Tmc1 function in the saccule of mutant larvae that depend just on this Tmc protein to hear. We demonstrate that hair cells that use Tmc1 are found in the posterior region of the saccule, within a single axis of best sensitivity, and hair bundles with opposite orientations retain function. Overall, we determine that Tmc reliance in the ear is dependent on the organ, subtype of hair cell, position within the ear, and axis of best sensitivity.
Collapse
Affiliation(s)
- Shaoyuan Zhu
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Case Western Reserve University, Cleveland, OH, United States.,Department of Biology, Case Western Reserve University, Cleveland, OH, United States
| | - Zongwei Chen
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Case Western Reserve University, Cleveland, OH, United States.,Department of Biology, Case Western Reserve University, Cleveland, OH, United States
| | - Haoming Wang
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Case Western Reserve University, Cleveland, OH, United States.,Department of Biology, Case Western Reserve University, Cleveland, OH, United States
| | - Brian M McDermott
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Case Western Reserve University, Cleveland, OH, United States.,Department of Biology, Case Western Reserve University, Cleveland, OH, United States.,Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, United States.,Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
85
|
Li S, Yan Z. Mechanotransduction Ion Channels in Hearing and Touch. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1349:371-385. [DOI: 10.1007/978-981-16-4254-8_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
86
|
Jiang F, Takagi Y, Shams A, Heissler SM, Friedman TB, Sellers JR, Bird JE. The ATPase mechanism of myosin 15, the molecular motor mutated in DFNB3 human deafness. J Biol Chem 2021; 296:100243. [PMID: 33372036 PMCID: PMC7948958 DOI: 10.1074/jbc.ra120.014903] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 11/18/2022] Open
Abstract
Cochlear hair cells each possess an exquisite bundle of actin-based stereocilia that detect sound. Unconventional myosin 15 (MYO15) traffics and delivers critical molecules required for stereocilia development and thus is essential for building the mechanosensory hair bundle. Mutations in the human MYO15A gene interfere with stereocilia trafficking and cause hereditary hearing loss, DFNB3, but the impact of these mutations is not known, as MYO15 itself is poorly characterized. To learn more, we performed a kinetic study of the ATPase motor domain to characterize its mechanochemical cycle. Using the baculovirus-Sf9 system, we purified a recombinant minimal motor domain (S1) by coexpressing the mouse MYO15 ATPase, essential and regulatory light chains that bind its IQ domains, and UNC45 and HSP90A chaperones required for correct folding of the ATPase. MYO15 purified with either UNC45A or UNC45B coexpression had similar ATPase activities (kcat = ∼ 6 s-1 at 20 °C). Using stopped-flow and quenched-flow transient kinetic analyses, we measured the major rate constants describing the ATPase cycle, including ATP, ADP, and actin binding; hydrolysis; and phosphate release. Actin-attached ADP release was the slowest measured transition (∼12 s-1 at 20 °C), although this did not rate-limit the ATPase cycle. The kinetic analysis shows the MYO15 motor domain has a moderate duty ratio (∼0.5) and weak thermodynamic coupling between ADP and actin binding. These findings are consistent with MYO15 being kinetically adapted for processive motility when oligomerized. Our kinetic characterization enables future studies into how deafness-causing mutations affect MYO15 and disrupt stereocilia trafficking necessary for hearing.
Collapse
Affiliation(s)
- Fangfang Jiang
- Department of Pharmacology and Therapeutics, and the Myology Institute, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Yasuharu Takagi
- Laboratory of Molecular Physiology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Arik Shams
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, USA
| | - Sarah M Heissler
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Thomas B Friedman
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, USA
| | - James R Sellers
- Laboratory of Molecular Physiology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jonathan E Bird
- Department of Pharmacology and Therapeutics, and the Myology Institute, University of Florida College of Medicine, Gainesville, Florida, USA.
| |
Collapse
|
87
|
Marcovich I, Holt JR. Evolution and function of Tmc genes in mammalian hearing. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.06.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
88
|
Wu CJ, Li X, Sommers CL, Kurima K, Huh S, Bugos G, Dong L, Li W, Griffith AJ, Samelson LE. Expression of a TMC6-TMC8-CIB1 heterotrimeric complex in lymphocytes is regulated by each of the components. J Biol Chem 2020; 295:16086-16099. [PMID: 32917726 DOI: 10.1074/jbc.ra120.013045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 09/09/2020] [Indexed: 11/06/2022] Open
Abstract
The TMC genes encode a set of homologous transmembrane proteins whose functions are not well understood. Biallelic mutations in either TMC6 or TMC8 are detected in more than half of cases of the pre-malignant skin disease epidermodysplasia verruciformis (EV). It is controversial whether EV induced by mutations in TMC6 or TMC8 originates from keratinocyte or lymphocyte defects. Quantification of TMC6 and TMC8 RNA levels in various organs revealed that lymphoid tissues have the highest levels of expression of both genes, and custom antibodies confirmed protein expression in mouse lymphocytes. To study the function of these proteins we generated mice with targeted deletion mutant alleles of Tmc6 or Tmc8 Either TMC6 or TMC8 deficiency induced a reduction in apparent molecular weight and/or amount of the other TMC molecule. Co-immunoprecipitation experiments indicated that TMC6 and TMC8 formed a protein complex in mouse and human T cells. MS and biochemical analysis demonstrated that TMC6 and TMC8 additionally interacted with the CIB1 protein to form TMC6-TMC8-CIB1 trimers. We demonstrated that TMC6 and TMC8 regulated CIB1 levels by protecting CIB1 from ubiquitination and proteasomal degradation. Reciprocally, CIB1 was needed for stabilizing TMC6 and TMC8 levels. These results suggest why inactivating mutations in any of the three human genes leads to similar clinical presentations. We also demonstrated that TMC6 and TMC8 levels are drastically lower and the proteins are less active in regulating CIB1 in keratinocytes than in T cells. Our study suggests that defects in lymphocytes may contribute to the etiology and pathogenesis of EV.
Collapse
Affiliation(s)
- Chuan-Jin Wu
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Xing Li
- Molecular Biology and Genetics Section, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Rockville, Maryland, USA
| | - Connie L Sommers
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Kiyoto Kurima
- Molecular Biology and Genetics Section, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Rockville, Maryland, USA
| | - Sunmee Huh
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Grace Bugos
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Lijin Dong
- Genetic Engineering Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Wenmei Li
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Andrew J Griffith
- Molecular Biology and Genetics Section, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Rockville, Maryland, USA
| | - Lawrence E Samelson
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.
| |
Collapse
|
89
|
Deafness mutation D572N of TMC1 destabilizes TMC1 expression by disrupting LHFPL5 binding. Proc Natl Acad Sci U S A 2020; 117:29894-29903. [PMID: 33168709 DOI: 10.1073/pnas.2011147117] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Transmembrane channel-like protein 1 (TMC1) and lipoma HMGIC fusion partner-like 5 (LHFPL5) are recognized as two critical components of the mechanotransduction complex in inner-ear hair cells. However, the physical and functional interactions of TMC1 and LHFPL5 remain largely unexplored. We examined the interaction between TMC1 and LHFPL5 by using multiple approaches, including our recently developed ultrasensitive microbead-based single-molecule pulldown (SiMPull) assay. We demonstrate that LHFPL5 physically interacts with and stabilizes TMC1 in both heterologous expression systems and in the soma and hair bundle of hair cells. Moreover, the semidominant deafness mutation D572N in human TMC1 (D569N in mouse TMC1) severely disrupted LHFPL5 binding and destabilized TMC1 expression. Thus, our findings reveal previously unrecognized physical and functional interactions of TMC1 and LHFPL5 and provide insights into the molecular mechanism by which the D572N mutation causes deafness. Notably, these findings identify a missing link in the currently known physical organization of the mechanotransduction macromolecular complex. Furthermore, this study has demonstrated the power of the microbead-based SiMPull assay for biochemical investigation of rare cells such as hair cells.
Collapse
|
90
|
George SS, Steele CR, Ricci AJ. Rat Auditory Inner Hair Cell Mechanotransduction and Stereociliary Membrane Diffusivity Are Similarly Modulated by Calcium. iScience 2020; 23:101773. [PMID: 33294782 PMCID: PMC7689183 DOI: 10.1016/j.isci.2020.101773] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/03/2020] [Accepted: 11/03/2020] [Indexed: 11/16/2022] Open
Abstract
The lipid bilayer plays a pivotal role in force transmission to many mechanically-gated channels. We developed the technology to monitor membrane diffusivity in order to test the hypothesis positing that Ca2+ regulates open probability (P o) of cochlear hair cell mechanotransduction (MET) channels via the plasma membrane. The stereociliary membrane was more diffusive (9x) than the basolateral membrane. Elevating intracellular Ca2+ buffering or lowering extracellular Ca2+ reduced stereociliary diffusivity and increased MET P o. In contrast, prolonged depolarization increased stereociliary diffusivity and reduced MET P o. No comparable effects were noted for soma measurements. Although MET channels are located in the shorter stereocilia rows, both rows had similar baseline diffusivity and showed similar responses to Ca2+ manipulations and MET channel blocks, suggesting that diffusivity is independent of MET. Together, these data suggest that the stereociliary membrane is a component of a calcium-modulated viscoelastic-like element regulating hair cell mechanotransduction.
Collapse
Affiliation(s)
- Shefin S George
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University, 240 Pasteur Drive, Stanford, CA 94305, USA
| | - Charles R Steele
- Department of Mechanical Engineering, Building 520, 440 Escondido Mall, Stanford University, CA 94305, USA
| | - Anthony J Ricci
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Stanford University, 240 Pasteur Drive, Stanford, CA 94305, USA.,Department of Molecular and Cellular Physiology, School of Medicine, Stanford University, 291 Campus Drive, Stanford, CA 94305, USA
| |
Collapse
|
91
|
Whatley M, Francis A, Ng ZY, Khoh XE, Atlas MD, Dilley RJ, Wong EYM. Usher Syndrome: Genetics and Molecular Links of Hearing Loss and Directions for Therapy. Front Genet 2020; 11:565216. [PMID: 33193648 PMCID: PMC7642844 DOI: 10.3389/fgene.2020.565216] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/21/2020] [Indexed: 12/19/2022] Open
Abstract
Usher syndrome (USH) is an autosomal recessive (AR) disorder that permanently and severely affects the senses of hearing, vision, and balance. Three clinically distinct types of USH have been identified, decreasing in severity from Type 1 to 3, with symptoms of sensorineural hearing loss (SNHL), retinitis pigmentosa (RP), and vestibular dysfunction. There are currently nine confirmed and two suspected USH-causative genes, and a further three candidate loci have been mapped. The proteins encoded by these genes form complexes that play critical roles in the development and maintenance of cellular structures within the inner ear and retina, which have minimal capacity for repair or regeneration. In the cochlea, stereocilia are located on the apical surface of inner ear hair cells (HC) and are responsible for transducing mechanical stimuli from sound pressure waves into chemical signals. These signals are then detected by the auditory nerve fibers, transmitted to the brain and interpreted as sound. Disease-causing mutations in USH genes can destabilize the tip links that bind the stereocilia to each other, and cause defects in protein trafficking and stereocilia bundle morphology, thereby inhibiting mechanosensory transduction. This review summarizes the current knowledge on Usher syndrome with a particular emphasis on mutations in USH genes, USH protein structures, and functional analyses in animal models. Currently, there is no cure for USH. However, the genetic therapies that are rapidly developing will benefit from this compilation of detailed genetic information to identify the most effective strategies for restoring functional USH proteins.
Collapse
Affiliation(s)
- Meg Whatley
- Ear Science Institute Australia, Nedlands, WA, Australia
| | - Abbie Francis
- Ear Science Institute Australia, Nedlands, WA, Australia
- Emergency Medicine, The University of Western Australia, Nedlands, WA, Australia
| | - Zi Ying Ng
- Ear Science Institute Australia, Nedlands, WA, Australia
| | - Xin Ee Khoh
- Ear Science Institute Australia, Nedlands, WA, Australia
- School of Human Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Marcus D. Atlas
- Ear Science Institute Australia, Nedlands, WA, Australia
- Ear Sciences Centre, The University of Western Australia, Nedlands, WA, Australia
| | - Rodney J. Dilley
- Ear Science Institute Australia, Nedlands, WA, Australia
- Ear Sciences Centre, The University of Western Australia, Nedlands, WA, Australia
- Centre for Cell Therapy and Regenerative Medicine, The University of Western Australia, Perth, WA, Australia
| | - Elaine Y. M. Wong
- Ear Science Institute Australia, Nedlands, WA, Australia
- Ear Sciences Centre, The University of Western Australia, Nedlands, WA, Australia
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin University, Bentley, WA, Australia
| |
Collapse
|
92
|
Nolen RM, Hufnagel RB, Friedman TB, Turriff AE, Brewer CC, Zalewski CK, King KA, Wafa TT, Griffith AJ, Brooks BP, Zein WM. Atypical and ultra-rare Usher syndrome: a review. Ophthalmic Genet 2020; 41:401-412. [PMID: 32372680 DOI: 10.1080/13816810.2020.1747090] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Usher syndrome has classically been described as a combination of hearing loss and rod-cone dystrophy; vestibular dysfunction is present in many patients. Three distinct clinical subtypes were documented in the late 1970s. Genotyping efforts have led to the identification of several genes associated with the disease. Recent literature has seen multiple publications referring to "atypical" Usher syndrome presentations. This manuscript reviews the molecular etiology of Usher syndrome, highlighting rare presentations and molecular causes. Reports of "atypical" disease are summarized noting the wide discrepancy in the spectrum of phenotypic deviations from the classical presentation. Guidelines for establishing a clear nomenclature system are suggested.
Collapse
Affiliation(s)
- Rosalie M Nolen
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health , Bethesda, MD, USA
| | - Robert B Hufnagel
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health , Bethesda, MD, USA
| | - Thomas B Friedman
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health , Bethesda, MD, USA
| | - Amy E Turriff
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health , Bethesda, MD, USA
| | - Carmen C Brewer
- Otolaryngology Branch, National Institute of Deafness and Other Communication Disorders, National Institutes of Health , Bethesda, MD, USA
| | - Christopher K Zalewski
- Otolaryngology Branch, National Institute of Deafness and Other Communication Disorders, National Institutes of Health , Bethesda, MD, USA
| | - Kelly A King
- Otolaryngology Branch, National Institute of Deafness and Other Communication Disorders, National Institutes of Health , Bethesda, MD, USA
| | - Talah T Wafa
- Otolaryngology Branch, National Institute of Deafness and Other Communication Disorders, National Institutes of Health , Bethesda, MD, USA
| | - Andrew J Griffith
- Otolaryngology Branch, National Institute of Deafness and Other Communication Disorders, National Institutes of Health , Bethesda, MD, USA
| | - Brian P Brooks
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health , Bethesda, MD, USA
| | - Wadih M Zein
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health , Bethesda, MD, USA
| |
Collapse
|
93
|
Li S, Mecca A, Kim J, Caprara GA, Wagner EL, Du TT, Petrov L, Xu W, Cui R, Rebustini IT, Kachar B, Peng AW, Shin JB. Myosin-VIIa is expressed in multiple isoforms and essential for tensioning the hair cell mechanotransduction complex. Nat Commun 2020; 11:2066. [PMID: 32350269 PMCID: PMC7190839 DOI: 10.1038/s41467-020-15936-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 04/01/2020] [Indexed: 11/09/2022] Open
Abstract
Mutations in myosin-VIIa (MYO7A) cause Usher syndrome type 1, characterized by combined deafness and blindness. MYO7A is proposed to function as a motor that tensions the hair cell mechanotransduction (MET) complex, but conclusive evidence is lacking. Here we report that multiple MYO7A isoforms are expressed in the mouse cochlea. In mice with a specific deletion of the canonical isoform (Myo7a-ΔC mouse), MYO7A is severely diminished in inner hair cells (IHCs), while expression in outer hair cells is affected tonotopically. IHCs of Myo7a-ΔC mice undergo normal development, but exhibit reduced resting open probability and slowed onset of MET currents, consistent with MYO7A's proposed role in tensioning the tip link. Mature IHCs of Myo7a-ΔC mice degenerate over time, giving rise to progressive hearing loss. Taken together, our study reveals an unexpected isoform diversity of MYO7A expression in the cochlea and highlights MYO7A's essential role in tensioning the hair cell MET complex.
Collapse
Affiliation(s)
- Sihan Li
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA.,Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
| | - Andrew Mecca
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jeewoo Kim
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Giusy A Caprara
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Elizabeth L Wagner
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA.,Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
| | - Ting-Ting Du
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Leonid Petrov
- Department of Mathematics, University of Virginia, Charlottesville, VA, USA
| | - Wenhao Xu
- Genetically Engineered Murine Model (GEMM) Core, University of Virginia, Charlottesville, VA, USA
| | - Runjia Cui
- National Institute for Deafness and Communications Disorders, National Institute of Health, Bethesda, MD, USA
| | - Ivan T Rebustini
- National Institute for Deafness and Communications Disorders, National Institute of Health, Bethesda, MD, USA
| | - Bechara Kachar
- National Institute for Deafness and Communications Disorders, National Institute of Health, Bethesda, MD, USA
| | - Anthony W Peng
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Jung-Bum Shin
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA. .,Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
94
|
Xu Z, Miyata H, Kaneda Y, Castaneda JM, Lu Y, Morohoshi A, Yu Z, Matzuk MM, Ikawa M. CIB4 is essential for the haploid phase of spermatogenesis in mice†. Biol Reprod 2020; 103:235-243. [PMID: 32430498 PMCID: PMC7401386 DOI: 10.1093/biolre/ioaa059] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/17/2020] [Accepted: 04/22/2020] [Indexed: 02/03/2023] Open
Abstract
Spermatogenesis is a complex developmental process that involves the proliferation of diploid cells, meiotic division, and haploid differentiation. Many genes are shown to be essential for male fertility using knockout (KO) mice; however, there still remain genes to be analyzed to elucidate their molecular mechanism and their roles in spermatogenesis. Calcium- and integrin-binding protein 1 (CIB1) is a ubiquitously expressed protein that possesses three paralogs: CIB2, CIB3, and CIB4. It is reported that Cib1 KO male mice are sterile due to impaired haploid differentiation. In this study, we discovered that Cib4 is expressed strongly in mouse and human testis and begins expression during the haploid phase of spermatogenesis in mice. To analyze the function of CIB4 in vivo, we generated Cib4 KO mice using the CRISPR/Cas9 system. Cib4 KO male mice are sterile due to impaired haploid differentiation, phenocopying Cib1 KO male mice. Spermatogenic cells isolated from seminiferous tubules demonstrate an essential function of CIB4 in the formation of the apical region of the sperm head. Further analysis of CIB4 function may shed light on the etiology of male infertility caused by spermatogenesis defects, and CIB4 could be a target for male contraceptives because of its dominant expression in the testis.
Collapse
Affiliation(s)
- Zoulan Xu
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.,Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Haruhiko Miyata
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Yuki Kaneda
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.,Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Julio M Castaneda
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Yonggang Lu
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Akane Morohoshi
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.,Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Zhifeng Yu
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, USA.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Martin M Matzuk
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, USA.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Masahito Ikawa
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.,Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan.,Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| |
Collapse
|
95
|
Tang YQ, Lee SA, Rahman M, Vanapalli SA, Lu H, Schafer WR. Ankyrin Is An Intracellular Tether for TMC Mechanotransduction Channels. Neuron 2020; 107:112-125.e10. [PMID: 32325031 PMCID: PMC7343241 DOI: 10.1016/j.neuron.2020.03.026] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 02/17/2020] [Accepted: 03/24/2020] [Indexed: 12/15/2022]
Abstract
Mechanotransduction channels have been proposed as force sensors in various physiological processes, such as hearing and touch. In particular, TMC1 has been shown to constitute the pore of hair cell mechanotransduction channels, but little is known about how force is sensed by TMC channels. Here, we identify UNC-44/ankyrin as an essential component of the TMC-1 mechanotransduction channel complex in the sensory cilia of Caenorhabditis elegans mechanoreceptor neurons. Ankyrin binds indirectly to TMC-1 via evolutionarily conserved CIB proteins, which are required for TMC-1-mediated mechanosensation in C. elegans OLQ neurons and body wall muscles. Mechanosensory activity conferred by ectopically expressed TMCs in mechanoinsensitive neurons depends on both ankyrin and CIB proteins, indicating that the ankyrin-CIB subcomplex is required for TMC mechanosensitivity. Our work indicates that ankyrin is a long-sought intracellular tether that transmits force to TMC mechanotransduction channels. TMC-1 functions as a mechanosensor in C. elegans neurons and muscles UNC-44/ankyrin binds indirectly to TMC-1 via CALM-1 CALM-1 and ankyrin are required for TMC-1-mediated mechanosensation Ankyrin acts as an intracellular tether to confer mechanosensitivity to TMC channels
Collapse
Affiliation(s)
- Yi-Quan Tang
- Neurobiology Division, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, UK.
| | - Sol Ah Lee
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332-0100, USA
| | - Mizanur Rahman
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX, USA
| | - Siva A Vanapalli
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX, USA
| | - Hang Lu
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332-0100, USA
| | - William R Schafer
- Neurobiology Division, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, UK; Department of Biology, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
96
|
Myosin-XVa Controls Both Staircase Architecture and Diameter Gradation of Stereocilia Rows in the Auditory Hair Cell Bundles. J Assoc Res Otolaryngol 2020; 21:121-135. [PMID: 32152769 DOI: 10.1007/s10162-020-00745-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 02/04/2020] [Indexed: 02/08/2023] Open
Abstract
Mammalian hair cells develop their mechanosensory bundles through consecutive phases of stereocilia elongation, thickening, and retraction of supernumerary stereocilia. Many molecules involved in stereocilia elongation have been identified, including myosin-XVa. Significantly less is known about molecular mechanisms of stereocilia thickening and retraction. Here, we used scanning electron microscopy (SEM) to quantify postnatal changes in number and diameters of the auditory hair cell stereocilia in shaker-2 mice (Myo15sh2) that lack both "long" and "short" isoforms of myosin-XVa, and in mice lacking only the "long" myosin-XVa isoform (Myo15∆N). Previously, we observed large mechanotransduction current in young postnatal inner (IHC) and outer (OHC) hair cells of both these strains. Stereocilia counts showed nearly identical developmental retraction of supernumerary stereocilia in control heterozygous, Myo15sh2/sh2, and Myo15∆N/∆N mice, suggesting that this retraction is largely unaffected by myosin-XVa deficiency. However, myosin-XVa deficiency does affect stereocilia diameters. In control, the first (tallest) and second row stereocilia grow in diameter simultaneously. However, the third row stereocilia in IHCs grow only until postnatal day 1-2 and then become thinner. In OHCs, they also grow slower than taller stereocilia, forming a stereocilia diameter gradation within a hair bundle. The sh2 mutation disrupts this gradation and makes all stereocilia nearly identical in thickness in both IHCs and OHCs, with only subtle residual diameter differences. All Myo15sh2/sh2 stereocilia grow postnatally including the third row, which is not a part of normal development. Serial sections with focused ion beam (FIB)-SEM confirmed that diameter changes of Myo15sh2/sh2 IHC and OHC stereocilia resulted from corresponding changes of their actin cores. In contrast to Myo15sh2/sh2, Myo15∆N/∆N hair cells develop prominent stereocilia diameter gradation. Thus, besides building the staircase, the short isoform of myosin-XVa is essential for controlling the diameter of the third row stereocilia and formation of the stereocilia diameter gradation in a hair bundle.
Collapse
|
97
|
Lee S, Dondzillo A, Gubbels SP, Raphael Y. Practical aspects of inner ear gene delivery for research and clinical applications. Hear Res 2020; 394:107934. [PMID: 32204962 DOI: 10.1016/j.heares.2020.107934] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 02/26/2020] [Accepted: 02/28/2020] [Indexed: 12/24/2022]
Abstract
The application of gene therapy is widely expanding in research and continuously improving in preparation for clinical applications. The inner ear is an attractive target for gene therapy for treating environmental and genetic diseases in both the auditory and vestibular systems. With the lack of spontaneous cochlear hair cell replacement, hair cell regeneration in adult mammals is among the most important goals of gene therapy. In addition, correcting gene defects can open up a new era for treating inner ear diseases. The relative isolation and small size of the inner ear dictate local administration routes and carefully calculated small volumes of reagents. In the current review, we will cover effective timing, injection routes and types of vectors for successful gene delivery to specific target cells within the inner ear. Differences between research purposes and clinical applications are also discussed.
Collapse
Affiliation(s)
- Sungsu Lee
- Kresge Hearing Research Institute, Department of Otolaryngology, Head and Neck Surgery, Michigan Medicine, Ann Arbor, MI, USA
| | - Anna Dondzillo
- Department of Otolaryngology, Head and Neck Surgery, University of Colorado School of Medicine, Aurora, CO, USA
| | - Samuel P Gubbels
- Department of Otolaryngology, Head and Neck Surgery, University of Colorado School of Medicine, Aurora, CO, USA
| | - Yehoash Raphael
- Kresge Hearing Research Institute, Department of Otolaryngology, Head and Neck Surgery, Michigan Medicine, Ann Arbor, MI, USA.
| |
Collapse
|
98
|
Erickson T, Pacentine IV, Venuto A, Clemens R, Nicolson T. The lhfpl5 Ohnologs lhfpl5a and lhfpl5b Are Required for Mechanotransduction in Distinct Populations of Sensory Hair Cells in Zebrafish. Front Mol Neurosci 2020; 12:320. [PMID: 32009898 PMCID: PMC6974483 DOI: 10.3389/fnmol.2019.00320] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 12/16/2019] [Indexed: 01/20/2023] Open
Abstract
Hair cells sense and transmit auditory, vestibular, and hydrodynamic information by converting mechanical stimuli into electrical signals. This process of mechano-electrical transduction (MET) requires a mechanically gated channel localized in the apical stereocilia of hair cells. In mice, lipoma HMGIC fusion partner-like 5 (LHFPL5) acts as an auxiliary subunit of the MET channel whose primary role is to correctly localize PCDH15 and TMC1 to the mechanotransduction complex. Zebrafish have two lhfpl5 genes (lhfpl5a and lhfpl5b), but their individual contributions to MET channel assembly and function have not been analyzed. Here we show that the zebrafish lhfpl5 genes are expressed in discrete populations of hair cells: lhfpl5a expression is restricted to auditory and vestibular hair cells in the inner ear, while lhfpl5b expression is specific to hair cells of the lateral line organ. Consequently, lhfpl5a mutants exhibit defects in auditory and vestibular function, while disruption of lhfpl5b affects hair cells only in the lateral line neuromasts. In contrast to previous reports in mice, localization of Tmc1 does not depend upon Lhfpl5 function in either the inner ear or lateral line organ. In both lhfpl5a and lhfpl5b mutants, GFP-tagged Tmc1 and Tmc2b proteins still localize to the stereocilia of hair cells. Using a stably integrated GFP-Lhfpl5a transgene, we show that the tip link cadherins Pcdh15a and Cdh23, along with the Myo7aa motor protein, are required for correct Lhfpl5a localization at the tips of stereocilia. Our work corroborates the evolutionarily conserved co-dependence between Lhfpl5 and Pcdh15, but also reveals novel requirements for Cdh23 and Myo7aa to correctly localize Lhfpl5a. In addition, our data suggest that targeting of Tmc1 and Tmc2b proteins to stereocilia in zebrafish hair cells occurs independently of Lhfpl5 proteins.
Collapse
Affiliation(s)
- Timothy Erickson
- Department of Biology, East Carolina University, Greenville, NC, United States.,Oregon Hearing Research Center and Vollum Institute, Oregon Health and Science University, Portland, OR, United States
| | - Itallia V Pacentine
- Oregon Hearing Research Center and Vollum Institute, Oregon Health and Science University, Portland, OR, United States
| | - Alexandra Venuto
- Department of Biology, East Carolina University, Greenville, NC, United States
| | - Rachel Clemens
- Oregon Hearing Research Center and Vollum Institute, Oregon Health and Science University, Portland, OR, United States
| | - Teresa Nicolson
- Oregon Hearing Research Center and Vollum Institute, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
99
|
Richardson GP, Petit C. Hair-Bundle Links: Genetics as the Gateway to Function. Cold Spring Harb Perspect Med 2019; 9:cshperspect.a033142. [PMID: 30617060 DOI: 10.1101/cshperspect.a033142] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Up to five distinct cell-surface specializations interconnect the stereocilia and the kinocilium of the mature hair bundle in some species: kinocilial links, tip links, top connectors, shaft connectors, and ankle links. In developing hair bundles, transient lateral links are prominent. Mutations in genes encoding proteins associated with these links cause Usher deafness/blindness syndrome or nonsyndromic (isolated) forms of human hereditary deafness, and mice with constitutive or conditional alleles of these genes have provided considerable insight into the molecular composition and function of the different links. We describe the structure of these links and review evidence showing CDH23 and PCDH15 are components of the tip, kinocilial, and transient-lateral links, that stereocilin (STRC) and protein tyrosine phosphatase (PTPRQ) are associated with top and shaft connectors, respectively, and that USH2A and ADGRV1 are associated with the ankle links. Whereas tip links are required for mechanoelectrical transduction, all link proteins play key roles in the normal development and/or the maintenance of hair bundle structure and function. Recent crystallographic and single-particle analyses of PCDH15 and CDH23 provide insight as to how the structure of tip link may contribute to the elastic element predicted to lie in series with the hair cell's mechanoelectrical transducer channel.
Collapse
Affiliation(s)
- Guy P Richardson
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, United Kingdom
| | - Christine Petit
- Institut Pasteur, 75724 Paris Cedex 15, France.,Collège de France, 75231 Paris Cedex 05, France
| |
Collapse
|
100
|
Jia Y, Zhao Y, Kusakizako T, Wang Y, Pan C, Zhang Y, Nureki O, Hattori M, Yan Z. TMC1 and TMC2 Proteins Are Pore-Forming Subunits of Mechanosensitive Ion Channels. Neuron 2019; 105:310-321.e3. [PMID: 31761710 DOI: 10.1016/j.neuron.2019.10.017] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 09/05/2019] [Accepted: 10/09/2019] [Indexed: 11/28/2022]
Abstract
Transmembrane channel-like (TMC) 1 and 2 are required for the mechanotransduction of mouse inner ear hair cells and localize to the site of mechanotransduction in mouse hair cell stereocilia. However, it remains unclear whether TMC1 and TMC2 are indeed ion channels and whether they can sense mechanical force directly. Here we express TMC1 from the green sea turtle (CmTMC1) and TMC2 from the budgerigar (MuTMC2) in insect cells, purify and reconstitute the proteins, and show that liposome-reconstituted CmTMC1 and MuTMC2 proteins possess ion channel activity. Furthermore, by applying pressure to proteoliposomes, we demonstrate that both CmTMC1 and MuTMC2 proteins can indeed respond to mechanical stimuli. In addition, CmTMC1 mutants corresponding to human hearing loss mutants exhibit reduced or no ion channel activity. Taken together, our results show that the CmTMC1 and MuTMC2 proteins are pore-forming subunits of mechanosensitive ion channels, supporting TMC1 and TMC2 as hair cell transduction channels.
Collapse
Affiliation(s)
- Yanyan Jia
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurosurgery at Huashan Hospital, Human Phenome Institute, Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center of Genetics and Development, Institute of Brain Science, Department of Physiology and Biophysics, School of Life Sciences, Fudan University, 2005 Songhu Road, Yangpu District, Shanghai 200438, China
| | - Yimeng Zhao
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Multiscale Research Institute for Complex Systems, Department of Physiology and Biophysics, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Tsukasa Kusakizako
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yao Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Multiscale Research Institute for Complex Systems, Department of Physiology and Biophysics, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Chengfang Pan
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurosurgery at Huashan Hospital, Human Phenome Institute, Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center of Genetics and Development, Institute of Brain Science, Department of Physiology and Biophysics, School of Life Sciences, Fudan University, 2005 Songhu Road, Yangpu District, Shanghai 200438, China
| | - Yuwei Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurosurgery at Huashan Hospital, Human Phenome Institute, Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center of Genetics and Development, Institute of Brain Science, Department of Physiology and Biophysics, School of Life Sciences, Fudan University, 2005 Songhu Road, Yangpu District, Shanghai 200438, China
| | - Osamu Nureki
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| | - Motoyuki Hattori
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Multiscale Research Institute for Complex Systems, Department of Physiology and Biophysics, School of Life Sciences, Fudan University, Shanghai 200438, China.
| | - Zhiqiang Yan
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurosurgery at Huashan Hospital, Human Phenome Institute, Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center of Genetics and Development, Institute of Brain Science, Department of Physiology and Biophysics, School of Life Sciences, Fudan University, 2005 Songhu Road, Yangpu District, Shanghai 200438, China.
| |
Collapse
|