1
|
Liao X, Tung CY, Krey JF, Behnammanesh G, Cirilo JA, Colpan M, Yengo CM, Barr-Gillespie PG, Bird JE, Perrin BJ. Myosin-dependent short actin filaments contribute to peripheral widening in developing stereocilia. RESEARCH SQUARE 2024:rs.3.rs-5448262. [PMID: 39678325 PMCID: PMC11643313 DOI: 10.21203/rs.3.rs-5448262/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
In the auditory and vestibular systems, stereocilia are actin-based protrusions that convert mechanical stimuli into electrical signals. During development, stereocilia elongate and widen by adding filamentous actin (F-actin), attaining their mature shape necessary for mechanosensitive function. Myosin motors, including MYO3A/B and MYO15A, are required for normal stereocilia growth, but the regulation of actin and the impact of myosins on actin assembly remain unclear. We focused on stereocilia widening, which requires the addition of new filaments to the bundle of linear F-actin comprising the initial stereocilia core. Our findings revealed that newly expressed actin incorporates at the stereocilia tip first, then along the shaft to promote stereocilia widening. The newly incorporated F-actin surrounded the existing F-actin core, suggesting that the core is stable once formed, with additional actin adding only to the periphery. To better understand the nature of incorporating actin, we used several probes to detect globular (G-) actin, F-actin barbed ends, and F-actin pointed ends. While F-actin core filaments are parallel and thought to present only barbed ends at stereocilia tips, we also detected F-actin pointed ends, indicating a previously undetected population of short actin filaments. Overexpression of actin resulted in abundant F-actin pointed and barbed ends along the periphery of the stereocilia shaft, suggesting that short actin filaments contribute to stereocilia widening. Short actin filament levels correlated with the levels of MYO3A/B and MYO15A at stereocilia tips, suggesting these myosins generate or stabilize short actin filaments essential for stereocilia widening and elongation.
Collapse
Affiliation(s)
- Xiayi Liao
- Department of Biology, Indiana University, Indianapolis, IN
| | - Chun-Yu Tung
- Department of Biology, Indiana University, Indianapolis, IN
| | - Jocelyn F Krey
- Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR
- Vollum Institute, Oregon Health & Science University, Portland, OR
| | | | - Joseph A Cirilo
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA
| | - Mert Colpan
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ
| | - Christopher M Yengo
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA
| | - Peter G Barr-Gillespie
- Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR
- Vollum Institute, Oregon Health & Science University, Portland, OR
| | - Jonathan E Bird
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL
| | | |
Collapse
|
2
|
Cirilo JA, Liao X, Perrin BJ, Yengo CM. The dynamics of actin protrusions can be controlled by tip-localized myosin motors. J Biol Chem 2024; 300:105516. [PMID: 38042485 PMCID: PMC10801316 DOI: 10.1016/j.jbc.2023.105516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 12/04/2023] Open
Abstract
Class III myosins localize to inner ear hair cell stereocilia and are thought to be crucial for stereocilia length regulation. Mutations within the motor domain of MYO3A that disrupt its intrinsic motor properties have been associated with non-syndromic hearing loss, suggesting that the motor properties of MYO3A are critical for its function within stereocilia. In this study, we investigated the impact of a MYO3A hearing loss mutation, H442N, using both in vitro motor assays and cell biological studies. Our results demonstrate the mutation causes a dramatic increase in intrinsic motor properties, actin-activated ATPase and in vitro actin gliding velocity, as well as an increase in actin protrusion extension velocity. We propose that both "gain of function" and "loss of function" mutations in MYO3A can impair stereocilia length regulation, which is crucial for stereocilia formation during development and normal hearing. Furthermore, we generated chimeric MYO3A constructs that replace the MYO3A motor and neck domain with the motor and neck domain of other myosins. We found that duty ratio, fraction of ATPase cycle myosin is strongly bound to actin, is a critical motor property that dictates the ability to tip localize within filopodia. In addition, in vitro actin gliding velocities correlated extremely well with filopodial extension velocities over a wide range of gliding and extension velocities. Taken together, our data suggest a model in which tip-localized myosin motors exert force that slides the membrane tip-ward, which can combat membrane tension and enhance the actin polymerization rate that ultimately drives protrusion elongation.
Collapse
Affiliation(s)
- Joseph A Cirilo
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Xiayi Liao
- Department of Biology, Indiana University - Purdue University, Indianapolis, Indiana, USA
| | - Benjamin J Perrin
- Department of Biology, Indiana University - Purdue University, Indianapolis, Indiana, USA
| | - Christopher M Yengo
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA.
| |
Collapse
|
3
|
Park J, Bird JE. The actin cytoskeleton in hair bundle development and hearing loss. Hear Res 2023; 436:108817. [PMID: 37300948 PMCID: PMC10408727 DOI: 10.1016/j.heares.2023.108817] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/18/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023]
Abstract
Inner ear hair cells assemble mechanosensitive hair bundles on their apical surface that transduce sounds and accelerations. Each hair bundle is comprised of ∼ 100 individual stereocilia that are arranged into rows of increasing height and width; their specific and precise architecture being necessary for mechanoelectrical transduction (MET). The actin cytoskeleton is fundamental to establishing this architecture, not only by forming the structural scaffold shaping each stereocilium, but also by composing rootlets and the cuticular plate that together provide a stable foundation supporting each stereocilium. In concert with the actin cytoskeleton, a large assortment of actin-binding proteins (ABPs) function to cross-link actin filaments into specific topologies, as well as control actin filament growth, severing, and capping. These processes are individually critical for sensory transduction and are all disrupted in hereditary forms of human hearing loss. In this review, we provide an overview of actin-based structures in the hair bundle and the molecules contributing to their assembly and functional properties. We also highlight recent advances in mechanisms driving stereocilia elongation and how these processes are tuned by MET.
Collapse
Affiliation(s)
- Jinho Park
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, United States; Myology Institute, University of Florida, Gainesville, FL 32610, United States
| | - Jonathan E Bird
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, United States; Myology Institute, University of Florida, Gainesville, FL 32610, United States.
| |
Collapse
|
4
|
Nakamura T, Sakaguchi H, Mohri H, Ninoyu Y, Goto A, Yamaguchi T, Hishikawa Y, Matsuda M, Saito N, Ueyama T. Dispensable role of Rac1 and Rac3 after cochlear hair cell specification. J Mol Med (Berl) 2023; 101:843-854. [PMID: 37204479 PMCID: PMC10300165 DOI: 10.1007/s00109-023-02317-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 04/03/2023] [Accepted: 04/11/2023] [Indexed: 05/20/2023]
Abstract
Rac small GTPases play important roles during embryonic development of the inner ear; however, little is known regarding their function in cochlear hair cells (HCs) after specification. Here, we revealed the localization and activation of Racs in cochlear HCs using GFP-tagged Rac plasmids and transgenic mice expressing a Rac1-fluorescence resonance energy transfer (FRET) biosensor. Furthermore, we employed Rac1-knockout (Rac1-KO, Atoh1-Cre;Rac1flox/flox) and Rac1 and Rac3 double KO (Rac1/Rac3-DKO, Atoh1-Cre;Rac1flox/flox;Rac3-/-) mice, under the control of the Atoh1 promoter. However, both Rac1-KO and Rac1/Rac3-DKO mice exhibited normal cochlear HC morphology at 13 weeks of age and normal hearing function at 24 weeks of age. No hearing vulnerability was observed in young adult (6-week-old) Rac1/Rac3-DKO mice even after intense noise exposure. Consistent with prior reports, the results from Atoh1-Cre;tdTomato mice confirmed that the Atoh1 promoter became functional only after embryonic day 14 when the sensory HC precursors exit the cell cycle. Taken together, these findings indicate that although Rac1 and Rac3 contribute to the early development of sensory epithelia in cochleae, as previously shown, they are dispensable for the maturation of cochlear HCs in the postmitotic state or for hearing maintenance following HC maturation. KEY MESSAGES: Mice with Rac1 and Rac3 deletion were generated after HC specification. Knockout mice exhibit normal cochlear hair cell morphology and hearing. Racs are dispensable for hair cells in the postmitotic state after specification. Racs are dispensable for hearing maintenance after HC maturation.
Collapse
Affiliation(s)
- Takashi Nakamura
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, 657-8501, Japan
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Hirofumi Sakaguchi
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Hiroaki Mohri
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, 657-8501, Japan
| | - Yuzuru Ninoyu
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, 657-8501, Japan
| | - Akihiro Goto
- Graduate School of Biostudies, Kyoto University, Kyoto, 606-8315, Japan
| | - Taro Yamaguchi
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, 573-0101, Japan
| | - Yoshitaka Hishikawa
- Department of Anatomy, Histochemistry and Cell Biology, Faculty of Medicine, University of Miyazaki, Miyazaki, 889-1692, Japan
| | - Michiyuki Matsuda
- Graduate School of Biostudies, Kyoto University, Kyoto, 606-8315, Japan
| | - Naoaki Saito
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, 657-8501, Japan
| | - Takehiko Ueyama
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, 657-8501, Japan.
| |
Collapse
|
5
|
Fitz GN, Weck ML, Bodnya C, Perkins OL, Tyska MJ. Protrusion growth driven by myosin-generated force. Dev Cell 2023; 58:18-33.e6. [PMID: 36626869 PMCID: PMC9940483 DOI: 10.1016/j.devcel.2022.12.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/10/2022] [Accepted: 11/29/2022] [Indexed: 01/11/2023]
Abstract
Actin-based protrusions extend from the surface of all eukaryotic cells, where they support diverse activities essential for life. Models of protrusion growth hypothesize that actin filament assembly exerts force for pushing the plasma membrane outward. However, membrane-associated myosin motors are also abundant in protrusions, although their potential for contributing, growth-promoting force remains unexplored. Using an inducible system that docks myosin motor domains to membrane-binding modules with temporal control, we found that application of myosin-generated force to the membrane is sufficient for driving robust protrusion elongation in human, mouse, and pig cell culture models. Protrusion growth scaled with motor accumulation, required barbed-end-directed force, and was independent of cargo delivery or recruitment of canonical elongation factors. Application of growth-promoting force was also supported by structurally distinct myosin motors and membrane-binding modules. Thus, myosin-generated force can drive protrusion growth, and this mechanism is likely active in diverse biological contexts.
Collapse
Affiliation(s)
- Gillian N Fitz
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Meredith L Weck
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Caroline Bodnya
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Olivia L Perkins
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Matthew J Tyska
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
6
|
Souissi A, Abdelmalek Driss D, Chakchouk I, Ben Said M, Ben Ayed I, Mosrati MA, Elloumi I, Tlili A, Aifa S, Masmoudi S. Molecular insights into MYO3A kinase domain variants explain variability in both severity and progression of DFNB30 hearing impairment. J Biomol Struct Dyn 2022; 40:10940-10951. [PMID: 34423747 DOI: 10.1080/07391102.2021.1953600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Hereditary hearing impairment (HI) is a common disease with the highest incidence among sensory defects. Several genes have been identified to affect stereocilia structure causing HI, including the unconventional myosin3A. Interestingly, we noticed that variants in MYO3A gene have been previously found to cause variable HI onset and severity. Using clinical exome sequencing, we identified a novel pathogenic variant p.(Lys50Arg) in the MYO3A kinase domain (MYO3A-KD). Previous in vitro studies supported its damaging effect as a 'kinase-dead' mutant. We further analyzed this variation through molecular dynamics which predicts that changes in flexibility of MYO3A structure would influence the protein-ATP binding properties. This Lys50Arg mutation segregated with congenital profound non-syndromic HI. To better investigate this variability, we collected previously identified MYO3A-KDs variants, p.(Tyr129Cys), p.(His142Gln) and p.(Pro189Thr), and built both wild type and mutant 3 D MYO3A-KD models to assess their impact on the protein structure and function. Our results suggest that KD mutations could either cause a congenital profound form of HI, when particularly affecting the kinase activity and preventing the auto-phosphorylation of the motor, or a late onset and progressive form, when partially or completely inactivating the MYO3A protein. In conclusion, we report a novel pathogenic variant affecting the ATP-binding site within the MYO3A-KD causing congenital profound HI. Through computational approaches we provide a deeper understanding on the correlation between the effects of MYO3A-KD mutations and the variable hearing phenotypes. To the best of our knowledge this is the first study to correlate mutations' genotypes with the variable phenotypes of DFNB30.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Amal Souissi
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Dorra Abdelmalek Driss
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Imen Chakchouk
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
| | - Mariem Ben Said
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Ikhlas Ben Ayed
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia.,Medical Genetic Department, University Hedi Chaker Hospital of Sfax, Sfax, Tunisia
| | - Mohamed Ali Mosrati
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Ines Elloumi
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Abdelaziz Tlili
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah, United Arab Emirates.,Human Genetics and Stem Cell Laboratory, Research Institute of Sciences and Engineering, University of Sharjah, Sharjah, United Arab Emirates
| | - Sami Aifa
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Saber Masmoudi
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| |
Collapse
|
7
|
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
|
8
|
Selective binding and transport of protocadherin 15 isoforms by stereocilia unconventional myosins in a heterologous expression system. Sci Rep 2022; 12:13764. [PMID: 35962067 PMCID: PMC9374675 DOI: 10.1038/s41598-022-17757-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 07/30/2022] [Indexed: 11/09/2022] Open
Abstract
During hair cell development, the mechanoelectrical transduction (MET) apparatus is assembled at the stereocilia tips, where it coexists with the stereocilia actin regulatory machinery. While the myosin-based tipward transport of actin regulatory proteins is well studied, isoform complexity and built-in redundancies in the MET apparatus have limited our understanding of how MET components are transported. We used a heterologous expression system to elucidate the myosin selective transport of isoforms of protocadherin 15 (PCDH15), the protein that mechanically gates the MET apparatus. We show that MYO7A selectively transports the CD3 isoform while MYO3A and MYO3B transports the CD2 isoform. Furthermore, MYO15A showed an insignificant role in the transport of PCDH15, and none of the myosins tested transport PCDH15-CD1. Our data suggest an important role for MYO3A, MYO3B, and MYO7A in the MET apparatus formation and highlight the intricate nature of MET and actin regulation during development and functional maturation of the stereocilia bundle.
Collapse
|
9
|
Deafness-related protein PDZD7 forms complex with the C-terminal tail of FCHSD2. Biochem J 2022; 479:1393-1405. [PMID: 35695292 PMCID: PMC9317961 DOI: 10.1042/bcj20220147] [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: 03/26/2022] [Revised: 06/09/2022] [Accepted: 06/13/2022] [Indexed: 11/24/2022]
Abstract
In cochlea, deafness-related protein PDZD7 is an indispensable component of the ankle link complex, which is critical for the maturation of inner-ear hair cell for sound perception. Ankle links, connecting the different rows of cochlear stereocilia, are essential for the staircase-like development of stereocilia. However, the molecular mechanism of how PDZD7 governs stereociliary development remains unknown. Here, we reported a novel PDZD7-binding partner, FCHSD2, identified by yeast two-hybrid screening. FCHSD2 was reported to be expressed in hair cell, where it co-operated with CDC42 and N-WASP to regulate the formation of cell protrusion. The association between FCHSD2 and PDZD7 was further confirmed in COS-7 cells. More importantly, we solved the complex structure of FCHSD2 tail with PDZD7 PDZ3 domain at 2.0 Å resolution. The crystal structure shows that PDZD7 PDZ3 adopts a typical PDZ domain topology, comprising five β strands and two α helixes. The PDZ-binding motif of FCHSD2 tail stretches through the αB/βB groove of PDZD7 PDZ3. Our study not only uncovers the interaction between FCHSD2 tail and PDZD7 PDZ3 at the atomic level, but also provides clues of connecting the ankle link complex with cytoskeleton dynamics for exploiting the molecular mechanism of stereociliary development.
Collapse
|
10
|
Frequency and origin of the c.2090T>G p.(Leu697Trp) MYO3A variant associated with autosomal dominant hearing loss. Eur J Hum Genet 2022; 30:13-21. [PMID: 33953343 PMCID: PMC8738757 DOI: 10.1038/s41431-021-00891-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 03/15/2021] [Accepted: 04/08/2021] [Indexed: 02/07/2023] Open
Abstract
We recently described a novel missense variant [c.2090T>G:p.(Leu697Trp)] in the MYO3A gene, found in two Brazilian families with late-onset autosomal dominant nonsyndromic hearing loss (ADNSHL). Since then, with the objective of evaluating its contribution to ADNSHL in Brazil, the variant was screened in additional 101 pedigrees with probable ADNSHL without conclusive molecular diagnosis. The variant was found in three additional families, explaining 3/101 (~3%) of cases with ADNSHL in our Brazilian pedigree collection. In order to identify the origin of the variant, 21 individuals from the five families were genotyped with a high-density SNP array (~600 K SNPs- Axiom Human Origins; ThermoFisher). The identity by descent (IBD) approach revealed that many pairs of individuals from the different families have a kinship coefficient equivalent to that of second cousins, and all share a minimum haplotype of ~607 kb which includes the c.2090T>G variant suggesting it probably arose in a common ancestor. We inferred that the mutation occurred in a chromosomal segment of European ancestry and the time since the most common ancestor was estimated in 1100 years (CI = 775-1425). This variant was also reported in a Dutch family, which shares a 87,121 bp haplotype with the Brazilian samples, suggesting that Dutch colonists may have brought it to Northeastern Brazil in the 17th century. Therefore, the present study opens new avenues to investigate this variant not only in Brazilians but also in European families with ADNSHL.
Collapse
|
11
|
Pacentine IV, Barr-Gillespie PG. Cy3-ATP labeling of unfixed, permeabilized mouse hair cells. Sci Rep 2021; 11:23855. [PMID: 34903829 PMCID: PMC8668996 DOI: 10.1038/s41598-021-03365-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/01/2021] [Indexed: 12/02/2022] Open
Abstract
ATP-utilizing enzymes play key roles in hair bundles, the mechanically sensitive organelles of sensory hair cells in the inner ear. We used a fluorescent ATP analog, EDA-ATP-Cy3 (Cy3-ATP), to label ATP-binding proteins in two different preparations of unfixed hair-cell stereocilia of the mouse. In the first preparation, we lightly permeabilized dissected cochleas, then labeled them with Cy3-ATP. Hair cells and their stereocilia remained intact, and stereocilia tips in rows 1 and 2 were labeled particularly strongly with Cy3-ATP. In many cases, vanadate (Vi) traps nucleotides at the active site of myosin isoforms and presents nucleotide dissociation. Co-application with Vi enhanced the tip labeling, which is consistent with myosin isoforms being responsible. By contrast, the actin polymerization inhibitors latrunculin A and cytochalasin D had no effect, suggesting that actin turnover at stereocilia tips was not involved. Cy3-ATP labeling was substantially reduced—but did not disappear altogether—in mutant cochleas lacking MYO15A; by contrast, labeling remained robust in cochleas lacking MYO7A. In the second preparation, used to quantify Cy3-ATP labeling, we labeled vestibular stereocilia that had been adsorbed to glass, which demonstrated that tip labeling was higher in longer stereocilia. We found that tip signal was reduced by ~ 50% in Myo15ash2/sh2 stereocilia as compared to Myo15ash2/+stereocilia. These results suggest that MYO15A accounts for a substantial fraction of the Cy3-ATP tip labeling in vestibular hair cells, and so this novel preparation could be utilized to examine the control of MYO15A ATPase activity in situ.
Collapse
Affiliation(s)
- Itallia V Pacentine
- Oregon Hearing Research Center & Vollum Institute, Mail Code L335A, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Peter G Barr-Gillespie
- Oregon Hearing Research Center & Vollum Institute, Mail Code L335A, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA.
| |
Collapse
|
12
|
Gunther LK, Cirilo JA, Desetty R, Yengo CM. Deafness mutation in the MYO3A motor domain impairs actin protrusion elongation mechanism. Mol Biol Cell 2021; 33:ar5. [PMID: 34788109 PMCID: PMC8886822 DOI: 10.1091/mbc.e21-05-0232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Class III myosins are actin-based motors proposed to transport cargo to the distal tips of stereocilia in the inner ear hair cells and/or to participate in stereocilia length regulation, which is especially important during development. Mutations in the MYO3A gene are associated with delayed onset deafness. A previous study demonstrated that L697W, a dominant deafness mutation, disrupts MYO3A ATPase and motor properties but does not impair its ability to localize to the tips of actin protrusions. In the current study, we characterized the transient kinetic mechanism of the L697W motor ATPase cycle. Our kinetic analysis demonstrates that the mutation slows the ADP release and ATP hydrolysis steps, which results in a slight reduction in the duty ratio and slows detachment kinetics. Fluorescence recovery after photobleaching (FRAP) of filopodia tip localized L697W and WT MYO3A in COS-7 cells revealed that the mutant does not alter turnover or average intensity at the actin protrusion tips. We demonstrate that the mutation slows filopodia extension velocity in COS-7 cells which correlates with its twofold slower in vitro actin gliding velocity. Overall, this work allowed us to propose a model for how the motor properties of MYO3A are crucial for facilitating actin protrusion length regulation.
Collapse
Affiliation(s)
- Laura K Gunther
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA, 17033
| | - Joseph A Cirilo
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA, 17033
| | - Rohini Desetty
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA, 17033
| | - Christopher M Yengo
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA, 17033
| |
Collapse
|
13
|
Revilla-i-Domingo R, Rajan VBV, Waldherr M, Prohaczka G, Musset H, Orel L, Gerrard E, Smolka M, Stockinger A, Farlik M, Lucas RJ, Raible F, Tessmar-Raible K. Characterization of cephalic and non-cephalic sensory cell types provides insight into joint photo- and mechanoreceptor evolution. eLife 2021; 10:e66144. [PMID: 34350831 PMCID: PMC8367381 DOI: 10.7554/elife.66144] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 08/04/2021] [Indexed: 12/11/2022] Open
Abstract
Rhabdomeric opsins (r-opsins) are light sensors in cephalic eye photoreceptors, but also function in additional sensory organs. This has prompted questions on the evolutionary relationship of these cell types, and if ancient r-opsins were non-photosensory. A molecular profiling approach in the marine bristleworm Platynereis dumerilii revealed shared and distinct features of cephalic and non-cephalic r-opsin1-expressing cells. Non-cephalic cells possess a full set of phototransduction components, but also a mechanosensory signature. Prompted by the latter, we investigated Platynereis putative mechanotransducer and found that nompc and pkd2.1 co-expressed with r-opsin1 in TRE cells by HCR RNA-FISH. To further assess the role of r-Opsin1 in these cells, we studied its signaling properties and unraveled that r-Opsin1 is a Gαq-coupled blue light receptor. Profiling of cells from r-opsin1 mutants versus wild-types, and a comparison under different light conditions reveals that in the non-cephalic cells light - mediated by r-Opsin1 - adjusts the expression level of a calcium transporter relevant for auditory mechanosensation in vertebrates. We establish a deep-learning-based quantitative behavioral analysis for animal trunk movements and identify a light- and r-Opsin-1-dependent fine-tuning of the worm's undulatory movements in headless trunks, which are known to require mechanosensory feedback. Our results provide new data on peripheral cell types of likely light sensory/mechanosensory nature. These results point towards a concept in which such a multisensory cell type evolved to allow for fine-tuning of mechanosensation by light. This implies that light-independent mechanosensory roles of r-opsins may have evolved secondarily.
Collapse
Affiliation(s)
- Roger Revilla-i-Domingo
- Max Perutz Labs, University of Vienna, Vienna BioCenterViennaAustria
- Research Platform “Rhythms of Life”, University of Vienna, Vienna BioCenterViennaAustria
- Research Platform "Single-Cell Regulation of Stem Cells", University of Vienna, Vienna BioCenterViennaAustria
| | - Vinoth Babu Veedin Rajan
- Max Perutz Labs, University of Vienna, Vienna BioCenterViennaAustria
- Research Platform “Rhythms of Life”, University of Vienna, Vienna BioCenterViennaAustria
| | - Monika Waldherr
- Max Perutz Labs, University of Vienna, Vienna BioCenterViennaAustria
- Research Platform “Rhythms of Life”, University of Vienna, Vienna BioCenterViennaAustria
| | - Günther Prohaczka
- Max Perutz Labs, University of Vienna, Vienna BioCenterViennaAustria
- Research Platform “Rhythms of Life”, University of Vienna, Vienna BioCenterViennaAustria
| | - Hugo Musset
- Max Perutz Labs, University of Vienna, Vienna BioCenterViennaAustria
- Research Platform “Rhythms of Life”, University of Vienna, Vienna BioCenterViennaAustria
| | - Lukas Orel
- Max Perutz Labs, University of Vienna, Vienna BioCenterViennaAustria
- Research Platform “Rhythms of Life”, University of Vienna, Vienna BioCenterViennaAustria
| | - Elliot Gerrard
- Division of Neuroscience & Experimental Psychology, University of ManchesterManchesterUnited Kingdom
| | - Moritz Smolka
- Max Perutz Labs, University of Vienna, Vienna BioCenterViennaAustria
- Research Platform “Rhythms of Life”, University of Vienna, Vienna BioCenterViennaAustria
- Center for Integrative Bioinformatics Vienna, Max Perutz Labs, University of Vienna and Medical University of ViennaViennaAustria
| | - Alexander Stockinger
- Max Perutz Labs, University of Vienna, Vienna BioCenterViennaAustria
- Research Platform “Rhythms of Life”, University of Vienna, Vienna BioCenterViennaAustria
- Research Platform "Single-Cell Regulation of Stem Cells", University of Vienna, Vienna BioCenterViennaAustria
| | - Matthias Farlik
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
- Department of Dermatology, Medical University of ViennaViennaAustria
| | - Robert J Lucas
- Division of Neuroscience & Experimental Psychology, University of ManchesterManchesterUnited Kingdom
| | - Florian Raible
- Max Perutz Labs, University of Vienna, Vienna BioCenterViennaAustria
- Research Platform “Rhythms of Life”, University of Vienna, Vienna BioCenterViennaAustria
- Research Platform "Single-Cell Regulation of Stem Cells", University of Vienna, Vienna BioCenterViennaAustria
| | - Kristin Tessmar-Raible
- Max Perutz Labs, University of Vienna, Vienna BioCenterViennaAustria
- Research Platform “Rhythms of Life”, University of Vienna, Vienna BioCenterViennaAustria
| |
Collapse
|
14
|
Abstract
Filopodia, microvilli and stereocilia represent an important group of plasma membrane protrusions. These specialized projections are supported by parallel bundles of actin filaments and have critical roles in sensing the external environment, increasing cell surface area, and acting as mechanosensors. While actin-associated proteins are essential for actin-filament elongation and bundling in these protrusions, myosin motors have a surprising role in the formation and extension of filopodia and stereocilia and in the organization of microvilli. Actin regulators and specific myosins collaborate in controlling the length of these structures. Myosins can transport cargoes along the length of these protrusions, and, in the case of stereocilia and microvilli, interactions with adaptors and cargoes can also serve to anchor adhesion receptors to the actin-rich core via functionally conserved motor-adaptor complexes. This review highlights recent progress in understanding the diverse roles myosins play in filopodia, microvilli and stereocilia.
Collapse
Affiliation(s)
- Anne Houdusse
- Structural Motility, Institut Curie, Paris Université Sciences et Lettres, Sorbonne Université, CNRS UMR144, 75005 Paris, France.
| | - Margaret A Titus
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
15
|
Fu X, An Y, Wang H, Li P, Lin J, Yuan J, Yue R, Jin Y, Gao J, Chai R. Deficiency of Klc2 Induces Low-Frequency Sensorineural Hearing Loss in C57BL/6 J Mice and Human. Mol Neurobiol 2021; 58:4376-4391. [PMID: 34014435 DOI: 10.1007/s12035-021-02422-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/05/2021] [Indexed: 12/19/2022]
Abstract
The transport system in cochlear hair cells (HCs) is important for their function, and the kinesin family of proteins transports numerous cellular cargos via the microtubule network in the cytoplasm. Here, we found that Klc2 (kinesin light chain 2), the light chain of kinesin-1 that mediates cargo binding and regulates kinesin-1 motility, is essential for cochlear function. We generated mice lacking Klc2, and they suffered from low-frequency hearing loss as early as 1 month of age. We demonstrated that deficiency of Klc2 resulted in abnormal transport of mitochondria and the down-regulation of the GABAA receptor family. In addition, whole-genome sequencing (WGS) of patient showed that KLC2 was related to low-frequency hearing in human. Hence, to explore therapeutic approaches, we developed adeno-associated virus containing the Klc2 wide-type cDNA sequence, and Klc2-null mice delivered virus showed apparent recovery, including decreased ABR threshold and reduced out hair cell (OHC) loss. In summary, we show that the kinesin transport system plays an indispensable and special role in cochlear HC function in mice and human and that mitochondrial localization is essential for HC survival.
Collapse
Affiliation(s)
- Xiaolong Fu
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China.,College of Laboratory Animal & Shandong Laboratory Animal Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yachun An
- School of Life Science, Shandong University, Qingdao, China
| | - Hongyang Wang
- College of Otolaryngology, Head and Neck Surgery, Institute of Otolaryngology, Chinese PLA General Hospital, Beijing, China
| | - Peipei Li
- School of Life Science, Shandong University, Qingdao, China
| | - Jing Lin
- Waksman Institute, the State University of New Jersey, RutgersNew Brunswick, NJ, USA
| | - Jia Yuan
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China
| | - Rongyu Yue
- Department of Otolaryngology-Head and Neck Surgery, Provincial Hospital Affiliated To Shandong University, Jinan, China
| | - Yecheng Jin
- School of Life Science, Shandong University, Qingdao, China
| | - Jiangang Gao
- College of Laboratory Animal & Shandong Laboratory Animal Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
| | - Renjie Chai
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China. .,College of Laboratory Animal & Shandong Laboratory Animal Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China. .,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China. .,Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing, China.
| |
Collapse
|
16
|
Cirilo JA, Gunther LK, Yengo CM. Functional Role of Class III Myosins in Hair Cells. Front Cell Dev Biol 2021; 9:643856. [PMID: 33718386 PMCID: PMC7947357 DOI: 10.3389/fcell.2021.643856] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/01/2021] [Indexed: 11/14/2022] Open
Abstract
Cytoskeletal motors produce force and motion using the energy from ATP hydrolysis and function in a variety of mechanical roles in cells including muscle contraction, cargo transport, and cell division. Actin-based myosin motors have been shown to play crucial roles in the development and function of the stereocilia of auditory and vestibular inner ear hair cells. Hair cells can contain hundreds of stereocilia, which rely on myosin motors to elongate, organize, and stabilize their structure. Mutations in many stereocilia-associated myosins have been shown to cause hearing loss in both humans and animal models suggesting that each myosin isoform has a specific function in these unique parallel actin bundle-based protrusions. Here we review what is known about the classes of myosins that function in the stereocilia, with a special focus on class III myosins that harbor point mutations associated with delayed onset hearing loss. Much has been learned about the role of the two class III myosin isoforms, MYO3A and MYO3B, in maintaining the precise stereocilia lengths required for normal hearing. We propose a model for how class III myosins play a key role in regulating stereocilia lengths and demonstrate how their motor and regulatory properties are particularly well suited for this function. We conclude that ongoing studies on class III myosins and other stereocilia-associated myosins are extremely important and may lead to novel therapeutic strategies for the treatment of hearing loss due to stereocilia degeneration.
Collapse
Affiliation(s)
- Joseph A Cirilo
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Laura K Gunther
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Christopher M Yengo
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| |
Collapse
|
17
|
Abstract
Myosins constitute a superfamily of actin-based molecular motor proteins that mediates a variety of cellular activities including muscle contraction, cell migration, intracellular transport, the formation of membrane projections, cell adhesion, and cell signaling. The 12 myosin classes that are expressed in humans share sequence similarities especially in the N-terminal motor domain; however, their enzymatic activities, regulation, ability to dimerize, binding partners, and cellular functions differ. It is becoming increasingly apparent that defects in myosins are associated with diseases including cardiomyopathies, colitis, glomerulosclerosis, neurological defects, cancer, blindness, and deafness. Here, we review the current state of knowledge regarding myosins and disease.
Collapse
|
18
|
Doll J, Hofrichter MAH, Bahena P, Heihoff A, Segebarth D, Müller T, Dittrich M, Haaf T, Vona B. A novel missense variant in MYO3A is associated with autosomal dominant high-frequency hearing loss in a German family. Mol Genet Genomic Med 2020; 8:e1343. [PMID: 32519820 PMCID: PMC7434730 DOI: 10.1002/mgg3.1343] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/16/2020] [Accepted: 05/19/2020] [Indexed: 12/30/2022] Open
Abstract
Background MYO3A, encoding the myosin IIIA protein, is associated with autosomal recessive and autosomal dominant nonsyndromic hearing loss. To date, only two missense variants located in the motor‐head domain of MYO3A have been described in autosomal dominant families with progressive, mild‐to‐profound sensorineural hearing loss. These variants alter the ATPase activity of myosin IIIA. Methods Exome sequencing of a proband from a three‐generation German family with prelingual, moderate‐to‐profound, high‐frequency hearing loss was performed. Segregation analysis confirmed a dominant inheritance pattern. Regression analysis of mean hearing level thresholds per individual and ear was performed at high‐, mid‐, and low‐frequencies. Results A novel heterozygous missense variant c.716T>C, p.(Leu239Pro) in the kinase domain of MYO3A was identified that is predicted in silico as disease causing. High‐frequency, progressive hearing loss was identified. Conclusion Correlation analysis of pure‐tone hearing thresholds revealed progressive hearing loss, especially in the high‐frequencies. In the present study, we report the first dominant likely pathogenic variant in MYO3A in a European family and further support MYO3A as an autosomal dominant hearing loss gene.
Collapse
Affiliation(s)
- Julia Doll
- Institute of Human Genetics, Julius Maximilians University, Würzburg, Germany
| | | | - Paulina Bahena
- Institute of Human Genetics, Julius Maximilians University, Würzburg, Germany
| | | | - Dennis Segebarth
- Institute of Clinical Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Tobias Müller
- Institute of Bioinformatics, Julius Maximilians University, Würzburg, Germany
| | - Marcus Dittrich
- Institute of Human Genetics, Julius Maximilians University, Würzburg, Germany.,Institute of Bioinformatics, Julius Maximilians University, Würzburg, Germany
| | - Thomas Haaf
- Institute of Human Genetics, Julius Maximilians University, Würzburg, Germany
| | - Barbara Vona
- Institute of Human Genetics, Julius Maximilians University, Würzburg, Germany.,Tübingen Hearing Research Centre, Department of Otolaryngology - Head and Neck Surgery, Eberhard Karls University, Tübingen, Germany
| |
Collapse
|
19
|
Song J, Patterson R, Metlagel Z, Krey JF, Hao S, Wang L, Ng B, Sazzed S, Kovacs J, Wriggers W, He J, Barr-Gillespie PG, Auer M. A cryo-tomography-based volumetric model of the actin core of mouse vestibular hair cell stereocilia lacking plastin 1. J Struct Biol 2020; 210:107461. [PMID: 31962158 PMCID: PMC7067663 DOI: 10.1016/j.jsb.2020.107461] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/11/2020] [Accepted: 01/14/2020] [Indexed: 12/11/2022]
Abstract
Electron cryo-tomography allows for high-resolution imaging of stereocilia in their native state. Because their actin filaments have a higher degree of order, we imaged stereocilia from mice lacking the actin crosslinker plastin 1 (PLS1). We found that while stereocilia actin filaments run 13 nm apart in parallel for long distances, there were gaps of significant size that were stochastically distributed throughout the actin core. Actin crosslinkers were distributed through the stereocilium, but did not occupy all possible binding sites. At stereocilia tips, protein density extended beyond actin filaments, especially on the side of the tip where a tip link is expected to anchor. Along the shaft, repeating density was observed that corresponds to actin-to-membrane connectors. In the taper region, most actin filaments terminated near the plasma membrane. The remaining filaments twisted together to make a tighter bundle than was present in the shaft region; the spacing between them decreased from 13 nm to 9 nm, and the apparent filament diameter decreased from 6.4 to 4.8 nm. Our models illustrate detailed features of distinct structural domains that are present within the stereocilium.
Collapse
Affiliation(s)
- Junha Song
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Roma Patterson
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Zoltan Metlagel
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Jocelyn F Krey
- Oregon Hearing Research Center & Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Samantha Hao
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Linshanshan Wang
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Brian Ng
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Salim Sazzed
- Department of Computer Science, Old Dominion University, Norfolk, VA, USA
| | - Julio Kovacs
- Department of Mechanical and Aerospace Engineering, Old Dominion University, Norfolk, VA, USA
| | - Willy Wriggers
- Department of Mechanical and Aerospace Engineering, Old Dominion University, Norfolk, VA, USA
| | - Jing He
- Department of Computer Science, Old Dominion University, Norfolk, VA, USA
| | - Peter G Barr-Gillespie
- Oregon Hearing Research Center & Vollum Institute, Oregon Health & Science University, Portland, OR, USA.
| | - Manfred Auer
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| |
Collapse
|
20
|
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
|
21
|
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
|
22
|
Friedman TB, Belyantseva IA, Frolenkov GI. Myosins and Hearing. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1239:317-330. [DOI: 10.1007/978-3-030-38062-5_13] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
23
|
Unconventional Myosins: How Regulation Meets Function. Int J Mol Sci 2019; 21:ijms21010067. [PMID: 31861842 PMCID: PMC6981383 DOI: 10.3390/ijms21010067] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 12/16/2019] [Accepted: 12/18/2019] [Indexed: 01/24/2023] Open
Abstract
Unconventional myosins are multi-potent molecular motors that are assigned important roles in fundamental cellular processes. Depending on their mechano-enzymatic properties and structural features, myosins fulfil their roles by acting as cargo transporters along the actin cytoskeleton, molecular anchors or tension sensors. In order to perform such a wide range of roles and modes of action, myosins need to be under tight regulation in time and space. This is achieved at multiple levels through diverse regulatory mechanisms: the alternative splicing of various isoforms, the interaction with their binding partners, their phosphorylation, their applied load and the composition of their local environment, such as ions and lipids. This review summarizes our current knowledge of how unconventional myosins are regulated, how these regulatory mechanisms can adapt to the specific features of a myosin and how they can converge with each other in order to ensure the required tight control of their function.
Collapse
|
24
|
Yizhar-Barnea O, Valensisi C, Jayavelu ND, Kishore K, Andrus C, Koffler-Brill T, Ushakov K, Perl K, Noy Y, Bhonker Y, Pelizzola M, Hawkins RD, Avraham KB. DNA methylation dynamics during embryonic development and postnatal maturation of the mouse auditory sensory epithelium. Sci Rep 2018; 8:17348. [PMID: 30478432 PMCID: PMC6255903 DOI: 10.1038/s41598-018-35587-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 11/08/2018] [Indexed: 12/17/2022] Open
Abstract
The inner ear is a complex structure responsible for hearing and balance, and organ pathology is associated with deafness and balance disorders. To evaluate the role of epigenomic dynamics, we performed whole genome bisulfite sequencing at key time points during the development and maturation of the mouse inner ear sensory epithelium (SE). Our single-nucleotide resolution maps revealed variations in both general characteristics and dynamics of DNA methylation over time. This allowed us to predict the location of non-coding regulatory regions and to identify several novel candidate regulatory factors, such as Bach2, that connect stage-specific regulatory elements to molecular features that drive the development and maturation of the SE. Constructing in silico regulatory networks around sites of differential methylation enabled us to link key inner ear regulators, such as Atoh1 and Stat3, to pathways responsible for cell lineage determination and maturation, such as the Notch pathway. We also discovered that a putative enhancer, defined as a low methylated region (LMR), can upregulate the GJB6 gene and a neighboring non-coding RNA. The study of inner ear SE methylomes revealed novel regulatory regions in the hearing organ, which may improve diagnostic capabilities, and has the potential to guide the development of therapeutics for hearing loss by providing multiple intervention points for manipulation of the auditory system.
Collapse
Affiliation(s)
- Ofer Yizhar-Barnea
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Cristina Valensisi
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Naresh Doni Jayavelu
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Kamal Kishore
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia, Milano, 20139, Italy
| | - Colin Andrus
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Tal Koffler-Brill
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Kathy Ushakov
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Kobi Perl
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Yael Noy
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Yoni Bhonker
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Mattia Pelizzola
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia, Milano, 20139, Italy
| | - R David Hawkins
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA, 98195, USA.
| | - Karen B Avraham
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel.
| |
Collapse
|
25
|
Li P, Wen Z, Zhang G, Zhang A, Fu X, Gao J. Knock-In Mice with Myo3a Y137C Mutation Displayed Progressive Hearing Loss and Hair Cell Degeneration in the Inner Ear. Neural Plast 2018; 2018:4372913. [PMID: 30123247 PMCID: PMC6079384 DOI: 10.1155/2018/4372913] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 05/28/2018] [Indexed: 12/02/2022] Open
Abstract
Myo3a is expressed in cochlear hair cells and retinal cells and is responsible for human recessive hereditary nonsyndromic deafness (DFNB30). To investigate the mechanism of DFNB30-type deafness, we established a mouse model of Myo3a kinase domain Y137C mutation by using CRISPR/Cas9 system. No difference in hearing between 2-month-old Myo3a mutant mice and wild-type mice was observed. The hearing threshold of the ≥6-month-old mutant mice was significantly elevated compared with that of the wild-type mice. We observed degeneration in the inner ear hair cells of 6-month-old Myo3a mutant mice, and the degeneration became more severe at the age of 12 months. We also found structural abnormality in the cochlear hair cell stereocilia. Our results showed that Myo3a is essential for normal hearing by maintaining the intact structure of hair cell stereocilia, and the kinase domain plays a critical role in the normal functions of Myo3a. This mouse line is an excellent model for studying DFNB30-type deafness in humans.
Collapse
Affiliation(s)
- Peipei Li
- School of Life Science, Shandong University, Jinan 250100, China
| | - Zongzhuang Wen
- School of Life Science, Shandong University, Jinan 250100, China
| | - Guangkai Zhang
- School of Life Science, Shandong University, Jinan 250100, China
| | - Aizhen Zhang
- School of Life Science, Shandong University, Jinan 250100, China
| | - Xiaolong Fu
- School of Life Science, Shandong University, Jinan 250100, China
| | - Jiangang Gao
- School of Life Science, Shandong University, Jinan 250100, China
| |
Collapse
|
26
|
Characterization of a novel MYO3A missense mutation associated with a dominant form of late onset hearing loss. Sci Rep 2018; 8:8706. [PMID: 29880844 PMCID: PMC5992146 DOI: 10.1038/s41598-018-26818-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 05/18/2018] [Indexed: 01/08/2023] Open
Abstract
Whole-exome sequencing of samples from affected members of two unrelated families with late-onset non-syndromic hearing loss revealed a novel mutation (c.2090 T > G; NM_017433) in MYO3A. The mutation was confirmed in 36 affected individuals, showing autosomal dominant inheritance. The mutation alters a single residue (L697W or p.Leu697Trp) in the motor domain of the stereocilia protein MYO3A, leading to a reduction in ATPase activity, motility, and an increase in actin affinity. MYO3A-L697W showed reduced filopodial actin protrusion initiation in COS7 cells, and a predominant tipward accumulation at filopodia and stereocilia when coexpressed with wild-type MYO3A and espin-1, an actin-regulatory MYO3A cargo. The combined higher actin affinity and duty ratio of the mutant myosin cause increased retention time at stereocilia tips, resulting in the displacement of the wild-type MYO3A protein, which may impact cargo transport, stereocilia length, and mechanotransduction. The dominant negative effect of the altered myosin function explains the dominant inheritance of deafness.
Collapse
|
27
|
Abstract
The delivery of intracellular material within cells is crucial for maintaining normal function. Myosins transport a wide variety of cargo, ranging from vesicles to ribonuclear protein particles (RNPs), in plants, fungi, and metazoa. The properties of a given myosin transporter are adapted to move on different actin filament tracks, either on the disordered actin networks at the cell cortex or along highly organized actin bundles to distribute their cargo in a localized manner or move it across long distances in the cell. Transport is controlled by selective recruitment of the myosin to its cargo that also plays a role in activation of the motor.
Collapse
Affiliation(s)
- Margaret A Titus
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| |
Collapse
|
28
|
Fettiplace R. Hair Cell Transduction, Tuning, and Synaptic Transmission in the Mammalian Cochlea. Compr Physiol 2017; 7:1197-1227. [PMID: 28915323 DOI: 10.1002/cphy.c160049] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sound pressure fluctuations striking the ear are conveyed to the cochlea, where they vibrate the basilar membrane on which sit hair cells, the mechanoreceptors of the inner ear. Recordings of hair cell electrical responses have shown that they transduce sound via submicrometer deflections of their hair bundles, which are arrays of interconnected stereocilia containing the mechanoelectrical transducer (MET) channels. MET channels are activated by tension in extracellular tip links bridging adjacent stereocilia, and they can respond within microseconds to nanometer displacements of the bundle, facilitated by multiple processes of Ca2+-dependent adaptation. Studies of mouse mutants have produced much detail about the molecular organization of the stereocilia, the tip links and their attachment sites, and the MET channels localized to the lower end of each tip link. The mammalian cochlea contains two categories of hair cells. Inner hair cells relay acoustic information via multiple ribbon synapses that transmit rapidly without rundown. Outer hair cells are important for amplifying sound-evoked vibrations. The amplification mechanism primarily involves contractions of the outer hair cells, which are driven by changes in membrane potential and mediated by prestin, a motor protein in the outer hair cell lateral membrane. Different sound frequencies are separated along the cochlea, with each hair cell being tuned to a narrow frequency range; amplification sharpens the frequency resolution and augments sensitivity 100-fold around the cell's characteristic frequency. Genetic mutations and environmental factors such as acoustic overstimulation cause hearing loss through irreversible damage to the hair cells or degeneration of inner hair cell synapses. © 2017 American Physiological Society. Compr Physiol 7:1197-1227, 2017.
Collapse
Affiliation(s)
- Robert Fettiplace
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
29
|
Zmurchok C, Small T, Ward MJ, Edelstein-Keshet L. Application of Quasi-Steady-State Methods to Nonlinear Models of Intracellular Transport by Molecular Motors. Bull Math Biol 2017; 79:1923-1978. [PMID: 28707220 DOI: 10.1007/s11538-017-0314-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 06/22/2017] [Indexed: 01/29/2023]
Abstract
Molecular motors such as kinesin and dynein are responsible for transporting material along microtubule networks in cells. In many contexts, motor dynamics can be modelled by a system of reaction-advection-diffusion partial differential equations (PDEs). Recently, quasi-steady-state (QSS) methods have been applied to models with linear reactions to approximate the behaviour of the full PDE system. Here, we extend this QSS reduction methodology to certain nonlinear reaction models. The QSS method relies on the assumption that the nonlinear binding and unbinding interactions of the cellular motors occur on a faster timescale than the spatial diffusion and advection processes. The full system dynamics are shown to be well approximated by the dynamics on the slow manifold. The slow manifold is parametrized by a single scalar quantity that satisfies a scalar nonlinear PDE, called the QSS PDE. We apply the QSS method to several specific nonlinear models for the binding and unbinding of molecular motors, and we use the resulting approximations to draw conclusions regarding the parameter dependence of the spatial distribution of motors for these models.
Collapse
Affiliation(s)
- Cole Zmurchok
- Department of Mathematics, University of British Columbia, Vancouver, V6T 1Z2, Canada.
| | - Tim Small
- Department of Mathematics, University of British Columbia, Vancouver, V6T 1Z2, Canada
| | - Michael J Ward
- Department of Mathematics, University of British Columbia, Vancouver, V6T 1Z2, Canada
| | - Leah Edelstein-Keshet
- Department of Mathematics, University of British Columbia, Vancouver, V6T 1Z2, Canada
| |
Collapse
|
30
|
Defective Gpsm2/Gα i3 signalling disrupts stereocilia development and growth cone actin dynamics in Chudley-McCullough syndrome. Nat Commun 2017; 8:14907. [PMID: 28387217 PMCID: PMC5385604 DOI: 10.1038/ncomms14907] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 02/13/2017] [Indexed: 01/14/2023] Open
Abstract
Mutations in GPSM2 cause Chudley-McCullough syndrome (CMCS), an autosomal recessive neurological disorder characterized by early-onset sensorineural deafness and brain anomalies. Here, we show that mutation of the mouse orthologue of GPSM2 affects actin-rich stereocilia elongation in auditory and vestibular hair cells, causing deafness and balance defects. The G-protein subunit Gαi3, a well-documented partner of Gpsm2, participates in the elongation process, and its absence also causes hearing deficits. We show that Gpsm2 defines an ∼200 nm nanodomain at the tips of stereocilia and this localization requires the presence of Gαi3, myosin 15 and whirlin. Using single-molecule tracking, we report that loss of Gpsm2 leads to decreased outgrowth and a disruption of actin dynamics in neuronal growth cones. Our results elucidate the aetiology of CMCS and highlight a new molecular role for Gpsm2/Gαi3 in the regulation of actin dynamics in epithelial and neuronal tissues. Mutations in GPSM2 cause a rare disease characterized by deafness and brain abnormalities. Here the authors show that Gpsm2 forms a molecular complex with a heterotrimeric G-protein subunit, whirlin and a myosin motor to regulate actin dynamics in neurons and auditory hair cell stereocilia.
Collapse
|
31
|
Pinkoviezky I, Gov NS. Exclusion and Hierarchy of Time Scales Lead to Spatial Segregation of Molecular Motors in Cellular Protrusions. PHYSICAL REVIEW LETTERS 2017; 118:018102. [PMID: 28106430 DOI: 10.1103/physrevlett.118.018102] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Indexed: 06/06/2023]
Abstract
Molecular motors that carry cargo along biopolymer filaments within cells play a crucial role in the functioning of the cell. In particular, these motors are essential for the formation and maintenance of the cellular protrusions that play key roles in motility and specific functionalities, such as the stereocilia in hair cells. Typically, there are several species of motors, carrying different cargos, that share the same track. Furthermore, it was observed that in the mature stereocilia, the different motors occupy well-segregated bands as a function of distance from the tip. We use a totally asymmetric exclusion process model with two- and three-motor species, to study the conditions that give rise to such spatial patterns. We find that the well-segregated bands appear for motors with a strong hierarchy of attachment or detachment rates. This is a striking example of pattern formation in nonequilibrium, low-dimensional systems.
Collapse
Affiliation(s)
- I Pinkoviezky
- Department of Chemical Physics, Weizmann Institute of Science, P.O. Box 26, Rehovot 76100, Israel
| | - N S Gov
- Department of Chemical Physics, Weizmann Institute of Science, P.O. Box 26, Rehovot 76100, Israel
| |
Collapse
|
32
|
Masters TA, Kendrick-Jones J, Buss F. Myosins: Domain Organisation, Motor Properties, Physiological Roles and Cellular Functions. Handb Exp Pharmacol 2017; 235:77-122. [PMID: 27757761 DOI: 10.1007/164_2016_29] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Myosins are cytoskeletal motor proteins that use energy derived from ATP hydrolysis to generate force and movement along actin filaments. Humans express 38 myosin genes belonging to 12 classes that participate in a diverse range of crucial activities, including muscle contraction, intracellular trafficking, cell division, motility, actin cytoskeletal organisation and cell signalling. Myosin malfunction has been implicated a variety of disorders including deafness, hypertrophic cardiomyopathy, Usher syndrome, Griscelli syndrome and cancer. In this chapter, we will first discuss the key structural and kinetic features that are conserved across the myosin family. Thereafter, we summarise for each member in turn its unique functional and structural adaptations, cellular roles and associated pathologies. Finally, we address the broad therapeutic potential for pharmacological interventions that target myosin family members.
Collapse
Affiliation(s)
- Thomas A Masters
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK.
| | | | - Folma Buss
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK
| |
Collapse
|
33
|
Global Analysis of Protein Expression of Inner Ear Hair Cells. J Neurosci 2016; 37:1320-1339. [PMID: 28039372 DOI: 10.1523/jneurosci.2267-16.2016] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 11/29/2016] [Accepted: 12/19/2016] [Indexed: 11/21/2022] Open
Abstract
The mammalian inner ear (IE) subserves auditory and vestibular sensations via highly specialized cells and proteins. Sensory receptor hair cells (HCs) are necessary for transducing mechanical inputs and stimulating sensory neurons by using a host of known and as yet unknown protein machinery. To understand the protein composition of these unique postmitotic cells, in which irreversible protein degradation or damage can lead to impaired hearing and balance, we analyzed IE samples by tandem mass spectrometry to generate an unbiased, shotgun-proteomics view of protein identities and abundances. By using Pou4f3/eGFP-transgenic mice in which HCs express GFP driven by Pou4f3, we FACS purified a population of HCs to analyze and compare the HC proteome with other IE subproteomes from sensory epithelia and whole IE. We show that the mammalian HC proteome comprises hundreds of uniquely or highly expressed proteins. Our global proteomic analysis of purified HCs extends the existing HC transcriptome, revealing previously undetected gene products and isoform-specific protein expression. Comparison of our proteomic data with mouse and human databases of genetic auditory/vestibular impairments confirms the critical role of the HC proteome for normal IE function, providing a cell-specific pool of candidates for novel, important HC genes. Several proteins identified exclusively in HCs by proteomics and verified by immunohistochemistry map to human genetic deafness loci, potentially representing new deafness genes. SIGNIFICANCE STATEMENT Hearing and balance rely on specialized sensory hair cells (HCs) in the inner ear (IE) to convey information about sound, acceleration, and orientation to the brain. Genetically and environmentally induced perturbations to HC proteins can result in deafness and severe imbalance. We used transgenic mice with GFP-expressing HCs, coupled with FACS sorting and tandem mass spectrometry, to define the most complete HC and IE proteome to date. We show that hundreds of proteins are uniquely identified or enriched in HCs, extending previous gene expression analyses to reveal novel HC proteins and isoforms. Importantly, deafness-linked proteins were significantly enriched in HCs, suggesting that this in-depth proteomic analysis of IE sensory cells may hold potential for deafness gene discovery.
Collapse
|
34
|
Raval MH, Quintero OA, Weck ML, Unrath WC, Gallagher JW, Cui R, Kachar B, Tyska MJ, Yengo CM. Impact of the Motor and Tail Domains of Class III Myosins on Regulating the Formation and Elongation of Actin Protrusions. J Biol Chem 2016; 291:22781-22792. [PMID: 27582493 PMCID: PMC5077211 DOI: 10.1074/jbc.m116.733741] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 08/29/2016] [Indexed: 11/06/2022] Open
Abstract
Class III myosins (MYO3A and MYO3B) are proposed to function as transporters as well as length and ultrastructure regulators within stable actin-based protrusions such as stereocilia and calycal processes. MYO3A differs from MYO3B in that it contains an extended tail domain with an additional actin-binding motif. We examined how the properties of the motor and tail domains of human class III myosins impact their ability to enhance the formation and elongation of actin protrusions. Direct examination of the motor and enzymatic properties of human MYO3A and MYO3B revealed that MYO3A is a 2-fold faster motor with enhanced ATPase activity and actin affinity. A chimera in which the MYO3A tail was fused to the MYO3B motor demonstrated that motor activity correlates with formation and elongation of actin protrusions. We demonstrate that removal of individual exons (30-34) in the MYO3A tail does not prevent filopodia tip localization but abolishes the ability to enhance actin protrusion formation and elongation in COS7 cells. Interestingly, our results demonstrate that MYO3A slows filopodia dynamics and enhances filopodia lifetime in COS7 cells. We also demonstrate that MYO3A is more efficient than MYO3B at increasing formation and elongation of stable microvilli on the surface of cultured epithelial cells. We propose that the unique features of MYO3A, enhanced motor activity, and an extended tail with tail actin-binding motif, allow it to play an important role in stable actin protrusion length and ultrastructure maintenance.
Collapse
Affiliation(s)
- Manmeet H Raval
- From the Department of Cellular and Molecular Physiology, Pennsylvania State University, College of Medicine, Hershey, Pennsylvania 17033
| | - Omar A Quintero
- the Department of Biology, University of Richmond, Richmond, Virginia 23173
| | - Meredith L Weck
- the Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - William C Unrath
- From the Department of Cellular and Molecular Physiology, Pennsylvania State University, College of Medicine, Hershey, Pennsylvania 17033
| | - James W Gallagher
- the Department of Biology, Lincoln University, Philadelphia, Pennsylvania 19104, and
| | - Runjia Cui
- the Laboratory of Cell Structure and Dynamics, NIDCD, National Institutes of Health, Bethesda, Maryland 20892
| | - Bechara Kachar
- the Laboratory of Cell Structure and Dynamics, NIDCD, National Institutes of Health, Bethesda, Maryland 20892
| | - Matthew J Tyska
- the Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Christopher M Yengo
- From the Department of Cellular and Molecular Physiology, Pennsylvania State University, College of Medicine, Hershey, Pennsylvania 17033,
| |
Collapse
|
35
|
Qu R, Sang Q, Xu Y, Feng R, Jin L, He L, Wang L. Identification of a novel homozygous mutation in MYO3A in a Chinese family with DFNB30 non-syndromic hearing impairment. Int J Pediatr Otorhinolaryngol 2016; 84:43-7. [PMID: 27063751 DOI: 10.1016/j.ijporl.2016.02.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/25/2016] [Accepted: 02/26/2016] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Hearing loss is a common sensory impairment. Several genetic loci or genes responsible for non-syndrome hearing loss have been identified, including the well-known deafness genes GJB2, MT-RNR1 and SLC26A4. MYO3A belongs to the myosin superfamily. Previously only three mutations in this gene have been found in an Isreali family with DFNB30, in which patients demonstrated progressive hearing loss. METHODS In this study, we characterized a consanguineous Kazakh family with congenital hearing loss. By targeted sequence capture and next-generation sequencing, we identified a homozygous mutation and did bioinformatics analysis to this mutation. RESULTS A homozygous mutation, MYO3A:c.1841C>T (p.S614F), was identified to be responsible for the disease. Ser614 is located in the motor domain of MYO3A that is highly conserved among different species. Molecular modeling predicts that the conserved Ser614 may play an important role in maintaining the stability of β-sheet and the interaction between neighboring β-strand. CONCLUSIONS This is the second report on MYO3A mutations in deafness and the first report in China. The finding help facilitate establishing a better relationship between MYO3A mutation and hearing phenotypes.
Collapse
Affiliation(s)
- Ronggui Qu
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Qing Sang
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yao Xu
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Ruizhi Feng
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Lin He
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China; Bio-X Center, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Wang
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
36
|
Ebrahim S, Avenarius MR, Grati M, Krey JF, Windsor AM, Sousa AD, Ballesteros A, Cui R, Millis BA, Salles FT, Baird MA, Davidson MW, Jones SM, Choi D, Dong L, Raval MH, Yengo CM, Barr-Gillespie PG, Kachar B. Stereocilia-staircase spacing is influenced by myosin III motors and their cargos espin-1 and espin-like. Nat Commun 2016; 7:10833. [PMID: 26926603 PMCID: PMC4773517 DOI: 10.1038/ncomms10833] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 01/25/2016] [Indexed: 12/12/2022] Open
Abstract
Hair cells tightly control the dimensions of their stereocilia, which are actin-rich protrusions with graded heights that mediate mechanotransduction in the inner ear. Two members of the myosin-III family, MYO3A and MYO3B, are thought to regulate stereocilia length by transporting cargos that control actin polymerization at stereocilia tips. We show that eliminating espin-1 (ESPN-1), an isoform of ESPN and a myosin-III cargo, dramatically alters the slope of the stereocilia staircase in a subset of hair cells. Furthermore, we show that espin-like (ESPNL), primarily present in developing stereocilia, is also a myosin-III cargo and is essential for normal hearing. ESPN-1 and ESPNL each bind MYO3A and MYO3B, but differentially influence how the two motors function. Consequently, functional properties of different motor-cargo combinations differentially affect molecular transport and the length of actin protrusions. This mechanism is used by hair cells to establish the required range of stereocilia lengths within a single cell.
Collapse
Affiliation(s)
- Seham Ebrahim
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Matthew R Avenarius
- Oregon Hearing Research Center and Vollum Institute, Oregon Health &Science University, Portland, Oregon 97239, USA
| | - M'hamed Grati
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Jocelyn F Krey
- Oregon Hearing Research Center and Vollum Institute, Oregon Health &Science University, Portland, Oregon 97239, USA
| | - Alanna M Windsor
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Aurea D Sousa
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Angela Ballesteros
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Runjia Cui
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Bryan A Millis
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Felipe T Salles
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Michelle A Baird
- National High Magnetic Field Laboratory and Department of Biological Science, Florida State University, Tallahassee, Florida 32310, USA
| | - Michael W Davidson
- National High Magnetic Field Laboratory and Department of Biological Science, Florida State University, Tallahassee, Florida 32310, USA
| | - Sherri M Jones
- Department of Special Education and Communication Disorders, University of Nebraska-Lincoln, Lincoln, Nebraska 68583, USA
| | - Dongseok Choi
- Department of Public Health and Preventive Medicine, Oregon Health and Science University, Portland, Oregon 97239, USA
| | - Lijin Dong
- Genetic Engineering Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Manmeet H Raval
- Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, Pennsylvania 17033, USA
| | - Christopher M Yengo
- Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, Pennsylvania 17033, USA
| | - Peter G Barr-Gillespie
- Oregon Hearing Research Center and Vollum Institute, Oregon Health &Science University, Portland, Oregon 97239, USA
| | - Bechara Kachar
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
37
|
Grati M, Yan D, Raval MH, Walsh T, Ma Q, Chakchouk I, Kannan-Sundhari A, Mittal R, Masmoudi S, Blanton SH, Tekin M, King MC, Yengo CM, Liu XZ. MYO3A Causes Human Dominant Deafness and Interacts with Protocadherin 15-CD2 Isoform. Hum Mutat 2016; 37:481-7. [PMID: 26841241 DOI: 10.1002/humu.22961] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 01/15/2016] [Indexed: 01/10/2023]
Abstract
Hereditary hearing loss (HL) is characterized by both allelic and locus genetic heterogeneity. Both recessive and dominant forms of HL may be caused by different mutations in the same deafness gene. In a family with post-lingual progressive non-syndromic deafness, whole-exome sequencing of genomic DNA from five hearing-impaired relatives revealed a single variant, p.Gly488Glu (rs145970949:G>A) in MYO3A, co-segregating with HL as an autosomal dominant trait. This amino acid change, predicted to be pathogenic, alters a highly conserved residue in the motor domain of MYO3A. The mutation severely alters the ATPase activity and motility of the protein in vitro, and the mutant protein fails to accumulate in the filopodia tips in COS7 cells. However, the mutant MYO3A was able to reach the tips of organotypic inner ear culture hair cell stereocilia, raising the possibility of a local effect on positioning of the mechanoelectrical transduction (MET) complex at the stereocilia tips. To address this hypothesis, we investigated the interaction of MYO3A with the cytosolic tail of the integral tip-link protein protocadherin 15 (PCDH15), a core component of MET complex. Interestingly, we uncovered a novel interaction between MYO3A and PCDH15 shedding new light on the function of myosin IIIA at stereocilia tips.
Collapse
Affiliation(s)
- M'hamed Grati
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Denise Yan
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Manmeet H Raval
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Tom Walsh
- Departments of Medicine and Genome Sciences, University of Washington, Seattle, Washington
| | - Qi Ma
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Imen Chakchouk
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida.,Laboratoire Procédés de Criblage Moléculaire et Cellulaire, Centre de Biotechnologie de Sfax, Université de Sfax, Sfax, Tunisie
| | | | - Rahul Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Saber Masmoudi
- Laboratoire Procédés de Criblage Moléculaire et Cellulaire, Centre de Biotechnologie de Sfax, Université de Sfax, Sfax, Tunisie
| | - Susan H Blanton
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida.,Dr. John T. Macdonald Foundation, Department of Human Genetics, and John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida
| | - Mustafa Tekin
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida.,Dr. John T. Macdonald Foundation, Department of Human Genetics, and John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida
| | - Mary-Claire King
- Departments of Medicine and Genome Sciences, University of Washington, Seattle, Washington
| | - Christopher M Yengo
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Xue Zhong Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida.,Dr. John T. Macdonald Foundation, Department of Human Genetics, and John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida.,Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
38
|
Liu H, Li J, Raval MH, Yao N, Deng X, Lu Q, Nie S, Feng W, Wan J, Yengo CM, Liu W, Zhang M. Myosin III-mediated cross-linking and stimulation of actin bundling activity of Espin. eLife 2016; 5. [PMID: 26785147 PMCID: PMC4758956 DOI: 10.7554/elife.12856] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 01/18/2016] [Indexed: 11/13/2022] Open
Abstract
Class III myosins (Myo3) and actin-bundling protein Espin play critical roles in regulating the development and maintenance of stereocilia in vertebrate hair cells, and their defects cause hereditary hearing impairments. Myo3 interacts with Espin1 through its tail homology I motif (THDI), however it is not clear how Myo3 specifically acts through Espin1 to regulate the actin bundle assembly and stabilization. Here we discover that Myo3 THDI contains a pair of repeat sequences capable of independently and strongly binding to the ankyrin repeats of Espin1, revealing an unexpected Myo3-mediated cross-linking mechanism of Espin1. The structures of Myo3 in complex with Espin1 not only elucidate the mechanism of the binding, but also reveal a Myo3-induced release of Espin1 auto-inhibition mechanism. We also provide evidence that Myo3-mediated cross-linking can further promote actin fiber bundling activity of Espin1.
Collapse
Affiliation(s)
- Haiyang Liu
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Jianchao Li
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
| | - Manmeet H Raval
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, United States
| | - Ningning Yao
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Xiaoying Deng
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Qing Lu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
| | - Si Nie
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Wei Feng
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jun Wan
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China.,Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
| | - Christopher M Yengo
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, United States
| | - Wei Liu
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China.,Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
| | - Mingjie Zhang
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China.,Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China.,Center of Systems Biology and Human Health, School of Science and Institute for Advanced Study, Hong Kong University of Science and Technology, Hong Kong, China
| |
Collapse
|
39
|
Pollock LM, Chou SW, McDermott BM. My oh my(osin): Insights into how auditory hair cells count, measure, and shape. J Cell Biol 2016; 212:135-7. [PMID: 26754648 PMCID: PMC4721532 DOI: 10.1083/jcb.201512086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 12/30/2015] [Indexed: 11/22/2022] Open
Abstract
The mechanisms underlying mechanosensory hair bundle formation in auditory sensory cells are largely mysterious. In this issue, Lelli et al. (2016. J. Cell Biol.http://dx.doi.org/10.1083/jcb.201509017) reveal that a pair of molecular motors, myosin IIIa and myosin IIIb, is involved in the hair bundle’s morphology and hearing.
Collapse
Affiliation(s)
- Lana M Pollock
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Case Western Reserve University, Cleveland, OH 44106 Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Shih-Wei Chou
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Case Western Reserve University, Cleveland, OH 44106 Department of Biology, Case Western Reserve University, Cleveland, OH 44106
| | - Brian M McDermott
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Case Western Reserve University, Cleveland, OH 44106 Department of Biology, Case Western Reserve University, Cleveland, OH 44106 Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106 Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| |
Collapse
|
40
|
Lelli A, Michel V, Boutet de Monvel J, Cortese M, Bosch-Grau M, Aghaie A, Perfettini I, Dupont T, Avan P, El-Amraoui A, Petit C. Class III myosins shape the auditory hair bundles by limiting microvilli and stereocilia growth. J Cell Biol 2016; 212:231-44. [PMID: 26754646 PMCID: PMC4738386 DOI: 10.1083/jcb.201509017] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 12/07/2015] [Indexed: 11/22/2022] Open
Abstract
Analysis of mice deficient for myosin IIIa and myosin IIIb shows that class III myosins limit the elongation of stereocilia and of subsequently regressing microvilli, thus contributing to the early hair bundle shaping. The precise architecture of hair bundles, the arrays of mechanosensitive microvilli-like stereocilia crowning the auditory hair cells, is essential to hearing. Myosin IIIa, defective in the late-onset deafness form DFNB30, has been proposed to transport espin-1 to the tips of stereocilia, thereby promoting their elongation. We show that Myo3a−/−Myo3b−/− mice lacking myosin IIIa and myosin IIIb are profoundly deaf, whereas Myo3a-cKO Myo3b−/− mice lacking myosin IIIb and losing myosin IIIa postnatally have normal hearing. Myo3a−/−Myo3b−/− cochlear hair bundles display robust mechanoelectrical transduction currents with normal kinetics but show severe embryonic abnormalities whose features rapidly change. These include abnormally tall and numerous microvilli or stereocilia, ungraded stereocilia bundles, and bundle rounding and closure. Surprisingly, espin-1 is properly targeted to Myo3a−/−Myo3b−/− stereocilia tips. Our results uncover the critical role that class III myosins play redundantly in hair-bundle morphogenesis; they unexpectedly limit the elongation of stereocilia and of subsequently regressing microvilli, thus contributing to the early hair bundle shaping.
Collapse
Affiliation(s)
- Andrea Lelli
- Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, 75724 Paris, Cedex 15, France Unité Mixte de Recherche UMRS1120, Institut National de la Santé et de la Recherche Médicale, 75015 Paris, France Sorbonne Universités, Université Pierre et Marie Curie (UPMC Paris VI), Complexité du Vivant, 75005 Paris, France
| | - Vincent Michel
- Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, 75724 Paris, Cedex 15, France Unité Mixte de Recherche UMRS1120, Institut National de la Santé et de la Recherche Médicale, 75015 Paris, France Sorbonne Universités, Université Pierre et Marie Curie (UPMC Paris VI), Complexité du Vivant, 75005 Paris, France
| | - Jacques Boutet de Monvel
- Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, 75724 Paris, Cedex 15, France Unité Mixte de Recherche UMRS1120, Institut National de la Santé et de la Recherche Médicale, 75015 Paris, France Sorbonne Universités, Université Pierre et Marie Curie (UPMC Paris VI), Complexité du Vivant, 75005 Paris, France
| | - Matteo Cortese
- Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, 75724 Paris, Cedex 15, France Unité Mixte de Recherche UMRS1120, Institut National de la Santé et de la Recherche Médicale, 75015 Paris, France Sorbonne Universités, Université Pierre et Marie Curie (UPMC Paris VI), Complexité du Vivant, 75005 Paris, France
| | - Montserrat Bosch-Grau
- Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, 75724 Paris, Cedex 15, France Unité Mixte de Recherche UMRS1120, Institut National de la Santé et de la Recherche Médicale, 75015 Paris, France Sorbonne Universités, Université Pierre et Marie Curie (UPMC Paris VI), Complexité du Vivant, 75005 Paris, France
| | - Asadollah Aghaie
- Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, 75724 Paris, Cedex 15, France Unité Mixte de Recherche UMRS1120, Institut National de la Santé et de la Recherche Médicale, 75015 Paris, France Sorbonne Universités, Université Pierre et Marie Curie (UPMC Paris VI), Complexité du Vivant, 75005 Paris, France Syndrome de Usher et Autres Atteintes Rétino-Cochléaires, Institut de la Vision, 75012 Paris, France
| | - Isabelle Perfettini
- Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, 75724 Paris, Cedex 15, France Unité Mixte de Recherche UMRS1120, Institut National de la Santé et de la Recherche Médicale, 75015 Paris, France Sorbonne Universités, Université Pierre et Marie Curie (UPMC Paris VI), Complexité du Vivant, 75005 Paris, France
| | - Typhaine Dupont
- Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, 75724 Paris, Cedex 15, France Unité Mixte de Recherche UMRS1120, Institut National de la Santé et de la Recherche Médicale, 75015 Paris, France Sorbonne Universités, Université Pierre et Marie Curie (UPMC Paris VI), Complexité du Vivant, 75005 Paris, France
| | - Paul Avan
- Laboratoire de Biophysique Sensorielle, Faculté de Médecine, Université d'Auvergne; Biophysique Médicale, Centre Jean Perrin, 63000 Clermont-Ferrand, France
| | - Aziz El-Amraoui
- Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, 75724 Paris, Cedex 15, France Unité Mixte de Recherche UMRS1120, Institut National de la Santé et de la Recherche Médicale, 75015 Paris, France Sorbonne Universités, Université Pierre et Marie Curie (UPMC Paris VI), Complexité du Vivant, 75005 Paris, France
| | - Christine Petit
- Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, 75724 Paris, Cedex 15, France Unité Mixte de Recherche UMRS1120, Institut National de la Santé et de la Recherche Médicale, 75015 Paris, France Sorbonne Universités, Université Pierre et Marie Curie (UPMC Paris VI), Complexité du Vivant, 75005 Paris, France Syndrome de Usher et Autres Atteintes Rétino-Cochléaires, Institut de la Vision, 75012 Paris, France Collège de France, 75005 Paris, France
| |
Collapse
|
41
|
Kim J. Unconventional mechanics of lipid membranes: a potential role for mechanotransduction of hair cell stereocilia. Biophys J 2015; 108:610-21. [PMID: 25650928 DOI: 10.1016/j.bpj.2014.12.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 11/15/2014] [Accepted: 12/10/2014] [Indexed: 01/20/2023] Open
Abstract
A force-conveying role of the lipid membrane across various mechanoreceptors is now an accepted hypothesis. However, such a mechanism is still not fully understood for mechanotransduction in the hair bundle of auditory sensory hair cells. A major goal of this theoretical assessment was to investigate the role of the lipid membrane in auditory mechanotransduction, especially in generating nonlinear bundle force versus displacement measurements, one of the main features of auditory mechanotransduction. To this end, a hair bundle model that generates lipid membrane tented deformation in the stereocilia was developed. A computational analysis of the model not only reproduced nonlinear bundle force measurements but also generated membrane energy that is potentially sufficient to activate the mechanosensitive ion channel of the hair cell. In addition, the model provides biophysical insight into 1) the likelihood that the channel must be linked in some way to the tip link; 2) how the interplay of the bending and stretching of the lipid bilayer may be responsible for the nonlinear force versus displacement response; 3) how measurements of negative stiffness may be a function of the rotational stiffness of the rootlets; and 4) how the standing tension of the tip link is required to interpret migration of the nonlinear force versus displacement and activation curves. These are all features of hair cell mechanotransduction, but the underlying biophysical mechanism has proved elusive for the last three decades.
Collapse
Affiliation(s)
- Jichul Kim
- Department of Mechanical Engineering, Stanford University, Stanford, California.
| |
Collapse
|
42
|
Zhao B, Müller U. The elusive mechanotransduction machinery of hair cells. Curr Opin Neurobiol 2015; 34:172-9. [PMID: 26342686 DOI: 10.1016/j.conb.2015.08.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 08/06/2015] [Accepted: 08/18/2015] [Indexed: 11/18/2022]
Abstract
Hair cells in the mammalian cochlea are specialized sensory cells that convert mechanical signals evoked by sound waves into electrochemical signals. Several integral membrane proteins have recently been identified that are closely linked to the mechanotransduction process. Efforts are under way to determine the extent to which they are subunits of the long thought-after mechanotransduction channel. Recent findings also suggest that mechanotransduction may have a role in fine tuning the length of the stereocilia and thus in the regulation of morphological features of hair cells that are inherently linked to the mechanotransduction process.
Collapse
Affiliation(s)
- Bo Zhao
- Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, United States
| | - Ulrich Müller
- Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, United States.
| |
Collapse
|
43
|
Orly G, Naoz M, Gov NS. Physical model for the geometry of actin-based cellular protrusions. Biophys J 2015; 107:576-587. [PMID: 25099797 DOI: 10.1016/j.bpj.2014.05.040] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 05/09/2014] [Accepted: 05/28/2014] [Indexed: 11/17/2022] Open
Abstract
Actin-based cellular protrusions are a ubiquitous feature of cell morphology, e.g., filopodia and microvilli, serving a huge variety of functions. Despite this, there is still no comprehensive model for the mechanisms that determine the geometry of these protrusions. We present here a detailed computational model that addresses a combination of multiple biochemical and physical processes involved in the dynamic regulation of the shape of these protrusions. We specifically explore the role of actin polymerization in determining both the height and width of the protrusions. Furthermore, we show that our generalized model can explain multiple morphological features of these systems, and account for the effects of specific proteins and mutations.
Collapse
Affiliation(s)
- G Orly
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot, Israel
| | - M Naoz
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot, Israel
| | - N S Gov
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
44
|
Mecklenburg KL, Freed SA, Raval M, Quintero OA, Yengo CM, O'Tousa JE. Invertebrate and vertebrate class III myosins interact with MORN repeat-containing adaptor proteins. PLoS One 2015; 10:e0122502. [PMID: 25822849 PMCID: PMC4379085 DOI: 10.1371/journal.pone.0122502] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 02/18/2015] [Indexed: 12/22/2022] Open
Abstract
In Drosophila photoreceptors, the NINAC-encoded myosin III is found in a complex with a small, MORN-repeat containing, protein Retinophilin (RTP). Expression of these two proteins in other cell types showed NINAC myosin III behavior is altered by RTP. NINAC deletion constructs were used to map the RTP binding site within the proximal tail domain of NINAC. In vertebrates, the RTP ortholog is MORN4. Co-precipitation experiments demonstrated that human MORN4 binds to human myosin IIIA (MYO3A). In COS7 cells, MORN4 and MYO3A, but not MORN4 and MYO3B, co-localize to actin rich filopodia extensions. Deletion analysis mapped the MORN4 binding to the proximal region of the MYO3A tail domain. MYO3A dependent MORN4 tip localization suggests that MYO3A functions as a motor that transports MORN4 to the filopodia tips and MORN4 may enhance MYO3A tip localization by tethering it to the plasma membrane at the protrusion tips. These results establish conserved features of the RTP/MORN4 family: they bind within the tail domain of myosin IIIs to control their behavior.
Collapse
Affiliation(s)
- Kirk L. Mecklenburg
- Department of Biology, Indiana University South Bend, South Bend, Indiana, United States of America
| | - Stephanie A. Freed
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Manmeet Raval
- Department of Cellular and Molecular Physiology, Pennsylvania State University, College of Medicine, Hershey, Pennsylvania, United States of America
| | - Omar A. Quintero
- Department of Biology, University of Richmond, Richmond, Virginia, United States of America
| | - Christopher M. Yengo
- Department of Cellular and Molecular Physiology, Pennsylvania State University, College of Medicine, Hershey, Pennsylvania, United States of America
- * E-mail: (CY); (JO)
| | - Joseph. E. O'Tousa
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
- * E-mail: (CY); (JO)
| |
Collapse
|
45
|
Nishio SY, Hattori M, Moteki H, Tsukada K, Miyagawa M, Naito T, Yoshimura H, Iwasa YI, Mori K, Shima Y, Sakuma N, Usami SI. Gene expression profiles of the cochlea and vestibular endorgans: localization and function of genes causing deafness. Ann Otol Rhinol Laryngol 2015; 124 Suppl 1:6S-48S. [PMID: 25814645 DOI: 10.1177/0003489415575549] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVES We sought to elucidate the gene expression profiles of the causative genes as well as the localization of the encoded proteins involved in hereditary hearing loss. METHODS Relevant articles (as of September 2014) were searched in PubMed databases, and the gene symbols of the genes reported to be associated with deafness were located on the Hereditary Hearing Loss Homepage using localization, expression, and distribution as keywords. RESULTS Our review of the literature allowed us to systematize the gene expression profiles for genetic deafness in the inner ear, clarifying the unique functions and specific expression patterns of these genes in the cochlea and vestibular endorgans. CONCLUSIONS The coordinated actions of various encoded molecules are essential for the normal development and maintenance of auditory and vestibular function.
Collapse
Affiliation(s)
- Shin-Ya Nishio
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto, Japan
| | - Mitsuru Hattori
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hideaki Moteki
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Keita Tsukada
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Maiko Miyagawa
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takehiko Naito
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hidekane Yoshimura
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yoh-Ichiro Iwasa
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Kentaro Mori
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yutaka Shima
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Naoko Sakuma
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan Department of Otorhinolaryngology and Head and Neck Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Shin-Ichi Usami
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|
46
|
An BC, Sakai T, Komaba S, Kishi H, Kobayashi S, Kim JY, Ikebe R, Ikebe M. Phosphorylation of the kinase domain regulates autophosphorylation of myosin IIIA and its translocation in microvilli. Biochemistry 2014; 53:7835-45. [PMID: 25402663 PMCID: PMC4270376 DOI: 10.1021/bi501247z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
![]()
Motor activity of myosin III is regulated
by autophosphorylation.
To investigate the role of the kinase activity on the transporter
function of myosin IIIA (Myo3A), we identified the phosphorylation
sites of kinase domain (KD), which is responsible for the regulation
of kinase activity and thus motor function. Using mass spectrometry,
we identified six phosphorylation sites in the KD, which are highly
conserved among class III myosins and Ste20-related misshapen (Msn)
kinases. Two predominant sites, Thr184 and Thr188, in KD are important for phosphorylation of the KD as well as the
motor domain, which regulates the affinity for actin. In the Caco2
cells, the full-length human Myo3A (hMyo3AFull) markedly enlarged
the microvilli, although it did not show discrete localization within
the microvilli. On the other hand, hMyo3AFull(T184A) and hMyo3AFull(T188A)
both showed clear localization at the microvilli tips. Our results
suggest that Myo3A induces large actin bundle formation to form microvilli,
and phosphorylation of KD at Thr184 and Thr188 is critical for the kinase activity of Myo3A, and regulation of
Myo3A translocation to the tip of microvilli. Retinal extracts potently
dephosphorylate both KD and motor domain without IQ motifs (MDIQo),
which was inhibited by okadaic acid (OA) with nanomolar range and
by tautomycetin (TMC) with micromolar range. The results suggest that
Myo3A phosphatase is protein phosphatase type 2A (PP2A). Supporting
this result, recombinant PP2Ac potently dephosphorylates both KD and
MDIQo. We propose that the phosphorylation–dephosphorylation
mechanism plays an essential role in mediating the transport and actin
bundle formation and stability functions of hMyo3A.
Collapse
Affiliation(s)
- Byung Chull An
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School , Worcester, Massachusetts 01605, United States
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Najmabadi H, Kahrizi K. Genetics of non-syndromic hearing loss in the Middle East. Int J Pediatr Otorhinolaryngol 2014; 78:2026-36. [PMID: 25281338 DOI: 10.1016/j.ijporl.2014.08.036] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 08/24/2014] [Accepted: 08/25/2014] [Indexed: 12/23/2022]
Abstract
Hearing impairment is the most common sensory disorder, present 1 in every 500 newborns. About 80% of genetic HL is classified as non-syndromic deafness. To date, over 115 non-syndromic loci have been identified of which fifty associated with autosomal recessive non-syndromic hearing loss (ARNSHL). In this review article, we represent the 40 genes function and contribution to genetic deafness in different Middle Eastern populations as well as gene frequencies and mutation spectrum. The wide variety of mutations have so far detected in 19 countries reflects the heterogeneity of the genes involved in HL in this region. The deafness genes can cause dysfunction of cochlear homeostasis, cellular organization, neuronal transmission, cell growth, differentiation, and survival, some coding for tectorial membrane-associated proteins, and the remaining with unknown functions. Non-syndromic deafness is highly heterogeneous and mutations in the GJB2 are responsible for almost 30-50% in northwest to as low as 0-5% in south and southeast of the Middle East, it remain as major gene in ARNSHL in Middle East. The other genes contributing to AR/ADNSHL in some countries have been determined while for many other countries in the Middle East have not been studied or little study has been done. With the advancement of next generation sequencing one could expect in next coming year many of the remaining genes to be determine and to understand their function in the inner ear.
Collapse
Affiliation(s)
- Hossein Najmabadi
- Genetics Research Centre (GRC), University of Social Welfare and Rehabilitation Sciences, Tehran, Iran.
| | - Kimia Kahrizi
- Genetics Research Centre (GRC), University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| |
Collapse
|
48
|
Fettiplace R, Kim KX. The physiology of mechanoelectrical transduction channels in hearing. Physiol Rev 2014; 94:951-86. [PMID: 24987009 DOI: 10.1152/physrev.00038.2013] [Citation(s) in RCA: 193] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Much is known about the mechanotransducer (MT) channels mediating transduction in hair cells of the vertrbrate inner ear. With the use of isolated preparations, it is experimentally feasible to deliver precise mechanical stimuli to individual cells and record the ensuing transducer currents. This approach has shown that small (1-100 nm) deflections of the hair-cell stereociliary bundle are transmitted via interciliary tip links to open MT channels at the tops of the stereocilia. These channels are cation-permeable with a high selectivity for Ca(2+); two channels are thought to be localized at the lower end of the tip link, each with a large single-channel conductance that increases from the low- to high-frequency end of the cochlea. Ca(2+) influx through open channels regulates their resting open probability, which may contribute to setting the hair cell resting potential in vivo. Ca(2+) also controls transducer fast adaptation and force generation by the hair bundle, the two coupled processes increasing in speed from cochlear apex to base. The molecular intricacy of the stereocilary bundle and the transduction apparatus is reflected by the large number of single-gene mutations that are linked to sensorineural deafness, especially those in Usher syndrome. Studies of such mutants have led to the discovery of many of the molecules of the transduction complex, including the tip link and its attachments to the stereociliary core. However, the MT channel protein is still not firmly identified, nor is it known whether the channel is activated by force delivered through accessory proteins or by deformation of the lipid bilayer.
Collapse
Affiliation(s)
- Robert Fettiplace
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kyunghee X Kim
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
49
|
Abstract
Uniquely among human senses, hearing is not simply a passive response to stimulation. Our auditory system is instead enhanced by an active process in cochlear hair cells that amplifies acoustic signals several hundred-fold, sharpens frequency selectivity and broadens the ear's dynamic range. Active motility of the mechanoreceptive hair bundles underlies the active process in amphibians and some reptiles; in mammals, this mechanism operates in conjunction with prestin-based somatic motility. Both individual hair bundles and the cochlea as a whole operate near a dynamical instability, the Hopf bifurcation, which accounts for the cardinal features of the active process.
Collapse
|
50
|
Genetics of auditory mechano-electrical transduction. Pflugers Arch 2014; 467:49-72. [PMID: 24957570 PMCID: PMC4281357 DOI: 10.1007/s00424-014-1552-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 06/04/2014] [Accepted: 06/05/2014] [Indexed: 12/29/2022]
Abstract
The hair bundles of cochlear hair cells play a central role in the auditory mechano-electrical transduction (MET) process. The identification of MET components and of associated molecular complexes by biochemical approaches is impeded by the very small number of hair cells within the cochlea. In contrast, human and mouse genetics have proven to be particularly powerful. The study of inherited forms of deafness led to the discovery of several essential proteins of the MET machinery, which are currently used as entry points to decipher the associated molecular networks. Notably, MET relies not only on the MET machinery but also on several elements ensuring the proper sound-induced oscillation of the hair bundle or the ionic environment necessary to drive the MET current. Here, we review the most significant advances in the molecular bases of the MET process that emerged from the genetics of hearing.
Collapse
|