1
|
Schubert NMA, Reijntjes DOJ, van Tuinen M, Vijayakumar S, Jones TA, Jones SM, Pyott SJ. Pathophysiological processes underlying hidden hearing loss revealed in Kcnt1/2 double knockout mice. Aging Cell 2024; 23:e14243. [PMID: 39049179 PMCID: PMC11488318 DOI: 10.1111/acel.14243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/25/2024] [Accepted: 05/30/2024] [Indexed: 07/27/2024] Open
Abstract
Presbycusis is a prevalent condition in older adults characterized by the progressive loss of hearing due to age-related changes in the cochlea, the auditory portion of the inner ear. Many adults also struggle with understanding speech in noise despite having normal auditory thresholds, a condition termed "hidden" hearing loss because it evades standard audiological assessments. Examination of animal models and postmortem human tissue suggests that hidden hearing loss is also associated with age-related changes in the cochlea and may, therefore, precede overt age-related hearing loss. Nevertheless, the pathological mechanisms underlying hidden hearing loss are not understood, which hinders the development of diagnostic biomarkers and effective treatments for age-related hearing loss. To fill these gaps in knowledge, we leveraged a combination of tools, including transcriptomic profiling and morphological and functional assessments, to identify these processes and examine the transition from hidden to overt hearing loss. As a novel approach, we took advantage of a recently characterized model of hidden hearing loss: Kcnt1/2 double knockout mice. Using this model, we find that even before observable morphological pathology, hidden hearing loss is associated with significant alteration in several processes, notably proteostasis, in the cochlear sensorineural structures, and increased susceptibility to overt hearing loss in response to noise exposure and aging. Our findings provide the first insight into the pathophysiology associated with the earliest and, therefore, most treatable stages of hearing loss and provide critical insight directing future investigation of pharmaceutical strategies to slow and possibly prevent overt age-related hearing loss.
Collapse
Affiliation(s)
- Nick M A Schubert
- Department of Otorhinolaryngology/Head and Neck Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Graduate School of Medical Sciences Research School of Behavioural and Cognitive Neurosciences, University of Groningen, Groningen, The Netherlands
| | - Daniël O J Reijntjes
- The Center for Hearing and Balance, Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Marcel van Tuinen
- Department of Otorhinolaryngology/Head and Neck Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sarath Vijayakumar
- Department of Special Education and Communication Disorders, Barkley Memorial Center, University of Nebraska Lincoln, Lincoln, Nebraska, USA
| | - Timothy A Jones
- Department of Special Education and Communication Disorders, Barkley Memorial Center, University of Nebraska Lincoln, Lincoln, Nebraska, USA
| | - Sherri M Jones
- Department of Special Education and Communication Disorders, Barkley Memorial Center, University of Nebraska Lincoln, Lincoln, Nebraska, USA
| | - Sonja J Pyott
- Department of Otorhinolaryngology/Head and Neck Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Graduate School of Medical Sciences Research School of Behavioural and Cognitive Neurosciences, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
2
|
Fisch KM, Rosenthal SB, Mark A, Sasik R, Nasamran CA, Clifford R, Derebery MJ, Boussaty E, Jepsen K, Harris J, Friedman RA. The genomic landscape of Ménière's disease: a path to endolymphatic hydrops. BMC Genomics 2024; 25:646. [PMID: 38943082 PMCID: PMC11212243 DOI: 10.1186/s12864-024-10552-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 06/21/2024] [Indexed: 07/01/2024] Open
Abstract
BACKGROUND Ménière's disease (MD) is a disorder of the inner ear that causes episodic bouts of severe dizziness, roaring tinnitus, and fluctuating hearing loss. To date, no targeted therapy exists. As such, we have undertaken a large whole genome sequencing study on carefully phenotyped unilateral MD patients with the goal of gene/pathway discovery and a move towards targeted intervention. This study was a retrospective review of patients with a history of Ménière's disease. Genomic DNA, acquired from saliva samples, was purified and subjected to whole genome sequencing. RESULTS Stringent variant calling, performed on 511 samples passing quality checks, followed by gene-based filtering by recurrence and proximity in molecular interaction networks, led to 481 high priority MD genes. These high priority genes, including MPHOSPH8, MYO18A, TRIOBP, OTOGL, TNC, and MYO6, were previously implicated in hearing loss, balance, and cochlear function, and were significantly enriched in common variant studies of hearing loss. Validation in an independent MD cohort confirmed 82 recurrent genes. Pathway analysis pointed to cell-cell adhesion, extracellular matrix, and cellular energy maintenance as key mediators of MD. Furthermore, the MD-prioritized genes were highly expressed in human inner ear hair cells and dark/vestibular cells, and were differentially expressed in a mouse model of hearing loss. CONCLUSION By enabling the development of model systems that may lead to targeted therapies and MD screening panels, the genes and variants identified in this study will inform diagnosis and treatment of MD.
Collapse
Affiliation(s)
- Kathleen M Fisch
- Center for Computational Biology & Bioinformatics, University of California, San Diego, La Jolla, CA, USA.
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Diego, La Jolla, CA, USA.
| | - Sara Brin Rosenthal
- Center for Computational Biology & Bioinformatics, University of California, San Diego, La Jolla, CA, USA
| | - Adam Mark
- Center for Computational Biology & Bioinformatics, University of California, San Diego, La Jolla, CA, USA
| | - Roman Sasik
- Center for Computational Biology & Bioinformatics, University of California, San Diego, La Jolla, CA, USA
| | - Chanond A Nasamran
- Center for Computational Biology & Bioinformatics, University of California, San Diego, La Jolla, CA, USA
| | - Royce Clifford
- Department of Otolaryngology, Head & Neck Surgery, University of California, San Diego, La Jolla, CA, USA
- Research Department, VA Hospitals, San Diego, CA, USA
| | | | - Ely Boussaty
- Department of Otolaryngology, Head & Neck Surgery, University of California, San Diego, La Jolla, CA, USA
| | - Kristen Jepsen
- Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jeffrey Harris
- Department of Otolaryngology, Head & Neck Surgery, University of California, San Diego, La Jolla, CA, USA
| | - Rick A Friedman
- Department of Otolaryngology, Head & Neck Surgery, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
3
|
Ghiselli S, Parmeggiani G, Zambonini G, Cuda D. Hearing Loss in Baraitser-Winter Syndrome: Case Reports and Review of the Literature. J Clin Med 2024; 13:1500. [PMID: 38592426 PMCID: PMC10935159 DOI: 10.3390/jcm13051500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/01/2024] [Accepted: 03/02/2024] [Indexed: 04/10/2024] Open
Abstract
Background: Baraitser-Winter Syndrome (BRWS) is a rare autosomal dominant condition associated with hearing loss (HL). In the literature, two types of this condition are reported, Baraitser-Winter type 1 (BRWS1) and type 2 (BRWS2) produced by specific pathogenetic variants of two different genes, ACTB for BRWS1 and ACTG1 for BRWS2. In addition to syndromic BRWS2, some pathogenic variants in ACTG1 are associated also to another pathologic entity, the "Autosomal dominant non-syndromic hearing loss 20/26". In these syndromes, typical craniofacial features, sensory impairment (vision and hearing) and intellectual disabilities are frequently present. Heart anomalies, renal and gastrointestinal involvement and seizure are also common. Wide inter- and intra-familial variety in the phenotypic spectrum is reported. Some phenotypic aspects of these syndromes are not yet fully described, such as the degree and progression of HL, and better knowledge of them could be useful for correct follow-up and treatment. Methods and Results: In this study, we report two cases of children with HL and diagnosis of BRWS and a review of the current literature on HL in these syndromes.
Collapse
Affiliation(s)
- Sara Ghiselli
- Department of Otorhinolaryngology, AUSL Piacenza, 29121 Piacenza, Italy; (G.Z.); (D.C.)
| | | | - Giulia Zambonini
- Department of Otorhinolaryngology, AUSL Piacenza, 29121 Piacenza, Italy; (G.Z.); (D.C.)
| | - Domenico Cuda
- Department of Otorhinolaryngology, AUSL Piacenza, 29121 Piacenza, Italy; (G.Z.); (D.C.)
- Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy
| |
Collapse
|
4
|
Boussaty EC, Ninoyu Y, Andrade LR, Li Q, Takeya R, Sumimoto H, Ohyama T, Wahlin KJ, Manor U, Friedman RA. Altered Fhod3 expression involved in progressive high-frequency hearing loss via dysregulation of actin polymerization stoichiometry in the cuticular plate. PLoS Genet 2024; 20:e1011211. [PMID: 38498576 PMCID: PMC10977885 DOI: 10.1371/journal.pgen.1011211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 03/28/2024] [Accepted: 03/05/2024] [Indexed: 03/20/2024] Open
Abstract
Age-related hearing loss (ARHL) is a common sensory impairment with complex underlying mechanisms. In our previous study, we performed a meta-analysis of genome-wide association studies (GWAS) in mice and identified a novel locus on chromosome 18 associated with ARHL specifically linked to a 32 kHz tone burst stimulus. Consequently, we investigated the role of Formin Homology 2 Domain Containing 3 (Fhod3), a newly discovered candidate gene for ARHL based on the GWAS results. We observed Fhod3 expression in auditory hair cells (HCs) primarily localized at the cuticular plate (CP). To understand the functional implications of Fhod3 in the cochlea, we generated Fhod3 overexpression mice (Pax2-Cre+/-; Fhod3Tg/+) (TG) and HC-specific conditional knockout mice (Atoh1-Cre+/-; Fhod3fl/fl) (KO). Audiological assessments in TG mice demonstrated progressive high-frequency hearing loss, characterized by predominant loss of outer hair cells, and a decreased phalloidin intensities of CP. Ultrastructural analysis revealed loss of the shortest row of stereocilia in the basal turn of the cochlea, and alterations in the cuticular plate surrounding stereocilia rootlets. Importantly, the hearing and HC phenotype in TG mice phenocopied that of the KO mice. These findings suggest that balanced expression of Fhod3 is critical for proper CP and stereocilia structure and function. Further investigation of Fhod3 related hearing impairment mechanisms may lend new insight towards the myriad mechanisms underlying ARHL, which in turn could facilitate the development of therapeutic strategies for ARHL.
Collapse
Affiliation(s)
- Ely Cheikh Boussaty
- Department of Otolaryngology–Head and Neck Surgery, University of California, San Diego, La Jolla, California, United States of America
| | - Yuzuru Ninoyu
- Department of Otolaryngology–Head and Neck Surgery, University of California, San Diego, La Jolla, California, United States of America
| | - Leonardo R. Andrade
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Qingzhong Li
- USC-Tina and Rick Caruso Department of Otolaryngology-Head & Neck Surgery, Zilkha Neurogenetic Institute, USC Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Ryu Takeya
- Department of Pharmacology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Hideki Sumimoto
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Takahiro Ohyama
- USC-Tina and Rick Caruso Department of Otolaryngology-Head & Neck Surgery, Zilkha Neurogenetic Institute, USC Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Karl J. Wahlin
- Shiley Eye Institute, University of California, San Diego, San Diego, California, United States of America
| | - Uri Manor
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, California, United States of America
- Department of Cell & Developmental Biology, School of Biological Sciences, University of California, San Diego, United States of America
| | - Rick A. Friedman
- Department of Otolaryngology–Head and Neck Surgery, University of California, San Diego, La Jolla, California, United States of America
| |
Collapse
|
5
|
王 子, 曹 麒, 胡 少, 范 新, 吕 俊, 王 会, 王 武, 李 华, 舒 易. [Study on gene therapy for DPOAE and ABR threshold changes in adult Otof-/- mice]. LIN CHUANG ER BI YAN HOU TOU JING WAI KE ZA ZHI = JOURNAL OF CLINICAL OTORHINOLARYNGOLOGY, HEAD, AND NECK SURGERY 2024; 38:49-56. [PMID: 38297849 PMCID: PMC11116155 DOI: 10.13201/j.issn.2096-7993.2024.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Indexed: 02/02/2024]
Abstract
Objective:This study aims to analyze the threshold changes in distortion product otoacoustic emissions(DPOAE) and auditory brainstem response(ABR) in adult Otof-/- mice before and after gene therapy, evaluating its effectiveness and exploring methods for assessing hearing recovery post-treatment. Methods:At the age of 4 weeks, adult Otof-/- mice received an inner ear injection of a therapeutic agent containing intein-mediated recombination of the OTOF gene, delivered via dual AAV vectors through the round window membrane(RWM). Immunofluorescence staining assessed the proportion of inner ear hair cells with restored otoferlin expression and the number of synapses.Statistical analysis was performed to compare the DPOAE and ABR thresholds before and after the treatment. Results:AAV-PHP. eB demonstrates high transduction efficiency in inner ear hair cells. The therapeutic regimen corrected hearing loss in adult Otof-/- mice without impacting auditory function in wild-type mice. The changes in DPOAE and ABR thresholds after gene therapy are significantly correlated at 16 kHz. Post-treatment,a slight increase in DPOAE was observeds,followed by a recovery trend at 2 months post-treatment. Conclusion:Gene therapy significantly restored hearing in adult Otof-/- mice, though the surgical delivery may cause transient hearing damage. Precise and gentle surgical techniques are essential to maximize gene therapy's efficacy.
Collapse
Affiliation(s)
- 子菁 王
- 南华大学附属第二医院耳鼻喉科(湖南衡阳,421001)Department of Otolaryngology, Second Affiliated Hospital of South China University Hengyang, 421001, China
- 复旦大学附属眼耳鼻喉科医院耳鼻喉科Otolaryngology Department of Fudan University Affiliated Eye, Ear, Nose and Throat Hospital
| | - 麒 曹
- 南华大学附属第二医院耳鼻喉科(湖南衡阳,421001)Department of Otolaryngology, Second Affiliated Hospital of South China University Hengyang, 421001, China
- 复旦大学附属眼耳鼻喉科医院耳鼻喉科Otolaryngology Department of Fudan University Affiliated Eye, Ear, Nose and Throat Hospital
| | - 少伟 胡
- 复旦大学附属眼耳鼻喉科医院耳鼻喉科Otolaryngology Department of Fudan University Affiliated Eye, Ear, Nose and Throat Hospital
| | - 新泰 范
- 复旦大学附属眼耳鼻喉科医院耳鼻喉科Otolaryngology Department of Fudan University Affiliated Eye, Ear, Nose and Throat Hospital
| | - 俊 吕
- 复旦大学附属眼耳鼻喉科医院耳鼻喉科Otolaryngology Department of Fudan University Affiliated Eye, Ear, Nose and Throat Hospital
| | - 会 王
- 复旦大学附属眼耳鼻喉科医院耳鼻喉科Otolaryngology Department of Fudan University Affiliated Eye, Ear, Nose and Throat Hospital
| | - 武庆 王
- 复旦大学附属眼耳鼻喉科医院耳鼻喉科Otolaryngology Department of Fudan University Affiliated Eye, Ear, Nose and Throat Hospital
| | - 华伟 李
- 复旦大学附属眼耳鼻喉科医院耳鼻喉科Otolaryngology Department of Fudan University Affiliated Eye, Ear, Nose and Throat Hospital
| | - 易来 舒
- 南华大学附属第二医院耳鼻喉科(湖南衡阳,421001)Department of Otolaryngology, Second Affiliated Hospital of South China University Hengyang, 421001, China
- 复旦大学附属眼耳鼻喉科医院耳鼻喉科Otolaryngology Department of Fudan University Affiliated Eye, Ear, Nose and Throat Hospital
| |
Collapse
|
6
|
Zhou LY, Jin CX, Wang WX, Song L, Shin JB, Du TT, Wu H. Differential regulation of hair cell actin cytoskeleton mediated by SRF and MRTFB. eLife 2023; 12:e90155. [PMID: 37982489 PMCID: PMC10703445 DOI: 10.7554/elife.90155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 11/17/2023] [Indexed: 11/21/2023] Open
Abstract
The MRTF-SRF pathway has been extensively studied for its crucial role in driving the expression of a large number of genes involved in actin cytoskeleton of various cell types. However, the specific contribution of MRTF-SRF in hair cells remains unknown. In this study, we showed that hair cell-specific deletion of Srf or Mrtfb, but not Mrtfa, leads to similar defects in the development of stereocilia dimensions and the maintenance of cuticular plate integrity. We used fluorescence-activated cell sorting-based hair cell RNA-Seq analysis to investigate the mechanistic underpinnings of the changes observed in Srf and Mrtfb mutants, respectively. Interestingly, the transcriptome analysis revealed distinct profiles of genes regulated by Srf and Mrtfb, suggesting different transcriptional regulation mechanisms of actin cytoskeleton activities mediated by Srf and Mrtfb. Exogenous delivery of calponin 2 using Adeno-associated virus transduction in Srf mutants partially rescued the impairments of stereocilia dimensions and the F-actin intensity of cuticular plate, suggesting the involvement of Cnn2, as an Srf downstream target, in regulating the hair bundle morphology and cuticular plate actin cytoskeleton organization. Our study uncovers, for the first time, the unexpected differential transcriptional regulation of actin cytoskeleton mediated by Srf and Mrtfb in hair cells, and also demonstrates the critical role of SRF-CNN2 in modulating actin dynamics of the stereocilia and cuticular plate, providing new insights into the molecular mechanism underlying hair cell development and maintenance.
Collapse
Affiliation(s)
- Ling-Yun Zhou
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Ear Institute, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose DiseasesShanghaiChina
| | - Chen-Xi Jin
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Ear Institute, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose DiseasesShanghaiChina
| | - Wen-Xiao Wang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Ear Institute, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose DiseasesShanghaiChina
| | - Lei Song
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Ear Institute, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose DiseasesShanghaiChina
| | - Jung-Bum Shin
- Department of Neuroscience, University of VirginiaCharlottesvilleUnited States
| | - Ting-Ting Du
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Ear Institute, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose DiseasesShanghaiChina
| | - Hao Wu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Ear Institute, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose DiseasesShanghaiChina
| |
Collapse
|
7
|
Zhu G, Huang Y, Zhang L, Yan K, Qiu C, He Y, Liu Q, Zhu C, Morín M, Moreno‐Pelayo MÁ, Zhu M, Cao X, Zhou H, Qian X, Xu Z, Chen J, Gao X, Wan G. Cingulin regulates hair cell cuticular plate morphology and is required for hearing in human and mouse. EMBO Mol Med 2023; 15:e17611. [PMID: 37691516 PMCID: PMC10630877 DOI: 10.15252/emmm.202317611] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 08/17/2023] [Accepted: 08/22/2023] [Indexed: 09/12/2023] Open
Abstract
Cingulin (CGN) is a cytoskeleton-associated protein localized at the apical junctions of epithelial cells. CGN interacts with major cytoskeletal filaments and regulates RhoA activity. However, physiological roles of CGN in development and human diseases are currently unknown. Here, we report a multi-generation family presenting with autosomal dominant non-syndromic hearing loss (ADNSHL) that co-segregates with a CGN heterozygous truncating variant, c.3330delG (p.Leu1110Leufs*17). CGN is normally expressed at the apical cell junctions of the organ of Corti, with enriched localization at hair cell cuticular plates and circumferential belts. In mice, the putative disease-causing mutation results in reduced expression and abnormal subcellular localization of the CGN protein, abolishes its actin polymerization activity, and impairs the normal morphology of hair cell cuticular plates and hair bundles. Hair cell-specific Cgn knockout leads to high-frequency hearing loss. Importantly, Cgn mutation knockin mice display noise-sensitive, progressive hearing loss and outer hair cell degeneration. In summary, we identify CGN c.3330delG as a pathogenic variant for ADNSHL and reveal essential roles of CGN in the maintenance of cochlear hair cell structures and auditory function.
Collapse
Affiliation(s)
- Guang‐Jie Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- MOE Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Yuhang Huang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- MOE Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
| | - Linqing Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- MOE Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
| | - Keji Yan
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life SciencesShandong UniversityQingdaoChina
| | - Cui Qiu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- MOE Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
| | - Yihan He
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- MOE Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
| | - Qing Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- MOE Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Chengwen Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- MOE Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Matías Morín
- Servicio de GenéticaHospital Universitario Ramón y Cajal, IRYCISMadridSpain
- Centro de Investigación Biomédica en Red de Enfermedades RarasInstituto de Salud Carlos III (CB06/07/0048; CIBERER‐ISCIII)MadridSpain
| | - Miguel Ángel Moreno‐Pelayo
- Servicio de GenéticaHospital Universitario Ramón y Cajal, IRYCISMadridSpain
- Centro de Investigación Biomédica en Red de Enfermedades RarasInstituto de Salud Carlos III (CB06/07/0048; CIBERER‐ISCIII)MadridSpain
| | - Min‐Sheng Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- MOE Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Xin Cao
- Department of Medical Genetics, School of Basic Medical ScienceNanjing Medical UniversityNanjingChina
| | - Han Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Xiaoyun Qian
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Zhigang Xu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life SciencesShandong UniversityQingdaoChina
| | - Jie Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Xia Gao
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Guoqiang Wan
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School, Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- MOE Key Laboratory of Model Animal for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| |
Collapse
|
8
|
Baxi AB, Nemes P, Moody SA. Time-resolved quantitative proteomic analysis of the developing Xenopus otic vesicle reveals putative congenital hearing loss candidates. iScience 2023; 26:107665. [PMID: 37670778 PMCID: PMC10475516 DOI: 10.1016/j.isci.2023.107665] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/16/2023] [Accepted: 08/14/2023] [Indexed: 09/07/2023] Open
Abstract
Over 200 genes are known to underlie human congenital hearing loss (CHL). Although transcriptomic approaches have identified candidate regulators of otic development, little is known about the abundance of their protein products. We used a multiplexed quantitative mass spectrometry-based proteomic approach to determine protein abundances over key stages of Xenopus otic morphogenesis to reveal a dynamic expression of cytoskeletal, integrin signaling, and extracellular matrix proteins. We correlated these dynamically expressed proteins to previously published lists of putative downstream targets of human syndromic hearing loss genes: SIX1 (BOR syndrome), CHD7 (CHARGE syndrome), and SOX10 (Waardenburg syndrome). We identified transforming growth factor beta-induced (Tgfbi), an extracellular integrin-interacting protein, as a putative target of Six1 that is required for normal otic vesicle formation. Our findings demonstrate the application of this Xenopus dataset to understanding the dynamic regulation of proteins during otic development and to discovery of additional candidates for human CHL.
Collapse
Affiliation(s)
- Aparna B. Baxi
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Peter Nemes
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA
| | - Sally A. Moody
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| |
Collapse
|
9
|
Boussaty EC, Ninoyu Y, Andrade L, Li Q, Takeya R, Sumimoto H, Ohyama T, Wahlin KJ, Manor U, Friedman RA. Altered Fhod3 Expression Involved in Progressive High-Frequency Hearing Loss via Dysregulation of Actin Polymerization Stoichiometry in The Cuticular Plate. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.20.549974. [PMID: 37546952 PMCID: PMC10401921 DOI: 10.1101/2023.07.20.549974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Age-related hearing loss (ARHL) is a common sensory impairment with comlex underlying mechanisms. In our previous study, we performed a meta-analysis of genome-wide association studies (GWAS) in mice and identified a novel locus on chromosome 18 associated with ARHL specifically linked to a 32 kHz tone burst stimulus. Consequently, we investigated the role of Formin Homology 2 Domain Containing 3 (Fhod3), a newly discovered candidate gene for ARHL based on the GWAS results. We observed Fhod3 expression in auditory hair cells (HCs) and primarily localized at the cuticular plate (CP). To understand the functional implications of Fhod3 in the cochlea, we generated Fhod3 overexpression mice (Pax2-Cre+/-; Fhod3Tg/+) (TG) and HC-specific conditional knockout mice (Atoh1-Cre+/-; Fhod3fl/fl) (KO). Audiological assessments in TG mice demonstrated progressive high-frequency hearing loss, characterized by predominant loss of outer HCs and decrease phalloidin intensities of CP. Ultrastructural analysis revealed shortened stereocilia in the basal turn cochlea. Importantly, the hearing and HC phenotype in TG mice were replicated in KO mice. These findings indicate that Fhod3 plays a critical role in regulating actin dynamics in CP and stereocilia. Further investigation of Fhod3-related hearing impairment mechanisms may facilitate the development of therapeutic strategies for ARHL in humans.
Collapse
|
10
|
Zhao Y, Zhang L, Wang D, Chen B, Shu Y. Approaches and Vectors for Efficient Cochlear Gene Transfer in Adult Mouse Models. Biomolecules 2022; 13:biom13010038. [PMID: 36671423 PMCID: PMC9855574 DOI: 10.3390/biom13010038] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/18/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Inner ear gene therapy using adeno-associated viral vectors (AAVs) in neonatal mice can alleviate hearing loss in mouse models of deafness. However, efficient and safe transgene delivery to the adult mouse cochlea is critical for the effectiveness of AAV-mediated therapy. Here, we examined three gene delivery approaches including posterior semicircular canal (PSCC) canalostomy, round window membrane (RWM) injection, and tubing-RWM+PSCC (t-RP) in adult mice. Transduction rates and survival rates of cochlear hair cells were analyzed, hearing function was recorded, AAV distribution in the sagittal brain sections was evaluated, and cochlear histopathologic images were appraised. We found that an injection volume of 1 μL AAV through the PSCC is safe and highly efficient and does not impair hearing function in adult mice, but local injection allows AAV vectors to spread slightly into the brain. We then tested five AAV serotypes (PHP.eB, IE, Anc80L65, AAV2, and PHP.s) in parallel and observed the most robust eGFP expression in inner hair cells, outer hair cells, and spiral ganglion neurons throughout the cochlea after AAV-Anc80L65 injection. Thus, PSCC-injected Anc80L65 provides a foundation for gene therapy in the adult cochlea and will facilitate the development of inner ear gene therapy.
Collapse
Affiliation(s)
- Yu Zhao
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Longlong Zhang
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Daqi Wang
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Bing Chen
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
- Correspondence: (B.C.); (Y.S.)
| | - Yilai Shu
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
- Correspondence: (B.C.); (Y.S.)
| |
Collapse
|
11
|
Gong R, Jiang F, Moreland ZG, Reynolds MJ, de los Reyes SE, Gurel P, Shams A, Heidings JB, Bowl MR, Bird JE, Alushin GM. Structural basis for tunable control of actin dynamics by myosin-15 in mechanosensory stereocilia. SCIENCE ADVANCES 2022; 8:eabl4733. [PMID: 35857845 PMCID: PMC9299544 DOI: 10.1126/sciadv.abl4733] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 06/03/2022] [Indexed: 05/12/2023]
Abstract
The motor protein myosin-15 is necessary for the development and maintenance of mechanosensory stereocilia, and mutations in myosin-15 cause hereditary deafness. In addition to transporting actin regulatory machinery to stereocilia tips, myosin-15 directly nucleates actin filament ("F-actin") assembly, which is disrupted by a progressive hearing loss mutation (p.D1647G, "jordan"). Here, we present cryo-electron microscopy structures of myosin-15 bound to F-actin, providing a framework for interpreting the impacts of deafness mutations on motor activity and actin nucleation. Rigor myosin-15 evokes conformational changes in F-actin yet maintains flexibility in actin's D-loop, which mediates inter-subunit contacts, while the jordan mutant locks the D-loop in a single conformation. Adenosine diphosphate-bound myosin-15 also locks the D-loop, which correspondingly blunts actin-polymerization stimulation. We propose myosin-15 enhances polymerization by bridging actin protomers, regulating nucleation efficiency by modulating actin's structural plasticity in a myosin nucleotide state-dependent manner. This tunable regulation of actin polymerization could be harnessed to precisely control stereocilium height.
Collapse
Affiliation(s)
- Rui Gong
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Fangfang Jiang
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Zane G. Moreland
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Matthew J. Reynolds
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | | | - Pinar Gurel
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Arik Shams
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - James B. Heidings
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Michael R. Bowl
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Campus, Oxfordshire, UK
- UCL Ear Institute, University College London, London, UK
| | - Jonathan E. Bird
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Gregory M. Alushin
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| |
Collapse
|
12
|
Trpchevska N, Freidin MB, Broer L, Oosterloo BC, Yao S, Zhou Y, Vona B, Bishop C, Bizaki-Vallaskangas A, Canlon B, Castellana F, Chasman DI, Cherny S, Christensen K, Concas MP, Correa A, Elkon R, Mengel-From J, Gao Y, Giersch ABS, Girotto G, Gudjonsson A, Gudnason V, Heard-Costa NL, Hertzano R, Hjelmborg JVB, Hjerling-Leffler J, Hoffman HJ, Kaprio J, Kettunen J, Krebs K, Kähler AK, Lallemend F, Launer LJ, Lee IM, Leonard H, Li CM, Lowenheim H, Magnusson PKE, van Meurs J, Milani L, Morton CC, Mäkitie A, Nalls MA, Nardone GG, Nygaard M, Palviainen T, Pratt S, Quaranta N, Rämö J, Saarentaus E, Sardone R, Satizabal CL, Schweinfurth JM, Seshadri S, Shiroma E, Shulman E, Simonsick E, Spankovich C, Tropitzsch A, Lauschke VM, Sullivan PF, Goedegebure A, Cederroth CR, Williams FMK, Nagtegaal AP. Genome-wide association meta-analysis identifies 48 risk variants and highlights the role of the stria vascularis in hearing loss. Am J Hum Genet 2022; 109:1077-1091. [PMID: 35580588 PMCID: PMC9247887 DOI: 10.1016/j.ajhg.2022.04.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 04/15/2022] [Indexed: 02/08/2023] Open
Abstract
Hearing loss is one of the top contributors to years lived with disability and is a risk factor for dementia. Molecular evidence on the cellular origins of hearing loss in humans is growing. Here, we performed a genome-wide association meta-analysis of clinically diagnosed and self-reported hearing impairment on 723,266 individuals and identified 48 significant loci, 10 of which are novel. A large proportion of associations comprised missense variants, half of which lie within known familial hearing loss loci. We used single-cell RNA-sequencing data from mouse cochlea and brain and mapped common-variant genomic results to spindle, root, and basal cells from the stria vascularis, a structure in the cochlea necessary for normal hearing. Our findings indicate the importance of the stria vascularis in the mechanism of hearing impairment, providing future paths for developing targets for therapeutic intervention in hearing loss.
Collapse
Affiliation(s)
- Natalia Trpchevska
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Maxim B Freidin
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Linda Broer
- Department of Internal Medicine, Erasmus Medical Center, 3015 CE Rotterdam, the Netherlands
| | - Berthe C Oosterloo
- Department of Otorhinolaryngology, Erasmus Medical Center, 3015 CE Rotterdam, the Netherlands
| | - Shuyang Yao
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Barbara Vona
- Institute of Human Genetics, University Medical Center Göttingen, 37073 Göttingen, Germany; Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany; Department of Otolaryngology-Head & Neck Surgery, University of Tübingen Medical Center, 72076 Tübingen, Germany
| | - Charles Bishop
- Department of Otolaryngology and Communicative Sciences, The University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Argyro Bizaki-Vallaskangas
- Department of Otolaryngology, University of Tampere, 33100 Tampere, Finland; Pirkanmaan Sairaanhoitopiiri, 33520 Tampere, Finland
| | - Barbara Canlon
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Fabio Castellana
- Unit of Data Sciences and Technology Innovation for Population Health, National Institute of Gastroenterology "Saverio de Bellis", Research Hospital, Castellana Grotte, 70124 Bari, Italy
| | - Daniel I Chasman
- Division of Preventative Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Stacey Cherny
- Department of Anatomy and Anthropology and Department of Epidemiology and Preventive Medicine, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Kaare Christensen
- The Danish Twin Registry, Department of Public Health, University of Southern Denmark, 5000 Odense C, Denmark; Department of Clinical Genetics, Odense University Hospital, 5000 Odense C, Denmark; Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, 5000 Odense C, Denmark
| | - Maria Pina Concas
- Institute for Maternal and Child Health - IRCCS, Burlo Garofolo, 34127 Trieste, Italy
| | - Adolfo Correa
- Jackson Heart Study, The University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Ran Elkon
- Department of Human Molecular Genetics & Biochemistry, Sackler School of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Jonas Mengel-From
- The Danish Twin Registry, Department of Public Health, University of Southern Denmark, 5000 Odense C, Denmark; Department of Clinical Genetics, Odense University Hospital, 5000 Odense C, Denmark
| | - Yan Gao
- Jackson Heart Study, The University of Mississippi Medical Center, Jackson, MS 39216, USA; Department of Population Health Science, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Anne B S Giersch
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Giorgia Girotto
- Institute for Maternal and Child Health - IRCCS, Burlo Garofolo, 34127 Trieste, Italy; Department of Medicine, Surgery and Health Sciences, University of Trieste, 34139 Trieste, Italy
| | | | - Vilmundur Gudnason
- Icelandic Heart Association, 201 Kopavogur, Iceland; Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Nancy L Heard-Costa
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA; Framingham Heart Study, Framingham, MA 01702, USA
| | - Ronna Hertzano
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland Baltimore, Baltimore, MD 21201, USA; Department of Anatomy and Neurobiology, University of Maryland Baltimore, Baltimore, MD 21201, USA; Institute for Genome Sciences, University of Maryland Baltimore, Baltimore, MD 21201, USA
| | - Jacob V B Hjelmborg
- The Danish Twin Registry, Department of Public Health, University of Southern Denmark, 5000 Odense C, Denmark
| | - Jens Hjerling-Leffler
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Howard J Hoffman
- Division of Scientific Programs, Epidemiology and Statistics Program, National Institute on Deafness and Other Communications Disorders (NIDCD), NIH, Bethesda, MD 20892, USA
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland
| | - Johannes Kettunen
- Computational Medicine, Center for Life Course Health Research, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland; Biocenter Oulu, University of Oulu, 90220 Oulu, Finland; Finnish Institute for Health and Welfare, 00271 Helsinki, Finland
| | - Kristi Krebs
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Anna K Kähler
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Francois Lallemend
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program National Institute on Aging, Bethesda, MD 20892, USA
| | - I-Min Lee
- Division of Preventative Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hampton Leonard
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA; Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA; Data Tecnica International, Glen Echo, MD 20812, USA
| | - Chuan-Ming Li
- Division of Scientific Programs, Epidemiology and Statistics Program, National Institute on Deafness and Other Communications Disorders (NIDCD), NIH, Bethesda, MD 20892, USA
| | - Hubert Lowenheim
- Department of Otolaryngology-Head & Neck Surgery, University of Tübingen Medical Center, 72076 Tübingen, Germany
| | - Patrik K E Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Joyce van Meurs
- Department of Internal Medicine, Erasmus Medical Center, 3015 CE Rotterdam, the Netherlands
| | - Lili Milani
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Cynthia C Morton
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Obstetrics and Gynecology and of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Manchester Centre for Audiology and Deafness, University of Manchester, Manchester M13 9PL, UK
| | - Antti Mäkitie
- Department of Otorhinolaryngology - Head and Neck Surgery, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland
| | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA; Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA; Data Tecnica International, Glen Echo, MD 20812, USA
| | | | - Marianne Nygaard
- The Danish Twin Registry, Department of Public Health, University of Southern Denmark, 5000 Odense C, Denmark; Department of Clinical Genetics, Odense University Hospital, 5000 Odense C, Denmark
| | - Teemu Palviainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland
| | - Sheila Pratt
- Department of Communication Science & Disorders, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Nicola Quaranta
- Otolaryngology Unit, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari Aldo Moro, 70121 Bari, Italy
| | - Joel Rämö
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland
| | - Elmo Saarentaus
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland
| | - Rodolfo Sardone
- Unit of Data Sciences and Technology Innovation for Population Health, National Institute of Gastroenterology "Saverio de Bellis", Research Hospital, Castellana Grotte, 70124 Bari, Italy
| | - Claudia L Satizabal
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA; Framingham Heart Study, Framingham, MA 01702, USA; Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases and Department of Population Health Sciences, University of Texas Health Sciences Center, San Antonio, TX 78229, USA
| | - John M Schweinfurth
- Department of Otolaryngology and Communicative Sciences, The University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA; Framingham Heart Study, Framingham, MA 01702, USA; Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases and Department of Population Health Sciences, University of Texas Health Sciences Center, San Antonio, TX 78229, USA
| | - Eric Shiroma
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Baltimore, MD 21224, USA
| | - Eldad Shulman
- Department of Human Molecular Genetics & Biochemistry, Sackler School of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Eleanor Simonsick
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD 21224, USA
| | - Christopher Spankovich
- Department of Otolaryngology and Communicative Sciences, The University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Anke Tropitzsch
- Department of Otolaryngology-Head & Neck Surgery, University of Tübingen Medical Center, 72076 Tübingen, Germany
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Patrick F Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 17177 Stockholm, Sweden; Department of Genetics, University of North Carolina, Chapel Hill, NC 27516, USA
| | - Andre Goedegebure
- Department of Otorhinolaryngology, Erasmus Medical Center, 3015 CE Rotterdam, the Netherlands
| | - Christopher R Cederroth
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden; National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust, Ropewalk House, NG1 5DU Nottingham, UK; Hearing Sciences, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, NG7 2UH Nottingham, UK.
| | - Frances M K Williams
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Andries Paul Nagtegaal
- Department of Otorhinolaryngology, Erasmus Medical Center, 3015 CE Rotterdam, the Netherlands
| |
Collapse
|
13
|
Chiereghin C, Robusto M, Massa V, Castorina P, Ambrosetti U, Asselta R, Soldà G. Role of Cytoskeletal Diaphanous-Related Formins in Hearing Loss. Cells 2022; 11:cells11111726. [PMID: 35681420 PMCID: PMC9179844 DOI: 10.3390/cells11111726] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 02/04/2023] Open
Abstract
Hearing relies on the proper functioning of auditory hair cells and on actin-based cytoskeletal structures. Diaphanous-related formins (DRFs) are evolutionarily conserved cytoskeletal proteins that regulate the nucleation of linear unbranched actin filaments. They play key roles during metazoan development, and they seem particularly pivotal for the correct physiology of the reproductive and auditory systems. Indeed, in Drosophila melanogaster, a single diaphanous (dia) gene is present, and mutants show sterility and impaired response to sound. Vertebrates, instead, have three orthologs of the diaphanous gene: DIAPH1, DIAPH2, and DIAPH3. In humans, defects in DIAPH1 and DIAPH3 have been associated with different types of hearing loss. In particular, heterozygous mutations in DIAPH1 are responsible for autosomal dominant deafness with or without thrombocytopenia (DFNA1, MIM #124900), whereas regulatory mutations inducing the overexpression of DIAPH3 cause autosomal dominant auditory neuropathy 1 (AUNA1, MIM #609129). Here, we provide an overview of the expression and function of DRFs in normal hearing and deafness.
Collapse
Affiliation(s)
- Chiara Chiereghin
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Milan, Italy; (C.C.); (R.A.)
| | - Michela Robusto
- Experimental Therapeutics Program, IFOM ETS—The AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy;
| | - Valentina Massa
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Via Di Rudinì 8, 20146 Milan, Italy;
| | | | - Umberto Ambrosetti
- Dipartimento di Scienze Cliniche e di Comunità, Università degli Studi di Milano and Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, UO Audiologia, Via F. Sforza 35, 20122 Milan, Italy;
| | - Rosanna Asselta
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Milan, Italy; (C.C.); (R.A.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Milan, Italy
| | - Giulia Soldà
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Milan, Italy; (C.C.); (R.A.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Milan, Italy
- Correspondence:
| |
Collapse
|
14
|
Miyoshi T, Belyantseva IA, Kitajiri SI, Miyajima H, Nishio SY, Usami SI, Kim BJ, Choi BY, Omori K, Shroff H, Friedman TB. Human deafness-associated variants alter the dynamics of key molecules in hair cell stereocilia F-actin cores. Hum Genet 2022; 141:363-382. [PMID: 34232383 PMCID: PMC11351816 DOI: 10.1007/s00439-021-02304-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/15/2021] [Indexed: 12/16/2022]
Abstract
Stereocilia protrude up to 100 µm from the apical surface of vertebrate inner ear hair cells and are packed with cross-linked filamentous actin (F-actin). They function as mechanical switches to convert sound vibration into electrochemical neuronal signals transmitted to the brain. Several genes encode molecular components of stereocilia including actin monomers, actin regulatory and bundling proteins, motor proteins and the proteins of the mechanotransduction complex. A stereocilium F-actin core is a dynamic system, which is continuously being remodeled while maintaining an outwardly stable architecture under the regulation of F-actin barbed-end cappers, severing proteins and crosslinkers. The F-actin cores of stereocilia also provide a pathway for motor proteins to transport cargos including components of tip-link densities, scaffolding proteins and actin regulatory proteins. Deficiencies and mutations of stereocilia components that disturb this "dynamic equilibrium" in stereocilia can induce morphological changes and disrupt mechanotransduction causing sensorineural hearing loss, best studied in mouse and zebrafish models. Currently, at least 23 genes, associated with human syndromic and nonsyndromic hearing loss, encode proteins involved in the development and maintenance of stereocilia F-actin cores. However, it is challenging to predict how variants associated with sensorineural hearing loss segregating in families affect protein function. Here, we review the functions of several molecular components of stereocilia F-actin cores and provide new data from our experimental approach to directly evaluate the pathogenicity and functional impact of reported and novel variants of DIAPH1 in autosomal-dominant DFNA1 hearing loss using single-molecule fluorescence microscopy.
Collapse
Affiliation(s)
- Takushi Miyoshi
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Porter Neuroscience Research Center, Room 1F-143A, Bethesda, MD, 20892, USA.
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan.
| | - Inna A Belyantseva
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Porter Neuroscience Research Center, Room 1F-143A, Bethesda, MD, 20892, USA
| | - Shin-Ichiro Kitajiri
- Department of Hearing Implant Sciences, Shinshu University School of Medicine, 390-8621, Matsumoto, Japan
| | - Hiroki Miyajima
- Department of Otolaryngology, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
- Department of Otolaryngology, Aizawa Hospital, Matsumoto, 390-8510, Japan
| | - Shin-Ya Nishio
- Department of Hearing Implant Sciences, Shinshu University School of Medicine, 390-8621, Matsumoto, Japan
| | - Shin-Ichi Usami
- Department of Hearing Implant Sciences, Shinshu University School of Medicine, 390-8621, Matsumoto, Japan
| | - Bong Jik Kim
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Chungnam National University Sejong Hospital, Sejong, 30099, South Korea
- Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, South Korea
| | - Byung Yoon Choi
- Department of Otorhinolaryngology, Seoul National University Bundang Hospital, Seongnam, 13620, South Korea
| | - Koichi Omori
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Hari Shroff
- Laboratory of High Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Thomas B Friedman
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Porter Neuroscience Research Center, Room 1F-143A, Bethesda, MD, 20892, USA
| |
Collapse
|
15
|
Lorente-Cánovas B, Eckrich S, Lewis MA, Johnson SL, Marcotti W, Steel KP. Grxcr1 regulates hair bundle morphogenesis and is required for normal mechanoelectrical transduction in mouse cochlear hair cells. PLoS One 2022; 17:e0261530. [PMID: 35235570 PMCID: PMC8890737 DOI: 10.1371/journal.pone.0261530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 12/03/2021] [Indexed: 12/02/2022] Open
Abstract
Tasmanian devil (tde) mice are deaf and exhibit circling behaviour. Sensory hair cells of mutants show disorganised hair bundles with abnormally thin stereocilia. The origin of this mutation is the insertion of a transgene which disrupts expression of the Grxcr1 (glutaredoxin cysteine rich 1) gene. We report here that Grxcr1 exons and transcript sequences are not affected by the transgene insertion in tde homozygous (tde/tde) mice. Furthermore, 5'RACE PCR experiments showed the presence of two different transcripts of the Grxcr1 gene, expressed in both tde/tde and in wild-type controls. However, quantitative analysis of Grxcr1 transcripts revealed a significantly decreased mRNA level in tde/tde mice. The key stereociliary proteins ESPN, MYO7A, EPS8 and PTPRQ were distributed in hair bundles of homozygous tde mutants in a similar pattern compared with control mice. We found that the abnormal morphology of the stereociliary bundle was associated with a reduction in the size and Ca2+-sensitivity of the mechanoelectrical transducer (MET) current. We propose that GRXCR1 is key for the normal growth of the stereociliary bundle prior to the onset of hearing, and in its absence hair cells are unable to mature into fully functional sensory receptors.
Collapse
Affiliation(s)
- Beatriz Lorente-Cánovas
- Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom
- Wellcome Sanger Institute, Hinxton, United Kingdom
| | - Stephanie Eckrich
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
| | - Morag A. Lewis
- Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom
- Wellcome Sanger Institute, Hinxton, United Kingdom
| | - Stuart L. Johnson
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
- Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom
| | - Walter Marcotti
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
- Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom
| | - Karen P. Steel
- Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom
- Wellcome Sanger Institute, Hinxton, United Kingdom
| |
Collapse
|
16
|
Duong HTT, Suzuki H, Katagiri S, Shibata M, Arai M, Yura K. Computational study of the impact of nucleotide variations on highly conserved proteins: In the case of actin. Biophys Physicobiol 2022; 19:e190025. [PMID: 36160324 PMCID: PMC9465404 DOI: 10.2142/biophysico.bppb-v19.0025] [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] [Received: 06/13/2022] [Accepted: 07/27/2022] [Indexed: 12/01/2022] Open
Abstract
Sequencing of individual human genomes enables studying relationship among nucleotide variations, amino acid substitutions, effect on protein structures and diseases. Many studies have found general tendencies, for instance, that pathogenic variations tend to be found in the buried regions of the protein structures, that benign variations tend to be found on the surface of the proteins, and that variations on evolutionary conserved residues tend to be pathogenic. These tendencies were deduced from globular proteins with standard evolutionary changes in amino acid sequences. In this study, we investigated the variation distribution on actin, one of the highly conserved proteins. Many nucleotide variations and three-dimensional structures of actin have been registered in databases. By combining those data, we found that variations buried inside the protein were rather benign and variations on the surface of the protein were pathogenic. This idiosyncratic distribution of the variation impact is likely ascribed to the extensive use of the surface of the protein for protein-protein interactions in actin.
Collapse
Affiliation(s)
- Ha T. T. Duong
- Graduate School of Humanities and Sciences, Ochanomizu University
| | - Hirofumi Suzuki
- Graduate School of Advanced Science and Engineering, Waseda University
| | - Saki Katagiri
- Graduate School of Humanities and Sciences, Ochanomizu University
| | - Mayu Shibata
- Graduate School of Humanities and Sciences, Ochanomizu University
| | - Misae Arai
- Graduate School of Humanities and Sciences, Ochanomizu University
| | - Kei Yura
- Graduate School of Humanities and Sciences, Ochanomizu University
| |
Collapse
|
17
|
Trigila AP, Pisciottano F, Franchini LF. Hearing loss genes reveal patterns of adaptive evolution at the coding and non-coding levels in mammals. BMC Biol 2021; 19:244. [PMID: 34784928 PMCID: PMC8594068 DOI: 10.1186/s12915-021-01170-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 10/21/2021] [Indexed: 11/26/2022] Open
Abstract
Background Mammals possess unique hearing capacities that differ significantly from those of the rest of the amniotes. In order to gain insights into the evolution of the mammalian inner ear, we aim to identify the set of genetic changes and the evolutionary forces that underlie this process. We hypothesize that genes that impair hearing when mutated in humans or in mice (hearing loss (HL) genes) must play important roles in the development and physiology of the inner ear and may have been targets of selective forces across the evolution of mammals. Additionally, we investigated if these HL genes underwent a human-specific evolutionary process that could underlie the evolution of phenotypic traits that characterize human hearing. Results We compiled a dataset of HL genes including non-syndromic deafness genes identified by genetic screenings in humans and mice. We found that many genes including those required for the normal function of the inner ear such as LOXHD1, TMC1, OTOF, CDH23, and PCDH15 show strong signatures of positive selection. We also found numerous noncoding accelerated regions in HL genes, and among them, we identified active transcriptional enhancers through functional enhancer assays in transgenic zebrafish. Conclusions Our results indicate that the key inner ear genes and regulatory regions underwent adaptive evolution in the basal branch of mammals and along the human-specific branch, suggesting that they could have played an important role in the functional remodeling of the cochlea. Altogether, our data suggest that morphological and functional evolution could be attained through molecular changes affecting both coding and noncoding regulatory regions. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01170-6.
Collapse
Affiliation(s)
- Anabella P Trigila
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Buenos Aires, Argentina
| | - Francisco Pisciottano
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Buenos Aires, Argentina.,Current address: Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Buenos Aires, Argentina
| | - Lucía F Franchini
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Buenos Aires, Argentina.
| |
Collapse
|
18
|
Essential Role of Sptan1 in Cochlear Hair Cell Morphology and Function Via Focal Adhesion Signaling. Mol Neurobiol 2021; 59:386-404. [PMID: 34708331 PMCID: PMC8786805 DOI: 10.1007/s12035-021-02551-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/31/2021] [Indexed: 12/05/2022]
Abstract
Hearing loss is the most common human sensory deficit. Hearing relies on stereocilia, inserted into the cuticular plate of hair cells (HCs), where they play an important role in the perception of sound and its transmission. Although numerous genes have been associated with hearing loss, the function of many hair cell genes has yet to be elucidated. Herein, we focused on nonerythroid spectrin αII (SPTAN1), abundant in the cuticular plate, surrounding the rootlets of stereocilia and along the plasma membrane. Interestingly, mice with HC-specific Sptan1 knockout exhibited rapid deafness, abnormal formation of stereocilia and cuticular plates, and loss of HCs from middle and apical turns of the cochlea during early postnatal stages. Additionally, Sptan1 deficiency led to the decreased spreading of House Ear Institute-Organ of Corti 1 cells, and induced abnormal formation of focal adhesions and integrin signaling in mouse HCs. Altogether, our findings highlight SPTAN1 as a critical molecule for HC stereocilia morphology and auditory function via regulation of focal adhesion signaling.
Collapse
|
19
|
DFNA20/26 and Other ACTG1-Associated Phenotypes: A Case Report and Review of the Literature. Audiol Res 2021; 11:582-593. [PMID: 34698053 PMCID: PMC8544197 DOI: 10.3390/audiolres11040052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/10/2021] [Accepted: 10/13/2021] [Indexed: 02/08/2023] Open
Abstract
Since the early 2000s, an ever-increasing subset of missense pathogenic variants in the ACTG1 gene has been associated with an autosomal-dominant, progressive, typically post-lingual non-syndromic hearing loss (NSHL) condition designed as DFNA20/26. ACTG1 gene encodes gamma actin, the predominant actin protein in the cytoskeleton of auditory hair cells; its normal expression and function are essential for the stereocilia maintenance. Different gain-of-function pathogenic variants of ACTG1 have been associated with two major phenotypes: DFNA20/26 and Baraitser-Winter syndrome, a multiple congenital anomaly disorder. Here, we report a novel ACTG1 variant [c.625G>A (p. Val209Met)] in an adult patient with moderate-severe NSHL characterized by a downsloping audiogram. The patient, who had a clinical history of slowly progressive NSHL and tinnitus, was referred to our laboratory for the analysis of a large panel of NSHL-associated genes by next generation sequencing. An extensive review of previously reported ACTG1 variants and their associated phenotypes was also performed.
Collapse
|
20
|
Abstract
Almost 25 years have passed since a mutation of a formin gene, DIAPH1, was identified as being responsible for a human inherited disorder: a form of sensorineural hearing loss. Since then, our knowledge of the links between formins and disease has deepened considerably. Mutations of DIAPH1 and six other formin genes (DAAM2, DIAPH2, DIAPH3, FMN2, INF2 and FHOD3) have been identified as the genetic cause of a variety of inherited human disorders, including intellectual disability, renal disease, peripheral neuropathy, thrombocytopenia, primary ovarian insufficiency, hearing loss and cardiomyopathy. In addition, alterations in formin genes have been associated with a variety of pathological conditions, including developmental defects affecting the heart, nervous system and kidney, aging-related diseases, and cancer. This review summarizes the most recent discoveries about the involvement of formin alterations in monogenic disorders and other human pathological conditions, especially cancer, with which they have been associated. In vitro results and experiments in modified animal models are discussed. Finally, we outline the directions for future research in this field.
Collapse
Affiliation(s)
| | - Miguel A. Alonso
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, 28049 Madrid, Spain;
| |
Collapse
|
21
|
Lakha R, Montero AM, Jabeen T, Costeas CC, Ma J, Vizcarra CL. Variable Autoinhibition among Deafness-Associated Variants of Diaphanous 1 (DIAPH1). Biochemistry 2021; 60:2320-2329. [PMID: 34279089 DOI: 10.1021/acs.biochem.1c00170] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
One of the earliest mapped human deafness genes, DIAPH1, encodes the formin DIAPH1. To date, at least three distinct mutations in the C-terminal domains and two additional mutations in the N-terminal region are associated with autosomal dominant hearing loss. The underlying molecular mechanisms are not known, and the role of formins in the inner ear is not well understood. In this study, we use biochemical assays to test the hypotheses that autoinhibition and/or actin assembly activities are disrupted by DFNA1 mutations. Our results indicate that C-terminal mutant forms of DIAPH1 are functional in vitro and promote actin filament assembly. Similarly, N-terminal mutants are well-folded and have quaternary structures and thermal stabilities similar to those of the wild-type (WT) protein. The strength of the autoinhibitory interactions varies widely among mutants, with the ttaa, A265S, and I530S mutations having an affinity similar to that of WT and the 1213x and Δag mutations completely abolishing autoinhibition. These data indicate that, in some cases, hearing loss may be linked to weakened inhibition of actin assembly.
Collapse
Affiliation(s)
- Rabina Lakha
- Department of Chemistry, Barnard College, New York, New York 10027, United States
| | - Angela M Montero
- Department of Chemistry, Barnard College, New York, New York 10027, United States
| | - Tayyaba Jabeen
- Department of Chemistry, Barnard College, New York, New York 10027, United States
| | - Christina C Costeas
- Department of Chemistry, Barnard College, New York, New York 10027, United States
| | - Jia Ma
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Christina L Vizcarra
- Department of Chemistry, Barnard College, New York, New York 10027, United States
| |
Collapse
|
22
|
Lin L, Shi Y, Wang M, Wang C, Lu Q, Zhu J, Zhang R. Phase separation-mediated condensation of Whirlin-Myo15-Eps8 stereocilia tip complex. Cell Rep 2021; 34:108770. [PMID: 33626355 DOI: 10.1016/j.celrep.2021.108770] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 01/05/2021] [Accepted: 01/28/2021] [Indexed: 12/19/2022] Open
Abstract
Stereocilia, the mechanosensory organelles on the apical surface of hair cells, are necessary to detect sound and carry out mechano-electrical transduction. An electron-dense matrix is located at the distal tips of stereocilia and plays crucial roles in the regulation of stereocilia morphology. Mutations of the components in this tip complex density (TCD) have been associated with profound deafness. However, the mechanism underlying the formation of the TCD is largely unknown. Here, we discover that the specific multivalent interactions among the Whirlin-myosin 15 (Myo15)-Eps8 complex lead to the formation of the TCD-like condensates through liquid-liquid phase separation. The reconstituted TCD-like condensates effectively promote actin bundling. A deafness-associated mutation of Myo15 interferes with the condensates formation and consequently impairs actin bundling. Therefore, our study not only suggests that the TCD in hair cell stereocilia may form via phase separation but it also provides important clues for the possible mechanism underlying hearing loss.
Collapse
Affiliation(s)
- Lin Lin
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yingdong Shi
- Ministry of Education Key Laboratory for Membrane-less Organelles & Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Mengli Wang
- Ministry of Education Key Laboratory for Membrane-less Organelles & Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Chao Wang
- Ministry of Education Key Laboratory for Membrane-less Organelles & Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Qing Lu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jinwei Zhu
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Rongguang Zhang
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
23
|
Labat-de-Hoz L, Alonso MA. The formin INF2 in disease: progress from 10 years of research. Cell Mol Life Sci 2020; 77:4581-4600. [PMID: 32451589 PMCID: PMC11104792 DOI: 10.1007/s00018-020-03550-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 05/04/2020] [Accepted: 05/13/2020] [Indexed: 02/07/2023]
Abstract
Formins are a conserved family of proteins that primarily act to form linear polymers of actin. Despite their importance to the normal functioning of the cytoskeleton, for a long time, the only two formin genes known to be a genetic cause of human disorders were DIAPH1 and DIAPH3, whose mutation causes two distinct forms of hereditary deafness. In the last 10 years, however, the formin INF2 has emerged as an important target of mutations responsible for the appearance of focal segmental glomerulosclerosis, which are histological lesions associated with glomerulus degeneration that often leads to end-stage renal disease. In some rare cases, focal segmental glomerulosclerosis concurs with Charcot-Marie-Tooth disease, which is a degenerative neurological disorder affecting peripheral nerves. All known INF2 gene mutations causing disease map to the exons encoding the amino-terminal domain. In this review, we summarize the structure, biochemical features and functions of INF2, conduct a systematic and comprehensive analysis of the pathogenic INF2 mutations, including a detailed study exon-by-exon of patient cases and mutations, address the impact of the pathogenic mutations on the structure, regulation and known functions of INF2, draw a series of conclusions that could be useful for INF2-related disease diagnosis, and suggest lines of research for future work on the molecular mechanisms by which INF2 causes disease.
Collapse
Affiliation(s)
- Leticia Labat-de-Hoz
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Miguel A Alonso
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
24
|
Begovich K, Yelon D, Wilhelm JE. PRPS polymerization influences lens fiber organization in zebrafish. Dev Dyn 2020; 249:1018-1031. [PMID: 32243675 DOI: 10.1002/dvdy.173] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 02/28/2020] [Accepted: 03/05/2020] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND The self-assembly of metabolic enzymes into filaments or foci highlights an intriguing mechanism for the regulation of metabolic activity. Recently, we identified the conserved polymerization of phosphoribosyl pyrophosphate synthetase (PRPS), which catalyzes the first step in purine nucleotide synthesis, in yeast and cultured mammalian cells. While previous work has revealed that loss of PRPS activity regulates retinal development in zebrafish, the extent to which PRPS filament formation affects tissue development remains unknown. RESULTS By generating novel alleles in the zebrafish PRPS paralogs, prps1a and prps1b, we gained new insight into the role of PRPS filaments during eye development. We found that mutations in prps1a alone are sufficient to generate abnormally small eyes along with defects in head size, pigmentation, and swim bladder inflation. Furthermore, a loss-of-function mutation that truncates the Prps1a protein resulted in the failure of PRPS filament assembly. Lastly, in mutants that fail to assemble PRPS filaments, we observed disorganization of the actin network in the lens fibers. CONCLUSIONS The truncation of Prps1a blocked PRPS filament formation and resulted in a disorganized lens fiber actin network. Altogether, these findings highlight a potential role for PRPS filaments during lens fiber organization in zebrafish.
Collapse
Affiliation(s)
- Kyle Begovich
- Howard Hughes Medical Institute (HHMI) Summer Institute, Marine Biological Laboratory, Woods Hole, Massachusetts, USA.,Division of Biological Sciences, University of California, San Diego, California, USA
| | - Deborah Yelon
- Division of Biological Sciences, University of California, San Diego, California, USA
| | - James E Wilhelm
- Howard Hughes Medical Institute (HHMI) Summer Institute, Marine Biological Laboratory, Woods Hole, Massachusetts, USA.,Division of Biological Sciences, University of California, San Diego, California, USA
| |
Collapse
|
25
|
Pisciottano F, Cinalli AR, Stopiello JM, Castagna VC, Elgoyhen AB, Rubinstein M, Gómez-Casati ME, Franchini LF. Inner Ear Genes Underwent Positive Selection and Adaptation in the Mammalian Lineage. Mol Biol Evol 2020; 36:1653-1670. [PMID: 31137036 DOI: 10.1093/molbev/msz077] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The mammalian inner ear possesses functional and morphological innovations that contribute to its unique hearing capacities. The genetic bases underlying the evolution of this mammalian landmark are poorly understood. We propose that the emergence of morphological and functional innovations in the mammalian inner ear could have been driven by adaptive molecular evolution. In this work, we performed a meta-analysis of available inner ear gene expression data sets in order to identify genes that show signatures of adaptive evolution in the mammalian lineage. We analyzed ∼1,300 inner ear expressed genes and found that 13% show signatures of positive selection in the mammalian lineage. Several of these genes are known to play an important function in the inner ear. In addition, we identified that a significant proportion of genes showing signatures of adaptive evolution in mammals have not been previously reported to participate in inner ear development and/or physiology. We focused our analysis in two of these genes: STRIP2 and ABLIM2 by generating null mutant mice and analyzed their auditory function. We found that mice lacking Strip2 displayed a decrease in neural response amplitudes. In addition, we observed a reduction in the number of afferent synapses, suggesting a potential cochlear neuropathy. Thus, this study shows the usefulness of pursuing a high-throughput evolutionary approach followed by functional studies to track down genes that are important for inner ear function. Moreover, this approach sheds light on the genetic bases underlying the evolution of the mammalian inner ear.
Collapse
Affiliation(s)
- Francisco Pisciottano
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Alejandro R Cinalli
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Juan Matías Stopiello
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Valeria C Castagna
- Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires,Argentina
| | - Ana Belén Elgoyhen
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Marcelo Rubinstein
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires, Buenos Aires,Argentina
| | - María Eugenia Gómez-Casati
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires,Argentina
| | - Lucía F Franchini
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
26
|
Li W, Feng Y, Chen A, Li T, Huang S, Liu J, Liu X, Liu Y, Gao J, Yan D, Sun J, Mei L, Liu X, Ling J. Elmod3 knockout leads to progressive hearing loss and abnormalities in cochlear hair cell stereocilia. Hum Mol Genet 2019; 28:4103-4112. [PMID: 31628468 PMCID: PMC7305813 DOI: 10.1093/hmg/ddz240] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 10/02/2019] [Accepted: 10/03/2019] [Indexed: 01/28/2023] Open
Abstract
ELMOD3, an ARL2 GTPase-activating protein, is implicated in causing hearing impairment in humans. However, the specific role of ELMOD3 in auditory function is still far from being elucidated. In the present study, we used the CRISPR/Cas9 technology to establish an Elmod3 knockout mice line in the C57BL/6 background (hereinafter referred to as Elmod3-/- mice) and investigated the role of Elmod3 in the cochlea and auditory function. Elmod3-/- mice started to exhibit hearing loss from 2 months of age, and the deafness progressed with aging, while the vestibular function of Elmod3-/- mice was normal. We also observed that Elmod3-/- mice showed thinning and receding hair cells in the organ of Corti and much lower expression of F-actin cytoskeleton in the cochlea compared with wild-type mice. The deafness associated with the mutation may be caused by cochlear hair cells dysfunction, which manifests with shortening and fusion of inner hair cells stereocilia and progressive degeneration of outer hair cells stereocilia. Our finding associates Elmod3 deficiencies with stereocilia dysmorphologies and reveals that they might play roles in the actin cytoskeleton dynamics in cochlear hair cells, and thus relate to hearing impairment.
Collapse
Affiliation(s)
- Wu Li
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Head and Neck Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yong Feng
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan, China
- Hunan Jiahui Genetics Hospital, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Anhai Chen
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan, China
| | - Taoxi Li
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan, China
- Hunan Jiahui Genetics Hospital, Changsha, Hunan, China
| | - Sida Huang
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan, China
| | - Jing Liu
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan, China
| | - Xianlin Liu
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan, China
| | - Yalan Liu
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan, China
| | - Jiangang Gao
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, Shandong, China
| | - Denise Yan
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jie Sun
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Lingyun Mei
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan, China
| | - Xuezhong Liu
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jie Ling
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Molecular Precision Medicine, Xiangya Hospital, Central South University and Hunan Key Laboratory of Molecular Precision Medicine, Changsha, Hunan, China
- Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, Hunan, China
| |
Collapse
|
27
|
Du TT, Dewey JB, Wagner EL, Cui R, Heo J, Park JJ, Francis SP, Perez-Reyes E, Guillot SJ, Sherman NE, Xu W, Oghalai JS, Kachar B, Shin JB. LMO7 deficiency reveals the significance of the cuticular plate for hearing function. Nat Commun 2019; 10:1117. [PMID: 30850599 PMCID: PMC6408450 DOI: 10.1038/s41467-019-09074-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 02/15/2019] [Indexed: 12/25/2022] Open
Abstract
Sensory hair cells, the mechanoreceptors of the auditory and vestibular systems, harbor two specialized elaborations of the apical surface, the hair bundle and the cuticular plate. In contrast to the extensively studied mechanosensory hair bundle, the cuticular plate is not as well understood. It is believed to provide a rigid foundation for stereocilia motion, but specifics about its function, especially the significance of its integrity for long-term maintenance of hair cell mechanotransduction, are not known. We discovered that a hair cell protein called LIM only protein 7 (LMO7) is specifically localized in the cuticular plate and the cell junction. Lmo7 KO mice suffer multiple cuticular plate deficiencies, including reduced filamentous actin density and abnormal stereociliar rootlets. In addition to the cuticular plate defects, older Lmo7 KO mice develop abnormalities in inner hair cell stereocilia. Together, these defects affect cochlear tuning and sensitivity and give rise to late-onset progressive hearing loss.
Collapse
MESH Headings
- Actins/metabolism
- Animals
- Cochlea/physiology
- Disease Models, Animal
- Hair Cells, Auditory/physiology
- Hair Cells, Auditory/ultrastructure
- Hair Cells, Auditory, Inner/physiology
- Hair Cells, Auditory, Inner/ultrastructure
- Hearing/genetics
- Hearing/physiology
- Hearing Loss/etiology
- Hearing Loss/genetics
- Hearing Loss/physiopathology
- LIM Domain Proteins/deficiency
- LIM Domain Proteins/genetics
- LIM Domain Proteins/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Inbred CBA
- Mice, Knockout
- Microscopy, Electron, Scanning
- Stereocilia/genetics
- Stereocilia/physiology
- Stereocilia/ultrastructure
- Transcription Factors/deficiency
- Transcription Factors/genetics
- Transcription Factors/physiology
Collapse
Affiliation(s)
- Ting-Ting Du
- Department of Neuroscience, University of Virginia, Charlottesville, VA, 22908, USA
| | - James B Dewey
- Caruso Department of Otolaryngology-Head and Neck Surgery, University of Southern California, Los Angeles, CA, 90033, USA
| | - Elizabeth L Wagner
- Department of Neuroscience, University of Virginia, Charlottesville, VA, 22908, USA
| | - Runjia Cui
- National Institute for Deafness and Communications Disorders, National Institute of Health, Bethesda, MD, 20892, USA
| | - Jinho Heo
- Center for Cell Signaling and Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Jeong-Jin Park
- Biomolecular Analysis Facility, University of Virginia, Charlottesville, VA, 22908, USA
| | - Shimon P Francis
- Department of Neuroscience, University of Virginia, Charlottesville, VA, 22908, USA
| | - Edward Perez-Reyes
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Stacey J Guillot
- Advanced Microscopy core, University of Virginia, Charlottesville, VA, 22908, USA
| | - Nicholas E Sherman
- Biomolecular Analysis Facility, University of Virginia, Charlottesville, VA, 22908, USA
| | - Wenhao Xu
- Genetically Engineered Murine Model (GEMM) core, University of Virginia, Charlottesville, VA, 22908, USA
| | - John S Oghalai
- Caruso Department of Otolaryngology-Head and Neck Surgery, University of Southern California, Los Angeles, CA, 90033, USA
| | - Bechara Kachar
- National Institute for Deafness and Communications Disorders, National Institute of Health, Bethesda, MD, 20892, USA
| | - Jung-Bum Shin
- Department of Neuroscience, University of Virginia, Charlottesville, VA, 22908, USA.
| |
Collapse
|
28
|
Men Y, Li X, Tu H, Zhang A, Fu X, Wang Z, Jin Y, Hou C, Zhang T, Zhang S, Zhou Y, Li B, Li J, Sun X, Wang H, Gao J. Tprn is essential for the integrity of stereociliary rootlet in cochlear hair cells in mice. Front Med 2018; 13:690-704. [PMID: 30159668 DOI: 10.1007/s11684-018-0638-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Accepted: 01/27/2018] [Indexed: 11/24/2022]
Abstract
Tprn encodes the taperin protein, which is concentrated in the tapered region of hair cell stereocilia in the inner ear. In humans, TPRN mutations cause autosomal recessive nonsyndromic deafness (DFNB79) by an unknown mechanism. To determine the role of Tprn in hearing, we generated Tprn-null mice by clustered regularly interspaced short palindromic repeat/Cas9 genome-editing technology from a CBA/CaJ background. We observed significant hearing loss and progressive degeneration of stereocilia in the outer hair cells of Tprn-null mice starting from postnatal day 30. Transmission electron microscopy images of stereociliary bundles in the mutant mice showed some stereociliary rootlets with curved shafts. The central cores of the stereociliary rootlets possessed hollow structures with surrounding loose peripheral dense rings. Radixin, a protein expressed at stereocilia tapering, was abnormally dispersed along the stereocilia shafts in Tprn-null mice. The expression levels of radixin and β-actin significantly decreased.We propose that Tprn is critical to the retention of the integrity of the stereociliary rootlet. Loss of Tprn in Tprn-null mice caused the disruption of the stereociliary rootlet, which resulted in damage to stereociliary bundles and hearing impairments. The generated Tprn-null mice are ideal models of human hereditary deafness DFNB79.
Collapse
Affiliation(s)
- Yuqin Men
- School of Life Science, Shandong University, Jinan, 250100, China
| | - Xiujuan Li
- Rizhao Polytechnic, Rizhao, 276826, China
| | - Hailong Tu
- 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
| | - Zhishuo Wang
- School of Life Science, Shandong University, Jinan, 250100, China
| | - Yecheng Jin
- School of Life Science, Shandong University, Jinan, 250100, China
| | - Congzhe Hou
- The Second Hospital of Shandong University, Jinan, 250033, China
| | - Tingting Zhang
- School of Life Science, Shandong University, Jinan, 250100, China
| | - Sen Zhang
- School of Life Science, Shandong University, Jinan, 250100, China
| | - Yichen Zhou
- School of Life Science, Shandong University, Jinan, 250100, China
| | - Boqin Li
- Electron Microscopy Laboratory, Shandong Institute of Otolaryngology, Jinan, 250022, China.,Laboratory of Electron Microscopy, Jinan WEI-YA Biotech Company, Jinan, 250100, China
| | - Jianfeng Li
- Department of Otolaryngology-Head and Neck Surgery, Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China
| | - Xiaoyang Sun
- School of Life Science, Shandong University, Jinan, 250100, China.
| | - Haibo Wang
- Department of Otolaryngology-Head and Neck Surgery, Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China.
| | - Jiangang Gao
- School of Life Science, Shandong University, Jinan, 250100, China.
| |
Collapse
|
29
|
Ellwanger DC, Scheibinger M, Dumont RA, Barr-Gillespie PG, Heller S. Transcriptional Dynamics of Hair-Bundle Morphogenesis Revealed with CellTrails. Cell Rep 2018; 23:2901-2914.e13. [PMID: 29874578 PMCID: PMC6089258 DOI: 10.1016/j.celrep.2018.05.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 03/19/2018] [Accepted: 05/01/2018] [Indexed: 11/30/2022] Open
Abstract
Protruding from the apical surface of inner ear sensory cells, hair bundles carry out mechanotransduction. Bundle growth involves sequential and overlapping cellular processes, which are concealed within gene expression profiles of individual cells. To dissect such processes, we developed CellTrails, a tool for uncovering, analyzing, and visualizing single-cell gene-expression dynamics. Utilizing quantitative gene-expression data for key bundle proteins from single cells of the developing chick utricle, we reconstructed de novo a bifurcating trajectory that spanned from progenitor cells to mature striolar and extrastriolar hair cells. Extraction and alignment of developmental trails and association of pseudotime with bundle length measurements linked expression dynamics of individual genes with bundle growth stages. Differential trail analysis revealed high-resolution dynamics of transcripts that control striolar and extrastriolar bundle development, including those that encode proteins that regulate [Ca2+]i or mediate crosslinking and lengthening of actin filaments.
Collapse
Affiliation(s)
- Daniel C Ellwanger
- Department of Otolaryngology, Head & Neck Surgery and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mirko Scheibinger
- Department of Otolaryngology, Head & Neck Surgery and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rachel A Dumont
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Peter G Barr-Gillespie
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Stefan Heller
- Department of Otolaryngology, Head & Neck Surgery and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
30
|
Li W, Sun J, Ling J, Li J, He C, Liu Y, Chen H, Men M, Niu Z, Deng Y, Li M, Li T, Wen J, Sang S, Li H, Wan Z, Richard EM, Chapagain P, Yan D, Liu XZ, Mei L, Feng Y. ELMOD3, a novel causative gene, associated with human autosomal dominant nonsyndromic and progressive hearing loss. Hum Genet 2018; 137:329-342. [PMID: 29713870 DOI: 10.1007/s00439-018-1885-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 04/16/2018] [Indexed: 11/26/2022]
Abstract
Autosomal dominant nonsyndromic hearing loss (ADNSHL) is a highly genetically heterogeneous disorder. Up to date only approximately 37 ADNSHL-causing genes have been identified. The goal of this study was to determine the causative gene in a five-generation Chinese family with ADNSHL. A Chinese family was ascertained. Simultaneously, two affected individuals and one normal hearing control from the family were analyzed by whole exome capture sequencing. To assess the functional effect of the identified variant, in-vitro studies were performed. novel missense variant, c.512A>G (p.His171Arg) in exon 8 of the ELMO domain-containing 3 (ELMOD3) gene, was identified as a causative variant in this family affected by late-onset and progressive ADNSHL. The variant was validated by Sanger sequencing and found to co-segregate with the phenotype within the pedigree and was absent in 500 ethnically matched unrelated normal hearing control subjects. To our knowledge, this is the first report of a family with ADNSHL caused by ELMOD3 mutation. Western blots and immunofluorescence staining demonstrated that p.His171Arg resulted in abnormal expression levels of ELMOD3 and abnormal subcellular localization. Furthermore, the analysis of the stability of the wild-type (WT) and mutant ELMOD3 protein shows that the decay of p.His171Arg is faster than that of the WT, suggesting a shorter halflife of the c.512A > G variant. A novel variant in the ELMOD3 gene, encoding a member of the engulfment and cell motility (ELMO) family of GTPase-activating proteins, was identified for the first time as responsible for ADNSHL.
Collapse
Affiliation(s)
- Wu Li
- Department of Otolaryngology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan, China
| | - Jie Sun
- Department of Otolaryngology, The Eight Affiliated Hospital, Sun Yat-sen University, 3025 Shennan Middle Road, Shenzhen, Guangdong, China
| | - Jie Ling
- Institute of Precision Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Jiada Li
- Center for Medical Genetics, Central South University, 110 Xiangya Road, Changsha, Hunan, China
- School of Life Sciences, Central South University of China, 110 Xiangya Road, Changsha, Hunan, China
| | - Chufeng He
- Department of Otolaryngology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan, China
| | - Yalan Liu
- Department of Otolaryngology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan, China
| | - Hongsheng Chen
- Department of Otolaryngology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan, China
| | - Meichao Men
- Health Management Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Zhijie Niu
- Department of Otolaryngology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan, China
| | - Yuyuan Deng
- Department of Otolaryngology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan, China
| | - Meng Li
- Department of Otolaryngology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan, China
| | - Taoxi Li
- Center for Medical Genetics, Central South University, 110 Xiangya Road, Changsha, Hunan, China
- School of Life Sciences, Central South University of China, 110 Xiangya Road, Changsha, Hunan, China
| | - Jie Wen
- Department of Otolaryngology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan, China
| | - Shushan Sang
- Department of Otolaryngology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan, China
| | - Haibo Li
- Department of Ophthalmology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
| | - Zhengqing Wan
- Center for Medical Genetics, Central South University, 110 Xiangya Road, Changsha, Hunan, China
- School of Life Sciences, Central South University of China, 110 Xiangya Road, Changsha, Hunan, China
| | - Elodie M Richard
- Department of Otorhinolaryngology Head and Neck Surgery, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Prem Chapagain
- Department of Physics, Florida International University, Miami, Florida, USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL, USA
| | - Denise Yan
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, USA
| | - Xue Zhong Liu
- Department of Otolaryngology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, USA
- Dr. John T. Macdonald Foundation, Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Lingyun Mei
- Department of Otolaryngology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China.
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan, China.
| | - Yong Feng
- Department of Otolaryngology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China.
- Province Key Laboratory of Otolaryngology Critical Diseases, Changsha, Hunan, China.
| |
Collapse
|
31
|
Wang L, Yan D, Qin L, Li T, Liu H, Li W, Mittal R, Yong F, Chapagain P, Liao S, Liu X. Amino acid 118 in the Deafness Causing (DFNA20/26) ACTG1 gene is a Mutational Hot Spot. GENE REPORTS 2018; 11:264-269. [PMID: 30599039 DOI: 10.1016/j.genrep.2018.04.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Background Hearing loss is an economically and socially important cause of human morbidity, affecting 360 million people (over 5% of the world's population), of whom 32 million are children. Of the estimated minimum of 50% of hereditary hearing loss, non-syndromic hearing loss (NSHL) accounts for more than 70%. The autosomal dominant non-syndromic hearing loss (ADNSHL) is highly heterogeneous. To date, 67 ADNSHL loci (DFNA1-67) have been mapped; however, only 35 causative genes have been cloned since 1997 (http://hereditaryhearingloss.org/). Methods To identify the genetic basis of hereditary hearing loss in a Chinese family with ADNSHL, we undertook a targeted sequencing of 180 genes using a custom capture panel (MiamiOtoGenes). Results The onset of hearing loss in the family occurred between the ages of 15 and 18 years. Hearing loss was bilateral, started in the high frequency and progressed to lower frequencies. The c.353A>T (K118M) in the AC TG1 gene was identified by panel and was confirmed by Sanger sequencing and was present in all affected family members. So far, five of the 23 DFNA20/26 families worldwide have been found to carry mutation involving the residue K118. Conclusions This is the first report of K118M mutation in the ACTG1 gene causing hearing loss in the Chinese population. The present data are in line with previous evidence to suggest that codon K118 of ACTG1 may represent a mutational hot spot that justifies a mutation screen for diagnostic purpose in the genetically heterogeneous group of DFNA20/26.
Collapse
Affiliation(s)
- Li Wang
- Institute of Medical Genetics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China.,Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, USA
| | - Denise Yan
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, USA
| | - Litao Qin
- Institute of Medical Genetics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Tao Li
- Institute of Medical Genetics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongjian Liu
- Department of Otorhinolaryngology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Wan Li
- Department of Otorhinolaryngology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Rahul Mittal
- Institute of Medical Genetics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Feng Yong
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, China
| | - Prem Chapagain
- Department of Physics, Florida International University, Miami, Florida.,Biomolecular Sciences Institute, Florida International University, Miami, Florida
| | - Shixiu Liao
- Institute of Medical Genetics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuezhong Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, USA
| |
Collapse
|
32
|
Apodaca G. Role of Polarity Proteins in the Generation and Organization of Apical Surface Protrusions. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a027813. [PMID: 28264821 DOI: 10.1101/cshperspect.a027813] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Protruding from the apical surfaces of epithelial cells are specialized structures, including cilia, microplicae, microvilli, and stereocilia. These contribute to epithelial function by cushioning the apical surface, by amplifying its surface area to facilitate nutrient absorption, and by promoting sensory transduction and barrier function. Despite these important roles, and the diseases that result when their formation is perturbed, there remain significant gaps in our understanding of the biogenesis of apical protrusions, or the pathways that promote their organization and orientation once at the apical surface. Here, I review some general aspects of these apical structures, and then discuss our current understanding of their formation and organization with respect to proteins that specify apicobasolateral polarity and planar cell polarity.
Collapse
Affiliation(s)
- Gerard Apodaca
- Department of Medicine Renal-Electrolyte Division and the Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| |
Collapse
|
33
|
Krey JF, Krystofiak ES, Dumont RA, Vijayakumar S, Choi D, Rivero F, Kachar B, Jones SM, Barr-Gillespie PG. Plastin 1 widens stereocilia by transforming actin filament packing from hexagonal to liquid. J Cell Biol 2016; 215:467-482. [PMID: 27811163 PMCID: PMC5119939 DOI: 10.1083/jcb.201606036] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 09/29/2016] [Accepted: 10/11/2016] [Indexed: 11/24/2022] Open
Abstract
With their essential role in inner ear function, stereocilia of sensory hair cells demonstrate the importance of cellular actin protrusions. Actin packing in stereocilia is mediated by cross-linkers of the plastin, fascin, and espin families. Although mice lacking espin (ESPN) have no vestibular or auditory function, we found that mice that either lacked plastin 1 (PLS1) or had nonfunctional fascin 2 (FSCN2) had reduced inner ear function, with double-mutant mice most strongly affected. Targeted mass spectrometry indicated that PLS1 was the most abundant cross-linker in vestibular stereocilia and the second most abundant protein overall; ESPN only accounted for ∼15% of the total cross-linkers in bundles. Mouse utricle stereocilia lacking PLS1 were shorter and thinner than wild-type stereocilia. Surprisingly, although wild-type stereocilia had random liquid packing of their actin filaments, stereocilia lacking PLS1 had orderly hexagonal packing. Although all three cross-linkers are required for stereocilia structure and function, PLS1 biases actin toward liquid packing, which allows stereocilia to grow to a greater diameter.
Collapse
Affiliation(s)
- Jocelyn F Krey
- Oregon Hearing Research Center, Oregon Health and Science University, Portland, OR 97239
- Vollum Institute, Oregon Health and Science University, Portland, OR 97239
| | - Evan S Krystofiak
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892
| | - Rachel A Dumont
- Oregon Hearing Research Center, Oregon Health and Science University, Portland, OR 97239
- Vollum Institute, Oregon Health and Science University, Portland, OR 97239
| | - Sarath Vijayakumar
- Department of Special Education and Communication Disorders, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - Dongseok Choi
- Oregon Health and Science University-Portland State University School of Public Health, Oregon Health and Science University, Portland, OR 97239
- Graduate School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Francisco Rivero
- Centre for Cardiovascular and Metabolic Research, The Hull York Medical School, University of Hull, Hull HU6 7RX, England, UK
| | - Bechara Kachar
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892
| | - Sherri M Jones
- Department of Special Education and Communication Disorders, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - Peter G Barr-Gillespie
- Oregon Hearing Research Center, Oregon Health and Science University, Portland, OR 97239
- Vollum Institute, Oregon Health and Science University, Portland, OR 97239
| |
Collapse
|
34
|
Ueyama T, Ninoyu Y, Nishio SY, Miyoshi T, Torii H, Nishimura K, Sugahara K, Sakata H, Thumkeo D, Sakaguchi H, Watanabe N, Usami SI, Saito N, Kitajiri SI. Constitutive activation of DIA1 (DIAPH1) via C-terminal truncation causes human sensorineural hearing loss. EMBO Mol Med 2016; 8:1310-1324. [PMID: 27707755 PMCID: PMC5090661 DOI: 10.15252/emmm.201606609] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
DIAPH1 encodes human DIA1, a formin protein that elongates unbranched actin. The c.3634+1G>T DIAPH1 mutation causes autosomal dominant nonsyndromic sensorineural hearing loss, DFNA1, characterized by progressive deafness starting in childhood. The mutation occurs near the C‐terminus of the diaphanous autoregulatory domain (DAD) of DIA1, which interacts with its N‐terminal diaphanous inhibitory domain (DID), and may engender constitutive activation of DIA1. However, the underlying pathogenesis that causes DFNA1 is unclear. We describe a novel patient‐derived DIAPH1 mutation (c.3610C>T) in two unrelated families, which results in early termination prior to a basic amino acid motif (RRKR1204–1207) at the DAD C‐terminus. The mutant DIA1(R1204X) disrupted the autoinhibitory DID‐DAD interaction and was constitutively active. This unscheduled activity caused increased rates of directional actin polymerization movement and induced formation of elongated microvilli. Mice expressing FLAG‐tagged DIA1(R1204X) experienced progressive deafness and hair cell loss at the basal turn and had various morphological abnormalities in stereocilia (short, fused, elongated, sparse). Thus, the basic region of the DAD mediates DIA1 autoinhibition; disruption of the DID‐DAD interaction and consequent activation of DIA1(R1204X) causes DFNA1.
Collapse
Affiliation(s)
- Takehiko Ueyama
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, Japan
| | - Yuzuru Ninoyu
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, Japan
| | - Shin-Ya Nishio
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takushi Miyoshi
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroko Torii
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koji Nishimura
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuma Sugahara
- Department of Otolaryngology, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | | | - Dean Thumkeo
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hirofumi Sakaguchi
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Naoki Watanabe
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Laboratory of Single-Molecule Cell Biology, Kyoto University Graduate School of Biostudies, Kyoto, Japan
| | - Shin-Ichi Usami
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Naoaki Saito
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, Japan
| | - Shin-Ichiro Kitajiri
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
35
|
Rehman AU, Bird JE, Faridi R, Shahzad M, Shah S, Lee K, Khan SN, Imtiaz A, Ahmed ZM, Riazuddin S, Santos-Cortez RLP, Ahmad W, Leal SM, Riazuddin S, Friedman TB. Mutational Spectrum of MYO15A and the Molecular Mechanisms of DFNB3 Human Deafness. Hum Mutat 2016; 37:991-1003. [PMID: 27375115 DOI: 10.1002/humu.23042] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 06/26/2016] [Indexed: 12/17/2022]
Abstract
Deafness in humans is a common neurosensory disorder and is genetically heterogeneous. Across diverse ethnic groups, mutations of MYO15A at the DFNB3 locus appear to be the third or fourth most common cause of autosomal-recessive, nonsyndromic deafness. In 49 of the 67 exons of MYO15A, there are currently 192 recessive mutations identified, including 14 novel mutations reported here. These mutations are distributed uniformly across MYO15A with one enigmatic exception; the alternatively spliced giant exon 2, encoding 1,233 residues, has 17 truncating mutations but no convincing deafness-causing missense mutations. MYO15A encodes three distinct isoform classes, one of which is 395 kDa (3,530 residues), the largest member of the myosin superfamily of molecular motors. Studies of Myo15 mouse models that recapitulate DFNB3 revealed two different pathogenic mechanisms of hearing loss. In the inner ear, myosin 15 is necessary both for the development and the long-term maintenance of stereocilia, mechanosensory sound-transducing organelles that extend from the apical surface of hair cells. The goal of this Mutation Update is to provide a comprehensive review of mutations and functions of MYO15A.
Collapse
Affiliation(s)
- Atteeq U Rehman
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, 20892
| | - Jonathan E Bird
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, 20892
| | - Rabia Faridi
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, 20892.,Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, 54550, Pakistan
| | - Mohsin Shahzad
- Department of Otorhinolaryngology Head & Neck Surgery, School of Medicine, University of Maryland, Baltimore, Maryland, 21201
| | - Sujay Shah
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, 20892
| | - Kwanghyuk Lee
- Center for Statistical Genetics, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030
| | - Shaheen N Khan
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, 54550, Pakistan
| | - Ayesha Imtiaz
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, 20892
| | - Zubair M Ahmed
- Department of Otorhinolaryngology Head & Neck Surgery, School of Medicine, University of Maryland, Baltimore, Maryland, 21201
| | - Saima Riazuddin
- Department of Otorhinolaryngology Head & Neck Surgery, School of Medicine, University of Maryland, Baltimore, Maryland, 21201
| | - Regie Lyn P Santos-Cortez
- Center for Statistical Genetics, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030
| | - Wasim Ahmad
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Suzanne M Leal
- Center for Statistical Genetics, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030
| | - Sheikh Riazuddin
- Allama Iqbal Medical Research Centre, Jinnah Hospital Complex, University of Health Sciences, Lahore, 54550, Pakistan
| | - Thomas B Friedman
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, 20892.
| |
Collapse
|
36
|
Andreini C, Banci L, Rosato A. Exploiting Bacterial Operons To Illuminate Human Iron–Sulfur Proteins. J Proteome Res 2016; 15:1308-22. [DOI: 10.1021/acs.jproteome.6b00045] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Claudia Andreini
- Magnetic Resonance Center and ‡Department of Chemistry, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Lucia Banci
- Magnetic Resonance Center and ‡Department of Chemistry, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Antonio Rosato
- Magnetic Resonance Center and ‡Department of Chemistry, University of Florence, 50019 Sesto Fiorentino, Italy
| |
Collapse
|
37
|
Yuan Y, Gao X, Huang B, Lu J, Wang G, Lin X, Qu Y, Dai P. Phenotypic Heterogeneity in a DFNA20/26 family segregating a novel ACTG1 mutation. BMC Genet 2016; 17:33. [PMID: 26832775 PMCID: PMC4736096 DOI: 10.1186/s12863-016-0333-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 01/14/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Genetic factors play an important role in hearing loss, contributing to approximately 60% of cases of congenital hearing loss. Autosomal dominant deafness accounts for approximately 20% of cases of hereditary hearing loss. Diseases with autosomal dominant inheritance often show pleiotropy, different degrees of penetrance, and variable expressivity. METHODS A three-generation Chinese family with autosomal dominant nonsyndromic hearing impairment (ADNSHI) was enrolled in this study. Audiometric data and blood samples were collected from the family. In total, 129 known human deafness genes were sequenced using next-generation sequencing (NGS) to identify the responsible gene mutation in the family. Whole Exome Sequencing (WES) was performed to exclude any other variant that cosegregated with the phenotype. RESULTS The age of onset of the affected family members was the second decade of life. The condition began with high-frequency hearing impairment in all family members excluding III:2. The novel ACTG1 c.638A > G (p.K213R) mutation was found in all affected family members and was not found in the unaffected family members. A heterozygous c.638A > G mutation in ACTG1 and homozygous c.109G > A (p.V37I) mutation in GJB2 were found in III:2, who was born with hearing loss. The WES result concurred with that of targeted sequencing of known deafness genes. CONCLUSIONS The novel mutation p.K213R in ACTG1 was found to be co-segregated with hearing loss and the genetic cause of ADNSHI in this family. A homozygous mutation associated with recessive inheritance only rarely co-acts with a dominant mutation to result in hearing loss in a dominant family. In such cases, the mutations in the two genes, as in ACTG1 and GJB2 in the present study, may result in a more severe phenotype. Targeted sequencing of known deafness genes is one of the best choices to identify the genetic cause in hereditary hearing loss families.
Collapse
Affiliation(s)
- Yongyi Yuan
- Department of Otolaryngology, Chinese PLA General Hospital, Beijing, 100853, People's Republic of China. .,Department of Otolaryngology, Hainan Branch of PLA General Hospital, Sanya, 572000, People's Republic of China.
| | - Xue Gao
- Department of Otolaryngology, Chinese PLA General Hospital, Beijing, 100853, People's Republic of China.
| | - Bangqing Huang
- Department of Otolaryngology, Hainan Branch of PLA General Hospital, Sanya, 572000, People's Republic of China.
| | - Jingqiao Lu
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, GA, 30322-3030, USA.
| | - Guojian Wang
- Department of Otolaryngology, Chinese PLA General Hospital, Beijing, 100853, People's Republic of China.
| | - Xi Lin
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, GA, 30322-3030, USA.
| | - Yan Qu
- Third hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050051, People's Republic of China.
| | - Pu Dai
- Department of Otolaryngology, Chinese PLA General Hospital, Beijing, 100853, People's Republic of China.
| |
Collapse
|
38
|
Gene Therapy Restores Hair Cell Stereocilia Morphology in Inner Ears of Deaf Whirler Mice. Mol Ther 2015; 24:17-25. [PMID: 26307667 DOI: 10.1038/mt.2015.150] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 08/10/2015] [Indexed: 01/09/2023] Open
Abstract
Hereditary deafness is one of the most common disabilities affecting newborns. Many forms of hereditary deafness are caused by morphological defects of the stereocilia bundles on the apical surfaces of inner ear hair cells, which are responsible for sound detection. We explored the effectiveness of gene therapy in restoring the hair cell stereocilia architecture in the whirlin mouse model of human deafness, which is deaf due to dysmorphic, short stereocilia. Wild-type whirlin cDNA was delivered via adeno-associated virus (AAV8) by injection through the round window of the cochleas in neonatal whirler mice. Subsequently, whirlin expression was detected in infected hair cells (IHCs), and normal stereocilia length and bundle architecture were restored. Whirlin gene therapy also increased inner hair cell survival in the treated ears compared to the contralateral nontreated ears. These results indicate that a form of inherited deafness due to structural defects in cochlear hair cells is amenable to restoration through gene therapy.
Collapse
|
39
|
Fang Q, Indzhykulian AA, Mustapha M, Riordan GP, Dolan DF, Friedman TB, Belyantseva IA, Frolenkov GI, Camper SA, Bird JE. The 133-kDa N-terminal domain enables myosin 15 to maintain mechanotransducing stereocilia and is essential for hearing. eLife 2015; 4. [PMID: 26302205 PMCID: PMC4592939 DOI: 10.7554/elife.08627] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 08/22/2015] [Indexed: 11/13/2022] Open
Abstract
The precise assembly of inner ear hair cell stereocilia into rows of increasing height is critical for mechanotransduction and the sense of hearing. Yet, how the lengths of actin-based stereocilia are regulated remains poorly understood. Mutations of the molecular motor myosin 15 stunt stereocilia growth and cause deafness. We found that hair cells express two isoforms of myosin 15 that differ by inclusion of an 133-kDa N-terminal domain, and that these isoforms can selectively traffic to different stereocilia rows. Using an isoform-specific knockout mouse, we show that hair cells expressing only the small isoform remarkably develop normal stereocilia bundles. However, a critical subset of stereocilia with active mechanotransducer channels subsequently retracts. The larger isoform with the 133-kDa N-terminal domain traffics to these specialized stereocilia and prevents disassembly of their actin core. Our results show that myosin 15 isoforms can navigate between functionally distinct classes of stereocilia, and are independently required to assemble and then maintain the intricate hair bundle architecture.
Collapse
Affiliation(s)
- Qing Fang
- Department of Human Genetics, University of Michigan, Ann Arbor, United States
| | | | - Mirna Mustapha
- Department of Human Genetics, University of Michigan, Ann Arbor, United States
| | - Gavin P Riordan
- Laboratory of Molecular Genetics, National Institutes of Health, Bethesda, United States
| | - David F Dolan
- Department of Otolaryngology, University of Michigan Medical School, Ann Arbor, United States
| | - Thomas B Friedman
- Laboratory of Molecular Genetics, National Institutes of Health, Bethesda, United States
| | - Inna A Belyantseva
- Laboratory of Molecular Genetics, National Institutes of Health, Bethesda, United States
| | | | - Sally A Camper
- Department of Human Genetics, University of Michigan, Ann Arbor, United States
| | - Jonathan E Bird
- Laboratory of Molecular Genetics, National Institutes of Health, Bethesda, United States
| |
Collapse
|
40
|
Andrade LR. Evidence for changes in beta- and gamma-actin proportions during inner ear hair cell life. Cytoskeleton (Hoboken) 2015; 72:282-91. [PMID: 26033950 DOI: 10.1002/cm.21227] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 05/11/2015] [Accepted: 05/21/2015] [Indexed: 12/18/2022]
Abstract
Cytoplasmic actin isoforms beta (β-) and gamma (γ-) perform crucial physiological roles in inner ear hair cells (HC). The stereocilium, which is structured by parallel actin filaments composed of both isoforms, is the responsive organelle to mechanical stimuli such as sound, gravity and head movements. Modifications in isoform proportions affect the function of the stereocilia as previously shown in genetic studies of mutant mice. Here, immunogold labeling TEM studies in mice showed that both β- and γ-actin isoforms colocalize throughout stereocilia actin filaments, adherens junctions and cuticular plates as early as embryonic stage 16.5. Gold-particle quantification indicated that there was 40% more γ- actin than β-actin at E16.5. In contrast, β- and γ-actin were equally concentrated in adult stereocilia of cochlear and vestibular HC. Interestingly, all actin-based structures presented almost five-fold more β-actin than γ-actin in 22 month- old mice, suggesting that γ-actin is probably under-expressed during the aging process. These data provide evidence of dynamic modifications of the actin isoforms in stereocilia, cuticular plates and cell junctions during the whole HC life.
Collapse
Affiliation(s)
- Leonardo R Andrade
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA.,Laboratory of Biomineralization, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| |
Collapse
|
41
|
Taylor R, Bullen A, Johnson SL, Grimm-Günter EM, Rivero F, Marcotti W, Forge A, Daudet N. Absence of plastin 1 causes abnormal maintenance of hair cell stereocilia and a moderate form of hearing loss in mice. Hum Mol Genet 2014; 24:37-49. [PMID: 25124451 PMCID: PMC4262491 DOI: 10.1093/hmg/ddu417] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Hearing relies on the mechanosensory inner and outer hair cells (OHCs) of the organ of Corti, which convert mechanical deflections of their actin-rich stereociliary bundles into electrochemical signals. Several actin-associated proteins are essential for stereocilia formation and maintenance, and their absence leads to deafness. One of the most abundant actin-bundling proteins of stereocilia is plastin 1, but its function has never been directly assessed. Here, we found that plastin 1 knock-out (Pls1 KO) mice have a moderate and progressive form of hearing loss across all frequencies. Auditory hair cells developed normally in Pls1 KO, but in young adult animals, the stereocilia of inner hair cells were reduced in width and length. The stereocilia of OHCs were comparatively less affected; however, they also showed signs of degeneration in ageing mice. The hair bundle stiffness and the acquisition of the electrophysiological properties of hair cells were unaffected by the absence of plastin 1, except for a significant change in the adaptation properties, but not the size of the mechanoelectrical transducer currents. These results show that in contrast to other actin-bundling proteins such as espin, harmonin or Eps8, plastin 1 is dispensable for the initial formation of stereocilia. However, the progressive hearing loss and morphological defects of hair cells in adult Pls1 KO mice point at a specific role for plastin 1 in the preservation of adult stereocilia and optimal hearing. Hence, mutations in the human PLS1 gene may be associated with relatively mild and progressive forms of hearing loss.
Collapse
Affiliation(s)
- Ruth Taylor
- Centre for Auditory Research, UCL Ear Institute, University College London, London, UK
| | - Anwen Bullen
- Centre for Auditory Research, UCL Ear Institute, University College London, London, UK
| | - Stuart L Johnson
- Department of Biomedical Science, University of Sheffield, Sheffield, UK and
| | - Eva-Maria Grimm-Günter
- Centre for Cardiovascular and Metabolic Research, The Hull York Medical School, University of Hull, Hull, UK
| | - Francisco Rivero
- Centre for Cardiovascular and Metabolic Research, The Hull York Medical School, University of Hull, Hull, UK
| | - Walter Marcotti
- Department of Biomedical Science, University of Sheffield, Sheffield, UK and
| | - Andrew Forge
- Centre for Auditory Research, UCL Ear Institute, University College London, London, UK
| | - Nicolas Daudet
- Centre for Auditory Research, UCL Ear Institute, University College London, London, UK
| |
Collapse
|
42
|
Chaperone-enhanced purification of unconventional myosin 15, a molecular motor specialized for stereocilia protein trafficking. Proc Natl Acad Sci U S A 2014; 111:12390-5. [PMID: 25114250 DOI: 10.1073/pnas.1409459111] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Unconventional myosin 15 is a molecular motor expressed in inner ear hair cells that transports protein cargos within developing mechanosensory stereocilia. Mutations of myosin 15 cause profound hearing loss in humans and mice; however, the properties of this motor and its regulation within the stereocilia organelle are unknown. To address these questions, we expressed a subfragment 1-like (S1) truncation of mouse myosin 15, comprising the predicted motor domain plus three light-chain binding sites. Following unsuccessful attempts to express functional myosin 15-S1 using the Spodoptera frugiperda (Sf9)-baculovirus system, we discovered that coexpression of the muscle-myosin-specific chaperone UNC45B, in addition to the chaperone heat-shock protein 90 (HSP90) significantly increased the yield of functional protein. Surprisingly, myosin 15-S1 did not bind calmodulin with high affinity. Instead, the IQ domains bound essential and regulatory light chains that are normally associated with class II myosins. We show that myosin 15-S1 is a barbed-end-directed motor that moves actin filaments in a gliding assay (∼ 430 nm · s(-1) at 30 °C), using a power stroke of 7.9 nm. The maximum ATPase rate (k(cat) ∼ 6 s(-1)) was similar to the actin-detachment rate (k(det) = 6.2 s(-1)) determined in single molecule optical trapping experiments, indicating that myosin 15-S1 was rate limited by transit through strongly actin-bound states, similar to other processive myosin motors. Our data further indicate that in addition to folding muscle myosin, UNC45B facilitates maturation of an unconventional myosin. We speculate that chaperone coexpression may be a simple method to optimize the purification of other myosin motors from Sf9 insect cells.
Collapse
|
43
|
Rubenstein PA, Wen KK. Insights into the effects of disease-causing mutations in human actins. Cytoskeleton (Hoboken) 2014; 71:211-29. [PMID: 24574087 DOI: 10.1002/cm.21169] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 02/13/2013] [Accepted: 02/19/2014] [Indexed: 01/04/2023]
Abstract
Mutations in all six actins in humans have now been shown to cause diseases. However, a number of factors have made it difficult to gain insight into how the changes in actin functions brought about by these pathogenic mutations result in the disease phenotype. These include the presence of multiple actins in the same cell, limited accessibility to pure mutant material, and complexities associated with the structures and their component cells that manifest the diseases. To try to circumvent these difficulties, investigators have turned to the use of model systems. This review describes these various approaches, the initial results obtained using them, and the insight they have provided into allosteric mechanisms that govern actin function. Although results so far have not explained a particular disease phenotype at the molecular level, they have provided valuable insight into actin function at the mechanistic level which can be utilized in the future to delineate the molecular bases of these different actinopathies.
Collapse
Affiliation(s)
- Peter A Rubenstein
- Department of Biochemistry, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | | |
Collapse
|
44
|
Rehman AU, Santos-Cortez RLP, Morell RJ, Drummond MC, Ito T, Lee K, Khan AA, Basra MAR, Wasif N, Ayub M, Ali RA, Raza SI, Nickerson DA, Shendure J, Bamshad M, Riazuddin S, Billington N, Khan SN, Friedman PL, Griffith AJ, Ahmad W, Riazuddin S, Leal SM, Friedman TB. Mutations in TBC1D24, a gene associated with epilepsy, also cause nonsyndromic deafness DFNB86. Am J Hum Genet 2014; 94:144-52. [PMID: 24387994 DOI: 10.1016/j.ajhg.2013.12.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/06/2013] [Indexed: 01/12/2023] Open
Abstract
Inherited deafness is clinically and genetically heterogeneous. We recently mapped DFNB86, a locus associated with nonsyndromic deafness, to chromosome 16p. In this study, whole-exome sequencing was performed with genomic DNA from affected individuals from three large consanguineous families in which markers linked to DFNB86 segregate with profound deafness. Analyses of these data revealed homozygous mutation c.208G>T (p.Asp70Tyr) or c.878G>C (p.Arg293Pro) in TBC1D24 as the underlying cause of deafness in the three families. Sanger sequence analysis of TBC1D24 in an additional large family in which deafness segregates with DFNB86 identified the c.208G>T (p.Asp70Tyr) substitution. These mutations affect TBC1D24 amino acid residues that are conserved in orthologs ranging from fruit fly to human. Neither variant was observed in databases of single-nucleotide variants or in 634 chromosomes from ethnically matched control subjects. TBC1D24 in the mouse inner ear was immunolocalized predominantly to spiral ganglion neurons, indicating that DFNB86 deafness might be an auditory neuropathy spectrum disorder. Previously, six recessive mutations in TBC1D24 were reported to cause seizures (hearing loss was not reported) ranging in severity from epilepsy with otherwise normal development to epileptic encephalopathy resulting in childhood death. Two of our four families in which deafness segregates with mutant alleles of TBC1D24 were available for neurological examination. Cosegregation of epilepsy and deafness was not observed in these two families. Although the causal relationship between genotype and phenotype is not presently understood, our findings, combined with published data, indicate that recessive alleles of TBC1D24 can cause either epilepsy or nonsyndromic deafness.
Collapse
Affiliation(s)
- Atteeq U Rehman
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Rockville, MD 20850, USA
| | - Regie Lyn P Santos-Cortez
- Center for Statistical Genetics, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Robert J Morell
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Rockville, MD 20850, USA
| | - Meghan C Drummond
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Rockville, MD 20850, USA
| | - Taku Ito
- Otolaryngology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Rockville, MD 20850, USA
| | - Kwanghyuk Lee
- Center for Statistical Genetics, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Asma A Khan
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore 54500, Pakistan
| | - Muhammad Asim R Basra
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore 54500, Pakistan
| | - Naveed Wasif
- Center for Research in Molecular Medicine, Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore 54000, Pakistan
| | - Muhammad Ayub
- Institute of Biochemistry, University of Baluchistan, Quetta 87300, Pakistan
| | - Rana A Ali
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore 54500, Pakistan
| | - Syed I Raza
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad 45320, Pakistan
| | - Deborah A Nickerson
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Michael Bamshad
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Saima Riazuddin
- Division of Pediatric Otolaryngology - Head and Neck Surgery, Cincinnati Children's Research Foundation, Cincinnati, OH 45229 USA; Department of Otolaryngology - Head and Neck Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Neil Billington
- Laboratory of Molecular Physiology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shaheen N Khan
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore 54500, Pakistan
| | | | - Andrew J Griffith
- Otolaryngology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Rockville, MD 20850, USA
| | - Wasim Ahmad
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad 45320, Pakistan
| | - Sheikh Riazuddin
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore 54500, Pakistan; Allama Iqbal Medical College and Jinnah Hospital Complex, University of Health Sciences, Lahore 54550, Pakistan
| | - Suzanne M Leal
- Center for Statistical Genetics, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Thomas B Friedman
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Rockville, MD 20850, USA.
| |
Collapse
|
45
|
LI ZHE, XU YAMING, ZHANG CAN, LIU XIN, JIANG LI, CHEN FUXUE. Mammalian diaphanous-related formin 1 is required for motility and invadopodia formation in human U87 glioblastoma cells. Int J Mol Med 2013; 33:383-91. [DOI: 10.3892/ijmm.2013.1577] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 11/13/2013] [Indexed: 11/06/2022] Open
|
46
|
An alteration in ELMOD3, an Arl2 GTPase-activating protein, is associated with hearing impairment in humans. PLoS Genet 2013; 9:e1003774. [PMID: 24039609 PMCID: PMC3764207 DOI: 10.1371/journal.pgen.1003774] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 07/24/2013] [Indexed: 12/30/2022] Open
Abstract
Exome sequencing coupled with homozygosity mapping was used to identify a transition mutation (c.794T>C; p.Leu265Ser) in ELMOD3 at the DFNB88 locus that is associated with nonsyndromic deafness in a large Pakistani family, PKDF468. The affected individuals of this family exhibited pre-lingual, severe-to-profound degrees of mixed hearing loss. ELMOD3 belongs to the engulfment and cell motility (ELMO) family, which consists of six paralogs in mammals. Several members of the ELMO family have been shown to regulate a subset of GTPases within the Ras superfamily. However, ELMOD3 is a largely uncharacterized protein that has no previously known biochemical activities. We found that in rodents, within the sensory epithelia of the inner ear, ELMOD3 appears most pronounced in the stereocilia of cochlear hair cells. Fluorescently tagged ELMOD3 co-localized with the actin cytoskeleton in MDCK cells and actin-based microvilli of LLC-PK1-CL4 epithelial cells. The p.Leu265Ser mutation in the ELMO domain impaired each of these activities. Super-resolution imaging revealed instances of close association of ELMOD3 with actin at the plasma membrane of MDCK cells. Furthermore, recombinant human GST-ELMOD3 exhibited GTPase activating protein (GAP) activity against the Arl2 GTPase, which was completely abolished by the p.Leu265Ser mutation. Collectively, our data provide the first insights into the expression and biochemical properties of ELMOD3 and highlight its functional links to sound perception and actin cytoskeleton. Autosomal recessive nonsyndromic hearing loss is a genetically heterogeneous disorder. Here, we report a severe-to-profound mixed hearing loss locus, DFNB88 on chromosome 2p12-p11.2. Exome enrichment followed by massive parallel sequencing revealed a c.794T>C transition mutation in ELMOD3 that segregated with DFNB88-associated hearing loss in a large Pakistani family. This transition mutation is predicted to substitute a highly invariant leucine residue with serine (p.Leu265Ser) in the engulfment and cell motility (ELMO) domain of the protein. No biological activity has been described previously for the ELMOD3 protein. We investigated the biochemical properties and ELMOD3 expression to gain mechanistic insights into the function of ELMOD3 in the inner ear. In rodent inner ears, ELMOD3 immunoreactivity was observed in the cochlear and vestibular hair cells and supporting cells. However, ELMOD3 appears most pronounced in the stereocilia of cochlear hair cells. Ex vivo, ELMOD3 is associated with actin-based structures, and this link is impaired by the DFNB88 mutation. ELMOD3 exhibited GAP activity against Arl2, a small GTPase, providing a potential functional link between Arf family signaling and stereocilia actin-based cytoskeletal architecture. Our study provides new insights into the molecules that are necessary for the development and/or function of inner ear sensory cells.
Collapse
|
47
|
Progressive hearing loss and gradual deterioration of sensory hair bundles in the ears of mice lacking the actin-binding protein Eps8L2. Proc Natl Acad Sci U S A 2013; 110:13898-903. [PMID: 23918390 DOI: 10.1073/pnas.1304644110] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mechanotransduction in the mammalian auditory system depends on mechanosensitive channels in the hair bundles that project from the apical surface of the sensory hair cells. Individual stereocilia within each bundle contain a core of tightly packed actin filaments, whose length is dynamically regulated during development and in the adult. We show that the actin-binding protein epidermal growth factor receptor pathway substrate 8 (Eps8)L2, a member of the Eps8-like protein family, is a newly identified hair bundle protein that is localized at the tips of stereocilia of both cochlear and vestibular hair cells. It has a spatiotemporal expression pattern that complements that of Eps8. In the cochlea, whereas Eps8 is essential for the initial elongation of stereocilia, Eps8L2 is required for their maintenance in adult hair cells. In the absence of both proteins, the ordered staircase structure of the hair bundle in the cochlea decays. In contrast to the early profound hearing loss associated with an absence of Eps8, Eps8L2 null-mutant mice exhibit a late-onset, progressive hearing loss that is directly linked to a gradual deterioration in hair bundle morphology. We conclude that Eps8L2 is required for the long-term maintenance of the staircase structure and mechanosensory function of auditory hair bundles. It complements the developmental role of Eps8 and is a candidate gene for progressive age-related hearing loss.
Collapse
|
48
|
Gasparini F, Caicci F, Rigon F, Zaniolo G, Burighel P, Manni L. Cytodifferentiation of hair cells during the development of a basal chordate. Hear Res 2013; 304:188-99. [PMID: 23876523 DOI: 10.1016/j.heares.2013.07.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 05/28/2013] [Accepted: 07/11/2013] [Indexed: 01/10/2023]
Abstract
Tunicates are unique animals for studying the origin and evolution of vertebrates because they are considered vertebrates' closest living relatives and share the vertebrate body plan and many specific features. Both possess neural placodes, transient thickenings of the cranial ectoderm that give rise to various types of sensory cells, including axonless secondary mechanoreceptors. In vertebrates, these are represented by the hair cells of the inner ear and the lateral line, which have an apical apparatus typically bearing cilia and stereovilli. In tunicates, they are found in the coronal organ, which is a mechanoreceptor located at the base of the oral siphon along the border of the velum and tentacles and is formed of cells bearing a row of cilia and short microvilli. The coronal organ represents the best candidate homolog for the vertebrate lateral line. To further understand the evolution of secondary sensory cells, we analysed the development and cytodifferentiation of coronal cells in the tunicate ascidian Ciona intestinalis for the first time. Here, coronal sensory cells can be identified as early as larval metamorphosis, before tentacles form, as cells with short cilia and microvilli. Sensory cells gradually differentiate, acquiring hair cell features with microvilli containing actin and myosin VIIa; in the meantime, the associated supporting cells develop. The coronal organ grows throughout the animal's lifespan, accompanying the growth of the tentacle crown. Anti-phospho Histone H3 immunostaining indicates that both hair cells and supporting cells can proliferate. This finding contributes to the understanding of the evolution of secondary sensory cells, suggesting that both ancestral cell types were able to proliferate and that this property was progressively restricted to supporting cells in vertebrates and definitively lost in mammals.
Collapse
Affiliation(s)
- Fabio Gasparini
- Dipartimento di Biologia, Università di Padova, Via U. Bassi 58/B, I-35121 Padova, Italy
| | | | | | | | | | | |
Collapse
|
49
|
|
50
|
Park G, Gim J, Kim AR, Han KH, Kim HS, Oh SH, Park T, Park WY, Choi BY. Multiphasic analysis of whole exome sequencing data identifies a novel mutation of ACTG1 in a nonsyndromic hearing loss family. BMC Genomics 2013; 14:191. [PMID: 23506231 PMCID: PMC3608096 DOI: 10.1186/1471-2164-14-191] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 03/04/2013] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND The genetic heterogeneity of sensorineural hearing loss is a major hurdle to the efficient discovery of disease-causing genes. We designed a multiphasic analysis of copy number variation (CNV), linkage, and single nucleotide variation (SNV) of whole exome sequencing (WES) data for the efficient discovery of mutations causing nonsyndromic hearing loss (NSHL). RESULTS From WES data, we identified five distinct CNV loci from a NSHL family, but they were not co-segregated among patients. Linkage analysis based on SNVs identified six candidate loci (logarithm of odds [LOD] >1.5). We selected 15 SNVs that co-segregated with NSHL in the family, which were located in six linkage candidate loci. Finally, the novel variant p.M305T in ACTG1 (DFNA20/26) was selected as a disease-causing variant. CONCLUSIONS Here, we present a multiphasic CNV, linkage, and SNV analysis of WES data for the identification of a candidate mutation causing NSHL. Our stepwise, multiphasic approach enabled us to expedite the discovery of disease-causing variants from a large number of patient variants.
Collapse
Affiliation(s)
- Gibeom Park
- Department of Biomedical Sciences, Seoul National University GraduateSchool, Seoul 110-799, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|