1
|
Li Q, Sun S, Zuo B, Lian C, Tang W, Xu H, Lu W. Novel Clinical Manifestation and Favorable Treatment Outcome of Cochlear Implant in a Chinese Family With Likely Pathogenic Variant of the P2RX2 Gene. Am J Med Genet A 2025; 197:e63877. [PMID: 39258340 DOI: 10.1002/ajmg.a.63877] [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: 12/19/2023] [Accepted: 08/29/2024] [Indexed: 09/12/2024]
Abstract
The rapid development and clinical application of sequencing technologies enable the genetic diagnosis of inherited deafness. P2RX2, as the gene responsible for autosomal dominant non-syndromic deafness-41 (DFNA41), has been proven to be essential for life-long normal hearing and for the protection of noise-induced hearing loss (NIHL). Our present study reports a missense variant in the P2RX2 gene (c.178G > T (p.V60L)), for the second time worldwide, in a five-generation kindred living in Henan, China. Despite carrying the same variant, the affected members in this family appear to present with earlier-onset hearing loss and poorer hearing compared to the original DFNA41 families. In addition, this study supplements some content that was not covered in previous reports. We quantitatively evaluated the pain perception ability of some members using the Pain Vision PS-2100 system, and further found an interesting clinical manifestation, that is, hyperalgesia, in heterozygotes for P2RX2 p.V60L. The cochlear implant (CI) was also provided for the proband of profound deafness, resulting in satisfactory clinical outcomes. Finally, we carried out a systematic review of recently published articles on the P2RX2 gene, which is beneficial for better understanding the role of the P2RX2 gene in the auditory system and the pathogenic mechanisms in sensorineural hearing loss (SNHL).
Collapse
Affiliation(s)
- Qiang Li
- Department of Otorhinolaryngology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuping Sun
- Department of Otorhinolaryngology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bin Zuo
- Department of Otorhinolaryngology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chengyu Lian
- Precision Medicine Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Wenxue Tang
- Precision Medicine Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- The Research and Application Center of Precision Medicine, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongen Xu
- Precision Medicine Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- The Research and Application Center of Precision Medicine, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Lu
- Department of Otorhinolaryngology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
2
|
Yu Y, Li Y, Wen C, Yang F, Chen X, Yi W, Deng L, Cheng X, Yu N, Huang L. High-frequency hearing vulnerability associated with the different supporting potential of Hensen's cells: SMART-Seq2 RNA sequencing. Biosci Trends 2024; 18:165-175. [PMID: 38583982 DOI: 10.5582/bst.2024.01044] [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] [Indexed: 04/09/2024]
Abstract
Hearing loss is the third most prevalent physical condition affecting communication, well-being, and healthcare costs. Sensorineural hearing loss often occurs first in the high-frequency region (basal turn), then towards the low-frequency region (apical turn). However, the mechanism is still unclear. Supporting cells play a critical role in the maintenance of normal cochlear function. The function and supporting capacity of these cells may be different from different frequency regions. Hensen's cells are one of the unique supporting cell types characterized by lipid droplets (LDs) in the cytoplasm. Here, we investigated the morphological and gene expression differences of Hensen's cells along the cochlear axis. We observed a gradient change in the morphological characteristics of Hensen's cells along the cochlear tonotopic axis, with larger and more abundant LDs observed in apical Hensen's cells. Smart-seq2 RNA-seq revealed differentially expressed genes (DEGs) between apical and basal Hensen's cells that clustered in several pathways, including unsaturated fatty acid biosynthesis, cholesterol metabolism, and fatty acid catabolism, which are associated with different energy storage capacities and metabolic potential. These findings suggest potential differences in lipid metabolism and oxidative energy supply between apical and basal Hensen's cells, which is consistent with the morphological differences of Hensen's cells. We also found differential expression patterns of candidate genes associated with hereditary hearing loss (HHL), noise-induced hearing loss (NIHL), and age-related hearing loss (ARHL). These findings indicate functional heterogeneity of SCs along the cochlear axis, contribute to our understanding of cochlear physiology and provide molecular basis evidence for future studies of hearing loss.
Collapse
Affiliation(s)
- Yiding Yu
- Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Otolaryngology, Beijing, China
- Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Beijing, China
| | - Yue Li
- Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Otolaryngology, Beijing, China
- Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Beijing, China
| | - Cheng Wen
- Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Otolaryngology, Beijing, China
- Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Beijing, China
| | - Fengbo Yang
- Otolaryngology Head and Neck Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xuemin Chen
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, China
| | - Wenqi Yi
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, China
| | - Lin Deng
- Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Otolaryngology, Beijing, China
- Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Beijing, China
| | - Xiaohua Cheng
- Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Otolaryngology, Beijing, China
- Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Beijing, China
| | - Ning Yu
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, China
| | - Lihui Huang
- Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Otolaryngology, Beijing, China
- Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Beijing, China
| |
Collapse
|
3
|
The purinergic receptors 2X3 on spiral ganglion neurons enhance the medial olivocochlear reflex in mice after long-term moderate noise exposure. Neuroreport 2022; 33:786-790. [PMID: 36367795 DOI: 10.1097/wnr.0000000000001846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Our purpose was to study the expression of purinergic receptors 2X2 (P2X2) and purinergic receptors 2X3 (P2X3) in spiral ganglion neurons (SGNs), the afferent nerves of medial olivocochlear (MOC) reflex, after long-term moderate noise exposure, and its relationship with the enhancement of MOC reflex. Mice were exposed a moderate broadband noise for 4 weeks consecutively. Then mouse hearing functions, including threshold auditory brainstem responses, distortion-product otoacoustic emissions, and MOC reflex, were evaluated and the expression of P2X2 and P2X3 on SGNs were assessed by cochlear immunofluorescence. AF-353 was injected before each noise exposure. Four weeks later, mice were also tested for hearing functions and expression of P2X2 and P2X3 on SGNs. The long-term moderate noise strengthened MOC reflex, and AF-353 reduced it in mice and P2X3 expression on SGNs increased after long-term moderate noise exposure, and AF-353 can downregulate it. The P2X3 on SGNs of mice increased after long-term moderate noise exposure, and the upregulation of it mediated the enhancement of MOC reflex.
Collapse
|
4
|
Chen J, Chen P, He B, Gong T, Li Y, Zhang J, Lv J, Mammano F, Hou S, Yang J. Connexin30-Deficiency Causes Mild Hearing Loss With the Reduction of Endocochlear Potential and ATP Release. Front Cell Neurosci 2022; 15:819194. [PMID: 35110999 PMCID: PMC8802669 DOI: 10.3389/fncel.2021.819194] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 12/22/2021] [Indexed: 12/26/2022] Open
Abstract
GJB2 and GJB6 are adjacent genes encoding connexin 26 (Cx26) and connexin 30 (Cx30), respectively, with overlapping expressions in the inner ear. Both genes are associated with the commonest monogenic hearing disorder, recessive isolated deafness DFNB1. Cx26 plays an important role in auditory development, while the role of Cx30 in hearing remains controversial. Previous studies found that Cx30 knockout mice had severe hearing loss along with a 90% reduction in Cx26, while another Cx30 knockout mouse model showed normal hearing with nearly half of Cx26 preserved. In this study, we used CRISPR/Cas9 technology to establish a new Cx30 knockout mouse model (Cx30−/−), which preserves approximately 70% of Cx26. We found that the 1, 3, and 6-month-old Cx30−/− mice showed mild hearing loss at full frequency. Immunofluorescence and HE staining suggested no significant differences in microstructure of the cochlea between Cx30−/− mice and wild-type mice. However, transmission electron microscopy showed slight cavity-like damage in the stria vascularis of Cx30−/− mice. And Cx30 deficiency reduced the production of endocochlear potential (EP) and the release of ATP, which may have induced hearing loss. Taken together, this study showed that lack of Cx30 can lead to hearing loss with an approximately 30% reduction of Cx26 in the present Cx30 knockout model. Hence, Cx30 may play an important rather than redundant role in hearing development.
Collapse
Affiliation(s)
- Junmin Chen
- Department of Otorhinolaryngology—Head & Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Penghui Chen
- Department of Otorhinolaryngology—Head & Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Baihui He
- Department of Otorhinolaryngology—Head & Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Tianyu Gong
- Department of Otorhinolaryngology—Head & Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Yue Li
- Department of Otorhinolaryngology—Head & Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Jifang Zhang
- Department of Otorhinolaryngology—Head & Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Jingrong Lv
- Department of Otorhinolaryngology—Head & Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Fabio Mammano
- Department of Physics and Astronomy “G. Galilei”, University of Padua, Padua, Italy
- Department of Biomedical Sciences, Institute of Cell Biology and Neurobiology, Italian National Research Council, Monterotondo, Italy
| | - Shule Hou
- Department of Otorhinolaryngology—Head & Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
- *Correspondence: Shule Hou Jun Yang
| | - Jun Yang
- Department of Otorhinolaryngology—Head & Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
- *Correspondence: Shule Hou Jun Yang
| |
Collapse
|
5
|
Identification of a Novel Stop Loss Mutation in P2RX2 Gene in an Iranian Family with Autosomal Nonsyndromic Hearing Loss. IRANIAN BIOMEDICAL JOURNAL 2021; 25:368-73. [PMID: 34425661 PMCID: PMC8487680 DOI: 10.52547/ibj.25.5.368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Background: Hearing loss, a congenital genetic disorder in human, is difficult to diagnose. WES is a powerful approach for ethiological disgnosis of such disorders. Methods: One Iranian family with two patients were attented in the study. Sequencing of known NSHL genes was carried out to recognize the genetic causes of HL. Results: Molecular analyses identified a novel stop loss mutation, c.1048T>G (p.Term350Glu), whitin the P2RX2 gene, causing a termination-site modification.This event would lead to continued translation into the 3' UTR of the gene, which in turn may result in a longer protein product. The mutation was segregating with the disease phenotype and predicted to be pathogenic by bioinformatic tools. Conclusion: This study is the first Iranian case report of a diagnosis of ADNSHL caused by P2RX2 mutation. The recognition of other causative mutations in P2RX2 gene more supports the probable function of this gene in causing ADNSHL.
Collapse
|
6
|
Cochlear homeostasis: a molecular physiological perspective on maintenance of sound transduction and auditory neurotransmission with noise and ageing. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.09.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
7
|
Holman HA, Wan Y, Rabbitt RD. Developmental GAD2 Expression Reveals Progenitor-like Cells with Calcium Waves in Mammalian Crista Ampullaris. iScience 2020; 23:101407. [PMID: 32771977 PMCID: PMC7415930 DOI: 10.1016/j.isci.2020.101407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/29/2020] [Accepted: 07/21/2020] [Indexed: 01/26/2023] Open
Abstract
Sense of motion, spatial orientation, and balance in vertebrates relies on sensory hair cells in the inner ear vestibular system. Vestibular supporting cells can regenerate hair cells that are lost from aging, ototoxicity, and trauma, although not all factors or specific cell types are known. Here we report a population of GAD2-positive cells in the mouse crista ampullaris and trace GAD2 progenitor-like cells that express pluripotent transcription factors SOX2, PROX1, and CTBP2. GAD2 progenitor-like cells organize into rosettes around a central branched structure in the eminentia cruciatum (EC) herein named the EC plexus. GCaMP5G calcium indicator shows spontaneous and acetylcholine-evoked whole-cell calcium waves in neonatal and adult mice. We present a hypothetical model that outlines the lineage and potential regenerative capacity of GAD2 cells in the mammalian vestibular neuroepithelium.
Collapse
Affiliation(s)
- Holly A Holman
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| | - Yong Wan
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Richard D Rabbitt
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA; Graduate Program in Neuroscience, University of Utah, Salt Lake City, UT 84112, USA; Department of Otolaryngology-Head & Neck Surgery, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
8
|
Jimenez JE, Nourbakhsh A, Colbert B, Mittal R, Yan D, Green CL, Nisenbaum E, Liu G, Bencie N, Rudman J, Blanton SH, Zhong Liu X. Diagnostic and therapeutic applications of genomic medicine in progressive, late-onset, nonsyndromic sensorineural hearing loss. Gene 2020; 747:144677. [PMID: 32304785 PMCID: PMC7244213 DOI: 10.1016/j.gene.2020.144677] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 04/12/2020] [Accepted: 04/14/2020] [Indexed: 02/06/2023]
Abstract
The progressive, late-onset, nonsyndromic, sensorineural hearing loss (PNSHL) is the most common cause of sensory impairment globally, with presbycusis affecting greater than a third of individuals over the age of 65. The etiology underlying PNSHL include presbycusis, noise-induced hearing loss, drug ototoxicity, and delayed-onset autosomal dominant hearing loss (AD PNSHL). The objective of this article is to discuss the potential diagnostic and therapeutic applications of genomic medicine in PNSHL. Genomic factors contribute greatly to PNSHL. The heritability of presbycusis ranges from 25 to 75%. Current therapies for PNSHL range from sound amplification to cochlear implantation (CI). PNSHL is an excellent candidate for genomic medicine approaches as it is common, has well-described pathophysiology, has a wide time window for treatment, and is amenable to local gene therapy by currently utilized procedural approaches. AD PNSHL is especially suited to genomic medicine approaches that can disrupt the expression of an aberrant protein product. Gene therapy is emerging as a potential therapeutic strategy for the treatment of PNSHL. Viral gene delivery approaches have demonstrated promising results in human clinical trials for two inherited causes of blindness and are being used for PNSHL in animal models and a human trial. Non-viral gene therapy approaches are useful in situations where a transient biologic effect is needed or for delivery of genome editing reagents (such as CRISPR/Cas9) into the inner ear. Many gene therapy modalities that have proven efficacious in animal trials have potential to delay or prevent PNSHL in humans. The development of new treatment modalities for PNSHL will lead to improved quality of life of many affected individuals and their families.
Collapse
Affiliation(s)
- Joaquin E Jimenez
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Aida Nourbakhsh
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Brett Colbert
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Human Genetics and John P. Hussman Institute of Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA; Medical Scientist Training Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Rahul Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Denise Yan
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Carlos L Green
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Eric Nisenbaum
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - George Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Nicole Bencie
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jason Rudman
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Susan H Blanton
- Department of Human Genetics and John P. Hussman Institute of Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Xue Zhong Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Human Genetics and John P. Hussman Institute of Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
9
|
Stokes L, Bidula S, Bibič L, Allum E. To Inhibit or Enhance? Is There a Benefit to Positive Allosteric Modulation of P2X Receptors? Front Pharmacol 2020; 11:627. [PMID: 32477120 PMCID: PMC7235284 DOI: 10.3389/fphar.2020.00627] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/21/2020] [Indexed: 12/15/2022] Open
Abstract
The family of ligand-gated ion channels known as P2X receptors were discovered several decades ago. Since the cloning of the seven P2X receptors (P2X1-P2X7), a huge research effort has elucidated their roles in regulating a range of physiological and pathophysiological processes. Transgenic animals have been influential in understanding which P2X receptors could be new therapeutic targets for disease. Furthermore, understanding how inherited mutations can increase susceptibility to disorders and diseases has advanced this knowledge base. There has been an emphasis on the discovery and development of pharmacological tools to help dissect the individual roles of P2X receptors and the pharmaceutical industry has been involved in pushing forward clinical development of several lead compounds. During the discovery phase, a number of positive allosteric modulators have been described for P2X receptors and these have been useful in assigning physiological roles to receptors. This review will consider the major physiological roles of P2X1-P2X7 and discuss whether enhancement of P2X receptor activity would offer any therapeutic benefit. We will review what is known about identified compounds acting as positive allosteric modulators and the recent identification of drug binding pockets for such modulators.
Collapse
Affiliation(s)
- Leanne Stokes
- School of Pharmacy, University of East Anglia, Norwich, United Kingdom
| | - Stefan Bidula
- School of Pharmacy, University of East Anglia, Norwich, United Kingdom
| | - Lučka Bibič
- School of Pharmacy, University of East Anglia, Norwich, United Kingdom
| | - Elizabeth Allum
- School of Pharmacy, University of East Anglia, Norwich, United Kingdom
| |
Collapse
|
10
|
Warren B, Fenton GE, Klenschi E, Windmill JFC, French AS. Physiological Basis of Noise-Induced Hearing Loss in a Tympanal Ear. J Neurosci 2020; 40:3130-3140. [PMID: 32144181 PMCID: PMC7141877 DOI: 10.1523/jneurosci.2279-19.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/19/2019] [Accepted: 12/08/2019] [Indexed: 11/30/2022] Open
Abstract
Acoustic overexposure, such as listening to loud music too often, results in noise-induced hearing loss. The pathologies of this prevalent sensory disorder begin within the ear at synapses of the primary auditory receptors, their postsynaptic partners and their supporting cells. The extent of noise-induced damage, however, is determined by overstimulation of primary auditory receptors, upstream of where the pathologies manifest. A systematic characterization of the electrophysiological function of the upstream primary auditory receptors is warranted to understand how noise exposure impacts on downstream targets, where the pathologies of hearing loss begin. Here, we used the experimentally-accessible locust ear (male, Schistocerca gregaria) to characterize a decrease in the auditory receptor's ability to respond to sound after noise exposure. Surprisingly, after noise exposure, the electrophysiological properties of the auditory receptors remain unchanged, despite a decrease in the ability to transduce sound. This auditory deficit stems from changes in a specialized receptor lymph that bathes the auditory receptors, revealing striking parallels with the mammalian auditory system.SIGNIFICANCE STATEMENT Noise exposure is the largest preventable cause of hearing loss. It is the auditory receptors that bear the initial brunt of excessive acoustic stimulation, because they must convert excessive sound-induced movements into electrical signals, but remain functional afterward. Here we use the accessible ear of an invertebrate to, for the first time in any animal, characterize changes in auditory receptors after noise overexposure. We find that their decreased ability to transduce sound into electrical signals is, most probably, due to changes in supporting (scolopale) cells that maintain the ionic composition of the ear. An emerging doctrine in hearing research is that vertebrate primary auditory receptors are surprisingly robust, something that we show rings true for invertebrate ears too.
Collapse
Affiliation(s)
- Ben Warren
- Department of Neuroscience, Psychology and Behavior, University of Leicester, Leicester, LE1 7RH, United Kingdom,
| | - Georgina E Fenton
- Department of Neuroscience, Psychology and Behavior, University of Leicester, Leicester, LE1 7RH, United Kingdom
| | - Elizabeth Klenschi
- Centre for Ultrasonic Engineering, Department of Electronic and Electrical Engineering, University of Strathclyde, Glasgow, G1 1XW, United Kingdom, and
| | - James F C Windmill
- Centre for Ultrasonic Engineering, Department of Electronic and Electrical Engineering, University of Strathclyde, Glasgow, G1 1XW, United Kingdom, and
| | - Andrew S French
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| |
Collapse
|
11
|
Warren B, Fenton GE, Klenschi E, Windmill JFC, French AS. Physiological Basis of Noise-Induced Hearing Loss in a Tympanal Ear. J Neurosci 2020. [PMID: 32144181 DOI: 10.3760/cma.j.cn112137-20200803-02267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Acoustic overexposure, such as listening to loud music too often, results in noise-induced hearing loss. The pathologies of this prevalent sensory disorder begin within the ear at synapses of the primary auditory receptors, their postsynaptic partners and their supporting cells. The extent of noise-induced damage, however, is determined by overstimulation of primary auditory receptors, upstream of where the pathologies manifest. A systematic characterization of the electrophysiological function of the upstream primary auditory receptors is warranted to understand how noise exposure impacts on downstream targets, where the pathologies of hearing loss begin. Here, we used the experimentally-accessible locust ear (male, Schistocerca gregaria) to characterize a decrease in the auditory receptor's ability to respond to sound after noise exposure. Surprisingly, after noise exposure, the electrophysiological properties of the auditory receptors remain unchanged, despite a decrease in the ability to transduce sound. This auditory deficit stems from changes in a specialized receptor lymph that bathes the auditory receptors, revealing striking parallels with the mammalian auditory system.SIGNIFICANCE STATEMENT Noise exposure is the largest preventable cause of hearing loss. It is the auditory receptors that bear the initial brunt of excessive acoustic stimulation, because they must convert excessive sound-induced movements into electrical signals, but remain functional afterward. Here we use the accessible ear of an invertebrate to, for the first time in any animal, characterize changes in auditory receptors after noise overexposure. We find that their decreased ability to transduce sound into electrical signals is, most probably, due to changes in supporting (scolopale) cells that maintain the ionic composition of the ear. An emerging doctrine in hearing research is that vertebrate primary auditory receptors are surprisingly robust, something that we show rings true for invertebrate ears too.
Collapse
Affiliation(s)
- Ben Warren
- Department of Neuroscience, Psychology and Behavior, University of Leicester, Leicester, LE1 7RH, United Kingdom,
| | - Georgina E Fenton
- Department of Neuroscience, Psychology and Behavior, University of Leicester, Leicester, LE1 7RH, United Kingdom
| | - Elizabeth Klenschi
- Centre for Ultrasonic Engineering, Department of Electronic and Electrical Engineering, University of Strathclyde, Glasgow, G1 1XW, United Kingdom, and
| | - James F C Windmill
- Centre for Ultrasonic Engineering, Department of Electronic and Electrical Engineering, University of Strathclyde, Glasgow, G1 1XW, United Kingdom, and
| | - Andrew S French
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| |
Collapse
|
12
|
Hearing loss mutations alter the functional properties of human P2X2 receptor channels through distinct mechanisms. Proc Natl Acad Sci U S A 2019; 116:22862-22871. [PMID: 31636190 DOI: 10.1073/pnas.1912156116] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Activation of P2X2 receptor channels by extracellular ATP is thought to play important roles in cochlear adaptation to elevated sound levels and protection from overstimulation. Each subunit of a trimeric P2X2 receptor is composed of intracellular N and C termini, a large extracellular domain containing the ATP binding site and 2 transmembrane helices (TM1 and TM2) that form a cation permeable pore. Whole-exome sequencing and linkage analysis have identified 3 hP2X2 receptor mutations (V60L, D273Y, and G353R) that cause dominantly inherited progressive sensorineural hearing loss (DFNA41). Available structures of related P2X receptors suggest that these 3 mutations localize to TM1 (V60L), TM2 (G353R), or the β-sheet linking the TMs to the extracellular ATP binding sites (D273Y). Previous studies have concluded that the V60L and G353R mutants are nonfunctional, whereas the D273Y mutant has yet to be studied. Here, we demonstrate that both V60L and G353R mutations do form functional channels, whereas the D273Y mutation prevents the expression of functional channels on the cell membrane. Our results show that the V60L mutant forms constitutively active channels that are insensitive to ATP or the antagonist suramin, suggesting uncoupling of the pore and the ligand binding domains. In contrast, the G353R mutant can be activated by ATP but exhibits alterations in sensitivity to ATP, inward rectification, and ion selectivity. Collectively, our results demonstrate that the loss of functional P2X2 receptors or distinct alterations of its functional properties lead to noise-induced hearing loss, highlighting the importance of these channels in preserving hearing.
Collapse
|
13
|
Berekméri E, Fekete Á, Köles L, Zelles T. Postnatal Development of the Subcellular Structures and Purinergic Signaling of Deiters' Cells along the Tonotopic Axis of the Cochlea. Cells 2019; 8:cells8101266. [PMID: 31627326 PMCID: PMC6830339 DOI: 10.3390/cells8101266] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/03/2019] [Accepted: 10/15/2019] [Indexed: 01/04/2023] Open
Abstract
Exploring the development of the hearing organ helps in the understanding of hearing and hearing impairments and it promotes the development of the regenerative approaches-based therapeutic efforts. The role of supporting cells in the development of the organ of Corti is much less elucidated than that of the cochlear sensory receptor cells. The use of our recently published method of single-cell electroporation loading of a fluorescent Ca2+ probe in the mouse hemicochlea preparation provided an appropriate means to investigate the Deiters’ cells at the subcellular level in two different cochlear turns (apical, middle). Deiters’ cell’s soma and process elongated, and the process became slimmer by maturation without tonotopic preference. The tonotopically heterogeneous spontaneous Ca2+ activity less frequently occurred by maturation and implied subcellular difference. The exogenous ATP- and UTP-evoked Ca2+ responses were maturation-dependent and showed P2Y receptor dominance in the apical turn. By monitoring the basic structural dimensions of this supporting cell type as well as its spontaneous and evoked purinergic Ca2+ signaling in the hemicochlea preparation in different stages in the critical postnatal P5-25 developmental period for the first time, we showed that the soma and the phalangeal process of the Deiters’ cells go through age- and tonotopy-dependent changes in the morphometric parameters and purinergic signaling.
Collapse
Affiliation(s)
- Eszter Berekméri
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4., 1089 Budapest, Hungary.
- Department of Ecology, University of Veterinary Medicine, Rottenbiller u. 50., 1077 Budapest, Hungary.
| | - Ádám Fekete
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, 555 University Ave, Toronto, ON M5G 1X8, Canada.
| | - László Köles
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4., 1089 Budapest, Hungary.
| | - Tibor Zelles
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4., 1089 Budapest, Hungary.
- Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony u. 43., 1083 Budapest, Hungary.
| |
Collapse
|
14
|
Liu XZ, Yan D, Mittal R, Ballard ME, Feng Y. Progressive Dominant Hearing Loss (Autosomal Dominant Deafness-41) and P2RX2 Gene Mutations: A Phenotype-Genotype Study. Laryngoscope 2019; 130:1657-1663. [PMID: 31593348 DOI: 10.1002/lary.28318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/29/2019] [Accepted: 08/28/2019] [Indexed: 01/14/2023]
Abstract
OBJECTIVES/HYPOTHESIS P2RX2 encoding P2X purinoreceptor 2 has been identified as the gene responsible for autosomal dominant deafness-41 (DFNA41) as well as mediating vulnerability to noise-induced hearing loss (NIHL). The objective of this study was to investigate the audiological and molecular characteristics of P2RX2-related deafness, with emphasis on its role in NIHL by determining the audiological characteristics of a previously reported six-generation DFNA41 family with a 10-year follow-up. We have also summarized phenotype-genotype correlations of P2RX2-related deafness in human and mouse models. STUDY DESIGN We describe clinical longitudinal follow-up in the DFNA41 family with P2RX2 (p.Val60Leu) mutation and perform a systematic literature search in PubMed and poster presentations on meeting/conference websites to identify current insights into P2RX2-mediated NIHL. METHODS Clinical and physical examinations of the family members were performed, and audiograms were obtained to assess the hearing thresholds. Clinical follow-up features in this DFNA41 family are presented along with correlation analyses of phenotype-genotype in all reported families with P2RX2-related deafness. RESULTS Progressive hearing impairment was confirmed by history and by audiological follow-up testing in all the patients. The onset of hearing loss was between age 25 and 35 years. All affected subjects had bilateral sensorineural hearing loss involving all frequencies with some significant gender differences. CONCLUSIONS Our study and the review of the literature suggest that P2RX2 plays a crucial role in predisposition to noise-induced and age-related hearing loss. A better knowledge about the P2RX2-associated genetic variants can help in developing novel therapeutic strategies. LEVEL OF EVIDENCE 2b Laryngoscope, 130:1657-1663, 2020.
Collapse
Affiliation(s)
- Xue Zhong Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, U.S.A.,Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, U.S.A
| | - Denise Yan
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, U.S.A
| | - Rahul Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, U.S.A
| | - Megan E Ballard
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, U.S.A
| | - Yong Feng
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
15
|
Köles L, Szepesy J, Berekméri E, Zelles T. Purinergic Signaling and Cochlear Injury-Targeting the Immune System? Int J Mol Sci 2019; 20:ijms20122979. [PMID: 31216722 PMCID: PMC6627352 DOI: 10.3390/ijms20122979] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/14/2019] [Accepted: 06/14/2019] [Indexed: 02/06/2023] Open
Abstract
Hearing impairment is the most common sensory deficit, affecting more than 400 million people worldwide. Sensorineural hearing losses currently lack any specific or efficient pharmacotherapy largely due to the insufficient knowledge of the pathomechanism. Purinergic signaling plays a substantial role in cochlear (patho)physiology. P2 (ionotropic P2X and the metabotropic P2Y) as well as adenosine receptors expressed on cochlear sensory and non-sensory cells are involved mostly in protective mechanisms of the cochlea. They are implicated in the sensitivity adjustment of the receptor cells by a K+ shunt and can attenuate the cochlear amplification by modifying cochlear micromechanics. Cochlear blood flow is also regulated by purines. Here, we propose to comprehend this field with the purine-immune interactions in the cochlea. The role of harmful immune mechanisms in sensorineural hearing losses has been emerging in the horizon of cochlear pathologies. In addition to decreasing hearing sensitivity and increasing cochlear blood supply, influencing the immune system can be the additional avenue for pharmacological targeting of purinergic signaling in the cochlea. Elucidating this complexity of purinergic effects on cochlear functions is necessary and it can result in development of new therapeutic approaches in hearing disabilities, especially in the noise-induced ones.
Collapse
Affiliation(s)
- László Köles
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary.
| | - Judit Szepesy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary.
| | - Eszter Berekméri
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary.
- Department of Ecology, University of Veterinary Medicine, H-1078 Budapest, Hungary.
| | - Tibor Zelles
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary.
- Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, H-1083 Budapest, Hungary.
| |
Collapse
|
16
|
Kaur T, Clayman AC, Nash AJ, Schrader AD, Warchol ME, Ohlemiller KK. Lack of Fractalkine Receptor on Macrophages Impairs Spontaneous Recovery of Ribbon Synapses After Moderate Noise Trauma in C57BL/6 Mice. Front Neurosci 2019; 13:620. [PMID: 31263398 PMCID: PMC6585312 DOI: 10.3389/fnins.2019.00620] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 05/29/2019] [Indexed: 01/08/2023] Open
Abstract
Noise trauma causes loss of synaptic connections between cochlear inner hair cells (IHCs) and the spiral ganglion neurons (SGNs). Such synaptic loss can trigger slow and progressive degeneration of SGNs. Macrophage fractalkine signaling is critical for neuron survival in the injured cochlea, but its role in cochlear synaptopathy is unknown. Fractalkine, a chemokine, is constitutively expressed by SGNs and signals via its receptor CX3CR1 that is expressed on macrophages. The present study characterized the immune response and examined the function of fractalkine signaling in degeneration and repair of cochlear synapses following noise trauma. Adult mice wild type, heterozygous and knockout for CX3CR1 on a C57BL/6 background were exposed for 2 h to an octave band noise at 90 dB SPL. Noise exposure caused temporary shifts in hearing thresholds without any evident loss of hair cells in CX3CR1 heterozygous mice that have intact fractalkine signaling. Enhanced macrophage migration toward the IHC-synaptic region was observed immediately after exposure in all genotypes. Synaptic immunolabeling revealed a rapid loss of ribbon synapses throughout the basal turn of the cochlea of all genotypes. The damaged synapses spontaneously recovered in mice with intact CX3CR1. However, CX3CR1 knockout (KO) animals displayed enhanced synaptic degeneration that correlated with attenuated suprathreshold neural responses at higher frequencies. Exposed CX3CR1 KO mice also exhibited increased loss of IHCs and SGN cell bodies compared to exposed heterozygous mice. These results indicate that macrophages can promote repair of damaged synapses after moderate noise trauma and that repair requires fractalkine signaling.
Collapse
Affiliation(s)
- Tejbeer Kaur
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States
| | - Anna C Clayman
- Program in Audiology and Communication Sciences, Washington University School of Medicine, St. Louis, MO, United States
| | - Andrew J Nash
- Program in Audiology and Communication Sciences, Washington University School of Medicine, St. Louis, MO, United States
| | - Angela D Schrader
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States
| | - Mark E Warchol
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States
| | - Kevin K Ohlemiller
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States.,Program in Audiology and Communication Sciences, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
17
|
Berekméri E, Szepesy J, Köles L, Zelles T. Purinergic signaling in the organ of Corti: Potential therapeutic targets of sensorineural hearing losses. Brain Res Bull 2019; 151:109-118. [PMID: 30721767 DOI: 10.1016/j.brainresbull.2019.01.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 01/10/2019] [Accepted: 01/25/2019] [Indexed: 01/04/2023]
Abstract
Purinergic signaling is deeply involved in the development, functions and protective mechanisms of the cochlea. Release of ATP and activation of purinergic receptors on sensory and supporting/epithelial cells play a substantial role in cochlear (patho)physiology. Both the ionotropic P2X and the metabotropic P2Y receptors are widely distributed on the inner and outer hair cells as well as on the different supporting cells in the organ of Corti and on other epithelial cells in the scala media. Among others, they are implicated in the sensitivity adjustment of the receptor cells by a K+ shunt and can attenuate the cochlear amplification by modifying cochlear micromechanics acting on outer hair cells and supporting cells. Cochlear blood flow is also regulated by purines. Sensorineural hearing losses currently lack any specific or efficient pharmacotherapy. Decreasing hearing sensitivity and increasing cochlear blood supply by pharmacological targeting of purinergic signaling in the cochlea are potential new therapeutic approaches in these hearing disabilities, especially in the noise-induced ones.
Collapse
Affiliation(s)
- Eszter Berekméri
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Judit Szepesy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - László Köles
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Tibor Zelles
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary.
| |
Collapse
|
18
|
Liu B, Cao W, Li J, Liu J. Lysosomal exocytosis of ATP is coupled to P2Y 2 receptor in marginal cells in the stria vascular in neonatal rats. Cell Calcium 2018; 76:62-71. [PMID: 30273839 DOI: 10.1016/j.ceca.2018.09.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/09/2018] [Accepted: 09/20/2018] [Indexed: 01/06/2023]
Abstract
Adenosine triphosphate (ATP) is stored as lysosomal vesicles in marginal cells of the stria vascular in neonatal rats, but the mechanisms of ATP release are unclear. Primary cultures of marginal cells from 1-day-old Sprague-Dawley rats were established. P2Y2 receptor and inositol 1,4,5-trisphosphate (IP3) receptor were immunolabelled in marginal cells of the stria vascular. We found that 30 μM ATP and 30 μM uridine triphosphate (UTP) evoked comparable significant increases in the intracellular Ca2+ concentration ([Ca2+]i) in the absence of extracellular Ca2+, whereas the response was suppressed by 100 μM suramin, 10 μM 1-(6-(17β-3-methoxyester-1,3,5(10)-trien-17-yl)amino)-hexyl)-1H-pyrrole-2,5-dione(U-73122), 100 μM 2-aminoethoxydiphenyl borate (2-APB) and 5 μM thapsigargin (TG), thus indicating that ATP coupled with the P2Y2R-PLC-IP3 pathway to evoke Ca2+ release from the endoplasmic reticulum (ER). Incubation with 200 μM Gly-Phe-β-naphthylamide (GPN) selectively disrupted lysosomes and caused significant increases in [Ca2+]I; this effect was partly inhibited by P2Y2R-PLC-IP3 pathway antagonists. After pre-treatment with 5 μM TG, [Ca2+]i was significantly lower than that after treatment with P2Y2R-PLC-IP3 pathway antagonists under the same conditions, thus indicating that lysosomal Ca2+ triggers Ca2+ release from ER Ca2+ stores. Baseline [Ca2+]i declined after treatment with the Ca2+ chelator 50 μM bis-(aminophenolxy) ethane-N,N,N',N'-tetra-acetic acid acetoxyme-thyl ester (BAPTA-AM) and 4 IU/ml apyrase. 30 μM ATP decrease of the number of quinacrine-positive vesicles via lysosome exocytosis, whereas the number of lysosomes did not change. However, lysosome exocytosis was significantly suppressed by pre-treatment with 5 μM vacuolin-1. Release of ATP and β-hexosaminidase both increased after treatment with 200 μM GPN and 5 μM TG, but decreased after incubation with 50 μM BAPTA-AM, 4 IU/ml apyrase and 5 μM vacuolin-1. We suggest that ATP triggers Ca2+ release from the ER, thereby contributing to secretion of lysosomal ATP via lysosomal exocytosis. Lysosomal stored Ca2+ triggers Ca2+ release from the ER directly though the IP3 receptors, and lysosomal ATP evokes Ca2+ signals indirectly via the P2Y2R-PLC-IP3 pathway.
Collapse
Affiliation(s)
- Bin Liu
- Department of Otorhinolaryngology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wanxin Cao
- Department of Otorhinolaryngology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jiping Li
- Department of Otorhinolaryngology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| | - Jun Liu
- Department of Otorhinolaryngology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
19
|
Bouzid A, Smeti I, Dhouib L, Roche M, Achour I, Khalfallah A, Gibriel AA, Charfeddine I, Ayadi H, Lachuer J, Ghorbel A, Petit C, Masmoudi S. Down-expression of P2RX2, KCNQ5, ERBB3 and SOCS3 through DNA hypermethylation in elderly women with presbycusis. Biomarkers 2018; 23:347-356. [PMID: 29325454 DOI: 10.1080/1354750x.2018.1427795] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
CONTEXT Presbycusis, an age-related hearing impairment (ARHI), represents the most common sensory disability in adults. Today, the molecular mechanisms underlying presbycusis remain unclear. This is in particular due to the fact that ARHI is a multifactorial complex disorder resulting from several genomic factors interacting with lifelong cumulative effects of: disease, diet, and environment. OBJECTIVE Identification of novel biomarkers for presbycusis. MATERIALS AND METHODS We selectively ascertained 18 elderly unrelated women lacking environmental and metabolic risk factors. Subsequently, we screened for methylation map changes in blood samples of women with presbycusis as compared to controls, using reduced representation bisulfite sequencing. We focused on hypermethylated cytosine bases located in gene promoters and the first two exons. To elucidate the related gene expression changes, we performed transcriptomic study using gene expression microarray. RESULTS Twenty-seven genes, known to be expressed in adult human cochlea, were found in the blood cells to be differentially hypermethylated with significant (p < 0.01) methylation differences (>30%) and down-expressed with fold change >1.2 (FDR <0.05). Functional annotation and qRT-PCR further identified P2RX2, KCNQ5, ERBB3 and SOCS3 to be associated with the progression of ARHI. DISCUSSION AND CONCLUSION Down-expressed genes associated with DNA hypermethylation could be used as biomarkers for understanding complex pathogenic mechanisms underlying presbycusis.
Collapse
Affiliation(s)
- Amal Bouzid
- a Laboratoire de Procédés de Criblage Moléculaire et Cellulaire, Centre de Biotechnologie de Sfax , Université de Sfax , Sfax , Tunisie
| | - Ibtihel Smeti
- a Laboratoire de Procédés de Criblage Moléculaire et Cellulaire, Centre de Biotechnologie de Sfax , Université de Sfax , Sfax , Tunisie
| | - Leila Dhouib
- b Service d'O.R.L, C.H.U. H. Bourguiba de Sfax , Sfax , Tunisie
| | - Magali Roche
- c Faculté de Médecine et de Pharmacie de Lyon, ISPBL, ProfileXpert-LCM , Lyon , France
| | - Imen Achour
- b Service d'O.R.L, C.H.U. H. Bourguiba de Sfax , Sfax , Tunisie
| | - Aida Khalfallah
- a Laboratoire de Procédés de Criblage Moléculaire et Cellulaire, Centre de Biotechnologie de Sfax , Université de Sfax , Sfax , Tunisie
| | - Abdullah Ahmed Gibriel
- d Department of Biochemistry and Molecular Biology, Faculty of Pharmacy , The British University in Egypt , Cairo , Egypt
| | | | - Hammadi Ayadi
- a Laboratoire de Procédés de Criblage Moléculaire et Cellulaire, Centre de Biotechnologie de Sfax , Université de Sfax , Sfax , Tunisie
| | - Joel Lachuer
- c Faculté de Médecine et de Pharmacie de Lyon, ISPBL, ProfileXpert-LCM , Lyon , France
| | | | - Christine Petit
- e Unité de Génétique et Physiologie de l'Audition , Institut Pasteur , Paris , France.,f UMRS 1120, Institut National de la Santé et de la Recherche Médicale (INSERM) , Paris , France.,g Collège de France , Paris , France
| | - Saber Masmoudi
- a Laboratoire de Procédés de Criblage Moléculaire et Cellulaire, Centre de Biotechnologie de Sfax , Université de Sfax , Sfax , Tunisie
| |
Collapse
|
20
|
Zhu Y, Beudez J, Yu N, Grutter T, Zhao HB. P2X2 Dominant Deafness Mutations Have No Negative Effect on Wild-Type Isoform: Implications for Functional Rescue and in Deafness Mechanism. Front Mol Neurosci 2017; 10:371. [PMID: 29180951 PMCID: PMC5693881 DOI: 10.3389/fnmol.2017.00371] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 10/26/2017] [Indexed: 11/13/2022] Open
Abstract
The P2X2 receptor is an ATP-gated ion channel, assembled by three subunits. Recently, it has been found that heterozygous mutations of P2X2 V60L and G353R can cause autosomal dominant nonsyndromic hearing loss. However, the underlying mechanism remains unclear. The fact that heterozygous mutations cause deafness suggests that the mutations may have dominant-negative effect (DNE) on wild-type (WT) P2X2 isoforms and/or other partners leading to hearing loss. In this study, the effect of these dominant deafness P2X2 mutations on WT P2X2 was investigated. We found that sole transfection of both V60L and G353R deafness mutants could efficiently target to the plasma membrane, like WT P2X2, but exhibit a significantly reduced response to ATP stimulation. Both mutants reduced the channel conductance, but G353R mutation also altered the voltage dependency. Co-expression with WT P2X2 could restore the response to ATP. As the ratio of WT P2X2 vs. mutants increased, the response to ATP was also increased. Computer modeling confirmed that both V60L and G353R dominant-deafness mutant subunits do not have any negative effect on WT P2X2 subunit, when assembled as a heterotrimer. Improper docking or defective gating is the more likely mechanism for impaired channel function by these P2X2 deafness mutations. These results suggest that P2X2 dominant deafness mutations do not have negative effects on WT P2X2 isoforms, and that adding additional WT P2X2 could rescue the lost channel function caused by the deafness mutations. These P2X2 dominant deafness mutations may have negative-effects on other partners leading to hearing loss.
Collapse
Affiliation(s)
- Yan Zhu
- Department of Otolaryngology, University of Kentucky Medical Center, Lexington, KY, United States
| | - Juline Beudez
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7199, Laboratoire de Conception et Application de Molécules Bioactives, Équipe de Chimie et Neurobiologie Moléculaire, Strasbourg, France.,Faculté de Pharmacie, Université de Strasbourg, Strasbourg, France
| | - Ning Yu
- Department of Otolaryngology, University of Kentucky Medical Center, Lexington, KY, United States.,Department of Otolaryngology, Institute of Otolaryngology, Chinese PLA General Hospital, Beijing, China
| | - Thomas Grutter
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7199, Laboratoire de Conception et Application de Molécules Bioactives, Équipe de Chimie et Neurobiologie Moléculaire, Strasbourg, France.,Faculté de Pharmacie, Université de Strasbourg, Strasbourg, France
| | - Hong-Bo Zhao
- Department of Otolaryngology, University of Kentucky Medical Center, Lexington, KY, United States
| |
Collapse
|
21
|
Kurabi A, Keithley EM, Housley GD, Ryan AF, Wong ACY. Cellular mechanisms of noise-induced hearing loss. Hear Res 2016; 349:129-137. [PMID: 27916698 PMCID: PMC6750278 DOI: 10.1016/j.heares.2016.11.013] [Citation(s) in RCA: 217] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 11/10/2016] [Accepted: 11/21/2016] [Indexed: 12/22/2022]
Abstract
Exposure to intense sound or noise can result in purely temporary threshold shift (TTS), or leave a residual permanent threshold shift (PTS) along with alterations in growth functions of auditory nerve output. Recent research has revealed a number of mechanisms that contribute to noise-induced hearing loss (NIHL). The principle cause of NIHL is damage to cochlear hair cells and associated synaptopathy. Contributions to TTS include reversible damage to hair cell (HC) stereocilia or synapses, while moderate TTS reflects protective purinergic hearing adaptation. PTS represents permanent damage to or loss of HCs and synapses. While the substrates of HC damage are complex, they include the accumulation of reactive oxygen species and the active stimulation of intracellular stress pathways, leading to programmed and/or necrotic cell death. Permanent damage to cochlear neurons can also contribute to the effects of NIHL, in addition to HC damage. These mechanisms have translational potential for pharmacological intervention and provide multiple opportunities to prevent HC damage or to rescue HCs and spiral ganglion neurons that have suffered injury. This paper reviews advances in our understanding of cellular mechanisms that contribute to NIHL and their potential for therapeutic manipulation.
Collapse
Affiliation(s)
- Arwa Kurabi
- Division of Otolaryngology, Department of Surgery, UCSD School of Medicine and San Diego VA Medical Center, La Jolla, CA, 92093, United States
| | - Elizabeth M Keithley
- Division of Otolaryngology, Department of Surgery, UCSD School of Medicine and San Diego VA Medical Center, La Jolla, CA, 92093, United States
| | - Gary D Housley
- Division of Otolaryngology, Department of Surgery, UCSD School of Medicine and San Diego VA Medical Center, La Jolla, CA, 92093, United States
| | - Allen F Ryan
- Division of Otolaryngology, Department of Surgery, UCSD School of Medicine and San Diego VA Medical Center, La Jolla, CA, 92093, United States.
| | - Ann C-Y Wong
- Division of Otolaryngology, Department of Surgery, UCSD School of Medicine and San Diego VA Medical Center, La Jolla, CA, 92093, United States
| |
Collapse
|
22
|
Abstract
Pannexin (Panx) is a gene family encoding gap junction proteins in vertebrates. So far, three isoforms (Panx1, 2 and 3) have been identified. All of three Panx isoforms express in the cochlea with distinct expression patterns. Panx1 expresses in the cochlea extensively, including the spiral limbus, the organ of Corti, and the cochlear lateral wall, whereas Panx2 and Panx3 restrict to the basal cells of the stria vascularis in the lateral wall and the cochlear bony structure, respectively. However, there is no pannexin expression in auditory sensory hair cells. Recent studies demonstrated that like connexin gap junction gene, Panx1 deficiency causes hearing loss. Panx1 channels dominate ATP release in the cochlea. Deletion of Panx1 abolishes ATP release in the cochlea and reduces endocochlear potential (EP), auditory receptor current/potential, and active cochlear amplification. Panx1 deficiency in the cochlea also activates caspase-3 cell apoptotic pathway leading to cell degeneration. These new findings suggest that pannexins have a critical role in the cochlea in regard to hearing. However, detailed information about pannexin function in the cochlea and Panx mutation induced hearing loss still remain largely undetermined. Further studies are required.
Collapse
Affiliation(s)
- Hong-Bo Zhao
- Department of Otolaryngology, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY, 40536, USA.
| |
Collapse
|
23
|
Mittal R, Chan B, Grati M, Mittal J, Patel K, Debs LH, Patel AP, Yan D, Chapagain P, Liu XZ. Molecular Structure and Regulation of P2X Receptors With a Special Emphasis on the Role of P2X2 in the Auditory System. J Cell Physiol 2015; 231:1656-70. [PMID: 26627116 DOI: 10.1002/jcp.25274] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 12/01/2015] [Indexed: 12/23/2022]
Abstract
The P2X purinergic receptors are cation-selective channels gated by extracellular adenosine 5'-triphosphate (ATP). These purinergic receptors are found in virtually all mammalian cell types and facilitate a number of important physiological processes. Within the past few years, the characterization of crystal structures of the zebrafish P2X4 receptor in its closed and open states has provided critical insights into the mechanisms of ligand binding and channel activation. Understanding of this gating mechanism has facilitated to design and interpret new modeling and structure-function experiments to better elucidate how different agonists and antagonists can affect the receptor with differing levels of potency. This review summarizes the current knowledge on the structure, activation, allosteric modulators, function, and location of the different P2X receptors. Moreover, an emphasis on the P2X2 receptors has been placed in respect to its role in the auditory system. In particular, the discovery of three missense mutations in P2X2 receptors could become important areas of study in the field of gene therapy to treat progressive and noise-induced hearing loss. J. Cell. Physiol. 231: 1656-1670, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Brandon Chan
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - M'hamed Grati
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Jeenu Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Kunal Patel
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Luca H Debs
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Amit P Patel
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Denise Yan
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Prem Chapagain
- Department of Physics, Florida International University, Miami, Florida.,Biomolecular Science Institute, Florida International University, Miami, Florida
| | - Xue Zhong Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida.,Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida.,Department of Biochemistry, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
24
|
Pannexin1 channels dominate ATP release in the cochlea ensuring endocochlear potential and auditory receptor potential generation and hearing. Sci Rep 2015; 5:10762. [PMID: 26035172 PMCID: PMC4451810 DOI: 10.1038/srep10762] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 04/28/2015] [Indexed: 01/04/2023] Open
Abstract
Pannexin1 (Panx1) is a gap junction gene in vertebrates whose proteins mainly function as non-junctional channels on the cell surface. Panx1 channels can release ATP under physiological conditions and play critical roles in many physiological and pathological processes. Here, we report that Panx1 deficiency can reduce ATP release and endocochlear potential (EP) generation in the cochlea inducing hearing loss. Panx1 extensively expresses in the cochlea, including the cochlear lateral wall. We found that deletion of Panx1 in the cochlear lateral wall almost abolished ATP release under physiological conditions. Positive EP is a driving force for current through hair cells to produce auditory receptor potential. EP generation requires ATP. In the Panx1 deficient mice, EP and auditory receptor potential as measured by cochlear microphonics (CM) were significantly reduced. However, no apparent hair cell loss was detected. Moreover, defect of connexin hemichannels by deletion of connexin26 (Cx26) and Cx30, which are predominant connexin isoforms in the cochlea, did not reduce ATP release under physiological conditions. These data demonstrate that Panx1 channels dominate ATP release in the cochlea ensuring EP and auditory receptor potential generation and hearing. Panx1 deficiency can reduce ATP release and EP generation causing hearing loss.
Collapse
|
25
|
Nishio SY, Hattori M, Moteki H, Tsukada K, Miyagawa M, Naito T, Yoshimura H, Iwasa YI, Mori K, Shima Y, Sakuma N, Usami SI. Gene expression profiles of the cochlea and vestibular endorgans: localization and function of genes causing deafness. Ann Otol Rhinol Laryngol 2015; 124 Suppl 1:6S-48S. [PMID: 25814645 DOI: 10.1177/0003489415575549] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVES We sought to elucidate the gene expression profiles of the causative genes as well as the localization of the encoded proteins involved in hereditary hearing loss. METHODS Relevant articles (as of September 2014) were searched in PubMed databases, and the gene symbols of the genes reported to be associated with deafness were located on the Hereditary Hearing Loss Homepage using localization, expression, and distribution as keywords. RESULTS Our review of the literature allowed us to systematize the gene expression profiles for genetic deafness in the inner ear, clarifying the unique functions and specific expression patterns of these genes in the cochlea and vestibular endorgans. CONCLUSIONS The coordinated actions of various encoded molecules are essential for the normal development and maintenance of auditory and vestibular function.
Collapse
Affiliation(s)
- Shin-Ya Nishio
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto, Japan
| | - Mitsuru Hattori
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hideaki Moteki
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Keita Tsukada
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Maiko Miyagawa
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takehiko Naito
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hidekane Yoshimura
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yoh-Ichiro Iwasa
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Kentaro Mori
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yutaka Shima
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Naoko Sakuma
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan Department of Otorhinolaryngology and Head and Neck Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Shin-Ichi Usami
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|
26
|
Moteki H, Azaiez H, Booth KT, Hattori M, Sato A, Sato Y, Motobayashi M, Sloan CM, Kolbe DL, Shearer AE, Smith RJH, Usami SI. Hearing loss caused by a P2RX2 mutation identified in a MELAS family with a coexisting mitochondrial 3243AG mutation. Ann Otol Rhinol Laryngol 2015; 124 Suppl 1:177S-83S. [PMID: 25788561 DOI: 10.1177/0003489415575045] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVES We present a family with a mitochondrial DNA 3243A>G mutation resulting in mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke-like episodes (MELAS), of which some members have hearing loss in which a novel mutation in the P2RX2 gene was identified. METHODS One hundred ninety-four (194) Japanese subjects from unrelated families were enrolled in the study. Targeted genomic enrichment and massively parallel sequencing of all known nonsyndromic hearing loss genes were performed to identify the genetic causes of hearing loss. RESULTS A novel mutation in the P2RX2 gene that corresponded to c.601G>A (p.Asp201Tyr) was identified. Two patients carried the mutation and had severe sensorineural hearing loss, while other members with MELAS (who did not carry the P2RX2 mutation) had normal hearing. CONCLUSION This is the first case report of a diagnosis of hearing loss caused by P2RX2 mutation in patients with MELAS. A potential explanation is that a decrease in adenosine triphosphate (ATP) production due to MELAS with a mitochondrial 3243A>G mutation might suppress activation of P2X2 receptors. We also suggest that hearing loss caused by the P2RX2 mutation might be influenced by the decrease in ATP production due to MELAS.
Collapse
Affiliation(s)
- Hideaki Moteki
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan Department of Otolaryngology-Head and Neck Surgery, Molecular Otolaryngology & Renal Research Labs, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hela Azaiez
- Department of Otolaryngology-Head and Neck Surgery, Molecular Otolaryngology & Renal Research Labs, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Kevin T Booth
- Department of Otolaryngology-Head and Neck Surgery, Molecular Otolaryngology & Renal Research Labs, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Mitsuru Hattori
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Ai Sato
- Division of Diabetes, Endocrinology and Metabolism: Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yoshihiko Sato
- Division of Diabetes, Endocrinology and Metabolism: Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - Mitsuo Motobayashi
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
| | - Christina M Sloan
- Department of Otolaryngology-Head and Neck Surgery, Molecular Otolaryngology & Renal Research Labs, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Diana L Kolbe
- Department of Otolaryngology-Head and Neck Surgery, Molecular Otolaryngology & Renal Research Labs, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - A Eliot Shearer
- Department of Otolaryngology-Head and Neck Surgery, Molecular Otolaryngology & Renal Research Labs, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Richard J H Smith
- Department of Otolaryngology-Head and Neck Surgery, Molecular Otolaryngology & Renal Research Labs, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Shin-Ichi Usami
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|
27
|
Jagger DJ, Forge A. The enigmatic root cell – Emerging roles contributing to fluid homeostasis within the cochlear outer sulcus. Hear Res 2013; 303:1-11. [DOI: 10.1016/j.heares.2012.10.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 10/19/2012] [Accepted: 10/26/2012] [Indexed: 12/20/2022]
|
28
|
Wong ACY, Froud KE, Hsieh YSY. Noise-induced hearing loss in the 21 st century: A research and translational update. World J Otorhinolaryngol 2013; 3:58-70. [DOI: 10.5319/wjo.v3.i3.58] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 07/19/2013] [Indexed: 02/06/2023] Open
Abstract
Millions of people worldwide are exposed to harmful levels of noise daily in their work and leisure environment. This makes noise-induced hearing loss (NIHL) a major occupational health risk globally. NIHL is the second most common form of acquired hearing loss after age-related hearing loss and is itself a major contributing factor to presbycusis. Temporary threshold shifts, once thought to be relatively harmless and recoverable, are now known to cause permanent cochlear injury leading to permanent loss of hearing sensitivity. This article reviews the current understanding of the cellular and molecular pathophysiology of NIHL with latest findings from animal models. Therapeutic approaches to protect against or to mitigate NIHL are discussed based on their proposed action against these known mechanisms of cochlear injury. Successes in identifying genes that predispose individuals to NIHL by candidate gene association studies are discussed with matched gene knockout animal models. This links to exciting developments in experimental gene therapy to replace and regenerate lost hair cells and post-noise otoprotective therapies currently being investigated in clinical trials. The aim is to provide new insights into current and projected future strategies to manage NIHL; bench to bedside treatment is foreseeable in the next 5 to 10 years.
Collapse
|
29
|
Abstract
The sense of hearing is remarkable for its auditory dynamic range, which spans more than 10(12) in acoustic intensity. The mechanisms that enable the cochlea to transduce high sound levels without damage are of key interest, particularly with regard to the broad impact of industrial, military, and recreational auditory overstimulation on hearing disability. We show that ATP-gated ion channels assembled from P2X2 receptor subunits in the cochlea are necessary for the development of temporary threshold shift (TTS), evident in auditory brainstem response recordings as sound levels rise. In mice null for the P2RX2 gene (encoding the P2X2 receptor subunit), sustained 85-dB noise failed to elicit the TTS that wild-type (WT) mice developed. ATP released from the tissues of the cochlear partition with elevation of sound levels likely activates the broadly distributed P2X2 receptors on epithelial cells lining the endolymphatic compartment. This purinergic signaling is supported by significantly greater noise-induced suppression of distortion product otoacoustic emissions derived from outer hair cell transduction and decreased suprathreshold auditory brainstem response input/output gain in WT mice compared with P2RX2-null mice. At higher sound levels (≥95 dB), additional processes dominated TTS, and P2RX2-null mice were more vulnerable than WT mice to permanent hearing loss due to hair cell synapse disruption. P2RX2-null mice lacked ATP-gated conductance across the cochlear partition, including loss of ATP-gated inward current in hair cells. These data indicate that a significant component of TTS represents P2X2 receptor-dependent purinergic hearing adaptation that underpins the upper physiological range of hearing.
Collapse
|
30
|
Mutation of the ATP-gated P2X(2) receptor leads to progressive hearing loss and increased susceptibility to noise. Proc Natl Acad Sci U S A 2013; 110:2228-33. [PMID: 23345450 DOI: 10.1073/pnas.1222285110] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Age-related hearing loss and noise-induced hearing loss are major causes of human morbidity. Here we used genetics and functional studies to show that a shared cause of these disorders may be loss of function of the ATP-gated P2X(2) receptor (ligand-gated ion channel, purinergic receptor 2) that is expressed in sensory and supporting cells of the cochlea. Genomic analysis of dominantly inherited, progressive sensorineural hearing loss DFNA41 in a six-generation kindred revealed a rare heterozygous allele, P2RX2 c.178G > T (p.V60L), at chr12:133,196,029, which cosegregated with fully penetrant hearing loss in the index family, and also appeared in a second family with the same phenotype. The mutation was absent from more than 7,000 controls. P2RX2 p.V60L abolishes two hallmark features of P2X(2) receptors: ATP-evoked inward current response and ATP-stimulated macropore permeability, measured as loss of ATP-activated FM1-43 fluorescence labeling. Coexpression of mutant and WT P2X(2) receptor subunits significantly reduced ATP-activated membrane permeability. P2RX2-null mice developed severe progressive hearing loss, and their early exposure to continuous moderate noise led to high-frequency hearing loss as young adults. Similarly, among family members heterozygous for P2RX2 p.V60L, noise exposure exacerbated high-frequency hearing loss in young adulthood. Our results suggest that P2X(2) function is required for life-long normal hearing and for protection from exposure to noise.
Collapse
|
31
|
Zhu Y, Zhao HB. ATP activates P2X receptors to mediate gap junctional coupling in the cochlea. Biochem Biophys Res Commun 2012; 426:528-32. [PMID: 22982314 DOI: 10.1016/j.bbrc.2012.08.119] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 08/24/2012] [Indexed: 10/27/2022]
Abstract
ATP is an important extracellular signaling molecule and can activate both ionotropic (P2X) and metabotropic purinergic (P2Y) receptors to influence cellular function in many aspects. Gap junction is an intercellular channel and plays a critical role in hearing. Here, we report that stimulation of ATP reduced gap junctional coupling between cochlear supporting cells. This uncoupling effect could be evoked by nanomolar physiological levels of ATP. A P2X receptor agonist benzoylbenzoyl-ATP (BzATP) but not a P2Y receptor agonist UTP stimulated this uncoupling effect. Application of P2X receptor antagonists pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid (PPADS, 50μM) or oxidized ATP (oATP, 0.1mM) eliminated this uncoupling effect. We further found that ATP activated P2X receptors in the cochlear supporting cells allowing Ca(2+) influxing, thereby increasing intracellular Ca(2+) concentration to mediate gap junctions. These data suggest that ATP can mediate cochlear gap junctions at the physiological level by the activation of P2X receptors rather than P2Y receptors. This P2X receptor-mediated purinergic control on the cochlear gap junctions may play an important role in the regulation of K(+)-recycling for ionic homeostasis in the cochlea and the reduction of hearing sensitivity under noise stress for protection.
Collapse
Affiliation(s)
- Yan Zhu
- Dept. of Otolaryngology, University of Kentucky Medical Center, Lexington, KY 40536 0293, United States
| | | |
Collapse
|
32
|
In pursuit of P2X3 antagonists: novel therapeutics for chronic pain and afferent sensitization. Purinergic Signal 2011; 8:3-26. [PMID: 22095157 PMCID: PMC3265711 DOI: 10.1007/s11302-011-9271-6] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Accepted: 09/19/2011] [Indexed: 12/20/2022] Open
Abstract
Treating pain by inhibiting ATP activation of P2X3-containing receptors heralds an exciting new approach to pain management, and Afferent's program marks the vanguard in a new class of drugs poised to explore this approach to meet the significant unmet needs in pain management. P2X3 receptor subunits are expressed predominately and selectively in so-called C- and Aδ-fiber primary afferent neurons in most tissues and organ systems, including skin, joints, and hollow organs, suggesting a high degree of specificity to the pain sensing system in the human body. P2X3 antagonists block the activation of these fibers by ATP and stand to offer an alternative approach to the management of pain and discomfort. In addition, P2X3 is expressed pre-synaptically at central terminals of C-fiber afferent neurons, where ATP further sensitizes transmission of painful signals. As a result of the selectivity of the expression of P2X3, there is a lower likelihood of adverse effects in the brain, gastrointestinal, or cardiovascular tissues, effects which remain limiting factors for many existing pain therapeutics. In the periphery, ATP (the factor that triggers P2X3 receptor activation) can be released from various cells as a result of tissue inflammation, injury or stress, as well as visceral organ distension, and stimulate these local nociceptors. The P2X3 receptor rationale has aroused a formidable level of investigation producing many reports that clarify the potential role of ATP as a pain mediator, in chronic sensitized states in particular, and has piqued the interest of pharmaceutical companies. P2X receptor-mediated afferent activation has been implicated in inflammatory, visceral, and neuropathic pain states, as well as in airways hyperreactivity, migraine, itch, and cancer pain. It is well appreciated that oftentimes new mechanisms translate poorly from models into clinical efficacy and effectiveness; however, the breadth of activity seen from P2X3 inhibition in models offers a realistic chance that this novel mechanism to inhibit afferent nerve sensitization may find its place in the sun and bring some merciful relief to the torment of persistent discomfort and pain. The development philosophy at Afferent is to conduct proof of concept patient studies and best identify target patient groups that may benefit from this new intervention.
Collapse
|
33
|
Bongartz EV, Rettinger J, Hausmann R. Aminoglycoside block of P2X2 receptors heterologously expressed in Xenopus laevis oocytes. Purinergic Signal 2010; 6:393-403. [PMID: 21437010 DOI: 10.1007/s11302-010-9204-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2009] [Accepted: 11/01/2010] [Indexed: 12/01/2022] Open
Abstract
Aminoglycosides are polycationic antibiotics that have been shown to block a variety of cation channels. The inhibitory effect of externally applied aminoglycosides on P2X2 receptor currents was examined after heterologous expression in Xenopus laevis oocytes using the two-electrode voltage-clamp technique. All of the aminoglycosides tested inhibited the ATP-evoked responses with potencies ranging from 71 μM to 2 mM (IC(50) values). The ranked order of potency was streptomycin > gentamicin > neomycin > paromomycin > kanamycin. The inhibition of P2X receptor currents was independent of the ATP concentration used for the activation, which is compatible with a noncompetitive mechanism. The inhibition was voltage-dependent and was reduced at more positive membrane potentials. To examine whether the current block was dependent on the receptor conformation, the aminoglycoside effect on a non-desensitizing P2X2-X1 receptor chimera was analyzed. The results from these measurements suggest that inhibition is caused by an open pore block that locks the P2X receptor chimera in an open nonconducting state from which the agonist dissociation is slow. We also demonstrate that the P2X2-X1 chimera can serve as a tool to directly test whether an antagonist acts competitively or not.
Collapse
|