1
|
Eckert MA, Harris KC, Lang H, Lewis MA, Schmiedt RA, Schulte BA, Steel KP, Vaden KI, Dubno JR. Translational and interdisciplinary insights into presbyacusis: A multidimensional disease. Hear Res 2021; 402:108109. [PMID: 33189490 PMCID: PMC7927149 DOI: 10.1016/j.heares.2020.108109] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 10/19/2020] [Accepted: 10/25/2020] [Indexed: 12/18/2022]
Abstract
There are multiple etiologies and phenotypes of age-related hearing loss or presbyacusis. In this review we summarize findings from animal and human studies of presbyacusis, including those that provide the theoretical framework for distinct metabolic, sensory, and neural presbyacusis phenotypes. A key finding in quiet-aged animals is a decline in the endocochlear potential (EP) that results in elevated pure-tone thresholds across frequencies with greater losses at higher frequencies. In contrast, sensory presbyacusis appears to derive, in part, from acute and cumulative effects on hair cells of a lifetime of environmental exposures (e.g., noise), which often result in pronounced high frequency hearing loss. These patterns of hearing loss in animals are recognizable in the human audiogram and can be classified into metabolic and sensory presbyacusis phenotypes, as well as a mixed metabolic+sensory phenotype. However, the audiogram does not fully characterize age-related changes in auditory function. Along with the effects of peripheral auditory system declines on the auditory nerve, primary degeneration in the spiral ganglion also appears to contribute to central auditory system aging. These inner ear alterations often correlate with structural and functional changes throughout the central nervous system and may explain suprathreshold speech communication difficulties in older adults with hearing loss. Throughout this review we highlight potential methods and research directions, with the goal of advancing our understanding, prevention, diagnosis, and treatment of presbyacusis.
Collapse
Affiliation(s)
- Mark A Eckert
- Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA.
| | - Kelly C Harris
- Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA
| | - Hainan Lang
- Medical University of South Carolina, Department of Pathology and Laboratory Medicine, Charleston, SC 29425, USA
| | - Morag A Lewis
- King's College London, Wolfson Centre for Age-Related Diseases, London SE1 1UL, United Kingdom
| | - Richard A Schmiedt
- Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA
| | - Bradley A Schulte
- Medical University of South Carolina, Department of Pathology and Laboratory Medicine, Charleston, SC 29425, USA; Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA
| | - Karen P Steel
- King's College London, Wolfson Centre for Age-Related Diseases, London SE1 1UL, United Kingdom
| | - Kenneth I Vaden
- Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA
| | - Judy R Dubno
- Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA; Medical University of South Carolina, Department of Pathology and Laboratory Medicine, Charleston, SC 29425, USA
| |
Collapse
|
2
|
Salt AN, Hirose K. Communication pathways to and from the inner ear and their contributions to drug delivery. Hear Res 2018; 362:25-37. [PMID: 29277248 PMCID: PMC5911243 DOI: 10.1016/j.heares.2017.12.010] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 11/08/2017] [Accepted: 12/05/2017] [Indexed: 01/04/2023]
Abstract
The environment of the inner ear is highly regulated in a manner that some solutes are permitted to enter while others are excluded or transported out. Drug therapies targeting the sensory and supporting cells of the auditory and vestibular systems require the agent to gain entry to the fluid spaces of the inner ear, perilymph or endolymph, which surround the sensory organs. Access to the inner ear fluids from the vasculature is limited by the blood-labyrinth barriers, which include the blood-perilymph and blood-strial barriers. Intratympanic applications provide an alternative approach in which drugs are applied locally. Drug from the applied solution enters perilymph through the round window membrane, through the stapes, and under some circumstances, through thin bone in the otic capsule. The amount of drug applied to the middle ear is always substantially more than the amount entering perilymph. As a result, significant amounts of the applied drug can pass to the digestive system, to the vasculature, and to the brain. Drugs in perilymph pass to the vasculature and to cerebrospinal fluid via the cochlear aqueduct. Conversely, drugs applied to cerebrospinal fluid, including those given intrathecally, can enter perilymph through the cochlear aqueduct. Other possible routes in or out of the ear include passage by neuronal pathways, passage via endolymph and the endolymphatic sac, and possibly via lymphatic pathways. A better understanding of the pathways for drug movements in and out of the ear will enable better intervention strategies.
Collapse
Affiliation(s)
- Alec N Salt
- Department of Otolaryngology, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, USA.
| | - Keiko Hirose
- Department of Otolaryngology, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, USA
| |
Collapse
|
3
|
Liu W, Schrott-Fischer A, Glueckert R, Benav H, Rask-Andersen H. The Human "Cochlear Battery" - Claudin-11 Barrier and Ion Transport Proteins in the Lateral Wall of the Cochlea. Front Mol Neurosci 2017; 10:239. [PMID: 28848383 PMCID: PMC5554435 DOI: 10.3389/fnmol.2017.00239] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 07/14/2017] [Indexed: 11/21/2022] Open
Abstract
Background: The cochlea produces an electric field potential essential for hair cell transduction and hearing. This biological “battery” is situated in the lateral wall of the cochlea and contains molecular machinery that secretes and recycles K+ ions. Its functioning depends on junctional proteins that restrict the para-cellular escape of ions. The tight junction protein Claudin-11 has been found to be one of the major constituents of this barrier that maintains ion gradients (Gow et al., 2004; Kitajiri et al., 2004a). We are the first to elucidate the human Claudin-11 framework and the associated ion transport machinery using super-resolution fluorescence illumination microscopy (SR-SIM). Methods: Archival cochleae obtained during meningioma surgery were used for SR-SIM together with transmission electron microscopy after ethical consent. Results: Claudin-11-expressing cells formed parallel tight junction lamellae that insulated the epithelial syncytium of the stria vascularis and extended to the suprastrial region. Intercellular gap junctions were found between the barrier cells and fibrocytes. Conclusion: Transmission electron microscopy, confocal microscopy and SR-SIM revealed exclusive cell specialization in the various subdomains of the lateral wall of the human cochlea. The Claudin-11-expressing cells exhibited both conductor and isolator characteristics, and these micro-porous separators may selectively mediate the movement of charged units to the intrastrial space in a manner that is analogous to a conventional electrochemical “battery.” The function and relevance of this battery for the development of inner ear disease are discussed.
Collapse
Affiliation(s)
- Wei Liu
- Department of Surgical Sciences, Section of Otolaryngology, Uppsala University HospitalUppsala, Sweden
| | | | - Rudolf Glueckert
- Department of Otolaryngology, Medical University of InnsbruckInnsbruck, Austria
| | | | - Helge Rask-Andersen
- Department of Surgical Sciences, Head and Neck Surgery, Section of Otolaryngology, Uppsala University HospitalUppsala, Sweden
| |
Collapse
|
4
|
Dalamon V, Fiori MC, Figueroa VA, Oliva CA, Del Rio R, Gonzalez W, Canan J, Elgoyhen AB, Altenberg GA, Retamal MA. Gap-junctional channel and hemichannel activity of two recently identified connexin 26 mutants associated with deafness. Pflugers Arch 2016; 468:909-18. [PMID: 26769242 DOI: 10.1007/s00424-016-1788-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 12/08/2015] [Accepted: 01/05/2016] [Indexed: 01/10/2023]
Abstract
Gap-junction channels (GJCs) are formed by head-to-head association of two hemichannels (HCs, connexin hexamers). HCs and GJCs are permeable to ions and hydrophilic molecules of up to Mr ~1 kDa. Hearing impairment of genetic origin is common, and mutations of connexin 26 (Cx26) are its major cause. We recently identified two novel Cx26 mutations in hearing-impaired subjects, L10P and G109V. L10P forms functional GJCs with slightly altered voltage dependence and HCs with decrease ATP/cationic dye selectivity. G109V does not form functional GJCs, but forms functional HCs with enhanced extracellular Ca(2+) sensitivity and subtle alterations in voltage dependence and ATP/cationic dye selectivity. Deafness associated with G109V could result from decreased GJCs activity, whereas deafness associated to L10P may have a more complex mechanism that involves changes in HC permeability.
Collapse
Affiliation(s)
- Viviana Dalamon
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Dr. Héctor N. Torres, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad Autónoma de Buenos Aires, Argentina
| | - Mariana C Fiori
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Vania A Figueroa
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Avenida Las Condes, 12438, Santiago, Chile
| | - Carolina A Oliva
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Avenida Las Condes, 12438, Santiago, Chile
| | - Rodrigo Del Rio
- Centro de Investigación Biomédica, Universidad Autónoma de Chile, Santiago, Chile
| | - Wendy Gonzalez
- Centro de Bioinformática y Simulación Molecular (CBSM), Universidad de Talca, Talca, Chile
| | - Jonathan Canan
- Centro de Bioinformática y Simulación Molecular (CBSM), Universidad de Talca, Talca, Chile
| | - Ana B Elgoyhen
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Dr. Héctor N. Torres, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad Autónoma de Buenos Aires, Argentina
- Departamento de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Guillermo A Altenberg
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Mauricio A Retamal
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Avenida Las Condes, 12438, Santiago, Chile.
| |
Collapse
|
5
|
Perilymph Kinetics of FITC-Dextran Reveals Homeostasis Dominated by the Cochlear Aqueduct and Cerebrospinal Fluid. J Assoc Res Otolaryngol 2015; 16:357-71. [PMID: 25801074 DOI: 10.1007/s10162-015-0512-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 02/13/2015] [Indexed: 02/07/2023] Open
Abstract
Understanding how drugs are distributed in perilymph following local applications is important as local drug therapies are increasingly used to treat disorders of the inner ear. The potential contribution of cerebrospinal fluid (CSF) entry to perilymph homeostasis has been controversial for over half a century, largely due to artifactual contamination of collected perilymph samples with CSF. Measures of perilymph flow and of drug distribution following round window niche applications have both suggested a slow, apically directed flow occurs along scala tympani (ST) in the normal, sealed cochlea. In the present study, we have used fluorescein isothiocyanate-dextran as a marker to study perilymph kinetics in guinea pigs. Dextran is lost from perilymph more slowly than other substances so far quantified. Dextran solutions were injected from pipettes sealed into the lateral semicircular canal (SCC), the cochlear apex, or the basal turn of ST. After varying delays, sequential perilymph samples were taken from the cochlear apex or lateral SCC, allowing dextran distribution along the perilymphatic spaces to be quantified. Variability was low and findings were consistent with the injection procedure driving volume flow towards the cochlear aqueduct, and with volume flow during perilymph sampling driven by CSF entry at the aqueduct. The decline of dextran with time in the period between injection and sampling was consistent with both a slow volume influx of CSF (~30 nL/min) entering the basal turn of ST at the cochlear aqueduct and a CSF-perilymph exchange driven by pressure-driven fluid oscillation across the cochlear aqueduct. Sample data also allowed contributions of other processes, such as communications with adjacent compartments, to be quantified. The study demonstrates that drug kinetics in the basal turn of ST is complex and is influenced by a considerable number of interacting processes.
Collapse
|
6
|
Hao X, Xing Y, Moore MW, Zhang J, Han D, Schulte BA, Dubno JR, Lang H. Sox10 expressing cells in the lateral wall of the aged mouse and human cochlea. PLoS One 2014; 9:e97389. [PMID: 24887110 PMCID: PMC4041576 DOI: 10.1371/journal.pone.0097389] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 04/17/2014] [Indexed: 12/20/2022] Open
Abstract
Age-related hearing loss (presbycusis) is a common human disorder, affecting one in three Americans aged 60 and over. Previous studies have shown that presbyacusis is associated with a loss of non-sensory cells in the cochlear lateral wall. Sox10 is a transcription factor crucial to the development and maintenance of neural crest-derived cells including some non-sensory cell types in the cochlea. Mutations of the Sox10 gene are known to cause various combinations of hearing loss and pigmentation defects in humans. This study investigated the potential relationship between Sox10 gene expression and pathological changes in the cochlear lateral wall of aged CBA/CaJ mice and human temporal bones from older donors. Cochlear tissues prepared from young adult (1–3 month-old) and aged (2–2.5 year-old) mice, and human temporal bone donors were examined using quantitative immunohistochemical analysis and transmission electron microscopy. Cells expressing Sox10 were present in the stria vascularis, outer sulcus and spiral prominence in mouse and human cochleas. The Sox10+ cell types included marginal and intermediate cells and outer sulcus cells, including those that border the scala media and those extending into root processes (root cells) in the spiral ligament. Quantitative analysis of immunostaining revealed a significant decrease in the number of Sox10+ marginal cells and outer sulcus cells in aged mice. Electron microscopic evaluation revealed degenerative alterations in the surviving Sox10+ cells in aged mice. Strial marginal cells in human cochleas from donors aged 87 and older showed only weak immunostaining for Sox10. Decreases in Sox10 expression levels and a loss of Sox10+ cells in both mouse and human aged ears suggests an important role of Sox10 in the maintenance of structural and functional integrity of the lateral wall. A loss of Sox10+ cells may also be associated with a decline in the repair capabilities of non-sensory cells in the aged ear.
Collapse
Affiliation(s)
- Xinping Hao
- Department of Otolaryngology – Head & Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Yazhi Xing
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Michael W. Moore
- Department of Otolaryngology – Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Jianning Zhang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Department of Otolaryngology, Shanghai Yueyang Integrated Medicine Hospital, Shanghai, China
| | - Demin Han
- Department of Otolaryngology – Head & Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- * E-mail: (HL); (DH)
| | - Bradley A. Schulte
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Department of Otolaryngology – Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Judy R. Dubno
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Department of Otolaryngology – Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Hainan Lang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- * E-mail: (HL); (DH)
| |
Collapse
|
7
|
Fiori MC, Reuss L, Cuello LG, Altenberg GA. Functional analysis and regulation of purified connexin hemichannels. Front Physiol 2014; 5:71. [PMID: 24611052 PMCID: PMC3933781 DOI: 10.3389/fphys.2014.00071] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 02/06/2014] [Indexed: 01/08/2023] Open
Abstract
Gap-junction channels (GJCs) are aqueous channels that communicate adjacent cells. They are formed by head-to-head association of two hemichannels (HCs), one from each of the adjacent cells. Functional HCs are connexin hexamers composed of one or more connexin isoforms. Deafness is the most frequent sensineural disorder, and mutations of Cx26 are the most common cause of genetic deafness. Cx43 is the most ubiquitous connexin, expressed in many organs, tissues, and cell types, including heart, brain, and kidney. Alterations in its expression and function play important roles in the pathophysiology of very frequent medical problems such as those related to cardiac and brain ischemia. There is extensive information on the relationship between phosphorylation and Cx43 targeting, location, and function from experiments in cells and organs in normal and pathological conditions. However, the molecular mechanisms of Cx43 regulation by phosphorylation are hard to tackle in complex systems. Here, we present the use of purified HCs as a model for functional and structural studies. Cx26 and Cx43 are the only isoforms that have been purified, reconstituted, and subjected to functional and structural analysis. Purified Cx26 and Cx43 HCs have properties compatible with those demonstrated in cells, and present methodologies for the functional analysis of purified HCs reconstituted in liposomes. We show that phosphorylation of serine 368 by PKC produces a partial closure of the Cx43 HCs, changing solute selectivity. We also present evidence that the effect of phosphorylation is highly cooperative, requiring modification of several connexin subunits, and that phosphorylation of serine 368 elicits conformational changes in the purified HCs. The use of purified HCs is starting to provide critical data to understand the regulation of HCs at the molecular level.
Collapse
Affiliation(s)
- Mariana C Fiori
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, Texas Tech University Health Sciences Center Lubbock, TX, USA
| | - Luis Reuss
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, Texas Tech University Health Sciences Center Lubbock, TX, USA
| | - Luis G Cuello
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, Texas Tech University Health Sciences Center Lubbock, TX, USA
| | - Guillermo A Altenberg
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, Texas Tech University Health Sciences Center Lubbock, TX, USA
| |
Collapse
|
8
|
Eckhard A, Müller M, Salt A, Smolders J, Rask-Andersen H, Löwenheim H. Water permeability of the mammalian cochlea: functional features of an aquaporin-facilitated water shunt at the perilymph-endolymph barrier. Pflugers Arch 2014; 466:1963-85. [PMID: 24385019 PMCID: PMC4081528 DOI: 10.1007/s00424-013-1421-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/03/2013] [Accepted: 12/03/2013] [Indexed: 11/02/2022]
Abstract
The cochlear duct epithelium (CDE) constitutes a tight barrier that effectively separates the inner ear fluids, endolymph and perilymph, thereby maintaining distinct ionic and osmotic gradients that are essential for auditory function. However, in vivo experiments have demonstrated that the CDE allows for rapid water exchange between fluid compartments. The molecular mechanism governing water permeation across the CDE remains elusive. We computationally determined the diffusional (PD) and osmotic (Pf) water permeability coefficients for the mammalian CDE based on in silico simulations of cochlear water dynamics integrating previously derived in vivo experimental data on fluid flow with expression sites of molecular water channels (aquaporins, AQPs). The PD of the entire CDE (PD = 8.18 × 10(-5) cm s(-1)) and its individual partitions including Reissner's membrane (PD = 12.06 × 10(-5) cm s(-1)) and the organ of Corti (PD = 10.2 × 10(-5) cm s(-1)) were similar to other epithelia with AQP-facilitated water permeation. The Pf of the CDE (Pf = 6.15 × 10(-4) cm s(-1)) was also in the range of other epithelia while an exceptionally high Pf was determined for an epithelial subdomain of outer sulcus cells in the cochlear apex co-expressing AQP4 and AQP5 (OSCs; Pf = 156.90 × 10(-3) cm s(-1)). The Pf/PD ratios of the CDE (Pf/PD = 7.52) and OSCs (Pf/PD = 242.02) indicate an aqueous pore-facilitated water exchange and reveal a high-transfer region or "water shunt" in the cochlear apex. This "water shunt" explains experimentally determined phenomena of endolymphatic longitudinal flow towards the cochlear apex. The water permeability coefficients of the CDE emphasise the physiological and pathophysiological relevance of water dynamics in the cochlea in particular for endolymphatic hydrops and Ménière's disease.
Collapse
Affiliation(s)
- A Eckhard
- Hearing Research Center, Department of Otorhinolaryngology-Head & Neck Surgery, University of Tübingen Medical Centre, Elfriede-Aulhorn-Strasse 5, 72076, Tübingen, Germany
| | | | | | | | | | | |
Collapse
|
9
|
|
10
|
Morphological and functional changes in a new animal model of Ménière's disease. J Transl Med 2013; 93:1001-11. [PMID: 23877650 DOI: 10.1038/labinvest.2013.91] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 06/20/2013] [Accepted: 06/22/2013] [Indexed: 11/09/2022] Open
Abstract
The purpose of this study was to clarify the underlying mechanism of vertiginous attacks in Ménière's disease (MD) while obtaining insight into water homeostasis in the inner ear using a new animal model. We conducted both histopathological and functional assessment of the vestibular system in the guinea-pig. In the first experiment, all animals were maintained 1 or 4 weeks after electrocauterization of the endolymphatic sac of the left ear and were given either saline or desmopressin (vasopressin type 2 receptor agonist). The temporal bones from both ears were harvested and the extent of endolymphatic hydrops was quantitatively assessed. In the second experiment, either 1 or 4 weeks after surgery, animals were assessed for balance disorders and nystagmus after the administration of saline or desmopressin. In the first experiment, the proportion of endolymphatic space in the cochlea and the saccule was significantly greater in ears that survived for 4 weeks after surgery and were given desmopressin compared with other groups. In the second experiment, all animals that underwent surgery and were given desmopressin showed spontaneous nystagmus and balance disorder, whereas all animals that had surgery but without desmopressin administration were asymptomatic. Our animal model induced severe endolymphatic hydrops in the cochlea and the saccule, and showed episodes of balance disorder along with spontaneous nystagmus. These findings suggest that administration of desmopressin can exacerbate endolymphatic hydrops because of acute V2 (vasopressin type 2 receptor)-mediated effects, and, when combined with endolymphathic sac dysfunction, can cause temporary vestibular abnormalities that are similar to the vertiginous attacks in patients with MD.
Collapse
|
11
|
Adeno-associated virus-mediated gene delivery into the scala media of the normal and deafened adult mouse ear. Gene Ther 2011; 18:569-78. [PMID: 21209625 PMCID: PMC3085601 DOI: 10.1038/gt.2010.175] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Murine models are ideal for studying cochlear gene transfer, as many hearing loss-related mutations have been discovered and mapped within the mouse genome. However, because of the small size and delicate nature, the membranous labyrinth of the mouse is a challenging target for the delivery of viral vectors. To minimize injection trauma, we developed a procedure for the controlled release of adeno-associated viruses (AAVs) into the scala media of adult mice. This procedure poses minimal risk of injury to structures of the cochlea and middle ear, and allows for near-complete preservation of low and middle frequency hearing. In this study, transduction efficiency and cellular specificity of AAV vectors (serotypes 1, 2, 5, 6 and 8) were investigated in normal and drug-deafened ears. Using the cytomegalovirus promoter to drive gene expression, a variety of cell types were transduced successfully, including sensory hair cells and supporting cells, as well as cells in the auditory nerve and spiral ligament. Among all five serotypes, inner hair cells were the most effectively transduced cochlear cell type. All five serotypes of AAV vectors transduced cells of the auditory nerve, though serotype 8 was the most efficient vector for transduction. Our findings indicate that efficient AAV inoculation (via the scala media) can be performed in adult mouse ears, with hearing preservation a realistic goal. The procedure we describe may also have applications for intra-endolymphatic drug delivery in many mouse models of human deafness.
Collapse
|
12
|
Abstract
Menière disease usually begins in adults from 20 to 60 years old, and occurs in more than 10% of patients older than 65. The treatment of Menière disease in the elderly represents a challenge because of polymedication. Antivertiginous drugs such as betahistine and cinnarizin give good results with minor secondary effects. In contrast, major vestibular suppressor drugs such as thiethylperazin must be avoided as long-term treatment because of their side effects. Definitive vestibular surgical deafferentations such as labyrinthectomy and selective vestibular neurectomy represent optional procedures but must be carefully evaluated from case to case. Ablative procedures remain the efficient treatment of drop attacks, which represent a high potential risk of severe injuries by older patients sometimes with important social consequences.
Collapse
Affiliation(s)
- Dominique Vibert
- Department of Otorhinolaryngology, Head and Neck Surgery, Inselspital, University of Berne, 3010 Berne, Switzerland.
| | | | | |
Collapse
|
13
|
Salt AN, Plontke SK. Endolymphatic hydrops: pathophysiology and experimental models. Otolaryngol Clin North Am 2010; 43:971-83. [PMID: 20713237 DOI: 10.1016/j.otc.2010.05.007] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
It is well established that endolymphatic hydrops plays a role in Ménière disease, even though the precise role is not fully understood and the presence of hydrops in the ear does not always result in symptoms of the disease. It nevertheless follows that a scientific understanding of how hydrops arises, how it affects the function of the ear, and how it can be manipulated or reversed could contribute to the development of effective treatments for the disease. Measurements in animal models in which endolymphatic hydrops has been induced have given numerous insights into the relationships between hydrops and other pathologic and electrophysiological changes, and how these changes influence the function of the ear. The prominent role of the endolymphatic sac in endolymph volume regulation, and the cascade of histopathological and electrophysiological changes that are associated with chronic endolymphatic hydrops, have now been established. An increasing number of models are now available that allow specific aspects of the interrelationships to be studied. The yclical nature of Ménière symptoms gives hope that treatments can be developed to maintain the ear in permanent state of remission, possibly by controlling endolymphatic hydrops, thereby avoiding the rogressive damage and secondary pathologic changes that may also contribute to the patient's symptoms.
Collapse
Affiliation(s)
- Alec N Salt
- Department of Otolaryngology, Washington University School of Medicine, Box 8115, 660 South Euclid Avenue, St Louis, MO 63110, USA.
| | | |
Collapse
|
14
|
Yum SW, Zhang J, Valiunas V, Kanaporis G, Brink PR, White TW, Scherer SS. Human connexin26 and connexin30 form functional heteromeric and heterotypic channels. Am J Physiol Cell Physiol 2007; 293:C1032-48. [PMID: 17615163 DOI: 10.1152/ajpcell.00011.2007] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mutations in GJB2 and GJB6, the genes that encode the human gap junction proteins connexin26 (Cx26) and connexin30 (Cx30), respectively, cause hearing loss. Cx26 and Cx30 are both expressed in the cochlea, leading to the potential formation of heteromeric hemichannels and heterotypic gap junction channels. To investigate their interactions, we expressed human Cx26 and Cx30 individually or together in HeLa cells. When they were expressed together, Cx26 and Cx30 appeared to interact directly (by their colocalization in gap junction plaques, by coimmunoprecipitation, and by fluorescence resonance energy transfer). Scrape-loading cells that express either Cx26 or Cx30 demonstrated that Cx26 homotypic channels robustly transferred both cationic and anionic tracers, whereas Cx30 homotypic channels transferred cationic but not anionic tracers. Cells expressing both Cx26 and Cx30 also transferred both cationic and anionic tracers by scrape loading, and the rate of calcein (an anionic tracer) transfer was intermediate between their homotypic counterparts by fluorescence recovery after photobleaching. Fluorescence recovery after photobleaching also showed that Cx26 and Cx30 form functional heterotypic channels, allowing the transfer of calcein, which did not pass the homotypic Cx30 channels. Electrophysiological recordings of cell pairs expressing different combinations of Cx26 and/or Cx30 demonstrated unique gating properties of cell pairs expressing both Cx26 and Cx30. These results indicate that Cx26 and Cx30 form functional heteromeric and heterotypic channels, whose biophysical properties and permeabilities are different from their homotypic counterparts.
Collapse
Affiliation(s)
- Sabrina W Yum
- Section of Neurology, St. Christopher's Hospital for Children, Erie Ave. at Front St., Philadelphia, PA 19134, USA.
| | | | | | | | | | | | | |
Collapse
|
15
|
Hordichok AJ, Steyger PS. Closure of supporting cell scar formations requires dynamic actin mechanisms. Hear Res 2007; 232:1-19. [PMID: 17716843 PMCID: PMC2665176 DOI: 10.1016/j.heares.2007.06.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2007] [Revised: 05/29/2007] [Accepted: 06/05/2007] [Indexed: 12/29/2022]
Abstract
In many vertebrate inner ear sensory epithelia, dying sensory hair cells are extruded, and the apices of surrounding supporting cells converge to re-seal the epithelial barrier between the electrochemically-distinct endolymph and perilymph. These cellular mechanisms remain poorly understood. Dynamic microtubular mechanisms have been proposed for hair cell extrusion; while contractile actomyosin-based mechanisms are required for cellular extrusion and closure in epithelial monolayers. The hypothesis that cytoskeletal mechanisms are required for hair cell extrusion and supporting cell scar formation was tested using bullfrog saccules incubated with gentamicin (6h), and allowed to recover (18h). Explants were then fixed, labeled for actin and cytokeratins, and viewed with confocal microscopy. To block dynamic cytoskeletal processes, disruption agents for microtubules (colchicine, paclitaxel) myosin (Y-27632, ML-9) or actin (cytochalasin D, latrunculin A) were added during treatment and recovery. Microtubule disruption agents had no effect on hair cell extrusion or supporting cell scar formation. Myosin disruption agents appeared to slow down scar formation but not hair cell extrusion. Actin disruption agents blocked scar formation, and largely prevented hair cell extrusion. These data suggest that actin-based cytoskeletal processes are required for hair cell extrusion and supporting cell scar formation in bullfrog saccules.
Collapse
Affiliation(s)
- Andrew J Hordichok
- Oregon Hearing Research Center, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | | |
Collapse
|
16
|
Esmaeili M, Bonyadi M, Nejadkazem M. Common mutation analysis of GJB2 and GJB6 genes in affected families with autosomal recessive non-syndromic hearing loss from Iran: simultaneous detection of two common mutations (35delG/del(GJB6-D13S1830)) in the DFNB1-related deafness. Int J Pediatr Otorhinolaryngol 2007; 71:869-73. [PMID: 17368814 DOI: 10.1016/j.ijporl.2007.02.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2006] [Revised: 02/05/2007] [Accepted: 02/17/2007] [Indexed: 11/28/2022]
Abstract
OBJECTIVE DFNB1 locus has been reported as a major cause of autosomal recessive non-syndromic hearing loss (ARNSHL) worldwide. 35delG and del(GJB6-D13S1830) are thought to be two common mutations in this locus among Caucasians. The aim of this study is to determine the significance of these two mutations in aetiology of ARNSHL in Iran. METHODS One hundred and thirty-three unrelated patients with ARNSHL were tested by using multiplex allele-specific PCR assay after validation by positive control samples. RESULTS The frequency of 35delG was about 18.5%, however, del(GJB6-D13S1830) was not found in the studied patients. Parental consanguinity was observed in 50% of 35delG-mutated families. CONCLUSIONS Our results support founder effect regarding these mutations.
Collapse
Affiliation(s)
- Mohsen Esmaeili
- Genetic Lab. Drug Applied Research Center, Biotechnology Research Center, Tabriz University of Medical Sciences, University Ave., Tabriz, Iran
| | | | | |
Collapse
|
17
|
Affiliation(s)
- S Plontke
- Klinik für Hals-, Nasen- und Ohrenheilkunde, Kopf- und Halschirurgie, Universitätsklinikum Tübingen, 72076, Tübingen.
| |
Collapse
|
18
|
Deng Y, Chen Y, Reuss L, Altenberg GA. Mutations of connexin 26 at position 75 and dominant deafness: essential role of arginine for the generation of functional gap-junctional channels. Hear Res 2006; 220:87-94. [PMID: 16945493 DOI: 10.1016/j.heares.2006.07.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2006] [Revised: 07/11/2006] [Accepted: 07/13/2006] [Indexed: 11/27/2022]
Abstract
Gap-junctional channels are large intercellular aqueous pores formed by head-to-head association of two gap-junctional hemichannels (connexin hexamers), one from each of the adjacent cells. The mechano-transduction of sound waves into electrical impulses occurs in the cochlea, which houses the organ of Corti. Hereditary deafness is frequent and mutations of connexin 26, the predominant connexin of the cochlea, are its most frequent cause. Mutations of R75 cause deafness and disrupt gap-junctional communication. Here, we determined the effects of substitutions of R75 with different residues (alanine, asparagine, aspartic acid, lysine, phenylalanine, tyrosine or tryptophan) on formation of gap-junctional channels and hemichannels. We show that connexin 26 R75 is essential for the formation of gap-junctional channels. Substitution of R75 with aromatic residues yields functional hemichannels that display altered voltage dependence, whereas substitution with other residues yields non-functional hemichannels. The expression of R75 mutants has a dominant negative effect on gap-junctional communication mediated by wild-type connexin 26, independently of the ability of the mutants to form functional gap-junctional hemichannels. Our results show that the arginine located at position 75 of connexin 26 is essential for function, and cannot be replaced by other residues.
Collapse
Affiliation(s)
- Yanqin Deng
- Department of Neuroscience and Cell Biology, and the Sealy Center for Structural Biology and Molecular Biophysics, The University of Texas Medical Branch, Galveston, TX 77555-0437, United States
| | | | | | | |
Collapse
|
19
|
Abstract
Non-syndromic deafness is a paradigm of genetic heterogeneity with 85 loci and 39 nuclear disease genes reported so far. Autosomal-recessive genes are responsible for about 80% of the cases of hereditary non-syndromic deafness of pre-lingual onset with 23 different genes identified to date. In the present article, we review these 23 genes, their function, and their contribution to genetic deafness in different populations. The wide range of functions of these DFNB genes reflects the heterogeneity of the genes involved in hearing and hearing loss. Several of these genes are involved in both recessive and dominant deafness, or in both non-syndromic and syndromic deafness. Mutations in the GJB2 gene encoding connexin 26 are responsible for as much as 50% of pre-lingual, recessive deafness. By contrast, mutations in most of the other DFNB genes have so far been detected in only a small number of families, and their contribution to deafness on a population scale might therefore be limited. Identification of all genes involved in hereditary hearing loss will help in our understanding of the basic mechanisms underlying normal hearing, in early diagnosis and therapy.
Collapse
Affiliation(s)
- M B Petersen
- Department of Genetics, Institute of Child Health, Aghia Sophia Children's Hospital, Athens, Greece.
| | | |
Collapse
|
20
|
Robertson NG, Cremers CWRJ, Huygen PLM, Ikezono T, Krastins B, Kremer H, Kuo SF, Liberman MC, Merchant SN, Miller CE, Nadol JB, Sarracino DA, Verhagen WIM, Morton CC. Cochlin immunostaining of inner ear pathologic deposits and proteomic analysis in DFNA9 deafness and vestibular dysfunction. Hum Mol Genet 2006; 15:1071-85. [PMID: 16481359 DOI: 10.1093/hmg/ddl022] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Seven missense mutations and one in-frame deletion mutation have been reported in the coagulation factor C homology (COCH) gene, causing the adult-onset, progressive sensorineural hearing loss and vestibular disorder at the DFNA9 locus. Prevalence of COCH mutations worldwide is unknown, as there is no systematic screening effort for late-onset hearing disorders; however, to date, COCH mutations have been found on four continents and the possibility of COCH playing an important role in presbycusis and disorders of imbalance has been considered. Cochlin (encoded by COCH) has also been shown as a major target antigen for autoimmune sensorineural hearing loss. In this report, we present histopathology, immunohistochemistry and proteomic analyses of inner ear tissues from post-mortem DFNA9 temporal bone samples of an individual from a large Dutch kindred segregating the P51S mutation and adult human unaffected controls, and wild-type (+/+) and Coch null (-/-) knock-out mice. DFNA9 is an inner ear disorder with a unique histopathology showing loss of cellularity and aggregation of abundant homogeneous acellular eosinophilic deposits in the cochlear and vestibular labyrinths, similar to protein aggregation in well-known neurodegenerative disorders. By immunohistochemistry on the DFNA9 temporal bone sections, we have shown cochlin staining of the characteristic cochlear and vestibular deposits, indicating aggregation of cochlin in the same structures in which it is normally expressed. Proteomic analysis identified cochlin as the most abundant protein in mouse and human cochleae. The high-level expression and stability of cochlin in the inner ear, even in the absence and severe atrophy of the fibrocytes that normally express COCH, are shown through these studies and further elucidate the pathobiologic events occurring in DFNA9 leading to hearing loss and vestibular dysfunction.
Collapse
Affiliation(s)
- Nahid G Robertson
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Lopez IA, Acuna D, Galbraith G, Bok D, Ishiyama A, Liu W, Kurtz I. Time course of auditory impairment in mice lacking the electroneutral sodium bicarbonate cotransporter NBC3 (slc4a7). BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2005; 160:63-77. [PMID: 16181686 DOI: 10.1016/j.devbrainres.2005.08.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2005] [Revised: 08/13/2005] [Accepted: 08/17/2005] [Indexed: 12/13/2022]
Abstract
Mice with a targeted disruption of the gene encoding the stilbene-insensitive electroneutral sodium bicarbonate cotransporter (NBC3; slc4a7) exhibit cochlear and retinal degeneration. To establish the progressive nature of sensory cells loss in slc4a7-/- deficient mice, we studied the morphology of cochleas of slc4a7-/- and slc4a7+/+ mice from postnatal day two (P2) to ninety (P90). Cell death was evaluated in slc4a7-/- cochleas using the TUNEL technique and caspase-3 immunoreactivity. The time course of NBC3 expression in the cochlea was assessed by immunohistochemistry using an antibody against NBC3. Between P2 and P8, slc4a7-/- mice cochlea exhibit normal morphology. There was a normal complement of inner and outer hair cells from the hook to the apical region. At P15, slc4a7-/- mice cochlea inner and outer hair cells were still present at the hook region, and vacuoles were seen underneath Hensen's cells. At P21, inner and outer hair cells were degenerated in this region. Between P30 and P90, there was a pronounced loss of hair cells and spiral ganglia neurons. Morphological analysis of the spiral ligament showed a progressive loss of type II and IV fibrocytes beginning at day 21. Transmission electron microscopy observations at P30 and P90 revealed that type II and IV fibrocytes showed shrinkage and vacuolization. In addition, hair cells were deteriorated with evidence of shrinkage and picnotic nuclei. TUNEL staining showed apoptotic cells at P8 in the organ of Corti at the basal region of the cochlea. At P15, caspase-3 immunoreactivity was present in supporting cells of the organ of Corti. NBC3 mild immunoreactivity was detected in the organ of Corti at P11. There was an increase in the expression of NBC3 in the spiral ligament between P17 and P19. From P21 to P90, NBC3 expression was confined to the spiral ligament and inner and outer sulcus cells. The vestibular sensory epithelia from slc4a7-/- mice were normal from P2 to P90. Damage of the sensory epithelia at the high frequency zone of the cochlea suggests that NBC3 may play an important physiological role in this region.
Collapse
Affiliation(s)
- Ivan A Lopez
- Department of Surgery, Division of Head and Neck, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| | | | | | | | | | | | | |
Collapse
|
22
|
Merchant SN, Adams JC, Nadol JB. Pathophysiology of Meniere's syndrome: are symptoms caused by endolymphatic hydrops? Otol Neurotol 2005; 26:74-81. [PMID: 15699723 DOI: 10.1097/00129492-200501000-00013] [Citation(s) in RCA: 349] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND The association of Meniere's syndrome with endolymphatic hydrops has led to the formation of a central hypothesis: many possible etiologic factors lead to hydrops, and hydrops in turn generates the symptoms. However, this hypothesis of hydrops as being the final common pathway has not been proven conclusively. SPECIFIC AIM To examine human temporal bones with respect to the role of hydrops in causing symptoms in Meniere's syndrome. If the central hypothesis were true, every case of Meniere's syndrome should have hydrops and every case of hydrops should show the typical symptoms. METHODS Review of archival temporal bone cases with a clinical diagnosis of Meniere's syndrome (28 cases) or a histopathologic diagnosis of hydrops (79 cases). RESULTS All 28 cases with classical symptoms of Meniere's syndrome showed hydrops in at least one ear. However, the reverse was not true. There were 9 cases with idiopathic hydrops and 10 cases with secondary hydrops, but the patients did not exhibit the classic symptoms of Meniere's syndrome. A review of the literature revealed cases with asymptomatic hydrops (similar to the current study), as well as cases where symptoms of Meniere's syndrome existed during life but no hydrops was observed on histology. We also review recent experimental data where obstruction of the endolymphatic duct in guinea pigs resulted in cytochemical abnormalities within fibrocytes of the spiral ligament before development of hydrops. This result is consistent with the hypothesis that hydrops resulted from disordered fluid homeostasis caused by disruption of regulatory elements within the spiral ligament. CONCLUSION Endolymphatic hydrops should be considered as a histologic marker for Meniere's syndrome rather than being directly responsible for its symptoms.
Collapse
Affiliation(s)
- Saumil N Merchant
- Otopathology Laboratory, Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts 02114-3096, USA.
| | | | | |
Collapse
|
23
|
Chen Y, Deng Y, Bao X, Reuss L, Altenberg GA. Mechanism of the defect in gap-junctional communication by expression of a connexin 26 mutant associated with dominant deafness. FASEB J 2005; 19:1516-8. [PMID: 16009703 DOI: 10.1096/fj.04-3491fje] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Gap-junctional channels (connexin oligomers) are large-diameter aqueous pores formed by head-to-head association of two gap-junctional hemichannels, one from each of the adjacent cells. Profound hearing loss of genetic origin is common, and mutations of connexin 26 (Cx26) are the most frequent cause of this disorder. The Cx26 R75W mutant has been associated with disruption of cell-to-cell communication and profound hearing loss, but the mechanism of the gap-junctional defect is unknown. Here, we show that Cx26 R75W forms gap-junctional hemichannels that display altered voltage dependency and reduced permeability, and which cannot form functional gap-junctional channels between neighboring cells. The R75W phenotype is dominant at the gap-junction channel but not at the hemichannel level. Therefore, the absence of gap-junctional communication caused by R75W expression is due to defective gap-junction formation by functional hemichannels.
Collapse
Affiliation(s)
- Y Chen
- Department of Neuroscience and Cell Biology, and Membrane Protein Laboratory of the Sealy Center for Structural Biology, The University of Texas Medical Branch Galveston, Texas 77555-0437, USA
| | | | | | | | | |
Collapse
|
24
|
Henzl MT, Thalmann I, Larson JD, Ignatova EG, Thalmann R. The cochlear F-box protein OCP1 associates with OCP2 and connexin 26. Hear Res 2005; 191:101-9. [PMID: 15109709 DOI: 10.1016/j.heares.2004.01.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2003] [Accepted: 01/01/2004] [Indexed: 11/18/2022]
Abstract
OCP1 and OCP2 are the most abundant proteins in the organ of Corti. Their distributions map identically to the epithelial gap-junction system, which unites the supporting cell population. Sequence data imply that OCP1 and OCP2 are subunits of an SCF E3 ubiquitin ligase. Consistent with that hypothesis, electrophoretic mobility-shift assays and pull-down assays with immobilized OCP1 demonstrate the formation of an OCP1-OCP2 complex. Sedimentation equilibrium data indicate that the complex is heterodimeric. The coincidence of the OCP1-OCP2 distribution and the epithelial gap-junction system suggests that one or more connexin isoforms may be targets of an SCF(OCP1) complex. Significantly, immobilized OCP1 binds (35)S-labeled connexin 26 (Cx26) produced by in vitro transcription-translation. Moreover, Cx26 can be co-immunoprecipitated from extracts of the organ of Corti by immobilized anti-OCP1, implying that OCP1 and Cx26 may associate in vivo. Given that lesions in the Cx26 gene (GJB2) are the most common cause of hereditary deafness, the OCP1-Cx26 interaction has substantial biomedical relevance.
Collapse
Affiliation(s)
- Michael T Henzl
- Department of Biochemistry, University of Missouri - Columbia, 117 Schweitzer Hall, Columbia, MO 65211, USA.
| | | | | | | | | |
Collapse
|
25
|
Samadi DS, Saunders JC, Crenshaw EB. Mutation of the POU-domain gene Brn4/Pou3f4 affects middle-ear sound conduction in the mouse. Hear Res 2005; 199:11-21. [PMID: 15574296 DOI: 10.1016/j.heares.2004.07.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2003] [Accepted: 07/13/2004] [Indexed: 11/19/2022]
Abstract
Mutagenesis of the POU-domain gene Brn4/Pou3f4 causes defects in the cochlear duct, semicircular canal, temporal bone and stapes footplate. The footplate defect suggested a middle-ear conductive component to the hearing loss associated with this mutation. This was examined by measuring velocity transfer functions at the umbo of wild type and knockout mice during sound stimulation of the tympanic membrane. When the median umbo velocity of test frequencies in the two groups were compared, the mid-range frequencies of the knockout mice showed a statistically reliable reduction in velocity (maximum of 13 dB) and high variability among animals. These results indicated that mutation of the POU-domain gene, Brn4, changed middle-ear sound conduction when measured at the umbo. The origin of the abnormal velocity response was sought by puncturing a hole in the pars flaccida (PF), and subsequently, measuring movements at the umbo and the head of the long arm of the incus. This hole permitted us to measure velocity at the tip of the incus long arm, just above the incudostapedial joint. The comparison of umbo behavior in both groups with PF perforated showed a loss of sensitivity in the mid-range frequencies of the knockout animals. A comparison of incus velocity in the two groups also exhibited a velocity reduction in the mid-range frequencies of the knockout animals. The reduction at the incus, however, was milder than observed at the umbo. The effect of the perforation in, and variability of, the knockout incus responses may have masked a more potent mid-range frequency effect. Nevertheless, evaluation of the stapes and oval window in knockout mice showed variable pathology from ear to ear. The presence of this pathology, the mid-frequency loss in incus sensitivity and the variability in incus velocity among animals suggested that abnormal stapes behavior in Brn4 deficient mice may determine the response of the ossicles, and thus account for the abnormal mid-frequency umbo behavior seen in knockout animals.
Collapse
Affiliation(s)
- Daniel S Samadi
- Mammalian Neurogenetics Group, Center for Childhood Communication, Division of Pediatric Otolaryngology, The Children's Hospital of Philadelphia, 712 Abramson Research Center, 34th and Civic Blvd., Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
26
|
Abstract
Gene transfer into the inner ear is an attractive technology for clinical applications. It offers the hope of preventing, arresting, reversing or curing vestibular or hearing disorders caused by hereditary diseases or environmental insults. There are currently very few treatment options for vestibular disorders and sensorineural hearing loss, and therefore, it is important to investigate and develop new technologies for inner ear disease. In utero gene transfer may indeed be an option, but this technology will have many technical and ethical issues that remain to be overcome.
Collapse
Affiliation(s)
- H Pau
- Royal Liverpool Children's Hospital, Alder Hey, Liverpool, UK.
| | | |
Collapse
|
27
|
Kusunoki T, Cureoglu S, Schachern PA, Baba K, Kariya S, Paparella MM. Age-related histopathologic changes in the human cochlea: a temporal bone study. Otolaryngol Head Neck Surg 2005; 131:897-903. [PMID: 15577787 DOI: 10.1016/j.otohns.2004.05.022] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVES Previous reports on aging of human cochlea included subjects with ear diseases or ototoxic drugs. We studied spiral ganglion cells, hair cells, and lateral wall of cochlea from subjects without ear disease or ototoxic drugs. STUDY DESIGN This study included 39 temporal bones from 24 subjects aged 1 day to 86 years. We assessed standard cytocochleograms, mean loss of fibrocytes in spiral ligament, and areas of stria vascularis. RESULTS Losses of outer hair cells and fibrocytes were significantly greater in children, adults, and the elderly compared with infants. Spiral ganglion cell loss was significantly greater in adults and elderly compared with infants and children. Areas of stria vascularis of infants were significantly larger than the elderly. CONCLUSIONS Degenerative changes of outer hair cells occur in children but spiral ganglion cells remain the same until around 20 years. Degeneration of stria vascularis due to aging appears to be slower than spiral ligament. EBM RATING C.
Collapse
|
28
|
Hildebrand MS, de Silva MG, Klockars T, Solares CA, Hirose K, Smith JD, Patel SC, Dahl HHM. Expression of the carrier protein apolipoprotein D in the mouse inner ear. Hear Res 2005; 200:102-14. [PMID: 15668042 DOI: 10.1016/j.heares.2004.08.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2004] [Accepted: 08/18/2004] [Indexed: 11/29/2022]
Abstract
The cochlear portion of the inner ear converts movements produced by sound waves into electrical impulses. Transcripts enriched in the cochlea are likely to have an important role in hearing. In this paper, we report that microarray analyses of the Soares NMIE inner ear library revealed cochlear enriched expression of apolipoprotein D (apoD), a glycoprotein and member of the lipocalin family that transport small hydrophobic ligands. The cochlear enriched expression of Apod was validated by quantitative real time PCR analysis. To investigate the function of apoD in the inner ear the transcript and protein were localised in the cochlea. Apod messenger RNA (mRNA) expression was localised to the spiral ligament and spiral limbus, particularly in the suprastrial and supralimbral regions. The apoD protein was detected in the spiral ligament, spiral limbus and also in the outer hair cells of the organ of Corti. Investigation of cell lines exhibiting characteristics of hair and supporting cells revealed no Apod mRNA expression in these cells. This suggests transport of the protein within the cochlea, followed by internalisation into outer hair cells. The spiral limbus and ligament contain subpopulations of fibrocytes that are intimately involved in regulation of ion balance in the cochlear fluids and type I, II and III fibrocytes of the spiral ligament were all shown to be positive for apoD protein. On the basis of these results it was hypothesised that apoD could be involved in maintaining cochlear fluid homeostasis. To determine whether the apoD gene product was important for normal auditory function the hearing ability of an apoD knockout mouse was tested. The mouse was found to have a hearing threshold that was not significantly different to the control strain.
Collapse
Affiliation(s)
- Michael S Hildebrand
- Department of Gene Identification and Expression, Murdoch Childrens Research Institute, Royal Children's Hospital, Flemington Road, Parkville, Melbourne, Vic. 3052, Australia
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Suzuki M, Kikuchi T, Ikeda K. Endocochlear potential and endolymphatic K+ changes induced by gap junction blockers. Acta Otolaryngol 2004; 124:902-6. [PMID: 15513524 DOI: 10.1080/00016480410017369] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
OBJECTIVE To examine the effects of gap junction blockers on the endocochlear potential (EP) and endolymphatic potassium concentration ([K(+)](e)). MATERIAL AND METHODS The EP and [K(+)](e) were monitored using double-barreled ion-selective microelectrodes in the second turn of the guinea pig cochlea during perilymphatic perfusion. RESULTS When the perilymphatic scalae of the cochlea were perfused with artificial perilymph containing 10 mM n-heptanol the EP was decreased by -8.8+/-1.4 mV (n=10), and this was accompanied by a decline in the [K(+)](e) of -6.7+/-2.1 mM (n=6). Perilymphatic application of 10 mM hexanol also produced declines in both the EP and [K(+)](e). In control studies, perilymphatic perfusion with 10 mM ethanol showed no remarkable changes in either the EP or [K(+)](e). Anoxia during perfusion with heptanol resulted in the generation of a negative EP, similar to the situation in controls. CONCLUSIONS A decline in the EP together with a lowering of [K(+)](e) induced by long-chain n-alkanols, which act as gap junction blockers, may be explained by an interruption in potassium ion transport related to a gap junction dysfunction.
Collapse
Affiliation(s)
- Masaaki Suzuki
- Department of Otorhinolaryngology-Head and Neck Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | | | | |
Collapse
|
30
|
Shen Z, Liang F, Hazen-Martin DJ, Schulte BA. BK channels mediate the voltage-dependent outward current in type I spiral ligament fibrocytes. Hear Res 2004; 187:35-43. [PMID: 14698085 DOI: 10.1016/s0378-5955(03)00345-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Recent experimental and clinical studies have provided considerable evidence to support the phenomenon of K(+) recycling in the mammalian cochlea. However, the precise cellular and molecular mechanisms underlying and regulating this process remain only partially understood. Here, we report that cultured type I spiral ligament fibrocytes (SLFs), a major component of the K(+) recycling pathway, have a dominant K(+) membrane conductance that is mediated by BK channels. The averaged half-maximal voltage-dependent membrane potential for the whole-cell currents was 70+/-1.2 mV at 1 nM intracellular free Ca(2+) and shifted to 38+/-0.2 mV at 20 microM intracellular free Ca(2+) (n=4-6). The reversal potential of whole-cell tail currents against different bath K(+) concentrations was 52 mV per decade (n=3-6). The sequence of relative ion permeability of the whole-cell conductance was K(+)>Rb(+)z.Gt;Cs(+)>Na(+) (n=5-17). The whole-cell currents were inhibited by extracellular tetraethylammonium and iberiotoxin (IbTx) with IC(50) values of 0.07 mM and 0.013 microM, respectively (n=3-7). The membrane potentials of type I SLFs measured with conventional zero-current whole-cell configuration were highly K(+)-selective and sensitive to IbTx (n=4-9). In addition, the BK channels in these cells exhibited voltage-dependent and incomplete inactivation properties and the recovery time was estimated to be approximately 6 s with repetitive voltage pulses from -70 to 80 mV (n=3). These data suggest that BK channels in type I SLFs play a major role in regulating the intracellular electrochemical gradient in the lateral wall syncytium responsible for facilitating the K(+) movement from perilymph to the stria vascularis.
Collapse
Affiliation(s)
- Zhijun Shen
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 165 Ashley Avenue, P.O. Box 250908, Charleston, SC 29425, USA.
| | | | | | | |
Collapse
|
31
|
Affiliation(s)
- James F Battey
- National Institute on Deafness and Other Communication Disorders, Bethesda, MD USA.
| |
Collapse
|
32
|
Pandya A, Arnos KS, Xia XJ, Welch KO, Blanton SH, Friedman TB, Garcia Sanchez G, Liu MD XZ, Morell R, Nance WE. Frequency and distribution of GJB2 (connexin 26) and GJB6 (connexin 30) mutations in a large North American repository of deaf probands. Genet Med 2003; 5:295-303. [PMID: 12865758 DOI: 10.1097/01.gim.0000078026.01140.68] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
PURPOSE Profound hearing loss occurs with a frequency of 1 in 1000 live births, half of which is genetic in etiology. The past decade has witnessed rapid advances in determining the pathogenesis of both syndromic and nonsyndromic deafness. The most significant clinical finding to date has been the discovery that mutations of GJB2 at the DFNB1 locus are the major cause of profound prelingual deafness in many countries. 1 More recently, GJB2 mutations have been shown to cause deafness when present with a deletion of the GJB6 gene. We report on the prevalence of GJB2 and GJB6 mutations in a large North American Repository of DNA from deaf probands and document the profound effects of familial ethnicity and parental mating types on the frequency of these mutations in the population. METHODS Deaf probands were ascertained through the Annual Survey of Deaf and Hard of Hearing Children and Youth, conducted at the Research Institute of Gallaudet University. Educational, etiologic, and audiologic information was collected after obtaining informed consent. DNA studies were performed for the GJB2 and GJB6 loci by sequencing and PCR methods. RESULTS GJB2 mutations accounted for 22.2% of deafness in the overall sample but differed significantly among Asians, African-Americans and Hispanics and for probands from deaf by deaf and deaf by hearing matings, as well as probands from simplex and multiplex sibships of hearing parents. In our sample, the overall incidence of GJB2/GJB6 deafness was 2.57%. CONCLUSION GJB2 mutations account for a large proportion of deafness in the US, with certain mutations having a high ethnic predilection. Heterozygotes at the GJB2 locus should be screened for the GJB6 deletion as a cause of deafness. Molecular testing for GJB2 and GJB6 should be offered to all patients with nonsyndromic hearing loss.
Collapse
Affiliation(s)
- Arti Pandya
- Department of Human Genetics, Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Merves M, Krane CM, Dou H, Greinwald JH, Menon AG, Choo D. Expression of aquaporin 1 and 5 in the developing mouse inner ear and audiovestibular assessment of an Aqp5 null mutant. J Assoc Res Otolaryngol 2003; 4:264-75. [PMID: 12943377 PMCID: PMC3202717 DOI: 10.1007/s10162-002-3033-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2002] [Accepted: 11/07/2002] [Indexed: 10/26/2022] Open
Abstract
To examine the potential roles of aquaporins 1 and 5 (AQP1 and AQP5, respectively) in inner ear development and function, we defined their spatial and temporal expression patterns in the developing mouse inner ear and examined the morphologic and physiologic effects of loss of Aqp5 function. Standard in situ hybridization (ISH) and immunohistochemical (IHC) assays were used for expression studies with routine morphologic, behavioral, and physiologic assessments of hearing and balance in Aqp5 null mutant mice. AQP1 was first detected at embryonic day 10.5 (E10.5) in the otocyst but eventually localized to specific nonsensory portions of the inner ear and connective tissue cells surrounding the membranous labyrinth. AQP5 displayed specific cochlear expression, first detectable at E15.5 in the nonsensory epithelium and later restricted to the lateral wall of the cochlear duct near the spiral prominence. AQP5 expression continued through postnatal periods with a change of expression domain to the stria vascularis between postnatal day 7 (P7) and P14. By in situ hybridization and immunohistochemical techniques, subtle differences between transcript and protein expression patterns were noted for both AQP1 and 5. Although AQP5 is dynamically expressed in the developing mouse inner ear, adult Aqp5 knockout mice show normal hearing when tested and normal inner ear structural development. These results suggest redundant or alternative mechanisms that likely regulate water homeostasis in the developing and mature inner ear.
Collapse
MESH Headings
- Aging/metabolism
- Animals
- Animals, Newborn/growth & development
- Animals, Newborn/metabolism
- Aquaporin 1
- Aquaporin 5
- Aquaporins/deficiency
- Aquaporins/metabolism
- Behavior, Animal
- Ear, Inner/embryology
- Ear, Inner/metabolism
- Ear, Inner/pathology
- Ear, Inner/physiopathology
- Embryo, Mammalian/metabolism
- Embryonic and Fetal Development
- Evoked Potentials, Auditory, Brain Stem
- Hearing
- Membrane Proteins
- Mice
- Mice, Inbred Strains
- Mice, Knockout
Collapse
Affiliation(s)
- Michele Merves
- Department of Pediatric Otolaryngology, Center for Hearing and Deafness Research, Cincinnati, OH 45229, USA
| | - Carissa M. Krane
- Department of Biology, University of Dayton, Dayton, OH 45404, USA
| | - Hongwei Dou
- Department of Pediatric Otolaryngology, Center for Hearing and Deafness Research, Cincinnati, OH 45229, USA
| | - John H. Greinwald
- Department of Pediatric Otolaryngology, Center for Hearing and Deafness Research, Cincinnati, OH 45229, USA
| | - Anil G. Menon
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Daniel Choo
- Department of Pediatric Otolaryngology, Center for Hearing and Deafness Research, Cincinnati, OH 45229, USA
| |
Collapse
|
34
|
Liang F, Niedzielski A, Schulte BA, Spicer SS, Hazen-Martin DJ, Shen Z. A voltage- and Ca2+-dependent big conductance K channel in cochlear spiral ligament fibrocytes. Pflugers Arch 2003; 445:683-92. [PMID: 12632188 DOI: 10.1007/s00424-002-0976-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2002] [Revised: 09/05/2002] [Accepted: 10/10/2002] [Indexed: 10/22/2022]
Abstract
Evidence is accruing that spiral ligament fibrocytes (SLFs) play an important role in cochlear K(+) homeostasis, but little direct physiological data is available to support this concept. Here we report the presence and characterization of a voltage- and Ca(2+)-dependent big-conductance K (BK) channel in type I SLFs cultured from the gerbil cochlea. A single-channel conductance of 298+/-5.6 pS (n=28) was measured under symmetrical K(+). Membrane potentials for half-maximal open probability (P(o)) were -67, -45 and 85 mV with cytosolic free-Ca(2+) levels of 0.7 mM, 10 microM and 1 microM, respectively (n=8-14). The Hill coefficient for Ca(2+) affinity was 1.9 at a membrane potential of 60 mV (n=6). The BK channel showed very low activity (P(o)=0.0019, n=5) under normal physiological conditions, suggesting a low resting intracellular free [Ca(2+)]. Pharmacological results fit well with the profile of classic BK channels. The estimated half-maximal inhibitory concentration and Hill coefficient for tetraethylammonium were 0.086+/-0.021 mM and 0.99, respectively (n=4-9). In whole cell recordings, the voltage-activated outward K current was inhibited 85.7+/-4.5% (n=6) by 0.1 microM iberiotoxin. A steady-state kinetic model with two open and two closed stages best described the BK gating process (tau(o1) 0.23+/-0.08 ms, tau(o2) 1.40+/-0.32 ms; tau(c1) 0.26+/-0.09 ms, tau(c2) 3.10+/-1.2 ms; n=11). RT-PCR analyses revealed a splice variant of the BK channel alpha subunit in cultured type I SLFs and freshly isolated spiral ligament tissues. The BK channel is likely to play a major role in regulating the membrane potential of type I SLFs, which may in turn influence K(+) recycling dynamics in the mammalian cochlea.
Collapse
Affiliation(s)
- F Liang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 171 Ashley Avenue, PO Box 250908, Charleston, SC 29425, USA
| | | | | | | | | | | |
Collapse
|
35
|
Affiliation(s)
- Elizabeth Quint
- MRC Institute of Hearing Research, University Park, Nottingham NG7 2RD, United Kingdom
| | | |
Collapse
|
36
|
Mhatre AN, Stern RE, Li J, Lalwani AK. Aquaporin 4 expression in the mammalian inner ear and its role in hearing. Biochem Biophys Res Commun 2002; 297:987-96. [PMID: 12359252 DOI: 10.1016/s0006-291x(02)02296-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Aquaporin 4 (Aqp4), a member of a family of water transport proteins, is a candidate for playing a critical role in inner ear fluid homeostasis. In this study, we assess cross-species Aqp4 expression in the inner ear, sequence conservation in the 5'-UTR, and hearing in Aqp4 knockout mice. A single Aqp4 isoform was expressed in a highly conserved pattern within the supporting epithelia surrounding the sensory cells of the auditory and vestibular sensory organs and the glial cells surrounding the auditory nerve of the mouse and rat. In the 5'-UTR of mouse and rat Aqp4 gene, sequence conservation was highest in the region spanning the transcription start site. Aqp4 knockout mice demonstrated impaired hearing, but normal neural conduction time. Similar Aqp4 expression pattern and regulatory sequence conservation across species suggest a highly conserved role for Aqp4 in the inner ear. In the Aqp4 deficient mouse, cochlear dysfunction is suggested as the primary cause of hearing impairment in the absence of neural conduction abnormality.
Collapse
Affiliation(s)
- Anand N Mhatre
- Laboratory of Molecular Otology, Epstein Laboratories, Department of Otolaryngology-Head & Neck Surgery, University of California-San Francisco, 533 Parnassus Avenue U490A, San Francisco, CA 94143-0526, USA.
| | | | | | | |
Collapse
|
37
|
Pampanos A, Economides J, Iliadou V, Neou P, Leotsakos P, Voyiatzis N, Eleftheriades N, Tsakanikos M, Antoniadi T, Hatzaki A, Konstantopoulou I, Yannoukakos D, Gronskov K, Brondum-Nielsen K, Grigoriadou M, Gyftodimou J, Iliades T, Skevas A, Petersen MB. Prevalence of GJB2 mutations in prelingual deafness in the Greek population. Int J Pediatr Otorhinolaryngol 2002; 65:101-8. [PMID: 12176179 DOI: 10.1016/s0165-5876(02)00177-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE Mutations in the gene encoding the gap junction protein connexin 26 (GJB2) have been shown as a major contributor to prelingual, sensorineural, nonsyndromic, recessive deafness. One specific mutation, 35delG, has accounted for the majority of the mutations detected in the GJB2 gene in Caucasian populations. The aim of our study was to determine the prevalence and spectrum of GJB2 mutations in prelingual deafness in the Greek population. METHODS In a collaboration with the major referral centers for childhood deafness in Greece, patients were examined by an extensive questionnaire to exclude syndromic forms and environmental causes of deafness and by allele-specific polymerase chain reaction (PCR) for the detection of the 35delG mutation. Patients heterozygous for the 35delG mutation were further analyzed by direct genomic sequencing of the coding region of the GJB2 gene. RESULTS The 35delG mutation was found in 42.2% of the chromosomes in 45 familial cases of prelingual, nonsyndromic deafness (18 homozygotes and 2 heterozygotes) and in 30.6% of the chromosomes in 165 sporadic cases (45 homozygotes and 11 heterozygotes). Direct genomic sequencing in heterozygous patients revealed the L90P (2 alleles), W24X (2 alleles), R184P (2 alleles), and 291insA (1 allele) mutations. CONCLUSION Mutations in the GJB2 gene are responsible for about one third of prelingual, sensorineural, nonsyndromic deafness in the Greek population, and allele-specific PCR is an easy screening method for the common 35delG mutation.
Collapse
Affiliation(s)
- Andreas Pampanos
- Department of Genetics, Institute of Child Health, Aghia Sophia Children's Hospital, GR-11527 Athens, Greece
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Non-syndromic deafness is a paradigm of genetic heterogeneity. More than 70 loci have been mapped, and 25 of the nuclear genes responsible for non-syndromic deafness have been identified. Autosomal-dominant genes are responsible for about 20% of the cases of hereditary non-syndromic deafness, with 16 different genes identified to date. In the present article we review these 16 genes, their function and their contribution to deafness in different populations. The complexity is underlined by the fact that several of the genes are involved in both dominant and recessive non-syndromic deafness or in both non-syndromic and syndromic deafness. Mutations in eight of the genes have so far been detected in only single dominant deafness families, and their contribution to deafness on a population base might therefore be limited, or is currently unknown. Identification of all genes involved in hereditary hearing loss will help in the understanding of the basic mechanisms underlying normal hearing, will facilitate early diagnosis and intervention and might offer opportunities for rational therapy.
Collapse
Affiliation(s)
- M B Petersen
- Department of Genetics, Institute of Child Health, Aghia Sophia Children's Hospital, GR-11527 Athens, Greece.
| |
Collapse
|
39
|
Xia AP, Kikuchi T, Minowa O, Katori Y, Oshima T, Noda T, Ikeda K. Late-onset hearing loss in a mouse model of DFN3 non-syndromic deafness: morphologic and immunohistochemical analyses. Hear Res 2002; 166:150-8. [PMID: 12062767 DOI: 10.1016/s0378-5955(02)00309-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Recently, we reported that homozygous males and females of a mouse model of DFN3 non-syndromic deafness generated by the deletion of Brn-4 transcription factor showed profound deafness due to severe alterations in the cochlear spiral ligament fibrocytes from the age of 11 weeks, whereas no hearing loss was recognized in young female heterozygotes. It is known that a part of obligate female carriers of DFN3 showed progressive hearing loss. In the present study, we examined the late-onset effect of Brn-4 deficiency on the hearing organ of the mouse. About one third of heterozygous female mice revealed late-onset profound deafness at the age of 1 year. Furthermore, in these deafened heterozygotes, characteristic abnormalities in Reissner's membrane attachment and type II fibrocytes in the suprastrial zone became evident under light microscope, similar to homozygous female mice. A significant reduction in the immunoreactivity of connexin 26 (Cx26), connexin 31 (Cx31), Na,K-ATPase and Na-K-Cl cotransporter in the spiral ligament fibrocytes was observed in aged heterozygotes showing late-onset profound deafness. The late-onset phenotype observed in heterozygous mutant mice, being consistent with the progressive deafness observed in human female heterozygotes, may be explained by alterations of the ion transport systems in the spiral ligament fibrocytes.
Collapse
Affiliation(s)
- An-Ping Xia
- Department of Otorhinolaryngology - Head and Neck Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | | | | | | | | | | | | |
Collapse
|
40
|
Jacobsen SD, Gronskov K, Brondum-Nielsen K, Parving A. Is there a relationship between U-shaped audiograms and mutations in connexin 26? SCANDINAVIAN AUDIOLOGY 2002; 30:184-8. [PMID: 11683456 DOI: 10.1080/010503901316914557] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The knowledge about gene mutations causing permanent hearing impairment (HI) is rapidly increasing, offering clinicians the possibility of analysing different gene mutations in relation to various phenotypes. This study examines a possible relationship between U-shaped audiograms and mutations in the GJB2-gene, coding for Connexin 26 (Cx 26). Thirty-eight subjects at a median age of 42 years, range 18-60 years with symmetric U-shaped audiograms classified as sensorineural were included in the genetic investigation. The gender distribution was 13 males and 25 females. No subjects had any indication of syndromic HI, and any possible exogenous factor that might cause HI was excluded. Three subjects had self-reported prelingual HI and 34 subjects had self-reported postlingual HI. Thirty-five subjects had one or more family members with HI. In 19 subjects the entire Cx 26 gene was examined, whereas 19 subjects were investigated for the 35delG mutation only. One female with mild HI and postlingual onset of the HI was heterogeneous for the L9OP-mutation in the Cx 26 gene. In all other subjects no mutations in the Cx 26 gene could be identified. Mutations of the Cx 26 gene are very rare among subjects exhibiting a U-shaped phenotype of the audiogram. However the majority of the investigated subjects (35/38) had a family history of HI and it seems therefore reasonable to ascribe U-shaped hearing deficit to genetic factors which has to be searched for in alternative gene mutations.
Collapse
Affiliation(s)
- S D Jacobsen
- Department of Audiology, Bispebjerg Hospital, Copenhagen, Denmark.
| | | | | | | |
Collapse
|
41
|
Szymko-Bennett YM, Russell LJ, Bale SJ, Griffith AJ. Auditory manifestations of Keratitis-Ichthyosis-Deafness (KID) syndrome. Laryngoscope 2002; 112:272-80. [PMID: 11889383 DOI: 10.1097/00005537-200202000-00014] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Evaluation of the auditory manifestations of Keratitis-Ichthyosis-Deafness (KID) syndrome, a rare genodermatosis characterized by follicular hyperkeratosis, vascularizing keratitis, and congenital hearing loss. STUDY DESIGN Five individuals with sporadic KID syndrome were evaluated in the outpatient audiology clinic at the Warren Grant Magnuson Clinical Center of the National Institutes of Health. METHODS Audiologic examinations included pure-tone audiometry, speech audiometry, and middle ear immittance testing. Auditory brainstem responses and otoacoustic emissions were analyzed in 2 subjects. RESULTS Four subjects had prelingual, bilateral, profound sensorineural hearing loss, whereas the fifth subject had significant residual hearing that exhibited no progression on serial audiograms. All 5 subjects had a history of non-erosive keratosis obturans and cutaneous cysts in the external ear canals that prevented continuous use of ear molds. CONCLUSIONS The sensorineural hearing loss in KID syndrome is generally prelingual and profound. This combination of auditory and cutaneous phenotypes is similar to those previously reported for KID syndrome. KID syndrome presents a difficult challenge for communication rehabilitation because keratitis may impair the perception of sign and spoken language, and the cutaneous manifestations routinely curtail use of external amplification devices.
Collapse
Affiliation(s)
- Yvonne M Szymko-Bennett
- National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | | | |
Collapse
|
42
|
Resendes BL, Williamson RE, Morton CC. At the speed of sound: gene discovery in the auditory system. Am J Hum Genet 2001; 69:923-35. [PMID: 11577373 PMCID: PMC1274369 DOI: 10.1086/324122] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2001] [Accepted: 08/29/2001] [Indexed: 11/03/2022] Open
Abstract
As auditory genes and deafness-associated mutations are discovered at a rapid rate, exciting opportunities have arisen to uncover the molecular mechanisms underlying hearing and hearing impairment. Single genes have been identified to be pathogenic for dominant or recessive forms of nonsyndromic hearing loss, syndromic hearing loss, and, in some cases, even multiple forms of hearing loss. Modifier loci and genes have been found, and investigations into their role in the hearing process will yield valuable insight into the fundamental processes of the auditory system.
Collapse
Affiliation(s)
- Barbara L. Resendes
- Departments of Obstetrics, Gynecology, and Reproductive Biology and Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston
| | - Robin E. Williamson
- Departments of Obstetrics, Gynecology, and Reproductive Biology and Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston
| | - Cynthia C. Morton
- Departments of Obstetrics, Gynecology, and Reproductive Biology and Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston
| |
Collapse
|
43
|
Smith SD. Relationships between neurologic disorders and hereditary hearing loss. Semin Pediatr Neurol 2001; 8:147-59. [PMID: 11575844 DOI: 10.1053/spen.2001.26448] [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: 11/11/2022]
Abstract
Hearing loss is a common disorder that often has a neurologic etiology. Recently, there has been significant progress in the discovery of the genes that cause sensorineural hearing loss, and this has led to increased understanding of the pathophysiology of both syndromic and nonsyndromic hearing problems. These genes cover the range of processes involved in neurologic development and function, including structural genes, transcription factors, and tumor suppressors; genes involved in signal transduction processes, such as ion homeostasis and intracellular transport; and mitochondrial genes responsible for oxidative phosphorylation and energy production. Interactions between genes as well as between genes and environmental factors have also been documented. Understanding of these processes should lead to earlier and more accurate diagnosis and more effective treatment for neurologic disorders and hearing loss.
Collapse
Affiliation(s)
- S D Smith
- Department of Pediatrics, Center for Human Molecular Genetics, Munroe Meyer Institute, University of Nebraska Medical Center, Omaha 68198-5455, USA
| |
Collapse
|
44
|
Abstract
A role for aquaporins (AQPs) in hearing has been suggested from the specific expression of aquaporins in inner ear and the need for precise volume regulation in epithelial cells involved in acoustic signal transduction. Using mice deficient in selected aquaporins as controls, we localized AQP1 in fibrocytes in the spiral ligament and AQP4 in supporting epithelial cells (Hensen's, Claudius, and inner sulcus cells) in the organ of Corti. To determine whether aquaporins play a role in hearing, auditory brain stem response (ABR) thresholds were compared in wild-type mice and transgenic null mice lacking (individually) AQP1, AQP3, AQP4, and AQP5. In 4-5-week-old mice in a CD1 genetic background, ABR thresholds in response to a click stimulus were remarkably increased by >12 db in AQP4 null mice compared with wild-type mice (p < 0.001), whereas ABR thresholds were not affected by AQP1, AQP3, or AQP5 deletion. In a C57/bl6 background, nearly all AQP4 null mice were deaf, whereas ABRs could be elicited in wild-type controls. ABRs in AQP4 null CD1 mice measured in response to tone bursts (4-20 kHz) indicated a frequency-independent hearing deficit. Light microscopy showed no differences in cochlear morphology of wild-type versus AQP4 null mice. These results provide the first direct evidence that an aquaporin water channel plays a role in hearing. AQP4 may facilitate rapid osmotic equilibration in epithelial cells in the organ of Corti, which are subject to large K(+) fluxes during mechano-electric signal transduction.
Collapse
Affiliation(s)
- J Li
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California 94143-0521, USA
| | | |
Collapse
|
45
|
Henzl MT, O'Neal J, Killick R, Thalmann I, Thalmann R. OCP1, an F-box protein, co-localizes with OCP2/SKP1 in the cochlear epithelial gap junction region. Hear Res 2001; 157:100-11. [PMID: 11470190 DOI: 10.1016/s0378-5955(01)00285-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Immunohistochemical data indicate that OCP1 co-localizes exactly with OCP2 in the epithelial gap junction region of the guinea pig organ of Corti (OC). Despite the abundance of OCP1 in the OC, gaining access to its coding sequence -- and, in particular, the 5' end of the coding sequence -- proved unexpectedly challenging. The putative full-length OCP1 cDNA -- 1180 nucleotides in length -- includes a 67 nucleotide 5' leader sequence, 300 codons (including initiation and termination signals), and a 216 nucleotide 3' untranslated region. The cDNA encodes a protein having a predicted molecular weight of 33,700. The inferred amino acid sequence harbors an F-box motif spanning residues 52--91, consistent with a role for OCP1 and OCP2 in the proteasome-mediated degradation of select OC proteins. Although OCP1 displays extensive homology to an F-box protein recently cloned from rat brain (NFB42), clustered sequence non-identities indicate that the two proteins are transcribed from distinct genes. The presumptive human OCP1 gene was identified in the human genome databank. Located on chromosome 1p35, the inferred translation product exhibits 94% identity with the guinea pig OCP1 coding sequence.
Collapse
Affiliation(s)
- M T Henzl
- Department of Biochemistry, University of Missouri-Columbia, 35211, USA.
| | | | | | | | | |
Collapse
|
46
|
Xia A, Katori Y, Oshima T, Watanabe K, Kikuchi T, Ikeda K. Expression of connexin 30 in the developing mouse cochlea. Brain Res 2001; 898:364-7. [PMID: 11306024 DOI: 10.1016/s0006-8993(01)02216-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Mutations in the GJB6 gene encoding connexin 30 (Cx30) can cause dominant forms of nonsyndromic deafness. By studying immunohistochemical localization of Cx30 in the mouse cochlea at different ages from 0 to 30 days after birth, we found that the expression of Cx30 is nearly the same as that of Cx26. These findings suggest that as well as Cx26, Cx30 may also contribute to the generation and maturation of endocochlear potential.
Collapse
Affiliation(s)
- A Xia
- Department of Otorhinolaryngology-Head and Neck Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, 980-8574, Sendai, Japan.
| | | | | | | | | | | |
Collapse
|
47
|
Casimiro MC, Knollmann BC, Ebert SN, Vary JC, Greene AE, Franz MR, Grinberg A, Huang SP, Pfeifer K. Targeted disruption of the Kcnq1 gene produces a mouse model of Jervell and Lange-Nielsen Syndrome. Proc Natl Acad Sci U S A 2001; 98:2526-31. [PMID: 11226272 PMCID: PMC30171 DOI: 10.1073/pnas.041398998] [Citation(s) in RCA: 176] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2000] [Indexed: 11/18/2022] Open
Abstract
KCNQ1 encodes KCNQ1, which belongs to a family of voltage-dependent K(+) ion channel proteins. KCNQ1 associates with a regulatory subunit, KCNE1, to produce the cardiac repolarizing current, I(Ks). Loss-of-function mutations in the human KCNQ1 gene have been linked to Jervell and Lange-Nielsen Syndrome (JLNS), a disorder characterized by profound bilateral deafness and a cardiac phenotype. To generate a mouse model for JLNS, we created a line of transgenic mice that have a targeted disruption in the Kcnq1 gene. Behavioral analysis revealed that the Kcnq1(-/-) mice are deaf and exhibit a shaker/waltzer phenotype. Histological analysis of the inner ear structures of Kcnq1(-/-) mice revealed gross morphological anomalies because of the drastic reduction in the volume of endolymph. ECGs recorded from Kcnq1(-/-) mice demonstrated abnormal T- and P-wave morphologies and prolongation of the QT and JT intervals when measured in vivo, but not in isolated hearts. These changes are indicative of cardiac repolarization defects that appear to be induced by extracardiac signals. Together, these data suggest that Kcnq1(-/-) mice are a potentially valuable animal model of JLNS.
Collapse
Affiliation(s)
- M C Casimiro
- Laboratory for Mammalian Genes and Development, National Institute of Child Health and Human Development/National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Metzler DE, Metzler CM, Sauke DJ. Chemical Communication Between Cells. Biochemistry 2001. [DOI: 10.1016/b978-012492543-4/50033-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
49
|
Armour G, Mhaskar Y, Rybak L, Dunaway G. Alteration of 6-phosphofructo-1-kinase subunits during neonatal maturation of the rat cochlear cells. Hear Res 2001; 151:149-156. [PMID: 11124462 DOI: 10.1016/s0378-5955(00)00222-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
During postnatal development of rat cochlear cells and the onset of hearing (10-23 days), the increasing endocochlear potential and energy requirements are largely provided by increased glucose utilization. It is well established that the ability of maturing rat tissues to use glucose is directly related to alteration of 6-phosphofructo-1-kinase (PFK) subunits. To gain insight into the alteration of PFK subunit levels in the cochlea from 6 to 60 days of age, PFK subunit types were measured in sections of paraffin-embedded temporal bone using IgG specific for each type of PFK subunit and quantified by computer image analysis. Although the L-type and C-type subunits did not exhibit statistically significant changes in the cochlear structures during maturation, the levels of M-type subunit in the stria vascularis cells, spiral ligament cell types I, II, and III, outer hair cells, inner hair cells, and support cells significantly increased. Also, the type IV and V spiral ligament fibrocytes during this period did not exhibit significant alterations of the M-type subunit. These data suggest that during neonatal development of the cochlear, the elevated levels of the M-type subunit are associated with increased glucose utilization and the onset of hearing.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Cochlea/cytology
- Cochlea/enzymology
- Cochlea/growth & development
- Energy Metabolism
- Glucose/metabolism
- Hair Cells, Auditory, Inner/cytology
- Hair Cells, Auditory, Inner/growth & development
- Hair Cells, Auditory, Inner/metabolism
- Hair Cells, Auditory, Outer/cytology
- Hair Cells, Auditory, Outer/growth & development
- Hair Cells, Auditory, Outer/metabolism
- Hearing/physiology
- Immunohistochemistry
- Phosphofructokinase-1/chemistry
- Phosphofructokinase-1/metabolism
- Protein Subunits
- Rats
- Rats, Inbred F344
- Stria Vascularis/cytology
- Stria Vascularis/metabolism
- Tissue Distribution
Collapse
Affiliation(s)
- G Armour
- Department of Pharmacology, Division of Otolaryngology, Southern Illinois University School of Medicine, Springfield 62794-9629, USA
| | | | | | | |
Collapse
|
50
|
Affiliation(s)
- J F Battey
- National Institute on Deafness and Other Communication Disorders, NIH Building 31, Room 3C02, 31 Convent Drive MSC 2320, 9000 Rockville Pike, Bethesda, Maryland 20892, USA.
| |
Collapse
|