1
|
De-la-Torre P, Martínez-García C, Gratias P, Mun M, Santana P, Akyuz N, González W, Indzhykulian AA, Ramírez D. Identification of druggable binding sites and small molecules as modulators of TMC1. Commun Biol 2025; 8:742. [PMID: 40360848 PMCID: PMC12075566 DOI: 10.1038/s42003-025-07943-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 03/17/2025] [Indexed: 05/15/2025] Open
Abstract
Our ability to hear and maintain balance relies on the proper functioning of inner ear sensory hair cells, which translate mechanical stimuli into electrical signals via mechano-electrical transducer (MET) channels, composed of TMC1/2 proteins. However, the therapeutic use of ototoxic drugs, such as aminoglycosides and cisplatin, which can enter hair cells through MET channels, often leads to profound auditory and vestibular dysfunction. To date, our understanding of how small-molecule modulators interact with TMCs remains limited, hampering the discovery of novel drugs. Here, we propose a structure-based drug screening approach, integrating 3D-pharmacophore modeling, molecular dynamics simulations of the TMC1 + CIB2 + TMIE complex, and experimental validation. Our pipeline successfully identified three potential drug-binding sites within the TMC1 pore, phospholipids, and key amino acids involved in the binding of several compounds, as well as FDA-approved drugs that reduced dye uptake in cultured cochlear explants. Our pipeline offers a broad application for discovering modulators for mechanosensitive ion channels.
Collapse
Affiliation(s)
- Pedro De-la-Torre
- Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School and Mass Eye and Ear, Boston, MA, USA.
- Facultad de Ciencias Básicas, Universidad del Atlántico, Barranquilla, Colombia.
- Life Sciences Research Center, Universidad Simón Bolívar, Barranquilla, Colombia.
| | - Claudia Martínez-García
- Departamento de Farmacología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Paul Gratias
- Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School and Mass Eye and Ear, Boston, MA, USA
| | - Matthew Mun
- Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School and Mass Eye and Ear, Boston, MA, USA
- Speech and Hearing Bioscience & Technology Program, Division of Medical Sciences, Harvard University, Boston, MA, USA
| | - Paula Santana
- Facultad de Ingeniería, Instituto de Ciencias Aplicadas, Universidad Autónoma de Chile, Santiago, Chile
| | - Nurunisa Akyuz
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Wendy González
- Center for Bioinformatics, Simulations and Modelling (CBSM), University of Talca, Talca, Chile
| | - Artur A Indzhykulian
- Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School and Mass Eye and Ear, Boston, MA, USA.
- Speech and Hearing Bioscience & Technology Program, Division of Medical Sciences, Harvard University, Boston, MA, USA.
| | - David Ramírez
- Departamento de Farmacología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.
| |
Collapse
|
2
|
Duan Y, Yao Y, Yang H, Yu Q, Yang W, Liang M, Xie L, Ni L, Yu Y, Zhang B. Decreased expressions of calcium- and integrin-binding protein 4 in the testes of patients with azoosperma and in the ejaculated spermatozoa of patients with abnormal semen parameters. Eur J Obstet Gynecol Reprod Biol 2025; 311:114045. [PMID: 40398143 DOI: 10.1016/j.ejogrb.2025.114045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 04/16/2025] [Accepted: 05/12/2025] [Indexed: 05/23/2025]
Abstract
OBJECTIVE Oligoasthenozoospermia (OAZS), asthenozoospermia (AZS), and azoospermia are the major causes of male infertility with no effective therapeutic treatment. To determine the expression and distribution of calcium- and integrin-binding protein 4 (CIB4) in the patients with azoospermia and OAZS, it is important to explore novel targets and treatment strategies for patients with abnormal semen parameters. DESIGN A small piece of testicular tissue was obtained by multi-site testicular biopsy from each of a total of 16 patients with azoospermia revealed in two groups, i.e., nonobstructive azoospermia (NOA; n = 8) and obstructive azoospermia (OA; n = 8). Sperm samples were collected from 24 infertile men categorized in two groups, OAZS (n = 11) and AZS (n = 13). The control group contained a total of 20 fertile men. Computer-assisted semen analysis (CASA) was performed to assess the sperm motility. Quantitative real-time PCR (RT-qPCR) and Western blot analyses were performed to examine the expression of CIB4. RESULTS The results of RT-qPCR and Western blotting analyses revealed the decreased expression levels of CIB4 mRNA and CIB4 protein in both the sperm of the OAZS patients and the testis tissue of NOA patients. The results of linear regression analysis of CIB4 mRNA expression with the clinical features of participants showed that the CIB4 mRNA level was positively correlated with sperm progressive motility (r = 0.397, P < 0.01) and sperm viability (r = 0.364, P < 0.05). CONCLUSIONS We conclude that decreased expression level of CIB4 in human testis and sperm, may be a contributor or a downstream indicator for some cases of azoospermia, especially OA and OAZS. The high expression level of CIB4 mRNA in patients with normal semen parameters suggests that CIB4 in human-ejaculated spermatozoa would potentially contribute to the diagnosis and treatment of male infertility caused by deficient sperm motility and count. These results provide novel insights into the molecular mechanisms underlying the male infertility and the clinical importance of CIB4 in the treatment of infertility.
Collapse
Affiliation(s)
- Yuyao Duan
- Reproductive Medical Center, The Second Hospital Affiliated to Shandong University of Traditional Chinese Medicine, Jinan 250001 Shandong, China
| | - Yi Yao
- Reproductive Medical Center, The Second Hospital Affiliated to Shandong University of Traditional Chinese Medicine, Jinan 250001 Shandong, China
| | - Huailiang Yang
- Reproductive Medical Center, The Second Hospital Affiliated to Shandong University of Traditional Chinese Medicine, Jinan 250001 Shandong, China
| | - Qing Yu
- Reproductive Medical Center, The Second Hospital Affiliated to Shandong University of Traditional Chinese Medicine, Jinan 250001 Shandong, China
| | - Weina Yang
- Reproductive Medical Center, The Second Hospital Affiliated to Shandong University of Traditional Chinese Medicine, Jinan 250001 Shandong, China
| | - Ming Liang
- Reproductive Medical Center, The Second Hospital Affiliated to Shandong University of Traditional Chinese Medicine, Jinan 250001 Shandong, China
| | - Linlin Xie
- Reproductive Medical Center, The Second Hospital Affiliated to Shandong University of Traditional Chinese Medicine, Jinan 250001 Shandong, China
| | - Linlin Ni
- Reproductive Medical Center, The Second Hospital Affiliated to Shandong University of Traditional Chinese Medicine, Jinan 250001 Shandong, China
| | - Yan Yu
- Reproductive Medical Center, The Second Hospital Affiliated to Shandong University of Traditional Chinese Medicine, Jinan 250001 Shandong, China.
| | - Bin Zhang
- Reproductive Medical Center, The Second Hospital Affiliated to Shandong University of Traditional Chinese Medicine, Jinan 250001 Shandong, China.
| |
Collapse
|
3
|
Deng S, Yan Z. TMC1 and TMC2 function as the mechano-electrical transduction ion channel in hearing. Curr Opin Neurobiol 2025; 93:103026. [PMID: 40280017 DOI: 10.1016/j.conb.2025.103026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/29/2025]
Abstract
Mechanotransduction within the specialized cochlea hair cells is fundamental to perceiving sound waves. This intricate mechanism converts mechanical vibrations into electrical signals that the brain can interpret as sound. The molecular identity of the mechanoelectrical transducer continues to be a subject of intense debate. Transmembrane channel-like protein 1 (TMC1) was initially recognized as a deafness gene in humans, with subsequent studies revealing the hearing loss phenotype in Tmc1 mutant mice. Mechanotransduction currents were lost in the hair cells of Tmc1;Tmc2 double knockout mice, indicating the involvement of TMC1/2 in auditory mechanotransduction. Both TMC1/2 are expressed at the tip of stereocilia in hair cells, the subcellular site of auditory mechanotransduction. Notably, recent in vitro studies have overcome long-standing technical barriers that TMC1/2 are not localized to the cell membrane in heterologous expression and provided compelling evidence that TMC1/2 are mechanically gated ion channels, finally fulfilling both the essential and necessary criteria they must meet as sensory transducers. In hair cells, tip-links possibly relay force to TMC1/2 by tether gating or membrane-tension gating, while the molecular mechanisms underlying each gating mechanism require further investigation.
Collapse
Affiliation(s)
- Siqi Deng
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China; Department of Life Sciences, Hong Kong University of Science and Technology, Hong Kong, China
| | - Zhiqiang Yan
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China; Department of Neurobiology, School of Basic Medicine, Capital Medical University, Beijing, China; Institute for Medical Physiology, Chinese Institutes for Medical Research, Beijing, China.
| |
Collapse
|
4
|
Thomas HB, Demain LAM, Cabrera-Orefice A, Schrauwen I, Shamseldin HE, Rea A, Bharadwaj T, Smith TB, Oláhová M, Thompson K, He L, Kaur N, Shukla A, Abukhalid M, Ansar M, Rehman S, Riazuddin S, Abdulwahab F, Smith JM, Stark Z, Mancilar H, Tumer S, Esen FN, Uctepe E, Topcu V, Yesilyurt A, Afzal E, Salari M, Carroll C, Zifarelli G, Bauer P, Kor D, Bulut FD, Houlden H, Maroofian R, Carrera S, Yue WW, Munro KJ, Alkuraya FS, Jamieson P, Ahmed ZM, Leal SM, Taylor RW, Wittig I, O'Keefe RT, Newman WG. Bi-allelic variants in MRPL49 cause variable clinical presentations, including sensorineural hearing loss, leukodystrophy, and ovarian insufficiency. Am J Hum Genet 2025; 112:952-962. [PMID: 40043708 DOI: 10.1016/j.ajhg.2025.02.005] [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: 10/09/2024] [Revised: 02/06/2025] [Accepted: 02/06/2025] [Indexed: 03/12/2025] Open
Abstract
Combined oxidative phosphorylation deficiency (COXPD) is a rare multisystem disorder that is clinically and genetically heterogeneous. Genome sequencing identified bi-allelic MRPL49 variants in individuals from nine unrelated families with presentations ranging from Perrault syndrome (primary ovarian insufficiency and sensorineural hearing loss) to severe childhood onset of leukodystrophy, learning disability, microcephaly, and retinal dystrophy. Complexome profiling of fibroblasts from affected individuals revealed reduced levels of the small mitochondrial ribosomal subunits and a more pronounced reduction of the large mitochondrial ribosomal subunits. There was no evidence of altered mitoribosomal assembly. The reductions in levels of oxidative phosphorylation (OXPHOS) enzyme complexes I and IV are consistent with a form of COXPD associated with bi-allelic MRPL49 variants, expanding the understanding of how disruption of the mitochondrial ribosomal large subunit results in multisystem phenotypes.
Collapse
Affiliation(s)
- Huw B Thomas
- Division of Evolution, Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester M13 9PL, UK; Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK
| | - Leigh A M Demain
- Division of Evolution, Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester M13 9PL, UK; Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK
| | - Alfredo Cabrera-Orefice
- Centre for Functional Proteomics, Institute for Cardiovascular Physiology, Medical Faculty, Goethe University, 60596 Frankfurt am Main, Germany; Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, 35392 Giessen, Germany
| | - Isabelle Schrauwen
- Department of Translational Neurosciences, University of Arizona College of Medicine Phoenix, Phoenix, AZ, USA
| | - Hanan E Shamseldin
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Alessandro Rea
- Division of Evolution, Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester M13 9PL, UK; Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK
| | - Thashi Bharadwaj
- Center for Statistical Genetics, Department of Neurology, Gertrude H. Sergievsky Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Thomas B Smith
- Division of Evolution, Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester M13 9PL, UK; Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK
| | - Monika Oláhová
- Department of Applied Sciences, Faculty of Health & Life Sciences, Northumbria University, Newcastle upon Tyne, UK; Mitochondrial Research Group, Clinical and Translational Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Kyle Thompson
- Mitochondrial Research Group, Clinical and Translational Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Langping He
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
| | - Namanpreet Kaur
- Department of Medical Genetics, Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Anju Shukla
- Department of Medical Genetics, Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Musaad Abukhalid
- Department of Neuroscience, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Muhammad Ansar
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad 45320, Pakistan
| | - Sakina Rehman
- Department of Otorhinolaryngology - Head & Neck Surgery, School of Medicine University of Maryland, Baltimore, MD, USA; Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Saima Riazuddin
- Department of Otorhinolaryngology - Head & Neck Surgery, School of Medicine University of Maryland, Baltimore, MD, USA; Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Firdous Abdulwahab
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Janine M Smith
- Specialty of Genomic Medicine, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2000, Australia; Western Sydney Genetics Program, Department of Clinical Genetics, Sydney Children's Hospitals Network, Westmead, NSW 2145, Australia
| | - Zornitza Stark
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Flemington Road, Melbourne, VIC, Australia; University of Melbourne, Melbourne, VIC, Australia
| | | | - Sait Tumer
- Acıbadem Labgen Genetic Diagnosis Center, Istanbul, Türkiye
| | - Fatma N Esen
- Acıbadem Labgen Genetic Diagnosis Center, Istanbul, Türkiye
| | - Eyyup Uctepe
- Acıbadem Labgen Genetic Diagnosis Center, Istanbul, Türkiye
| | - Vehap Topcu
- Acıbadem Labgen Genetic Diagnosis Center, Istanbul, Türkiye
| | | | - Erum Afzal
- Department of Development Pediatrics, The Children's Hospital and The Institute of Child Health, Multan, Pakistan
| | - Mehri Salari
- Department of Neurology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Christopher Carroll
- Genetics Section, Molecular and Clinical Sciences Research Institute, St. George's, University of London, London, UK
| | | | - Peter Bauer
- CENTOGENE GmbH, Am Strande 7, 18055 Rostock, Germany
| | - Deniz Kor
- Cukurova University, Medical Faculty, Department of Pediatric Metabolism and Nutrition, Adana, Turkey
| | - Fatma D Bulut
- Cukurova University, Medical Faculty, Department of Pediatric Metabolism and Nutrition, Adana, Turkey
| | - Henry Houlden
- Department of Neuromuscular Diseases, University College London, Queen Square, Institute of Neurology, London WC1N 3BG, UK
| | - Reza Maroofian
- Department of Neuromuscular Diseases, University College London, Queen Square, Institute of Neurology, London WC1N 3BG, UK
| | - Samantha Carrera
- Genome Editing Unit Core Facility, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Wyatt W Yue
- Newcastle University Biosciences Institute, Medical School, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Kevin J Munro
- Manchester Centre for Audiology and Deafness (ManCAD), School of Health Sciences, University of Manchester, Manchester, UK
| | - Fowzan S Alkuraya
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia; Department of Anatomy and Cell Biology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Peter Jamieson
- Department of Radiology, Manchester University Hospital NHS Foundation Trust, Manchester M13 9PW, UK
| | - Zubair M Ahmed
- Department of Otorhinolaryngology - Head & Neck Surgery, School of Medicine University of Maryland, Baltimore, MD, USA; Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Suzanne M Leal
- Center for Statistical Genetics, Department of Neurology, Gertrude H. Sergievsky Center, Columbia University Medical Center, New York, NY 10032, USA; Taub Institute for Alzheimer's Disease and the Aging Brain, and the Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Robert W Taylor
- Mitochondrial Research Group, Clinical and Translational Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
| | - Ilka Wittig
- Centre for Functional Proteomics, Institute for Cardiovascular Physiology, Medical Faculty, Goethe University, 60596 Frankfurt am Main, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, 60596 Frankfurt am Main, Germany
| | - Raymond T O'Keefe
- Division of Evolution, Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester M13 9PL, UK; Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK.
| | - William G Newman
- Division of Evolution, Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester M13 9PL, UK; Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK.
| |
Collapse
|
5
|
Li S, Park J, Phan TM, Egelman EH, Bird JE, Shin JB. Tonotopic Specialization of MYO7A Isoforms in Auditory Hair Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.01.646665. [PMID: 40236041 PMCID: PMC11996455 DOI: 10.1101/2025.04.01.646665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
1. Mutations in Myo7a cause Usher syndrome type 1B and non-syndromic deafness, but the precise function of MYO7A in sensory hair cells remains unclear. We identify and characterize a novel isoform, MYO7A-N, expressed in auditory hair cells alongside the canonical MYO7A-C. Isoform-specific knock-in mice reveal that inner hair cells primarily express MYO7A-C, while outer hair cells express both isoforms in opposing tonotopic gradients. Both localize to the upper tip-link insertion site, consistent with a role in the tip link for mechanotransduction. Loss of MYO7A-N leads to outer hair cell degeneration and progressive hearing loss. Cryo-EM structures reveal isoform-specific differences at actomyosin interfaces, correlating with distinct ATPase activities. These findings reveal an unexpected layer of molecular diversity within the mechanotransduction machinery. We propose that MYO7A isoform specialization enables fine-tuning of tip-link tension, thus hearing sensitivity, and contributes to the frequency-resolving power of the cochlea.
Collapse
|
6
|
Zhang H, Qiu X, Mittelstadt J, Müller U. Ankyrins are dispensable for mechanotransduction by cochlear hair cells. Hear Res 2025; 459:109224. [PMID: 40024092 PMCID: PMC11934226 DOI: 10.1016/j.heares.2025.109224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/07/2025] [Accepted: 02/18/2025] [Indexed: 03/04/2025]
Abstract
The mechanotransduction (MET) channels of cochlear hair cells is a heteromeric protein complex consisting of TMC1, TMIE and CIB2. The activity of this ion channel is thought to be regulated by a gating spring, a mechanical element that conveys sound-induced vibrations to the MET channel. In nematodes, orthologs of TMC-1, TMIE and CIB2 similarly assemble into a MET channel mediating light nose-touch. Studies in nematodes have suggested that nematode Unc-44, an ortholog of the mammalian ankyrins Ank1, 2, and 3, encodes a gating spring that tethers the nematode MET channel to the cytoskeleton. Here we show that mammalian ankyrins are expressed in cochlear hair cells. Using single and triple conditional knockout mice, we demonstrate that Ank1, 2, and 3 are dispensable for the function of cochlear hair cells. We concluded that Ank1, 2, and 3 are unlikely to be components of the gating spring that gates mechanotransduction channels in mammalian cochlear hair cells.
Collapse
Affiliation(s)
- Hong Zhang
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Meicine, Baltimore, MD 21205, USA
| | - Xufeng Qiu
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Meicine, Baltimore, MD 21205, USA
| | - Jonah Mittelstadt
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Meicine, Baltimore, MD 21205, USA
| | - Ulrich Müller
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Meicine, Baltimore, MD 21205, USA.
| |
Collapse
|
7
|
Yao X, Zhang Y, Hong X, Xing Y, Xu Z. Esrp1 and Esrp2 regulate the stability of tmc1/ 2a mRNAs in zebrafish sensory hair cells. J Neurosci 2025; 45:e0837242025. [PMID: 40086870 PMCID: PMC12019119 DOI: 10.1523/jneurosci.0837-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025] Open
Abstract
RNA-binding proteins (RBPs) are important for post-transcriptional RNA processing, including pre-mRNA alternative splicing, mRNA stability, and translation. Several RBPs have been shown to play pivotal roles in the inner ear, whose dysfunction leads to auditory and/or balance impairments. Epithelial splicing-regulatory protein 1 (ESRP1) regulates alternative splicing and mRNA stability, and mutations in ESRP1 gene have been associated with sensorineural hearing loss in humans. In Esrp1 knockout mouse embryos, alternative splicing of its target genes such as Fgfr2 is impaired, which eventually result in cochlear development deficits. However, Esrp1 knockout mice die soon after birth because of complications from cleft-lip and palate defects, impeding further investigations at later postnatal ages. In the present study, we explored the role of ESRP1 in hearing using zebrafish as a model. We showed that esrp1 and its paralog esrp2 are expressed in the inner ear and certain anterior lateral line (ALL) neuromasts. Furthermore, our data suggested that Esrp1 and Esrp2 are required for the mechano-electrical transduction (MET) function of hair cells. RNA sequencing results indicated a significant decrease in the levels of several mRNAs in esrp1/2 double knockout larvae. Among the dysregulated genes are tmc1 and tmc2a, which encode essential subunits of the MET complex. Further investigations demonstrated that Esrp1/2 could directly bind to tmc1 and tmc2a mRNAs and affect their stability. Taken together, we showed here that Esrp1 and Esrp2 regulate the MET function of zebrafish sensory hair cells by modulating the stability of tmc1 and tmc2a mRNAs.Significance statement ESRP1 is an important RNA-binding protein, whose malfunction has been associated with hearing loss in humans. Esrp1 knockout affects alternative splicing of its target mRNAs such as Fgfr2, eventually leading to cochlear development deficits in mice. However, Esrp1 knockout mice die soon after birth, precluding further investigations at later postnatal ages. In this study, we explored the role of ESRP1 in hearing using zebrafish as a model. Our results demonstrated that esrp1 and its paralog esrp2 are expressed in the zebrafish inner ear, and that esrp1/esrp2 double knockout compromised the mechano-electrical transduction (MET) function of hair cells. Additionally, we successfully identified tmc1 and tmc2a mRNAs as the targets of Esrp1/2, which encode essential subunits of the MET complex.
Collapse
Affiliation(s)
- Xuebo Yao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
| | - Yan Zhang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
- Department of Biochemistry and Department of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Xiaying Hong
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
| | - Yanyi Xing
- Women's Hospital, Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Zhigang Xu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
- Shandong Provincial Collaborative Innovation Center of Cell Biology, Shandong Normal University, Jinan, Shandong 250014, China
| |
Collapse
|
8
|
Lv Z, Sun L, Chen X, Guo P, Xie X, Yao X, Tian S, Wang C, Shao Y, Liu J. TMC7 is required for spermiogenesis and male fertility by regulating TGN-derived vesicles. Int J Biol Macromol 2025; 293:139070. [PMID: 39732242 DOI: 10.1016/j.ijbiomac.2024.139070] [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: 07/01/2024] [Revised: 08/05/2024] [Accepted: 12/19/2024] [Indexed: 12/30/2024]
Abstract
Infertility affects 10-12 % of couples worldwide, 50 % of which are male. Abnormal spermatogenesis is among the main causes of male infertility. We were curious about the possible role of transmembrane channel-like protein 7 (TMC7) in spermatogenesis because of its aberrant expression in several male infertility patients. In this study, we found that deletion of Tmc7, which is highly expressed during spermiogenesis, causes a human oligoasthenoteratozoospermia (OAT)-like phenotype in male mice. By histological analysis, TEM, RNA-seq and library-free data-independent acquisition mass spectrometry (DIA-MS) of TMC7-null mouse testes, we found that Tmc7 deletion caused abnormal swelling of trans-Golgi network (TGN) vesicles in elongated spermatids. Further immunofluorescence localization analysis revealed that these vesicles were defined by synaptophysin-like 1 (SYPL1). In addition, TMC7 may act as a potential chloride transport channel to regulate the size of transport vesicles. In conclusion, this study demonstrated that TMC7 is essential for male fertility and may be used as a potential protein for the identification and recognition of OAT. On the other hand, TMC7 may be a potential male contraceptive target.
Collapse
Affiliation(s)
- Zheng Lv
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Longjie Sun
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Xuexue Chen
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Peilan Guo
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Xiaomei Xie
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Xiaohong Yao
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Shuang Tian
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Chaofan Wang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Yujing Shao
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Jiali Liu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
9
|
Giese APJ, Parker A, Rehman S, Brown SDM, Riazuddin S, Vander Kooi CW, Bowl MR, Ahmed ZM. CIB2 function is distinct from that of whirlin in the organization of sterocilia architecture. Dis Model Mech 2025; 18:dmm052043. [PMID: 40083274 PMCID: PMC11992350 DOI: 10.1242/dmm.052043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 03/07/2025] [Indexed: 03/16/2025] Open
Abstract
Humans and mice with mutations in genes encoding CIB2 and whirlin (WHRN) are deaf. We previously reported that CIB2 binds to WHRN and is essential for stereocilia staircase architecture of cochlear hair cells. Here, we refine the interaction domains of both proteins and show that these proteins play unique roles in stereocilia bundle formation and organization. We found that the EF2 domain of CIB2 binds to the HHD2 region of WHRN. AlphaFold2 multimer independently identified the same interacting regions and gave a thorough structural model. Next, we investigated genetic interaction between murine Cib2 and Whrn. Hearing in mice double heterozygous for functionally null alleles (Cib2KO/+;Whrnwi/+) was similar to that in age-matched wild-type mice, indicating that partial deficiency for both Cib2 and Whrn does not impair hearing. Double homozygous mutant mice (Cib2KO/KO;Whrnwi/wi) were deaf, and their cochlear stereocilia exhibited a predominant phenotype seen in single Whrnwi/wi mutants. Overexpression of WHRN in Cib2KO/KO mice did not rescue the stereocilia morphology. These data suggest that CIB2 is multifunctional, with key independent functions in the development and/or maintenance of the stereocilia staircase pattern in auditory hair cells.
Collapse
Affiliation(s)
- Arnaud P. J. Giese
- Department of Otorhinolaryngology – Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Andrew Parker
- MRC Harwell Institute, Mammalian Genetics Unit, Harwell Campus, Oxfordshire OX11 0RD, UK
| | - Sakina Rehman
- Department of Otorhinolaryngology – Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Steve D. M. Brown
- MRC Harwell Institute, Mammalian Genetics Unit, Harwell Campus, Oxfordshire OX11 0RD, UK
| | - Saima Riazuddin
- Department of Otorhinolaryngology – Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21205, USA
| | - Craig W. Vander Kooi
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32603, USA
| | - Michael R. Bowl
- MRC Harwell Institute, Mammalian Genetics Unit, Harwell Campus, Oxfordshire OX11 0RD, UK
- UCL Ear Institute, University College London, London WC1X 8EE, UK
| | - Zubair M. Ahmed
- Department of Otorhinolaryngology – Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21205, USA
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Program in Neuroscience and Cognitive Science, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
10
|
Wang X, Yi Z, Shi M, Sun Y. The Diverse Functions of the Calcium- and Integrin-Binding Protein Family. Int J Mol Sci 2025; 26:2223. [PMID: 40076845 PMCID: PMC11900603 DOI: 10.3390/ijms26052223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
The calcium- and integrin-binding protein (CIB) family, comprising four evolutionarily conserved members (CIB1, CIB2, CIB3, and CIB4), is characterized by canonical EF-hand motifs. The functions of CIBs in the inner ear have been investigated, although further research is still necessary to gain a comprehensive understanding of them. Among the CIB family members, CIB2 is essential for auditory function. CIB3 and CIB2 jointly participate in the regulation of balance. Beyond their sensory roles, CIBs exhibit multifunctionality through calcium-dependent interactions with diverse molecular partners, contributing to the pathogenesis of various conditions, including neurological disorders, cardiovascular diseases, cancer, and male infertility. In this review, we discuss the conserved structure of the CIB family, highlighting its contributions to various biological functions. We also summarize the distribution and function of the CIB family, emphasizing the pivotal roles of CIB2 and CIB3 in hearing and balance.
Collapse
Affiliation(s)
- Xiaoying Wang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhangyi Yi
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Mengwen Shi
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yu Sun
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Institute of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinic Research Center for Deafness and Vertigo, Wuhan 430022, China
| |
Collapse
|
11
|
Li Y, Chen J, Jiang W, Ye J, Zhang X, Wang C. Structural insights into calcium-dependent CIB2-TMC1 interaction in hair cell mechanotransduction. Commun Biol 2025; 8:306. [PMID: 40000792 PMCID: PMC11861898 DOI: 10.1038/s42003-025-07761-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Calcium- and integrin-binding protein 2 (CIB2) plays a crucial role in mechanoelectrical transduction (MET) in cochlear hair cells, particularly in modulating the function and localization of the core components of MET channels TMC1/2. CIB2, along with its homolog CIB3, interacts with TMC1/2 through two distinct sites. Here, our study unveils CIB2/3's role as a calcium sensor in its interaction with TMC1. Utilizing X-ray crystallography, we elucidate the high-resolution structure of the mammalian CIB2-TMC1 complex. Structural analyses reveal that cation-bound CIB2 forms a negatively charged surface that aligns with a positively charged surface on the TMC1 N-terminus. Moreover, our data suggest that Ca²⁺ modulates CIB2's interaction with both the N-terminal domain and the loop 1 region of TMC1, and that Ca²⁺-bound CIB2 is capable of simultaneously binding to both regions of TMC1. Critically, we examine pathogenic variants of CIB2 associated with hearing loss, discovering that these variants have differential impacts on CIB2's interactions with TMC1's dual binding sites, displaying diminished calcium-binding affinities for several of these CIB2 mutations. These findings provide a deeper understanding of the molecular mechanisms underlying CIB2 function and its implications in hearing loss, offering potential avenues for therapeutic interventions in deafness.
Collapse
Affiliation(s)
- Yahong Li
- Department of Neurology, the First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jiasheng Chen
- Department of Neurology, the First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wenli Jiang
- Department of Neurology, the First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jin Ye
- Department of Neurology, the First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xuan Zhang
- Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China
| | - Chao Wang
- Department of Neurology, the First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Ministry of Education Key Laboratory for Membrane-less Organelles and Cellular Dynamics, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
12
|
Li J, Oh SI, Liu C, Zhao B. Inhibition of GABARAP or GABARAPL1 prevents aminoglycoside- induced hearing loss. Proc Natl Acad Sci U S A 2025; 122:e2416453122. [PMID: 39928869 PMCID: PMC11848329 DOI: 10.1073/pnas.2416453122] [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: 08/13/2024] [Accepted: 01/06/2025] [Indexed: 02/12/2025] Open
Abstract
Aminoglycosides (AGs) are highly potent, broad-spectrum antibiotics frequently used as first-line treatments for multiple life-threatening infections. Despite their severe ototoxicity, causing irreversible hearing loss in millions of people annually, no preventive therapy has been approved. We previously reported that GABARAP and several other central autophagy proteins are essential for AG-induced hearing loss. This finding opens avenues for the rational design and development of inhibitors that selectively target proteins in this pathway, thereby mitigating AG ototoxicity. In this study, we generated a mouse model with a targeted deletion of GABARAPL1, a homolog of GABARAP, and another model deficient in both GABARAP and GABARAPL1. We found that normal hearing is unaffected by the depletion of these proteins. Remarkably, both proteins are essential for AG-induced hearing loss, with GABARAP playing a more significant role. To further explore the therapeutic potential, we designed and validated short hairpin RNAs targeting the mouse and human GABARAP gene. By inhibiting GABARAP expression in inner ear hair cells using adeno-associated virus-mediated RNA interference, we successfully prevented AG-induced hair cell death and subsequent hearing loss. Our findings underscore the critical role of GABARAP in AG ototoxicity and highlight its potential as a therapeutic target for preventing AG-induced hearing loss.
Collapse
Affiliation(s)
- Jinan Li
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN46202
| | - Seung-Il Oh
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN46202
| | - Chang Liu
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN46202
| | - Bo Zhao
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN46202
| |
Collapse
|
13
|
Zou W, Fan Y, Liu J, Cheng H, Hong H, Al-Sheikh U, Li S, Zhu L, Li R, He L, Tang YQ, Zhao G, Zhang Y, Wang F, Zhan R, Zheng X, Kang L. Anoctamin-1 is a core component of a mechanosensory anion channel complex in C. elegans. Nat Commun 2025; 16:1680. [PMID: 39956854 PMCID: PMC11830769 DOI: 10.1038/s41467-025-56938-z] [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: 02/01/2024] [Accepted: 02/06/2025] [Indexed: 02/18/2025] Open
Abstract
Mechanotransduction channels are widely expressed in both vertebrates and invertebrates, mediating various physiological processes such as touch, hearing and blood-pressure sensing. While previously known mechanotransduction channels in metazoans are primarily cation-selective, we identified Anoctamin-1 (ANOH-1), the C. elegans homolog of mammalian calcium-activated chloride channel ANO1/TMEM16A, as an essential component of a mechanosensory channel complex that contributes to the nose touch mechanosensation in C. elegans. Ectopic expression of either C. elegans or human Anoctamin-1 confers mechanosensitivity to touch-insensitive neurons, suggesting a cell-autonomous role of ANOH-1/ANO1 in mechanotransduction. Additionally, we demonstrated that the mechanosensory function of ANOH-1/ANO1 relies on CIB (calcium- and integrin- binding) proteins. Thus, our results reveal an evolutionarily conserved chloride channel involved in mechanosensory transduction in metazoans, highlighting the importance of anion channels in mechanosensory processes.
Collapse
Affiliation(s)
- Wenjuan Zou
- Department of Neurosurgery of the First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Zhejiang, China.
- Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.
| | - Yuedan Fan
- Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jia Liu
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hankui Cheng
- Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Huitao Hong
- Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Umar Al-Sheikh
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shitian Li
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Linhui Zhu
- Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Rong Li
- Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Longyuan He
- Department of Neurosurgery of the First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Zhejiang, China
| | - Yi-Quan Tang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, ENT Institute and Otorhinolaryngology, Department of Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, Fudan University, Shanghai, China
| | - Guohua Zhao
- Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongming Zhang
- Department of Ophthalmology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Feng Wang
- Department of Neurosurgery of the First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Zhejiang, China
| | - Renya Zhan
- Department of Neurosurgery of the First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Zhejiang, China
| | - Xiujue Zheng
- Department of Neurosurgery of the First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Zhejiang, China
| | - Lijun Kang
- Department of Neurosurgery of the First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Zhejiang, China.
- Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
14
|
Fu S, Pan X, Lu M, Dong J, Yan Z. Human TMC1 and TMC2 are mechanically gated ion channels. Neuron 2025; 113:411-425.e4. [PMID: 39674179 DOI: 10.1016/j.neuron.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 09/23/2024] [Accepted: 11/14/2024] [Indexed: 12/16/2024]
Abstract
Mammalian transmembrane channel-like proteins 1 and 2 (TMC1 and TMC2) have emerged as very promising candidate mechanotransduction channels in hair cells. However, controversy persists because the heterogeneously expressed TMC1/2 in cultured cells lack evidence of mechanical gating, primarily due to their absence from the plasma membrane. By employing domain swapping with OSCA1.1 and subsequent point mutations, we successfully identified membrane-localized mouse TMC1/2 mutants, demonstrating that they are mechanically gated in heterologous cells. Further, whole-genome CRISPRi screening enabled wild-type human TMC1/2 localization in the plasma membrane, where they responded robustly to poking stimuli. In addition, wild-type human TMC1/2 showed stretch-activated currents and clear single-channel current activities. Deafness-related TMC1 mutations altered the reversal potential of TMC1, indicating that TMC1/2 are pore-forming mechanotransduction channels. In summary, our study provides evidence that human TMC1/2 are pore-forming, mechanically activated ion channels, supporting their roles as mechanotransduction channels in hair cells.
Collapse
Affiliation(s)
- Songdi Fu
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China; Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China
| | - Xueqi Pan
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China; Department of Neurobiology, School of Basic Medicine, Capital Medical University, Beijing, China
| | - Mingshun Lu
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jianying Dong
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China; Department of Neurobiology, School of Basic Medicine, Capital Medical University, Beijing, China
| | - Zhiqiang Yan
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China; Department of Neurobiology, School of Basic Medicine, Capital Medical University, Beijing, China; Institute for Medical Physiology, Chinese Institutes for Medical Research, Beijing, China.
| |
Collapse
|
15
|
Wu S, Lin L, Hu Q, Yao X, Wang H, Liu S, Liu Q, Xi Y, Lin Y, Gong J, Hu R, Zhan W, Luo Y, He G, Liu Z, Xiong W, Wang Q, Xu Z, Bai F, Lu Q. Mechano-electrical transduction components TMC1-CIB2 undergo a Ca 2+-induced conformational change linked to hearing loss. Dev Cell 2025:S1534-5807(25)00004-8. [PMID: 39889697 DOI: 10.1016/j.devcel.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/20/2024] [Accepted: 01/09/2025] [Indexed: 02/03/2025]
Abstract
TMC1, a unique causative gene associated with deafness, exhibits variants with autosomal dominant and recessive inheritance patterns. TMC1 codes for the transmembrane channel-like protein 1 (TMC1), a key component of the mechano-electrical transduction (MET) machinery for hearing. However, the molecular mechanism of Ca2+ regulation in MET remains unclear. Calcium and integrin-binding protein 2 (CIB2), another MET component associated with deafness, can bind with Ca2+. Our study shows that TMC1-CIB2 complex undergoes a Ca2+-induced conformational change. We identified a vertebrate-specific binding site on TMC1 that interacts with apo CIB2, linked with hearing loss. Using an ex vivo mouse organotypic cochlea model, we demonstrated that disruption of the calcium-binding site of CIB2 perturbs the MET channel conductivity. After systematically analyzing the hearing loss variants, we observed dominant mutations of TMC1 cluster around the putative ion pore or at the binding interfaces with CIB2. These findings elucidate the molecular mechanisms underlying TMC1-linked hearing loss.
Collapse
Affiliation(s)
- Shaoxuan Wu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Lin Lin
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Qiaoyu Hu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; Innovation Center for AI and Drug Discovery, School of Pharmacy, East China Normal University, Shanghai 200062, China
| | - Xuebo Yao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
| | - Hongyang Wang
- Senior Department of Otolaryngology, Head and Neck Surgery, Department of Audiology and Vestibular Medicine, Chinese PLA Institute of Otolaryngology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China
| | - Shuang Liu
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Qingling Liu
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Yuehui Xi
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
| | - Yuzhe Lin
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Jianqiao Gong
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ruixing Hu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Wei Zhan
- National Facility for Protein Science in Shanghai, Shanghai Advanced Research Institute, Chinese Academy of Science, Zhangjiang Lab, Shanghai 201210, China
| | - Yi Luo
- Senior Department of Otolaryngology, Head and Neck Surgery, Department of Audiology and Vestibular Medicine, Chinese PLA Institute of Otolaryngology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China
| | - Guang He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Zhijun Liu
- National Facility for Protein Science in Shanghai, Shanghai Advanced Research Institute, Chinese Academy of Science, Zhangjiang Lab, Shanghai 201210, China
| | - Wei Xiong
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Qiuju Wang
- Senior Department of Otolaryngology, Head and Neck Surgery, Department of Audiology and Vestibular Medicine, Chinese PLA Institute of Otolaryngology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China.
| | - Zhigang Xu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China.
| | - Fang Bai
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; School of Information Science and Technology, ShanghaiTech University, Shanghai 201210, China; Shanghai Clinical Research and Trial Center, Shanghai 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| | - Qing Lu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China; Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China.
| |
Collapse
|
16
|
Giese APJ, Weng WH, Kindt KS, Chang HHV, Montgomery JS, Ratzan EM, Beirl AJ, Aponte Rivera R, Lotthammer JM, Walujkar S, Foster MP, Zobeiri OA, Holt JR, Riazuddin S, Cullen KE, Sotomayor M, Ahmed ZM. Complexes of vertebrate TMC1/2 and CIB2/3 proteins form hair-cell mechanotransduction cation channels. eLife 2025; 12:RP89719. [PMID: 39773557 PMCID: PMC11709434 DOI: 10.7554/elife.89719] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Abstract
Calcium and integrin-binding protein 2 (CIB2) and CIB3 bind to transmembrane channel-like 1 (TMC1) and TMC2, the pore-forming subunits of the inner-ear mechano-electrical transduction (MET) apparatus. These interactions have been proposed to be functionally relevant across mechanosensory organs and vertebrate species. Here, we show that both CIB2 and CIB3 can form heteromeric complexes with TMC1 and TMC2 and are integral for MET function in mouse cochlea and vestibular end organs as well as in zebrafish inner ear and lateral line. Our AlphaFold 2 models suggest that vertebrate CIB proteins can simultaneously interact with at least two cytoplasmic domains of TMC1 and TMC2 as validated using nuclear magnetic resonance spectroscopy of TMC1 fragments interacting with CIB2 and CIB3. Molecular dynamics simulations of TMC1/2 complexes with CIB2/3 predict that TMCs are structurally stabilized by CIB proteins to form cation channels. Overall, our work demonstrates that intact CIB2/3 and TMC1/2 complexes are integral to hair-cell MET function in vertebrate mechanosensory epithelia.
Collapse
Affiliation(s)
- Arnaud PJ Giese
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of MedicineBaltimoreUnited States
| | - Wei-Hsiang Weng
- Department of Chemistry and Biochemistry, The Ohio State UniversityColumbusUnited States
- Biophysics Graduate Program, The Ohio State UniversityColumbusUnited States
| | - Katie S Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | | | - Jonathan S Montgomery
- Department of Chemistry and Biochemistry, The Ohio State UniversityColumbusUnited States
- Ohio State Biochemistry Program, The Ohio State UniversityColumbusUnited States
| | - Evan M Ratzan
- Departments of Otolaryngology and Neurology, F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Alisha J Beirl
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Roberto Aponte Rivera
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Jeffrey M Lotthammer
- Department of Chemistry and Biochemistry, The Ohio State UniversityColumbusUnited States
| | - Sanket Walujkar
- Department of Chemistry and Biochemistry, The Ohio State UniversityColumbusUnited States
| | - Mark P Foster
- Department of Chemistry and Biochemistry, The Ohio State UniversityColumbusUnited States
- Biophysics Graduate Program, The Ohio State UniversityColumbusUnited States
- Ohio State Biochemistry Program, The Ohio State UniversityColumbusUnited States
| | - Omid A Zobeiri
- Department of Biomedical Engineering, McGill UniversityMontrealCanada
| | - Jeffrey R Holt
- Departments of Otolaryngology and Neurology, F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Saima Riazuddin
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of MedicineBaltimoreUnited States
| | - Kathleen E Cullen
- Departments of Biomedical Engineering, Neuroscience, and Otolaryngology and Head and Neck Surgery, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Marcos Sotomayor
- Department of Chemistry and Biochemistry, The Ohio State UniversityColumbusUnited States
- Biophysics Graduate Program, The Ohio State UniversityColumbusUnited States
- Ohio State Biochemistry Program, The Ohio State UniversityColumbusUnited States
| | - Zubair M Ahmed
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of MedicineBaltimoreUnited States
- Department of Biochemistry and Molecular Biology, University of Maryland School of MedicineBaltimoreUnited States
- Department of Ophthalmology and Visual Sciences, University of Maryland School of MedicineBaltimoreUnited States
| |
Collapse
|
17
|
Olivieri G, Dal Cortivo G, Del Conte R, Zanzoni S, Marino V, Dell'Orco D, Cantini F. Structural dynamics of calcium and integrin-binding protein 2 (CIB2) reveal uncommon flexibility and heterogeneous calcium and magnesium loading. Int J Biol Macromol 2025; 286:138003. [PMID: 39586446 DOI: 10.1016/j.ijbiomac.2024.138003] [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: 09/25/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024]
Abstract
Calcium- and Integrin-Binding protein 2 (CIB2) is a widely expressed protein with an uncertain biological role. Two of its four EF-hand motifs bind Mg(II) and/or Ca(II), thus triggering conformational changes. Although previous studies suggested that CIB2 preferentially binds Mg(II) over Ca(II) under physiological conditions, an atomic level characterization of CIB2 in the presence of both cations was lacking. Based on a combination of solution NMR, exhaustive molecular dynamics simulations and isothermal titration and differential scanning calorimetry, we characterized the interaction of CIB2 with both Ca(II) and Mg(II) ions and elucidated the protein regions involved in the interaction with the α7B integrin target. Analysis of experimental amide nitrogen relaxation rates shows that the EF4 motif exhibits high mobility regardless of the specific bound metal ion and demonstrates that the Mg(II)- and Ca(II)-bound state of CIB2 is relatively floppy, with pico-nanosecond motions induced in a region involved in target recognition. Overall, our data indicate a preferential, thermodynamically stable but structurally flexible state for CIB2, in which a Mg(II) ion is bound to EF3 and a Ca(II) ion to EF4. These results unveil the role of metal binding events in CIB2 and offer new insights into the dynamic regulation of target recognition.
Collapse
Affiliation(s)
- Gabriele Olivieri
- Magnetic Resonance Center, Department of Chemistry, University of Florence, Sesto Fiorentino, Italy
| | - Giuditta Dal Cortivo
- Department of Neurosciences, Biomedicine and Movement Sciences, Biological Chemistry Section, University of Verona, Verona, Italy
| | - Rebecca Del Conte
- Magnetic Resonance Center, Department of Chemistry, University of Florence, Sesto Fiorentino, Italy
| | - Serena Zanzoni
- Centro Piattaforme Tecnologiche, University of Verona, Verona, Italy
| | - Valerio Marino
- Department of Neurosciences, Biomedicine and Movement Sciences, Biological Chemistry Section, University of Verona, Verona, Italy
| | - Daniele Dell'Orco
- Department of Neurosciences, Biomedicine and Movement Sciences, Biological Chemistry Section, University of Verona, Verona, Italy.
| | - Francesca Cantini
- Magnetic Resonance Center, Department of Chemistry, University of Florence, Sesto Fiorentino, Italy.
| |
Collapse
|
18
|
Oestreicher D, Chepurwar S, Kusch K, Rankovic V, Jung S, Strenzke N, Pangrsic T. CaBP1 and 2 enable sustained Ca V1.3 calcium currents and synaptic transmission in inner hair cells. eLife 2024; 13:RP93646. [PMID: 39718549 DOI: 10.7554/elife.93646] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024] Open
Abstract
To encode continuous sound stimuli, the inner hair cell (IHC) ribbon synapses utilize calcium-binding proteins (CaBPs), which reduce the inactivation of their CaV1.3 calcium channels. Mutations in the CABP2 gene underlie non-syndromic autosomal recessive hearing loss DFNB93. Besides CaBP2, the structurally related CaBP1 is highly abundant in the IHCs. Here, we investigated how the two CaBPs cooperatively regulate IHC synaptic function. In Cabp1/2 double-knockout mice, we find strongly enhanced CaV1.3 inactivation, slowed recovery from inactivation and impaired sustained exocytosis. Already mild IHC activation further reduces the availability of channels to trigger synaptic transmission and may effectively silence synapses. Spontaneous and sound-evoked responses of spiral ganglion neurons in vivo are strikingly reduced and strongly depend on stimulation rates. Transgenic expression of CaBP2 leads to substantial recovery of IHC synaptic function and hearing sensitivity. We conclude that CaBP1 and 2 act together to suppress voltage- and calcium-dependent inactivation of IHC CaV1.3 channels in order to support sufficient rate of exocytosis and enable fast, temporally precise and indefatigable sound encoding.
Collapse
Affiliation(s)
- David Oestreicher
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Shashank Chepurwar
- Auditory Systems Physiology Group, Institute for Auditory Neuroscience, InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
| | - Kathrin Kusch
- Functional Auditory Genomics, Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Vladan Rankovic
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Restorative Cochlear Genomics Group, Auditory Neuroscience and Optogenetics Laboratory, German Primate Cente, Göttingen, Germany
| | - Sangyong Jung
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Nicola Strenzke
- Auditory Systems Physiology Group, Institute for Auditory Neuroscience, InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
| | - Tina Pangrsic
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
- Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
19
|
López-Porras AI, Kruse AM, McClendon MT, Vélez-Ortega AC. Myosin XVA isoforms participate in the mechanotransduction-dependent remodeling of the actin cytoskeleton in auditory stereocilia. Front Neurol 2024; 15:1482892. [PMID: 39777322 PMCID: PMC11704364 DOI: 10.3389/fneur.2024.1482892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Auditory hair cells form precise and sensitive staircase-like actin protrusions known as stereocilia. These specialized microvilli detect deflections induced by sound through the activation of mechano-electrical transduction (MET) channels located at their tips. At rest, a small MET channel current results in a constant calcium influx which regulates the morphology of the actin cytoskeleton in the shorter 'transducing' stereocilia. However, the molecular mechanisms involved in this novel type of activity-driven plasticity in the stereocilium cytoskeleton are currently unknown. Here, we tested the contribution of myosin XVA (MYO15A) isoforms, given their known roles in the regulation of stereocilia dimensions during hair bundle development and the maintenance of transducing stereocilia in mature hair cells. We used electron microscopy to evaluate morphological changes in the cytoskeleton of auditory hair cell stereocilia after the pharmacological blockage of resting MET currents in cochlear explants from mice that lacked one or all isoforms of MYO15A. Hair cells lacking functional MYO15A isoforms did not exhibit MET-dependent remodeling in their stereocilia cytoskeleton. In contrast, hair cells lacking only the long isoform of MYO15A exhibited increased MET-dependent stereocilia remodeling, including remodeling in stereocilia from the tallest 'non-transducing' row of the bundle. We conclude that MYO15A isoforms both enable and fine-tune the MET-dependent remodeling of the actin cytoskeleton in transducing stereocilia, while also contributing to the stability of the tallest row.
Collapse
|
20
|
De-la-Torre P, Martínez-García C, Gratias P, Mun M, Santana P, Akyuz N, González W, Indzhykulian AA, Ramírez D. Identification of Druggable Binding Sites and Small Molecules as Modulators of TMC1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.05.583611. [PMID: 38826329 PMCID: PMC11142246 DOI: 10.1101/2024.03.05.583611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Our ability to hear and maintain balance relies on the proper functioning of inner ear sensory hair cells, which translate mechanical stimuli into electrical signals via mechano-electrical transducer (MET) channels, composed of TMC1/2 proteins. However, the therapeutic use of ototoxic drugs, such as aminoglycosides and cisplatin, which can enter hair cells through MET channels, often leads to profound auditory and vestibular dysfunction. Despite extensive research on otoprotective compounds targeting MET channels, our understanding of how small-molecule modulators interact with these channels remains limited, hampering the discovery of novel drugs. Here, we propose a structure-based screening approach, integrating 3D-pharmacophore modeling, molecular dynamics simulations of the TMC1+CIB2+TMIE complex, and experimental validation. Our pipeline successfully identified several novel compounds and FDA-approved drugs that reduced dye uptake in cultured cochlear explants, indicating MET-modulation activity. Simulations, molecular docking and free-energy estimations allowed us to identify three potential drug-binding sites within the channel pore, phospholipids, key amino acids involved in modulator interactions, and TMIE as a flexible component of the MET complex. We also identified shared ligand-binding features between TMC and structurally related TMEM16 proteins, providing novel insights into their distinct inhibition. Our pipeline offers a broad application for discovering modulators for mechanosensitive ion channels.
Collapse
Affiliation(s)
- Pedro De-la-Torre
- Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School and Mass Eye and Ear, Boston, MA, USA
| | - Claudia Martínez-García
- Departamento de Farmacología, Facultad de Ciencias Biológicas, Universidad de Concepción, Chile
| | - Paul Gratias
- Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School and Mass Eye and Ear, Boston, MA, USA
| | - Matthew Mun
- Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School and Mass Eye and Ear, Boston, MA, USA
| | - Paula Santana
- Facultad de Ingeniería, Instituto de Ciencias Químicas Aplicadas, Universidad Autónoma de Chile, Santiago, Chile
| | - Nurunisa Akyuz
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Wendy González
- Center for Bioinformatics and Molecular Simulations (CBSM), University of Talca, Talca 3460000, Chile
| | - Artur A. Indzhykulian
- Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School and Mass Eye and Ear, Boston, MA, USA
| | - David Ramírez
- Departamento de Farmacología, Facultad de Ciencias Biológicas, Universidad de Concepción, Chile
| |
Collapse
|
21
|
Cederroth CR, Dyhrfjeld-Johnsen J, Canlon B. Pharmacological Approaches to Hearing Loss. Pharmacol Rev 2024; 76:1063-1088. [PMID: 39164117 PMCID: PMC11549935 DOI: 10.1124/pharmrev.124.001195] [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: 02/14/2024] [Revised: 07/08/2024] [Accepted: 07/16/2024] [Indexed: 08/22/2024] Open
Abstract
Hearing disorders pose significant challenges to individuals experiencing them and their overall quality of life, emphasizing the critical need for advanced pharmacological approaches to address these conditions. Current treatment options often focus on amplification devices, cochlear implants, or other rehabilitative therapies, leaving a substantial gap regarding effective pharmacological interventions. Advancements in our understanding of the molecular and cellular mechanisms involved in hearing disorders induced by noise, aging, and ototoxicity have opened new avenues for drug development, some of which have led to numerous clinical trials, with promising results. The development of optimal drug delivery solutions in animals and humans can also enhance the targeted delivery of medications to the ear. Moreover, large genome studies contributing to a genetic understanding of hearing loss in humans combined with advanced molecular technologies in animal studies have shown a great potential to increase our understanding of the etiologies of hearing loss. The auditory system exhibits circadian rhythms and temporal variations in its physiology, its vulnerability to auditory insults, and its responsiveness to drug treatments. The cochlear clock rhythms are under the control of the glucocorticoid system, and preclinical evidence suggests that the risk/benefit profile of hearing disorder treatments using chronopharmacological approaches would be beneficial. If translatable to the bedside, such approaches may improve the outcome of clinical trials. Ongoing research into the molecular and genetic basis of auditory disorders, coupled with advancements in drug formulation and delivery as well as optimized timing of drug administration, holds great promise of more effective treatments. SIGNIFICANCE STATEMENT: Hearing disorders pose significant challenges to individuals and their overall quality of life, emphasizing the critical need for advanced pharmacological approaches to address these conditions. Ongoing research into the molecular and genetic basis of auditory disorders, coupled with advancements in drug delivery procedures and optimized timing of drug administration, holds the promise of more effective treatments.
Collapse
Affiliation(s)
- Christopher R Cederroth
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| | - Jonas Dyhrfjeld-Johnsen
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| | - Barbara Canlon
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| |
Collapse
|
22
|
Thomas HB, Demain LAM, Cabrera-Orefice A, Schrauwen I, Shamseldin HE, Rea A, Bharadwaj T, Smith TB, Oláhová M, Thompson K, He L, Kaur N, Shukla A, Abukhalid M, Ansar M, Rehman S, Riazuddin S, Abdulwahab F, Smith JM, Stark Z, Carrera S, Yue WW, Munro KJ, Alkuraya FS, Jamieson P, Ahmed ZM, Leal SM, Taylor RW, Wittig I, O'Keefe RT, Newman WG. Biallelic variants in MRPL49 cause variable clinical presentations, including sensorineural hearing loss, leukodystrophy, and ovarian insufficiency. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.10.24315152. [PMID: 39417135 PMCID: PMC11483032 DOI: 10.1101/2024.10.10.24315152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Combined oxidative phosphorylation deficiency (COXPD) is a rare multisystem disorder which is clinically and genetically heterogeneous. Genome sequencing identified biallelic MRPL49 variants in individuals from five unrelated families with presentations ranging from Perrault syndrome (primary ovarian insufficiency and sensorineural hearing loss) to severe childhood onset of leukodystrophy, learning disability, microcephaly and retinal dystrophy. Complexome profiling of fibroblasts from affected individuals revealed reduced levels of the small and, a more pronounced reduction of, the large mitochondrial ribosomal subunits. There was no evidence of altered mitoribosomal assembly. The reductions in levels of OXPHOS enzyme complexes I and IV are consistent with a form of COXPD associated with biallelic MRPL49 variants, expanding the understanding of how disruption of the mitochondrial ribosomal large subunit results in multi-system phenotypes.
Collapse
Affiliation(s)
- Huw B Thomas
- Division of Evolution, Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester, M13 9PL, UK
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK
| | - Leigh A M Demain
- Division of Evolution, Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester, M13 9PL, UK
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK
| | - Alfredo Cabrera-Orefice
- Centre for Functional Proteomics, Institute for Cardiovascular Physiology, Medical Faculty, Goethe University, 60596 Frankfurt am Main, Germany
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, 35392 Giessen, Germany
| | - Isabelle Schrauwen
- Department of Translational Neurosciences, University of Arizona College of Medicine Phoenix, Phoenix, AZ, USA
| | - Hanan E Shamseldin
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Alessandro Rea
- Division of Evolution, Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester, M13 9PL, UK
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK
| | - Thashi Bharadwaj
- Center for Statistical Genetics, Department of Neurology, Gertrude H. Sergievsky Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Thomas B Smith
- Division of Evolution, Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester, M13 9PL, UK
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK
| | - Monika Oláhová
- Department of Applied Sciences, Faculty of Health & Life Sciences, Northumbria University, Newcastle upon Tyne, UK
- Mitochondrial Research Group, Clinical and Translational Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Kyle Thompson
- Mitochondrial Research Group, Clinical and Translational Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Langping He
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, NE1 4LP, UK
| | - Namanpreet Kaur
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Anju Shukla
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Musaad Abukhalid
- Department of Neuroscience, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Muhammad Ansar
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, 45320, Pakistan
| | - Sakina Rehman
- Department of Otorhinolaryngology - Head & Neck Surgery, School of Medicine University of Maryland, Baltimore, United States
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Saima Riazuddin
- Department of Otorhinolaryngology - Head & Neck Surgery, School of Medicine University of Maryland, Baltimore, United States
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Firdous Abdulwahab
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Janine M Smith
- Specialty of Genomic Medicine, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2000, Australia
- Western Sydney Genetics Program, Department of Clinical Genetics, Sydney Children's Hospitals Network, Westmead, NSW 2145, Australia
| | - Zornitza Stark
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Flemington Road, Melbourne, VIC, Australia
- University of Melbourne, Melbourne, VIC, Australia
| | - Samantha Carrera
- Genome Editing Unit Core Facility, Faculty of Biology, Medicine and Health, University of Manchester, UK
| | - Wyatt W Yue
- Newcastle University Biosciences Institute, Medical School, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Kevin J Munro
- Manchester Centre for Audiology and Deafness (ManCAD), School of Health Sciences, University of Manchester, Manchester, UK
| | - Fowzan S Alkuraya
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- Department of Anatomy and Cell Biology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Peter Jamieson
- Department of Radiology, Manchester University Hospital NHS Foundation Trust, Manchester, M13 9PW, UK
| | - Zubair M Ahmed
- Department of Otorhinolaryngology - Head & Neck Surgery, School of Medicine University of Maryland, Baltimore, United States
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Suzanne M Leal
- Center for Statistical Genetics, Department of Neurology, Gertrude H. Sergievsky Center, Columbia University Medical Center, New York, NY 10032, USA
- Taub Institute for Alzheimer's Disease and the Aging Brain, and the Department of Neurology, Columbia University Medical Center, New York, NY
| | - Robert W Taylor
- Mitochondrial Research Group, Clinical and Translational Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, NE1 4LP, UK
| | - Ilka Wittig
- Centre for Functional Proteomics, Institute for Cardiovascular Physiology, Medical Faculty, Goethe University, 60596 Frankfurt am Main, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, 60596 Frankfurt am Main, Germany
| | - Raymond T O'Keefe
- Division of Evolution, Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester, M13 9PL, UK
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK
| | - William G Newman
- Division of Evolution, Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester, M13 9PL, UK
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK
| |
Collapse
|
23
|
Wang P, Miller KK, He E, Dhawan SS, Cunningham CL, Grillet N. LOXHD1 is indispensable for maintaining TMC1 auditory mechanosensitive channels at the site of force transmission. Nat Commun 2024; 15:7865. [PMID: 39256406 PMCID: PMC11387651 DOI: 10.1038/s41467-024-51850-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 08/20/2024] [Indexed: 09/12/2024] Open
Abstract
Hair cell bundles consist of stereocilia arranged in rows of increasing heights, connected by tip links that transmit sound-induced forces to shorter stereocilia tips. Auditory mechanotransduction channel complexes, composed of proteins TMC1/2, TMIE, CIB2, and LHFPL5, are located at the tips of shorter stereocilia. While most components can interact with the tip link in vitro, their ability to maintain the channel complexes at the tip link in vivo is uncertain. Return, using mouse models, we show that an additional component, LOXHD1, is essential for keeping TMC1-pore forming subunits at the tip link but is dispensable for TMC2. Using SUB-immunogold-SEM, we showed that TMC1 localizes near the tip link but mislocalizes without LOXHD1. LOXHD1 selectively interacts with TMC1, CIB2, LHFPL5, and tip-link protein PCDH15. Our results demonstrate that TMC1-driven mature auditory channels require LOXHD1 to stay connected to the tip link and remain functional, while TMC2-driven developmental channels do not.
Collapse
Affiliation(s)
- Pei Wang
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, 240 Pasteur Drive, Stanford, CA, USA
| | - Katharine K Miller
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, 240 Pasteur Drive, Stanford, CA, USA
| | - Enqi He
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, 240 Pasteur Drive, Stanford, CA, USA
| | - Siddhant S Dhawan
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, 240 Pasteur Drive, Stanford, CA, USA
| | - Christopher L Cunningham
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Nicolas Grillet
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, 240 Pasteur Drive, Stanford, CA, USA.
| |
Collapse
|
24
|
Zhou FM, Wang KK, Wang LH, Qiu JG, Wang W, Liu WJ, Wang L, Jiang BH. CIB2 mediates acquired gefitinib resistance by inducing ZEB1 expression and epithelial-mesenchymal transition. Aging (Albany NY) 2024; 16:12277-12292. [PMID: 39264588 PMCID: PMC11424576 DOI: 10.18632/aging.206086] [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: 02/20/2024] [Accepted: 08/01/2024] [Indexed: 09/13/2024]
Abstract
EGFR-TKIs have been used as frontline treatment in patients with advanced non-small cell lung cancer (NSCLC) suffering from the EGFR mutation. Gefitinib, the first-generation EGFR-TKI, has greatly improved survival rates in lung cancer patients, whereas acquired gefitinib resistance is still a critical issue that needs to be overcome. In our research, high expression levels of CIB2 were found in gefitinib-resistant lung cancer cells. CIB2 knockout rendered gefitinib-resistant cells more sensitive to gefitinib, and overexpression of CIB2 in parental cells was sufficient to induce more resistance to gefitinib. Inhibition of CIB2 in gefitinib-resistant lung cancer cells significantly induced cell apoptosis. To clarify the major molecular mechanism by which CIB2 increases gefitinib resistance, we demonstrated that raised CIB2 in lung cancer cells promoted epithelial-to-mesenchymal transition (EMT) through upregulation of ZEB1. Moreover, FOSL1 transcriptionally regulated CIB2 expression. Finally, CIB2 rendered tumors resistant to gefitinib treatment in vivo. Our results explored a new mechanism: upregulated CIB2 promoted EMT through ZEB1 to regulate gefitinib resistance, which could be a candidate therapeutic target for overcoming acquired resistance to EGFR-TKIs in NSCLC patients.
Collapse
Affiliation(s)
- Feng-Mei Zhou
- Academy of Medical Science, Zhengzhou University, Zhengzhou 450000, China
| | - Kun-Kun Wang
- Academy of Medical Science, Zhengzhou University, Zhengzhou 450000, China
| | - Li-Hong Wang
- Academy of Medical Science, Zhengzhou University, Zhengzhou 450000, China
| | - Jian-Ge Qiu
- Academy of Medical Science, Zhengzhou University, Zhengzhou 450000, China
| | - Wei Wang
- Academy of Medical Science, Zhengzhou University, Zhengzhou 450000, China
| | - Wen-Jing Liu
- The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Lin Wang
- Academy of Medical Science, Zhengzhou University, Zhengzhou 450000, China
| | - Bing-Hua Jiang
- Academy of Medical Science, Zhengzhou University, Zhengzhou 450000, China
| |
Collapse
|
25
|
Stanley OR, Swaminathan A, Wojahn E, Bao C, Ahmed ZM, Cullen KE. An open-source tool for automated human-level circling behavior detection. Sci Rep 2024; 14:20914. [PMID: 39245735 PMCID: PMC11381541 DOI: 10.1038/s41598-024-71665-z] [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: 06/15/2023] [Accepted: 08/29/2024] [Indexed: 09/10/2024] Open
Abstract
Quantitatively relating behavior to underlying biology is crucial in life science. Although progress in keypoint tracking tools has reduced barriers to recording postural data, identifying specific behaviors from this data remains challenging. Manual behavior coding is labor-intensive and inconsistent, while automatic methods struggle to explicitly define complex behaviors, even when they seem obvious to the human eye. Here, we demonstrate an effective technique for detecting circling in mice, a form of locomotion characterized by stereotyped spinning. Despite circling's extensive history as a behavioral marker, there currently exists no standard automated detection method. We developed a circling detection technique using simple postprocessing of keypoint data obtained from videos of freely-exploring (Cib2-/-;Cib3-/-) mutant mice, a strain previously found to exhibit circling behavior. Our technique achieves statistical parity with independent human observers in matching occurrence times based on human consensus, and it accurately distinguishes between videos of wild type mice and mutants. Our pipeline provides a convenient, noninvasive, quantitative tool for analyzing circling mouse models without the need for software engineering experience. Additionally, as the concepts underlying our approach are agnostic to the behavior being analyzed, and indeed to the modality of the recorded data, our results support the feasibility of algorithmically detecting specific research-relevant behaviors using readily-interpretable parameters tuned on the basis of human consensus.
Collapse
Affiliation(s)
- O R Stanley
- Department of Biomedical Engineering, Johns Hopkins University, 720 Rutland Ave, Traylor 504, Baltimore, MD, 21205-2109, USA
| | - A Swaminathan
- Department of Biomedical Engineering, Johns Hopkins University, 720 Rutland Ave, Traylor 504, Baltimore, MD, 21205-2109, USA
| | - E Wojahn
- Department of Biomedical Engineering, Johns Hopkins University, 720 Rutland Ave, Traylor 504, Baltimore, MD, 21205-2109, USA
| | - C Bao
- Department of Biomedical Engineering, Johns Hopkins University, 720 Rutland Ave, Traylor 504, Baltimore, MD, 21205-2109, USA
| | - Z M Ahmed
- Departments of Otorhinolaryngology-Head and Neck Surgery, Biochemistry and Molecular Biology, Ophthalmology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - K E Cullen
- Department of Biomedical Engineering, Johns Hopkins University, 720 Rutland Ave, Traylor 504, Baltimore, MD, 21205-2109, USA.
- Departments of Neuroscience, Otolaryngology-Head and Neck Surgery, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
26
|
Jiang Q, Zou W, Li S, Qiu X, Zhu L, Kang L, Müller U. Sequence variations and accessory proteins adapt TMC functions to distinct sensory modalities. Neuron 2024; 112:2922-2937.e8. [PMID: 38986620 PMCID: PMC11377162 DOI: 10.1016/j.neuron.2024.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 05/10/2024] [Accepted: 06/14/2024] [Indexed: 07/12/2024]
Abstract
Transmembrane channel-like (TMC) proteins are expressed throughout the animal kingdom and are thought to encode components of ion channels. Mammals express eight TMCs (mTMC1-8), two of which (mTMC1 and mTMC2) are subunits of mechanotransduction channels. C. elegans expresses two TMCs (TMC-1 and TMC-2), which mediate mechanosensation, egg laying, and alkaline sensing. The mechanisms by which nematode TMCs contribute to such diverse physiological processes and their functional relationship to mammalian mTMCs is unclear. Here, we show that association with accessory proteins tunes nematode TMC-1 to divergent sensory functions. In addition, distinct TMC-1 domains enable touch and alkaline sensing. Strikingly, these domains are segregated in mammals between mTMC1 and mTMC3. Consistent with these findings, mammalian mTMC1 can mediate mechanosensation in nematodes, while mTMC3 can mediate alkaline sensation. We conclude that sequence diversification and association with accessory proteins has led to the emergence of TMC protein complexes with diverse properties and physiological functions.
Collapse
Affiliation(s)
- Qiang Jiang
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Wenjuan Zou
- Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310053, Zhejiang, China
| | - Shitian Li
- Department of Neurology of the Fourth Affiliated Hospital and School of Brain Science and Brain Medicine, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Xufeng Qiu
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Linhui Zhu
- Department of Neurology of the Fourth Affiliated Hospital and School of Brain Science and Brain Medicine, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Lijun Kang
- Department of Neurology of the Fourth Affiliated Hospital and School of Brain Science and Brain Medicine, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Yiwu 322000, China.
| | - Ulrich Müller
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
27
|
Ebrahim S, Ballesteros A, Zheng WS, Mukherjee S, Hu G, Weng WH, Montgomery JS, Agyemang Y, Cui R, Sun W, Krystofiak E, Foster MP, Sotomayor M, Kachar B. Transmembrane channel-like 4 and 5 proteins at microvillar tips are potential ion channels and lipid scramblases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.22.609173. [PMID: 39229161 PMCID: PMC11370596 DOI: 10.1101/2024.08.22.609173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Microvilli-membrane bound actin protrusions on the surface of epithelial cells-are sites of critical processes including absorption, secretion, and adhesion. Increasing evidence suggests microvilli are mechanosensitive, but underlying molecules and mechanisms remain unknown. Here, we localize transmembrane channel-like proteins 4 and 5 (TMC4 and 5) and calcium and integrin binding protein 3 (CIB3) to microvillar tips in intestinal epithelial cells, near glycocalyx insertion sites. We find that TMC5 colocalizes with CIB3 in cultured cells and that a TMC5 fragment forms a complex with CIB3 in vitro. Homology and AlphaFold2 models reveal a putative ion permeation pathway in TMC4 and 5, and molecular dynamics simulations predict both proteins can conduct ions and perform lipid scrambling. These findings raise the possibility that TMC4 and 5 interact with CIB3 at microvillar tips to form a mechanosensitive complex, akin to TMC1 and 2, and CIB2 and 3, within the mechanotransduction channel complex at the tips of inner ear stereocilia.
Collapse
Affiliation(s)
- Seham Ebrahim
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA
| | - Angela Ballesteros
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
- Section on Sensory Physiology and Biophysics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - W. Sharon Zheng
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA
| | - Shounak Mukherjee
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Gaizun Hu
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA
| | - Wei-Hsiang Weng
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
- Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Jonathan S. Montgomery
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH 43210, USA
| | - Yaw Agyemang
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Runjia Cui
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Willy Sun
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Evan Krystofiak
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mark P. Foster
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
- Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH 43210, USA
| | - Marcos Sotomayor
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
- Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH 43210, USA
| | - Bechara Kachar
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
28
|
Giese APJ, Parker A, Rehman S, Brown SDM, Riazuddin S, Vander Kooi CW, Bowl MR, Ahmed ZM. CIB2 function is distinct from Whirlin in the development of cochlear stereocilia staircase pattern. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.30.605852. [PMID: 39131343 PMCID: PMC11312573 DOI: 10.1101/2024.07.30.605852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Variations in genes coding for calcium and integrin binding protein 2 (CIB2) and whirlin cause deafness both in humans and mice. We previously reported that CIB2 binds to whirlin, and is essential for normal staircase architecture of auditory hair cells stereocilia. Here, we refine the interacting domains between these proteins and provide evidence that both proteins have distinct role in the development and organization of stereocilia bundles required for auditory transduction. Using a series of CIB2 and whirlin deletion constructs and nanoscale pulldown (NanoSPD) assays, we localized the regions of CIB2 that are critical for interaction with whirlin. AlphaFold 2 multimer, independently identified the same interacting regions between CIB2 and whirlin proteins, providing a detailed structural model of the interaction between the CIB2 EF2 domain and whirlin HHD2 domain. Next, we investigated genetic interaction between murine Cib2 and Whrn using genetic approaches. Hearing in mice double heterozygous for functionally null alleles (Cib2 KO/+ ;Whrn wi/+ ) was similar to age-matched wild type mice, indicating that partial deficiency for both Cib2 and Whrn does not impair hearing. Double homozygous mutant mice (Cib2 KO/KO ;Whrn wi/wi ) had profound hearing loss and cochlear stereocilia exhibited a predominant phenotype seen in single Whrn wi/wi mutants. Furthermore, over-expression of Whrn in Cib2 KO/KO mice did not rescue the stereocilia morphology. These data suggest that, CIB2 is multifunctional, with key independent functions in development and/or maintenance of stereocilia staircase pattern in auditory hair cells.
Collapse
Affiliation(s)
- Arnaud P. J. Giese
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Andrew Parker
- MRC Harwell Institute, Mammalian Genetics Unit, Harwell Campus, Oxfordshire, OX11 0RD, UK
| | - Sakina Rehman
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Steve D. M. Brown
- MRC Harwell Institute, Mammalian Genetics Unit, Harwell Campus, Oxfordshire, OX11 0RD, UK
| | - Saima Riazuddin
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Craig W. Vander Kooi
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, USA
| | - Michael R. Bowl
- MRC Harwell Institute, Mammalian Genetics Unit, Harwell Campus, Oxfordshire, OX11 0RD, UK
- UCL Ear Institute, University College London, London, WC1X 8EE, UK
| | - Zubair M. Ahmed
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Program in Neuroscience & Cognitive Science, University of Maryland, College Park, MD, USA
| |
Collapse
|
29
|
Giese APJ, Weng WH, Kindt KS, Chang HHV, Montgomery JS, Ratzan EM, Beirl AJ, Rivera RA, Lotthammer JM, Walujkar S, Foster MP, Zobeiri OA, Holt JR, Riazuddin S, Cullen KE, Sotomayor M, Ahmed ZM. Complexes of vertebrate TMC1/2 and CIB2/3 proteins form hair-cell mechanotransduction cation channels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.26.542533. [PMID: 37398045 PMCID: PMC10312449 DOI: 10.1101/2023.05.26.542533] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Calcium and integrin-binding protein 2 (CIB2) and CIB3 bind to transmembrane channel-like 1 (TMC1) and TMC2, the pore-forming subunits of the inner-ear mechano-electrical transduction (MET) apparatus. These interactions have been proposed to be functionally relevant across mechanosensory organs and vertebrate species. Here we show that both CIB2 and CIB3 can form heteromeric complexes with TMC1 and TMC2 and are integral for MET function in mouse cochlea and vestibular end organs as well as in zebrafish inner ear and lateral line. Our AlphaFold 2 models suggest that vertebrate CIB proteins can simultaneously interact with at least two cytoplasmic domains of TMC1 and TMC2 as validated using nuclear magnetic resonance spectroscopy of TMC1 fragments interacting with CIB2 and CIB3. Molecular dynamics simulations of TMC1/2 complexes with CIB2/3 predict that TMCs are structurally stabilized by CIB proteins to form cation channels. Overall, our work demonstrates that intact CIB2/3 and TMC1/2 complexes are integral to hair-cell MET function in vertebrate mechanosensory epithelia.
Collapse
Affiliation(s)
- Arnaud P J Giese
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Wei-Hsiang Weng
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
- Biophysics Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Katie S Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | | | - Jonathan S Montgomery
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH, USA
| | - Evan M Ratzan
- Departments of Otolaryngology and Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alisha J Beirl
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Roberto Aponte Rivera
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey M Lotthammer
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
| | - Sanket Walujkar
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
| | - Mark P Foster
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
- Biophysics Graduate Program, The Ohio State University, Columbus, OH, USA
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH, USA
| | - Omid A Zobeiri
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Jeffrey R Holt
- Departments of Otolaryngology and Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Saima Riazuddin
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kathleen E Cullen
- Departments of Biomedical Engineering, Neuroscience, and Otolaryngology and Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marcos Sotomayor
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
- Biophysics Graduate Program, The Ohio State University, Columbus, OH, USA
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH, USA
| | - Zubair M Ahmed
- Department of Otorhinolaryngology - Head & Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
30
|
Wilkerson MD, Hupalo D, Gray JC, Zhang X, Wang J, Girgenti MJ, Alba C, Sukumar G, Lott NM, Naifeh JA, Aliaga P, Kessler RC, Turner C, Pollard HB, Dalgard CL, Ursano RJ, Stein MB. Uncommon Protein-Coding Variants Associated With Suicide Attempt in a Diverse Sample of U.S. Army Soldiers. Biol Psychiatry 2024; 96:15-25. [PMID: 38141912 DOI: 10.1016/j.biopsych.2023.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/02/2023] [Accepted: 12/05/2023] [Indexed: 12/25/2023]
Abstract
BACKGROUND Suicide is a societal and public health concern of global scale. Identifying genetic risk factors for suicide attempt can characterize underlying biology and enable early interventions to prevent deaths. Recent studies have described common genetic variants for suicide-related behaviors. Here, we advance this search for genetic risk by analyzing the association between suicide attempt and uncommon variation exome-wide in a large, ancestrally diverse sample. METHODS We sequenced whole genomes of 13,584 soldiers from the Army STARRS (Army Study to Assess Risk and Resilience in Servicemembers), including 979 individuals with a history of suicide attempt. Uncommon, nonsilent protein-coding variants were analyzed exome-wide for association with suicide attempt using gene-collapsed and single-variant analyses. RESULTS We identified 19 genes with variants enriched in individuals with history of suicide attempt, either through gene-collapsed or single-variant analysis (Bonferroni padjusted < .05). These genes were CIB2, MLF1, HERC1, YWHAE, RCN2, VWA5B1, ATAD3A, NACA, EP400, ZNF585A, LYST, RC3H2, PSD3, STARD9, SGMS1, ACTR6, RGS7BP, DIRAS2, and KRTAP10-1. Most genes had variants across multiple genomic ancestry groups. Seventeen of these genes were expressed in healthy brain tissue, with 9 genes expressed at the highest levels in the brain versus other tissues. Brains from individuals deceased from suicide aberrantly expressed RGS7BP (padjusted = .035) in addition to nominally significant genes including YWHAE and ACTR6, all of which have reported associations with other mental disorders. CONCLUSIONS These results advance the molecular characterization of suicide attempt behavior and support the utility of whole-genome sequencing for complementing the findings of genome-wide association studies in suicide research.
Collapse
Affiliation(s)
- Matthew D Wilkerson
- Center for Military Precision Health, Uniformed Services University, Bethesda, Maryland; Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, Maryland
| | - Daniel Hupalo
- Center for Military Precision Health, Uniformed Services University, Bethesda, Maryland
| | - Joshua C Gray
- Department of Medical and Clinical Psychology, Uniformed Services University, Bethesda, Maryland
| | - Xijun Zhang
- Center for Military Precision Health, Uniformed Services University, Bethesda, Maryland
| | - Jiawei Wang
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Matthew J Girgenti
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Camille Alba
- Center for Military Precision Health, Uniformed Services University, Bethesda, Maryland
| | - Gauthaman Sukumar
- Center for Military Precision Health, Uniformed Services University, Bethesda, Maryland
| | - Nathaniel M Lott
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, Maryland
| | - James A Naifeh
- Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University, Bethesda, Maryland
| | - Pablo Aliaga
- Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University, Bethesda, Maryland
| | - Ronald C Kessler
- Department of Health Care Policy, Harvard Medical School, Boston, Massachusetts
| | - Clesson Turner
- Department of Pediatrics, Uniformed Services University, Bethesda, Maryland
| | - Harvey B Pollard
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, Maryland
| | - Clifton L Dalgard
- Center for Military Precision Health, Uniformed Services University, Bethesda, Maryland; Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, Maryland
| | - Robert J Ursano
- Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University, Bethesda, Maryland
| | - Murray B Stein
- Department of Psychiatry, University of California San Diego, La Jolla, California; Herbert Wertheim School of Public Health, University of California San Diego, La Jolla, California; VA San Diego Healthcare System, San Diego, California.
| |
Collapse
|
31
|
Holt JR, Fettiplace R, Müller U. Sensory transduction in auditory hair cells-PIEZOs can't touch this. J Gen Physiol 2024; 156:e202413585. [PMID: 38727631 PMCID: PMC11090049 DOI: 10.1085/jgp.202413585] [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] [Indexed: 05/15/2024] Open
Abstract
In this Viewpoint, Holt, Fettiplace, and Müller weigh the evidence supporting a role for PIEZO and TMC channels in mechanosensory transduction in inner ear hair cells.
Collapse
Affiliation(s)
- Jeffrey R. Holt
- Departments of Otolaryngology and Neurology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Robert Fettiplace
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Ulrich Müller
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
32
|
Liu Z, Zhang H, Hong G, Bi X, Hu J, Zhang T, An Y, Guo N, Dong F, Xiao Y, Li W, Zhao X, Chu B, Guo S, Zhang X, Chai R, Fu X. Inhibition of Gpx4-mediated ferroptosis alleviates cisplatin-induced hearing loss in C57BL/6 mice. Mol Ther 2024; 32:1387-1406. [PMID: 38414247 PMCID: PMC11081921 DOI: 10.1016/j.ymthe.2024.02.029] [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: 10/23/2023] [Revised: 01/29/2024] [Accepted: 02/24/2024] [Indexed: 02/29/2024] Open
Abstract
Cisplatin-induced hearing loss is a common side effect of cancer chemotherapy in clinics; however, the mechanism of cisplatin-induced ototoxicity is still not completely clarified. Cisplatin-induced ototoxicity is mainly associated with the production of reactive oxygen species, activation of apoptosis, and accumulation of intracellular lipid peroxidation, which also is involved in ferroptosis induction. In this study, the expression of TfR1, a ferroptosis biomarker, was upregulated in the outer hair cells of cisplatin-treated mice. Moreover, several key ferroptosis regulator genes were altered in cisplatin-damaged cochlear explants based on RNA sequencing, implying the induction of ferroptosis. Ferroptosis-related Gpx4 and Fsp1 knockout mice were established to investigate the specific mechanisms associated with ferroptosis in cochleae. Severe outer hair cell loss and progressive damage of synapses in inner hair cells were observed in Atoh1-Gpx4-/- mice. However, Fsp1-/- mice showed no significant hearing phenotype, demonstrating that Gpx4, but not Fsp1, may play an important role in the functional maintenance of HCs. Moreover, findings showed that FDA-approved luteolin could specifically inhibit ferroptosis and alleviate cisplatin-induced ototoxicity through decreased expression of transferrin and intracellular concentration of ferrous ions. This study indicated that ferroptosis inhibition through the reduction of intracellular ferrous ions might be a potential strategy to prevent cisplatin-induced hearing loss.
Collapse
MESH Headings
- Animals
- Cisplatin/adverse effects
- Ferroptosis/drug effects
- Ferroptosis/genetics
- Mice
- Hearing Loss/chemically induced
- Hearing Loss/genetics
- Hearing Loss/metabolism
- Mice, Knockout
- Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism
- Phospholipid Hydroperoxide Glutathione Peroxidase/genetics
- Mice, Inbred C57BL
- Disease Models, Animal
- Receptors, Transferrin/metabolism
- Receptors, Transferrin/genetics
- Reactive Oxygen Species/metabolism
- Lipid Peroxidation/drug effects
- Hair Cells, Auditory, Outer/metabolism
- Hair Cells, Auditory, Outer/drug effects
- Hair Cells, Auditory, Outer/pathology
- Ototoxicity/etiology
- Ototoxicity/metabolism
- Antineoplastic Agents/adverse effects
- Apoptosis/drug effects
Collapse
Affiliation(s)
- Ziyi Liu
- Medical Science and Technology Innovation Center, Institute of Brain Science and Brain-inspired Research, Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Hanbing Zhang
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, NHC Key Laboratory of Otorhinolaryngology (Shandong University), Jinan, Shandong 250012, China
| | - Guodong Hong
- Medical Science and Technology Innovation Center, Institute of Brain Science and Brain-inspired Research, Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Xiuli Bi
- Medical Science and Technology Innovation Center, Institute of Brain Science and Brain-inspired Research, Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Jun Hu
- Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Tiancheng Zhang
- Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yachun An
- School of Life Science, Shandong University, Qingdao, Shandong 266237, China
| | - Na Guo
- Medical Science and Technology Innovation Center, Institute of Brain Science and Brain-inspired Research, Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Fengyue Dong
- School of Life Science, Shandong University, Qingdao, Shandong 266237, China
| | - Yu Xiao
- School of Life Science, Shandong University, Qingdao, Shandong 266237, China
| | - Wen Li
- Medical Science and Technology Innovation Center, Institute of Brain Science and Brain-inspired Research, Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Xiaoxu Zhao
- Medical Science and Technology Innovation Center, Institute of Brain Science and Brain-inspired Research, Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Bo Chu
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250102, China
| | - Siwei Guo
- School of Life Science, Shandong University, Qingdao, Shandong 266237, China
| | - Xiaohan Zhang
- Medical Science and Technology Innovation Center, Institute of Brain Science and Brain-inspired Research, Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Renjie Chai
- Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, School of Medicine, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, Jiangsu 210096, China; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China; Department of Neurology, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China; Southeast University Shenzhen Research Institute, Shenzhen, Guangdong 518063, China.
| | - Xiaolong Fu
- Medical Science and Technology Innovation Center, Institute of Brain Science and Brain-inspired Research, Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China.
| |
Collapse
|
33
|
Miyoshi T, Belyantseva IA, Sajeevadathan M, Friedman TB. Pathophysiology of human hearing loss associated with variants in myosins. Front Physiol 2024; 15:1374901. [PMID: 38562617 PMCID: PMC10982375 DOI: 10.3389/fphys.2024.1374901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 02/21/2024] [Indexed: 04/04/2024] Open
Abstract
Deleterious variants of more than one hundred genes are associated with hearing loss including MYO3A, MYO6, MYO7A and MYO15A and two conventional myosins MYH9 and MYH14. Variants of MYO7A also manifest as Usher syndrome associated with dysfunction of the retina and vestibule as well as hearing loss. While the functions of MYH9 and MYH14 in the inner ear are debated, MYO3A, MYO6, MYO7A and MYO15A are expressed in inner ear hair cells along with class-I myosin MYO1C and are essential for developing and maintaining functional stereocilia on the apical surface of hair cells. Stereocilia are large, cylindrical, actin-rich protrusions functioning as biological mechanosensors to detect sound, acceleration and posture. The rigidity of stereocilia is sustained by highly crosslinked unidirectionally-oriented F-actin, which also provides a scaffold for various proteins including unconventional myosins and their cargo. Typical myosin molecules consist of an ATPase head motor domain to transmit forces to F-actin, a neck containing IQ-motifs that bind regulatory light chains and a tail region with motifs recognizing partners. Instead of long coiled-coil domains characterizing conventional myosins, the tails of unconventional myosins have various motifs to anchor or transport proteins and phospholipids along the F-actin core of a stereocilium. For these myosins, decades of studies have elucidated their biochemical properties, interacting partners in hair cells and variants associated with hearing loss. However, less is known about how myosins traffic in a stereocilium using their motor function, and how each variant correlates with a clinical condition including the severity and onset of hearing loss, mode of inheritance and presence of symptoms other than hearing loss. Here, we cover the domain structures and functions of myosins associated with hearing loss together with advances, open questions about trafficking of myosins in stereocilia and correlations between hundreds of variants in myosins annotated in ClinVar and the corresponding deafness phenotypes.
Collapse
Affiliation(s)
- Takushi Miyoshi
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
- Division of Molecular and Integrative Physiology, Department of Biomedical Sciences, Southern Illinois University School of Medicine, Carbondale, IL, United States
| | - Inna A. Belyantseva
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Mrudhula Sajeevadathan
- Division of Molecular and Integrative Physiology, Department of Biomedical Sciences, Southern Illinois University School of Medicine, Carbondale, IL, United States
| | - Thomas B. Friedman
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
34
|
Clark S, Jeong H, Posert R, Goehring A, Gouaux E. The structure of the Caenorhabditis elegans TMC-2 complex suggests roles of lipid-mediated subunit contacts in mechanosensory transduction. Proc Natl Acad Sci U S A 2024; 121:e2314096121. [PMID: 38354260 PMCID: PMC10895266 DOI: 10.1073/pnas.2314096121] [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: 08/15/2023] [Accepted: 12/21/2023] [Indexed: 02/16/2024] Open
Abstract
Mechanotransduction is the process by which a mechanical force, such as touch, is converted into an electrical signal. Transmembrane channel-like (TMC) proteins are an evolutionarily conserved family of membrane proteins whose function has been linked to a variety of mechanosensory processes, including hearing and balance sensation in vertebrates and locomotion in Drosophila. TMC1 and TMC2 are components of ion channel complexes, but the molecular features that tune these complexes to diverse mechanical stimuli are unknown. Caenorhabditis elegans express two TMC homologs, TMC-1 and TMC-2, both of which are the likely pore-forming subunits of mechanosensitive ion channels but differ in their expression pattern and functional role in the worm. Here, we present the single-particle cryo-electron microscopy structure of the native TMC-2 complex isolated from C. elegans. The complex is composed of two copies of the pore-forming TMC-2 subunit, the calcium and integrin binding protein CALM-1 and the transmembrane inner ear protein TMIE. Comparison of the TMC-2 complex to the recently published cryo-EM structure of the C. elegans TMC-1 complex highlights conserved protein-lipid interactions, as well as a π-helical structural motif in the pore-forming helices, that together suggest a mechanism for TMC-mediated mechanosensory transduction.
Collapse
Affiliation(s)
- Sarah Clark
- Vollum Institute, Oregon Health and Science University, Portland, OR97239
| | - Hanbin Jeong
- Vollum Institute, Oregon Health and Science University, Portland, OR97239
| | - Rich Posert
- Vollum Institute, Oregon Health and Science University, Portland, OR97239
| | - April Goehring
- Vollum Institute, Oregon Health and Science University, Portland, OR97239
- HHMI, Oregon Health and Science University, Portland, OR97239
| | - Eric Gouaux
- Vollum Institute, Oregon Health and Science University, Portland, OR97239
- HHMI, Oregon Health and Science University, Portland, OR97239
| |
Collapse
|
35
|
Lee JH, Perez-Flores MC, Park S, Kim HJ, Chen Y, Kang M, Kersigo J, Choi J, Thai PN, Woltz RL, Perez-Flores DC, Perkins G, Sihn CR, Trinh P, Zhang XD, Sirish P, Dong Y, Feng WW, Pessah IN, Dixon RE, Sokolowski B, Fritzsch B, Chiamvimonvat N, Yamoah EN. The Piezo channel is a mechano-sensitive complex component in the mammalian inner ear hair cell. Nat Commun 2024; 15:526. [PMID: 38228630 PMCID: PMC10791687 DOI: 10.1038/s41467-023-44230-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 12/04/2023] [Indexed: 01/18/2024] Open
Abstract
The inner ear is the hub where hair cells (HCs) transduce sound, gravity, and head acceleration stimuli to the brain. Hearing and balance rely on mechanosensation, the fastest sensory signals transmitted to the brain. The mechanoelectrical transducer (MET) channel is the entryway for the sound-balance-brain interface, but the channel-complex composition is not entirely known. Here, we report that the mouse utilizes Piezo1 (Pz1) and Piezo2 (Pz2) isoforms as MET-complex components. The Pz channels, expressed in HC stereocilia, and cell lines are co-localized and co-assembled with MET complex partners. Mice expressing non-functional Pz1 and Pz2 at the ROSA26 locus have impaired auditory and vestibular traits that can only be explained if the Pzs are integral to the MET complex. We suggest that Pz subunits constitute part of the MET complex and that interactions with other MET complex components yield functional MET units to generate HC MET currents.
Collapse
Affiliation(s)
- Jeong Han Lee
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Maria C Perez-Flores
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Seojin Park
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
- Prestige Biopharma, 11-12F, 44, Myongjigukje7-ro, Gangseo-gu, Busan, 67264, South Korea
| | - Hyo Jeong Kim
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Yingying Chen
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Mincheol Kang
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
- Prestige Biopharma, 11-12F, 44, Myongjigukje7-ro, Gangseo-gu, Busan, 67264, South Korea
| | | | - Jinsil Choi
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Phung N Thai
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA, 95616, USA
| | - Ryan L Woltz
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA, 95616, USA
| | | | - Guy Perkins
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA, 92093, USA
| | - Choong-Ryoul Sihn
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Pauline Trinh
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA, 95616, USA
| | - Xiao-Dong Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA, 95616, USA
| | - Padmini Sirish
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA, 95616, USA
| | - Yao Dong
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, 1089 VM3B, Davis, CA, 95616, USA
| | - Wayne Wei Feng
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, 1089 VM3B, Davis, CA, 95616, USA
| | - Isaac N Pessah
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, 1089 VM3B, Davis, CA, 95616, USA
| | - Rose E Dixon
- Department of Physiology & Membrane Biology, Tupper Hall, One Shields Avenue, Davis, CA, 95616, USA
| | - Bernd Sokolowski
- Department of Otolaryngology-Head and Neck Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, IA, USA
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA, 95616, USA
- VA Northern California Healthcare System, Sacramento, USA
| | - Ebenezer N Yamoah
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA.
| |
Collapse
|
36
|
Beurg M, Schwalbach ET, Fettiplace R. LHFPL5 is a key element in force transmission from the tip link to the hair cell mechanotransducer channel. Proc Natl Acad Sci U S A 2024; 121:e2318270121. [PMID: 38194445 PMCID: PMC10801851 DOI: 10.1073/pnas.2318270121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 11/30/2023] [Indexed: 01/11/2024] Open
Abstract
During auditory transduction, sound-evoked vibrations of the hair cell stereociliary bundles open mechanotransducer (MET) ion channels via tip links extending from one stereocilium to its neighbor. How tension in the tip link is delivered to the channel is not fully understood. The MET channel comprises a pore-forming subunit, transmembrane channel-like protein (TMC1 or TMC2), aided by several accessory proteins, including LHFPL5 (lipoma HMGIC fusion partner-like 5). We investigated the role of LHFPL5 in transduction by comparing MET channel activation in outer hair cells of Lhfpl5-/- knockout mice with those in Lhfpl5+/- heterozygotes. The 10 to 90 percent working range of transduction in Tmc1+/+; Lhfpl5+/- was 52 nm, from which the single-channel gating force, Z, was evaluated as 0.34 pN. However, in Tmc1+/+; Lhfpl5-/- mice, the working range increased to 123 nm and Z more than halved to 0.13 pN, indicating reduced sensitivity. Tip link tension is thought to activate the channel via a gating spring, whose stiffness is inferred from the stiffness change on tip link destruction. The gating stiffness was ~40 percent of the total bundle stiffness in wild type but was virtually abolished in Lhfpl5-/-, implicating LHFPL5 as a principal component of the gating spring. The mutation Tmc1 p.D569N reduced the LHFPL5 immunolabeling in the stereocilia and like Lhfpl5-/- doubled the MET working range, but other deafness mutations had no effect on the dynamic range. We conclude that tip-link tension is transmitted to the channel primarily via LHFPL5; residual activation without LHFPL5 may occur by direct interaction between PCDH15 and TMC1.
Collapse
Affiliation(s)
- Maryline Beurg
- Department of Neuroscience, University of WisconsinSchool of Medicine and Public Health, Madison, WI53706
| | - Evan Travis Schwalbach
- Department of Neuroscience, University of WisconsinSchool of Medicine and Public Health, Madison, WI53706
| | - Robert Fettiplace
- Department of Neuroscience, University of WisconsinSchool of Medicine and Public Health, Madison, WI53706
| |
Collapse
|
37
|
Wang P, Miller KK, He E, Dhawan SS, Cunningham CL, Grillet N. LOXHD1 is indispensable for coupling auditory mechanosensitive channels to the site of force transmission. RESEARCH SQUARE 2024:rs.3.rs-3752492. [PMID: 38260480 PMCID: PMC10802736 DOI: 10.21203/rs.3.rs-3752492/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Hearing is initiated in hair cells by the mechanical activation of ion channels in the hair bundle. The hair bundle is formed by stereocilia organized into rows of increasing heights interconnected by tip links, which convey sound-induced forces to stereocilia tips. The auditory mechanosensitive channels are complexes containing at least four protein-subunits - TMC1/2, TMIE, CIB2, and LHFPL51-16 - and are located at the tips of shorter stereocilia at a yet-undetermined distance from the lower tip link insertion point17. While multiple auditory channel subunits appear to interact with the tip link, it remains unknown whether their combined interaction alone can resist the high-frequency mechanical stimulations owing to sound. Here we show that an unanticipated additional element, LOXHD1, is indispensable for maintaining the TMC1 pore-forming channel subunits coupled to the tip link. We demonstrate that LOXHD1 is a unique element of the auditory mechanotransduction complex that selectively affects the localization of TMC1, but not its close developmental paralogue TMC2. Taking advantage of our novel immunogold scanning electron microscopy method for submembranous epitopes (SUB-immunogold-SEM), we demonstrate that TMC1 normally concentrates within 100-nm of the tip link insertion point. In LOXHD1's absence, TMC1 is instead mislocalized away from this force transmission site. Supporting this finding, we found that LOXHD1 interacts selectively in vitro with TMC1 but not with TMC2 while also binding to channel subunits CIB2 and LHFPL5 and tip-link protein PCDH15. SUB-immunogold-SEM additionally demonstrates that LOXHD1 and TMC1 are physically connected to the lower tip-link complex in situ. Our results show that the TMC1-driven mature channels require LOXHD1 to stay coupled to the tip link and remain functional, but the TMC2-driven developmental channels do not. As both tip links and TMC1 remain present in hair bundles lacking LOXHD1, it opens the possibility to reconnect them and restore hearing for this form of genetic deafness.
Collapse
Affiliation(s)
- Pei Wang
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, 240 Pasteur Drive, Stanford, CA, USA
| | - Katharine K. Miller
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, 240 Pasteur Drive, Stanford, CA, USA
| | - Enqi He
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, 240 Pasteur Drive, Stanford, CA, USA
| | - Siddhant S. Dhawan
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, 240 Pasteur Drive, Stanford, CA, USA
| | - Christopher L. Cunningham
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Nicolas Grillet
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, 240 Pasteur Drive, Stanford, CA, USA
- Lead contact
| |
Collapse
|
38
|
Warren B, Eberl D. What can insects teach us about hearing loss? J Physiol 2024; 602:297-316. [PMID: 38128023 DOI: 10.1113/jp281281] [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: 05/26/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
Over the last three decades, insects have been utilized to provide a deep and fundamental understanding of many human diseases and disorders. Here, we present arguments for insects as models to understand general principles underlying hearing loss. Despite ∼600 million years since the last common ancestor of vertebrates and invertebrates, we share an overwhelming degree of genetic homology particularly with respect to auditory organ development and maintenance. Despite the anatomical differences between human and insect auditory organs, both share physiological principles of operation. We explain why these observations are expected and highlight areas in hearing loss research in which insects can provide insight. We start by briefly introducing the evolutionary journey of auditory organs, the reasons for using insect auditory organs for hearing loss research, and the tools and approaches available in insects. Then, the first half of the review focuses on auditory development and auditory disorders with a genetic cause. The second half analyses the physiological and genetic consequences of ageing and short- and long-term changes as a result of noise exposure. We finish with complex age and noise interactions in auditory systems. In this review, we present some of the evidence and arguments to support the use of insects to study mechanisms and potential treatments for hearing loss in humans. Obviously, insects cannot fully substitute for all aspects of human auditory function and loss of function, although there are many important questions that can be addressed in an animal model for which there are important ethical, practical and experimental advantages.
Collapse
Affiliation(s)
- Ben Warren
- Neurogenetics Group, College of Life Sciences, University of Leicester, Leicester, UK
| | - Daniel Eberl
- Department of Biology, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
39
|
Clark S, Mitra J, Elferich J, Goehring A, Ge J, Ha T, Gouaux E. Single molecule studies of the native hair cell mechanosensory transduction complex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.11.571162. [PMID: 38168376 PMCID: PMC10760052 DOI: 10.1101/2023.12.11.571162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Hearing and balance rely on the conversion of a mechanical stimulus into an electrical signal, a process known as mechanosensory transduction (MT). In vertebrates, this process is accomplished by an MT complex that is located in hair cells of the inner ear. While the past three decades of research have identified many subunits that are important for MT and revealed interactions between these subunits, the composition and organization of a functional complex remains unknown. The major challenge associated with studying the MT complex is its extremely low abundance in hair cells; current estimates of MT complex quantity range from 3-60 attomoles per cochlea or utricle, well below the detection limit of most biochemical assays that are used to characterize macromolecular complexes. Here we describe the optimization of two single molecule assays, single molecule pull-down (SiMPull) and single molecule array (SiMoA), to study the composition and quantity of native mouse MT complexes. We demonstrate that these assays are capable of detecting and quantifying low attomoles of the native MT subunits protocadherin-15 (PCDH15) and lipoma HMGIC fusion partner-like protein 5 (LHFPL5). Our results illuminate the stoichiometry of PCDH15- and LHFPL5-containing complexes and establish SiMPull and SiMoA as productive methods for probing the abundance, composition, and arrangement of subunits in the native MT complex.
Collapse
Affiliation(s)
- Sarah Clark
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
- Present address: Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Jaba Mitra
- Department of Materials Science and Engineering, University of Illinois Urbana-Champaign, IL 61801, USA
- Present address: Pacific Biosciences, Menlo Park, CA 94025, USA
| | - Johannes Elferich
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
- Present address: UMass Chan Medical School, Worcester, MA 01655, USA
| | - April Goehring
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Jingpeng Ge
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
- Present address: School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, 201210, China
| | - Taekjip Ha
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University, Baltimore, MD 21215, USA
- Present address: Howard Hughes Medical Institute, Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Present address: Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Eric Gouaux
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
- Howard Hughes Medical Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| |
Collapse
|
40
|
Smith ET, Sun P, Yu SK, Raible DW, Nicolson T. Differential expression of mechanotransduction complex genes in auditory/vestibular hair cells in zebrafish. Front Mol Neurosci 2023; 16:1274822. [PMID: 38035267 PMCID: PMC10682102 DOI: 10.3389/fnmol.2023.1274822] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/23/2023] [Indexed: 12/02/2023] Open
Abstract
Ciliated sensory cells such as photo- and olfactory receptors employ multiple types of opsins or hundreds of unique olfactory G-protein coupled receptors to respond to various wavelengths of light or odorants. With respect to hearing and balance, the mechanotransduction machinery involves fewer variants; however, emerging evidence suggests that specialization occurs at the molecular level. To address how the mechanotransduction complex varies in the inner ear, we characterized the expression of paralogous genes that encode components required for mechanotransduction in zebrafish hair cells using RNA-FISH and bioinformatic analysis. Our data indicate striking zonal differences in the expression of two components of the mechanotransduction complex which are known to physically interact, the transmembrane channel-like 1 and 2 (tmc1/2) family members and the calcium and integrin binding 2 and 3 (cib2/3) paralogues. tmc1, tmc2b, and cib3 are largely expressed in peripheral or extrastriolar hair cells, whereas tmc2a and cib2 are enriched in central or striolar hair cells. In addition, a gene implicated in deaf-blindness, ush1c, is highly enriched in a subset of extrastriolar hair cells. These results indicate that specific combinations of these components may optimize responses to mechanical stimuli in subtypes of sensory receptors within the inner ear.
Collapse
Affiliation(s)
- Eliot T. Smith
- Department of Otolaryngology-HNS, Stanford University, Stanford, CA, United States
| | - Peng Sun
- Department of Otolaryngology-HNS, Stanford University, Stanford, CA, United States
| | - Shengyang Kevin Yu
- Department of Otolaryngology-HNS and Biological Structure, Viginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, WA, United States
| | - David W. Raible
- Department of Otolaryngology-HNS and Biological Structure, Viginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, WA, United States
| | - Teresa Nicolson
- Department of Otolaryngology-HNS, Stanford University, Stanford, CA, United States
| |
Collapse
|
41
|
Derudas M, O’Reilly M, Kirkwood NK, Kenyon EJ, Grimsey S, Kitcher SR, Workman S, Bull JC, Ward SE, Kros CJ, Richardson GP. Charge and lipophilicity are required for effective block of the hair-cell mechano-electrical transducer channel by FM1-43 and its derivatives. Front Cell Dev Biol 2023; 11:1247324. [PMID: 37900280 PMCID: PMC10601989 DOI: 10.3389/fcell.2023.1247324] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 09/19/2023] [Indexed: 10/31/2023] Open
Abstract
The styryl dye FM1-43 is widely used to study endocytosis but behaves as a permeant blocker of the mechano-electrical transducer (MET) channel in sensory hair cells, loading rapidly and specifically into the cytoplasm of hair cells in a MET channel-dependent manner. Patch clamp recordings of mouse outer hair cells (OHCs) were used to determine how a series of structural modifications of FM1-43 affect MET channel block. Fluorescence microscopy was used to assess how the modifications influence hair-cell loading in mouse cochlear cultures and zebrafish neuromasts. Cochlear cultures were also used to evaluate otoprotective potential of the modified FM1-43 derivatives. Structure-activity relationships reveal that the lipophilic tail and the cationic head group of FM1-43 are both required for MET channel block in mouse cochlear OHCs; neither moiety alone is sufficient. The extent of MET channel block is augmented by increasing the lipophilicity/bulkiness of the tail, by reducing the number of positive charges in the head group from two to one, or by increasing the distance between the two charged head groups. Loading assays with zebrafish neuromasts and mouse cochlear cultures are broadly in accordance with these observations but reveal a loss of hair-cell specific labelling with increasing lipophilicity. Although FM1-43 and many of its derivatives are generally cytotoxic when tested on cochlear cultures in the presence of an equimolar concentration of the ototoxic antibiotic gentamicin (5 µM), at a 10-fold lower concentration (0.5 µM), two of the derivatives protect OHCs from cell death caused by 48 h-exposure to 5 µM gentamicin.
Collapse
Affiliation(s)
- Marco Derudas
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
- Sussex Drug Discovery Centre, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Molly O’Reilly
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
- Department of Experimental Cardiology, Academic Medical Center, Amsterdam, Netherlands
| | - Nerissa K. Kirkwood
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Emma J. Kenyon
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
- School of Medicine, Institute of Life Sciences, Swansea University, Swansea, United Kingdom
| | - Sybil Grimsey
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Siân R. Kitcher
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders NIH, Bethesda, MD, United States
| | - Shawna Workman
- Department of Biosciences, College of Science, Swansea University, Swansea, United Kingdom
| | - James C. Bull
- Department of Biosciences, College of Science, Swansea University, Swansea, United Kingdom
| | - Simon E. Ward
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
- Medicines Discovery Institute, Cardiff University, Cardiff, United Kingdom
| | - Corné J. Kros
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Guy P. Richardson
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| |
Collapse
|
42
|
Park J, Bird JE. The actin cytoskeleton in hair bundle development and hearing loss. Hear Res 2023; 436:108817. [PMID: 37300948 PMCID: PMC10408727 DOI: 10.1016/j.heares.2023.108817] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/18/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023]
Abstract
Inner ear hair cells assemble mechanosensitive hair bundles on their apical surface that transduce sounds and accelerations. Each hair bundle is comprised of ∼ 100 individual stereocilia that are arranged into rows of increasing height and width; their specific and precise architecture being necessary for mechanoelectrical transduction (MET). The actin cytoskeleton is fundamental to establishing this architecture, not only by forming the structural scaffold shaping each stereocilium, but also by composing rootlets and the cuticular plate that together provide a stable foundation supporting each stereocilium. In concert with the actin cytoskeleton, a large assortment of actin-binding proteins (ABPs) function to cross-link actin filaments into specific topologies, as well as control actin filament growth, severing, and capping. These processes are individually critical for sensory transduction and are all disrupted in hereditary forms of human hearing loss. In this review, we provide an overview of actin-based structures in the hair bundle and the molecules contributing to their assembly and functional properties. We also highlight recent advances in mechanisms driving stereocilia elongation and how these processes are tuned by MET.
Collapse
Affiliation(s)
- Jinho Park
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, United States; Myology Institute, University of Florida, Gainesville, FL 32610, United States
| | - Jonathan E Bird
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, United States; Myology Institute, University of Florida, Gainesville, FL 32610, United States.
| |
Collapse
|
43
|
Clark S, Jeong H, Posert R, Goehring A, Gouaux E. Structure of C. elegans TMC-2 complex suggests roles of lipid-mediated subunit contacts in mechanosensory transduction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.16.553618. [PMID: 37645790 PMCID: PMC10462014 DOI: 10.1101/2023.08.16.553618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Mechanotransduction is the process by which a mechanical force, such as touch, is converted into an electrical signal. Transmembrane channel-like (TMC) proteins are an evolutionarily-conserved family of ion channels whose function has been linked to a variety of mechanosensory processes, including hearing and balance sensation in vertebrates and locomotion in Drosophila. The molecular features that tune homologous TMC ion channel complexes to diverse mechanical stimuli are unknown. Caenorhabditis elegans express two TMC homologs, TMC-1 and TMC-2, both of which are the likely pore-forming subunits of mechanosensitive ion channels but differ in their expression pattern and functional role in the worm. Here we present the single particle cryo-electron microscopy structure of the native TMC-2 complex isolated from C. elegans. The complex is composed of two copies each of the pore-forming TMC-2 subunit, the calcium and integrin binding protein CALM-1 and the transmembrane inner ear protein TMIE. Comparison of the TMC-2 complex to the recently published cryo-EM structure of the C. elegans TMC-1 complex reveals differences in subunit composition and highlights conserved protein-lipid interactions, as well as other structural features, that together suggest a mechanism for TMC-mediated mechanosensory transduction.
Collapse
Affiliation(s)
- Sarah Clark
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Hanbin Jeong
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Rich Posert
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - April Goehring
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
- Howard Hughes Medical Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Eric Gouaux
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
- Howard Hughes Medical Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| |
Collapse
|
44
|
Aristizábal-Ramírez I, Dragich AK, Giese APJ, Sofia Zuluaga-Osorio K, Watkins J, Davies GK, Hadi SE, Riazuddin S, Vander Kooi CW, Ahmed ZM, Frolenkov GI. Calcium and Integrin-binding protein 2 (CIB2) controls force sensitivity of the mechanotransducer channels in cochlear outer hair cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.09.545606. [PMID: 37461484 PMCID: PMC10350036 DOI: 10.1101/2023.07.09.545606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Calcium and Integrin-Binding Protein 2 (CIB2) is an essential subunit of the mechano-electrical transduction (MET) complex in mammalian auditory hair cells. CIB2 binds to pore-forming subunits of the MET channel, TMC1/2 and is required for their transport and/or retention at the tips of mechanosensory stereocilia. Since genetic ablation of CIB2 results in complete loss of MET currents, the exact role of CIB2 in the MET complex remains elusive. Here, we generated a new mouse strain with deafness-causing p.R186W mutation in Cib2 and recorded small but still measurable MET currents in the cochlear outer hair cells. We found that R186W variant causes increase of the resting open probability of MET channels, steeper MET current dependence on hair bundle deflection (I-X curve), loss of fast adaptation, and increased leftward shifts of I-X curves upon hair cell depolarization. Combined with AlphaFold2 prediction that R186W disrupts one of the multiple interacting sites between CIB2 and TMC1/2, our data suggest that CIB2 mechanically constraints TMC1/2 conformations to ensure proper force sensitivity and dynamic range of the MET channels. Using a custom piezo-driven stiff probe deflecting the hair bundles in less than 10 µs, we also found that R186W variant slows down the activation of MET channels. This phenomenon, however, is unlikely to be due to direct effect on MET channels, since we also observed R186W-evoked disruption of the electron-dense material at the tips of mechanotransducing stereocilia and the loss of membrane-shaping BAIAP2L2 protein from the same location. We concluded that R186W variant of CIB2 disrupts force sensitivity of the MET channels and force transmission to these channels.
Collapse
|
45
|
Yamaguchi S, Kamino M, Hamamura M, Otsuguro KI. The cytosolic N-terminal region of heterologously-expressed transmembrane channel-like protein 1 (TMC1) can be cleaved in HEK293 cells. PLoS One 2023; 18:e0287249. [PMID: 37352201 PMCID: PMC10289374 DOI: 10.1371/journal.pone.0287249] [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: 03/13/2023] [Accepted: 06/01/2023] [Indexed: 06/25/2023] Open
Abstract
Transmembrane channel-like protein 1 (TMC1) is a transmembrane protein forming mechano-electrical transduction (MET) channel, which transduces mechanical stimuli into electrical signals at the top of stereocilia of hair cells in the inner ear. As an unexpected phenomenon, we found that the cytosolic N-terminal (Nt) region of heterologously-expressed mouse TMC1 (mTMC1) was localized in nuclei of a small population of the transfected HEK293 cells. This raised the possibility that the Nt region of heterologously-expressed mTMC1 was cleaved and transported into the nucleus. To confirm the cleavage, we performed western blot analyses. The results revealed that at least a fragment of the Nt region was produced from heterologously-expressed mTMC1. Site-directed mutagenesis experiments identified amino acid residues which were required to produce the fragment. The accumulation of the heterologously-expressed Nt fragment into the nuclei depended on nuclear localization signals within the Nt region. Furthermore, a structural comparison showed a similarity between the Nt region of mTMC1 and basic region leucine zipper (bZIP) transcription factors. However, transcriptome analyses using a next-generation sequencer showed that the heterologously-expression of the Nt fragment of mTMC1 hardly altered expression levels of genes. Although it is still unknown what is the precise mechanism and the physiological significance of this cleavage, these results showed that the cytosolic Nt region of heterologously-expressed mTMC1 could be cleaved in HEK293 cells. Therefore, it should be taken into account that the cleavage of Nt region might influence the functional analysis of TMC1 by the heterologous-expression system using HEK293 cells.
Collapse
Affiliation(s)
- Soichiro Yamaguchi
- Laboratory of Physiology, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Maho Kamino
- Laboratory of Pharmacology, Department of Basic Veterinary Sciences, School of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Maho Hamamura
- Laboratory of Pharmacology, Department of Basic Veterinary Sciences, School of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Ken-ichi Otsuguro
- Laboratory of Pharmacology, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
46
|
Stanley OR, Swaminathan A, Wojahn E, Ahmed ZM, Cullen KE. An Open-Source Tool for Automated Human-Level Circling Behavior Detection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.30.540066. [PMID: 37398316 PMCID: PMC10312579 DOI: 10.1101/2023.05.30.540066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Quantifying behavior and relating it to underlying biological states is of paramount importance in many life science fields. Although barriers to recording postural data have been reduced by progress in deep-learning-based computer vision tools for keypoint tracking, extracting specific behaviors from this data remains challenging. Manual behavior coding, the present gold standard, is labor-intensive and subject to intra- and inter-observer variability. Automatic methods are stymied by the difficulty of explicitly defining complex behaviors, even ones which appear obvious to the human eye. Here, we demonstrate an effective technique for detecting one such behavior, a form of locomotion characterized by stereotyped spinning, termed 'circling'. Though circling has an extensive history as a behavioral marker, at present there exists no standard automated detection method. Accordingly, we developed a technique to identify instances of the behavior by applying simple postprocessing to markerless keypoint data from videos of freely-exploring (Cib2-/-;Cib3-/-) mutant mice, a strain we previously found to exhibit circling. Our technique agrees with human consensus at the same level as do individual observers, and it achieves >90% accuracy in discriminating videos of wild type mice from videos of mutants. As using this technique requires no experience writing or modifying code, it also provides a convenient, noninvasive, quantitative tool for analyzing circling mouse models. Additionally, as our approach was agnostic to the underlying behavior, these results support the feasibility of algorithmically detecting specific, research-relevant behaviors using readily-interpretable parameters tuned on the basis of human consensus.
Collapse
Affiliation(s)
- O R Stanley
- Dept. Biomedical Engineering; Johns Hopkins University
| | - A Swaminathan
- Dept. Biomedical Engineering; Johns Hopkins University
| | - E Wojahn
- Dept. Biomedical Engineering; Johns Hopkins University
| | - Z M Ahmed
- Depts. Otorhinolaryngology-Head & Neck Surgery, Biochemistry & Molecular Biology, Ophthalmology; University of Maryland School of Medicine
| | - K E Cullen
- Dept. Biomedical Engineering; Johns Hopkins University
- Depts. Neuroscience, Otolaryngology-Head & Neck Surgery, Johns Hopkins University
| |
Collapse
|
47
|
Wang X, Liu S, Cheng Q, Qu C, Ren R, Du H, Li N, Yan K, Wang Y, Xiong W, Xu Z. CIB2 and CIB3 Regulate Stereocilia Maintenance and Mechanoelectrical Transduction in Mouse Vestibular Hair Cells. J Neurosci 2023; 43:3219-3231. [PMID: 37001993 PMCID: PMC10162464 DOI: 10.1523/jneurosci.1807-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 03/20/2023] [Accepted: 03/25/2023] [Indexed: 04/03/2023] Open
Abstract
The mechanoelectrical transduction (MET) protein complex in the inner-ear hair cells is essential for hearing and balance perception. Calcium and integrin-binding protein 2 (CIB2) has been reported to be a component of MET complex, and loss of CIB2 completely abolishes MET currents in auditory hair cells, causing profound congenital hearing loss. However, loss of CIB2 does not affect MET currents in vestibular hair cells (VHCs) as well as general balance function. Here, we show that CIB2 and CIB3 act redundantly to regulate MET in VHCs, as MET currents are completely abolished in the VHCs of Cib2/Cib3 double knock-out mice of either sex. Furthermore, we show that Cib2 and Cib3 transcripts have complementary expression patterns in the vestibular maculae, and that they play different roles in stereocilia maintenance in VHCs. Cib2 transcripts are highly expressed in the striolar region, and knock-out of Cib2 affects stereocilia maintenance in striolar VHCs. In contrast, Cib3 transcripts are highly expressed in the extrastriolar region, and knock-out of Cib3 mainly affects stereocilia maintenance in extrastriolar VHCs. Simultaneous knock-out of Cib2 and Cib3 affects stereocilia maintenance in all VHCs and leads to severe balance deficits. Taken together, our present work reveals that CIB2 and CIB3 are important for stereocilia maintenance as well as MET in mouse VHCs.SIGNIFICANCE STATEMENT Calcium and integrin-binding protein 2 (CIB2) is an important component of mechanoelectrical transduction (MET) complex, and loss of CIB2 completely abolishes MET in auditory hair cells. However, MET is unaffected in Cib2 knock-out vestibular hair cells (VHCs). In the present work, we show that CIB3 could compensate for the loss of CIB2 in VHCs, and Cib2/Cib3 double knock-out completely abolishes MET in VHCs. Interestingly, CIB2 and CIB3 could also regulate VHC stereocilia maintenance in a nonredundant way. Cib2 and Cib3 transcripts are highly expressed in the striolar and extrastriolar regions, respectively. Stereocilia maintenance and balance function are differently affected in Cib2 or Cib3 knock-out mice. In conclusion, our data suggest that CIB2 and CIB3 are important for stereocilia maintenance and MET in mouse VHCs.
Collapse
Affiliation(s)
- Xiaoying Wang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, People's Republic of China
| | - Shuang Liu
- School of Life Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, People's Republic of China
- Chinese Institute for Brain Research, Beijing 102206, People's Republic of China
| | - Qi Cheng
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, People's Republic of China
| | - Chengli Qu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, People's Republic of China
| | - Rui Ren
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, People's Republic of China
| | - Haibo Du
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, People's Republic of China
| | - Nana Li
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, People's Republic of China
| | - 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 Sciences, Shandong University, Qingdao, Shandong 266237, People's Republic of China
| | - Yanfei Wang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, People's Republic of China
| | - Wei Xiong
- School of Life Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, People's Republic of China
- Chinese Institute for Brain Research, Beijing 102206, People's Republic of China
| | - Zhigang Xu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, People's Republic of China
- Shandong Provincial Collaborative Innovation Center of Cell Biology, Shandong Normal University, Jinan, Shandong 250014, People's Republic of China
| |
Collapse
|
48
|
Davis MJ, Earley S, Li YS, Chien S. Vascular mechanotransduction. Physiol Rev 2023; 103:1247-1421. [PMID: 36603156 PMCID: PMC9942936 DOI: 10.1152/physrev.00053.2021] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno, Nevada
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
- Department of Medicine, University of California, San Diego, California
| |
Collapse
|
49
|
Krey JF, Chatterjee P, Halford J, Cunningham CL, Perrin BJ, Barr-Gillespie PG. Control of stereocilia length during development of hair bundles. PLoS Biol 2023; 21:e3001964. [PMID: 37011103 PMCID: PMC10101650 DOI: 10.1371/journal.pbio.3001964] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 04/13/2023] [Accepted: 03/14/2023] [Indexed: 04/05/2023] Open
Abstract
Assembly of the hair bundle, the sensory organelle of the inner ear, depends on differential growth of actin-based stereocilia. Separate rows of stereocilia, labeled 1 through 3 from tallest to shortest, lengthen or shorten during discrete time intervals during development. We used lattice structured illumination microscopy and surface rendering to measure dimensions of stereocilia from mouse apical inner hair cells during early postnatal development; these measurements revealed a sharp transition at postnatal day 8 between stage III (row 1 and 2 widening; row 2 shortening) and stage IV (final row 1 lengthening and widening). Tip proteins that determine row 1 lengthening did not accumulate simultaneously during stages III and IV; while the actin-bundling protein EPS8 peaked at the end of stage III, GNAI3 peaked several days later-in early stage IV-and GPSM2 peaked near the end of stage IV. To establish the contributions of key macromolecular assemblies to bundle structure, we examined mouse mutants that eliminated tip links (Cdh23v2J or Pcdh15av3J), transduction channels (TmieKO), or the row 1 tip complex (Myo15ash2). Cdh23v2J/v2J and Pcdh15av3J/av3J bundles had adjacent stereocilia in the same row that were not matched in length, revealing that a major role of these cadherins is to synchronize lengths of side-by-side stereocilia. Use of the tip-link mutants also allowed us to distinguish the role of transduction from effects of transduction proteins themselves. While levels of GNAI3 and GPSM2, which stimulate stereocilia elongation, were greatly attenuated at the tips of TmieKO/KO row 1 stereocilia, they accumulated normally in Cdh23v2J/v2J and Pcdh15av3J/av3J stereocilia. These results reinforced the suggestion that the transduction proteins themselves facilitate localization of proteins in the row 1 complex. By contrast, EPS8 concentrates at tips of all TmieKO/KO, Cdh23v2J/v2J, and Pcdh15av3J/av3J stereocilia, correlating with the less polarized distribution of stereocilia lengths in these bundles. These latter results indicated that in wild-type hair cells, the transduction complex prevents accumulation of EPS8 at the tips of shorter stereocilia, causing them to shrink (rows 2 and 3) or disappear (row 4 and microvilli). Reduced rhodamine-actin labeling at row 2 stereocilia tips of tip-link and transduction mutants suggests that transduction's role is to destabilize actin filaments there. These results suggest that regulation of stereocilia length occurs through EPS8 and that CDH23 and PCDH15 regulate stereocilia lengthening beyond their role in gating mechanotransduction channels.
Collapse
Affiliation(s)
- Jocelyn F. Krey
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Paroma Chatterjee
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Julia Halford
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Christopher L. Cunningham
- Pittsburgh Hearing Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Benjamin J. Perrin
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| | - Peter G. Barr-Gillespie
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| |
Collapse
|
50
|
The tetraspan LHFPL5 is critical to establish maximal force sensitivity of the mechanotransduction channel of cochlear hair cells. Cell Rep 2023; 42:112245. [PMID: 36917610 DOI: 10.1016/j.celrep.2023.112245] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/28/2023] [Accepted: 02/27/2023] [Indexed: 03/14/2023] Open
Abstract
The mechanoelectrical transduction (MET) channel of cochlear hair cells is gated by the tip link, but the mechanisms that establish the exquisite force sensitivity of this MET channel are not known. Here, we show that the tetraspan lipoma HMGIC fusion partner-like 5 (LHFPL5) directly couples the tip link to the MET channel. Disruption of these interactions severely perturbs MET. Notably, the N-terminal cytoplasmic domain of LHFPL5 binds to an amphipathic helix in TMC1, a critical gating domain conserved between different MET channels. Mutations in the amphipathic helix of TMC1 or in the N-terminus of LHFPL5 that perturb interactions of LHFPL5 with the amphipathic helix affect channel responses to mechanical force. We conclude that LHFPL5 couples the tip link to the MET channel and that channel gating depends on a structural element in TMC1 that is evolutionarily conserved between MET channels. Overall, our findings support a tether model for transduction channel gating by the tip link.
Collapse
|