201
|
Morozko EL, Nishio A, Ingham NJ, Chandra R, Fitzgerald T, Martelletti E, Borck G, Wilson E, Riordan GP, Wangemann P, Forge A, Steel KP, Liddle RA, Friedman TB, Belyantseva IA. ILDR1 null mice, a model of human deafness DFNB42, show structural aberrations of tricellular tight junctions and degeneration of auditory hair cells. Hum Mol Genet 2014; 24:609-24. [PMID: 25217574 PMCID: PMC4291242 DOI: 10.1093/hmg/ddu474] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In the mammalian inner ear, bicellular and tricellular tight junctions (tTJs) seal the paracellular space between epithelial cells. Tricellulin and immunoglobulin-like (Ig-like) domain containing receptor 1 (ILDR1, also referred to as angulin-2) localize to tTJs of the sensory and non-sensory epithelia in the organ of Corti and vestibular end organs. Recessive mutations of TRIC (DFNB49) encoding tricellulin and ILDR1 (DFNB42) cause human nonsyndromic deafness. However, the pathophysiology of DFNB42 deafness remains unknown. ILDR1 was recently reported to be a lipoprotein receptor mediating the secretion of the fat-stimulated cholecystokinin (CCK) hormone in the small intestine, while ILDR1 in EpH4 mouse mammary epithelial cells in vitro was shown to recruit tricellulin to tTJs. Here we show that two different mouse Ildr1 mutant alleles have early-onset severe deafness associated with a rapid degeneration of cochlear hair cells (HCs) but have a normal endocochlear potential. ILDR1 is not required for recruitment of tricellulin to tTJs in the cochlea in vivo; however, tricellulin becomes mislocalized in the inner ear sensory epithelia of ILDR1 null mice after the first postnatal week. As revealed by freeze-fracture electron microscopy, ILDR1 contributes to the ultrastructure of inner ear tTJs. Taken together, our data provide insight into the pathophysiology of human DFNB42 deafness and demonstrate that ILDR1 is crucial for normal hearing by maintaining the structural and functional integrity of tTJs, which are critical for the survival of auditory neurosensory HCs.
Collapse
Affiliation(s)
- Eva L Morozko
- National Institute on Deafness and Other Communication Disorders, Section on Human Genetics
| | - Ayako Nishio
- National Institute on Deafness and Other Communication Disorders, Molecular Biology and Genetics Section
| | - Neil J Ingham
- Wolfson Centre for Age-Related Diseases, King's College London, London SE1 1UL, UK
| | - Rashmi Chandra
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Tracy Fitzgerald
- National Institute on Deafness and Other Communication Disorders, Mouse Auditory Testing Core Facility, National Institutes of Health, Bethesda, MD 20892-3729, USA
| | - Elisa Martelletti
- Wolfson Centre for Age-Related Diseases, King's College London, London SE1 1UL, UK
| | - Guntram Borck
- Institute of Human Genetics, University of Ulm, Ulm 89081, Germany
| | - Elizabeth Wilson
- National Institute on Deafness and Other Communication Disorders, Section on Human Genetics
| | - Gavin P Riordan
- National Institute on Deafness and Other Communication Disorders, Section on Human Genetics
| | - Philine Wangemann
- Anatomy and Physiology Department, Kansas State University, Manhattan, KS 66506-5802, USA and
| | - Andrew Forge
- Centre for Auditory Research, University College London, London WC1X 8EE, UK
| | - Karen P Steel
- Wolfson Centre for Age-Related Diseases, King's College London, London SE1 1UL, UK
| | - Rodger A Liddle
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Thomas B Friedman
- National Institute on Deafness and Other Communication Disorders, Section on Human Genetics
| | - Inna A Belyantseva
- National Institute on Deafness and Other Communication Disorders, Section on Human Genetics
| |
Collapse
|
202
|
Yoshida T, Nin F, Ogata G, Uetsuka S, Kitahara T, Inohara H, Akazawa K, Komune S, Kurachi Y, Hibino H. NKCCs in the fibrocytes of the spiral ligament are silent on the unidirectional K⁺ transport that controls the electrochemical properties in the mammalian cochlea. Pflugers Arch 2014; 467:1577-1589. [PMID: 25143138 DOI: 10.1007/s00424-014-1597-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 07/08/2014] [Accepted: 08/07/2014] [Indexed: 11/26/2022]
Abstract
Unidirectional K(+) transport across the lateral cochlear wall contributes to the endocochlear potential (EP) of +80 mV in the endolymph, a property essential for hearing. The wall comprises two epithelial layers, the syncytium and the marginal cells. The basolateral surface of the former and the apical membranes of the latter face the perilymph and the endolymph, respectively. Intrastrial space (IS), an extracellular compartment between the two layers, exhibits low [K(+)] and a potential similar to the EP. This IS potential (ISP) dominates the EP and represents a K(+) diffusion potential elicited by a large K(+) gradient across the syncytial apical surface. The K(+) gradient depends on the unidirectional K(+) transport driven by Na(+),K(+)-ATPases on the basolateral surface of each layer and the concomitant Na(+),K(+),2Cl(-)-cotransporters (NKCCs) in the marginal cell layer. The NKCCs coexpressed with the Na(+),K(+)-ATPases in the syncytial layer also seem to participate in the K(+) transport. To test this hypothesis, we examined the electrochemical properties of the lateral wall with electrodes measuring [K(+)] and potential. Blocking NKCCs by perilymphatic perfusion of bumetanide suppressed the ISP. Unexpectedly and unlike the inhibition of the syncytial Na(+),K(+)-ATPases, the perfusion barely altered the electrochemical properties of the syncytium but markedly augmented [K(+)] of the IS. Consequently, the K(+) gradient decreased and the ISP declined. These observations resembled those when the marginal cells' Na(+),K(+)-ATPases or NKCCs were blocked with vascularly applied inhibitors. It is plausible that NKCCs in the marginal cells are affected by the perilymphatically perfused bumetanide, and these transporters, but not those in the syncytium, mediate the unidirectional K(+) transport.
Collapse
Affiliation(s)
- Takamasa Yoshida
- Department of Molecular Physiology, School of Medicine, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, Niigata, 951-8510, Japan
- Center for Transdisciplinary Research, Niigata University, Niigata, Japan
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Fumiaki Nin
- Department of Molecular Physiology, School of Medicine, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, Niigata, 951-8510, Japan
- Center for Transdisciplinary Research, Niigata University, Niigata, Japan
| | - Genki Ogata
- Department of Molecular Physiology, School of Medicine, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, Niigata, 951-8510, Japan
- Center for Transdisciplinary Research, Niigata University, Niigata, Japan
| | - Satoru Uetsuka
- Department of Molecular Physiology, School of Medicine, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, Niigata, 951-8510, Japan
- Center for Transdisciplinary Research, Niigata University, Niigata, Japan
- Department of Otorhinolaryngology-Head and Neck Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Tadashi Kitahara
- Department of Otorhinolaryngology-Head and Neck Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
- Department of Otorhinolaryngology-Head and Neck Surgery, Nara Medical University, Kashihara, Japan
| | - Hidenori Inohara
- Department of Otorhinolaryngology-Head and Neck Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Kohei Akazawa
- Department of Medical Informatics, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Shizuo Komune
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihisa Kurachi
- Division of Molecular and Cellular Pharmacology, Department of Pharmacology, Graduate School of Medicine, and The Center for Advanced Medical Engineering and Informatics, Osaka University, Suita, Japan
| | - Hiroshi Hibino
- Department of Molecular Physiology, School of Medicine, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, Niigata, 951-8510, Japan.
- Center for Transdisciplinary Research, Niigata University, Niigata, Japan.
| |
Collapse
|
203
|
Oron Y, Elgart K, Marom T, Roth Y. Cardiovascular risk factors as causes for hearing impairment. Audiol Neurootol 2014; 19:256-60. [PMID: 25073427 DOI: 10.1159/000363215] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 04/25/2014] [Indexed: 11/19/2022] Open
Abstract
The purpose of this paper is to provide a contemporary review of the correlation between cardiovascular risk factors (CVRFs) and hearing impairment (HI) . We conducted a comprehensive review of the literature in order to assess the effects of the different CVRFs on HI. We focused on the pathological findings in the inner ear and their correlation with cochlear function in population-based studies. We found that CVRFs adversely affect hearing acuity. HI diagnosis should be accompanied by detecting and treating CVRFs, according to the presented outline, which may augment hearing rehabilitation and improve the general health and the well-being of the patient. © 2014 S. Karger AG, Basel.
Collapse
Affiliation(s)
- Yahav Oron
- Department of Otolaryngology, Head and Neck Surgery, Edith Wolfson Medical Center, Sackler School of Medicine, Tel Aviv University, Holon, Israel
| | | | | | | |
Collapse
|
204
|
Oonk AM, Ekker MS, Huygen PL, Kunst HP, Kremer H, Schelhaas JJ, Pennings RJ. Intrafamilial Variable Hearing Loss in TRPV4 Induced Spinal Muscular Atrophy. Ann Otol Rhinol Laryngol 2014; 123:859-65. [DOI: 10.1177/0003489414539130] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective: Mutations in the transient receptor potential vanilloid 4 gene ( TRPV4) can induce a great diversity of neuropathies. Together with these neuropathies, hearing loss can occur. This study is focused on providing an audiometric phenotype description of a Dutch family with spinal muscular atrophy caused by a mutation in TRPV4. Methods: A neurological examination was repeated and pure tone and speech audiometry were performed. Results: A large variety in neurological symptoms as well as variation in audiometric characteristics was observed. The severity of hearing loss is mild to moderate and the audiogram configuration is highly variable. The hearing loss of these patients has a progressive nature in general. The frequencies that deteriorate significantly differ between family members. When compared to presbyacusis patients, speech recognition scores of patients with a TRPV4 mutation are not clearly different. Conclusion: The function of TRPV4 in the inner ear is still elusive but it is suggested that TRPV4 is required for maintenance of cochlear function in stress conditions, like acoustic injury. We can neither confirm nor reject this based on the results obtained in this family. Therefore, one might consider advising patients with a TRPV4 mutation to avoid exposure to environmental influences such as noise exposure.
Collapse
Affiliation(s)
- Anne M.M. Oonk
- Department of Otorhinolaryngology, Hearing & Genes, Radboud University Medical Centre, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Merel S. Ekker
- Department of Otorhinolaryngology, Hearing & Genes, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Patrick L.M. Huygen
- Department of Otorhinolaryngology, Hearing & Genes, Radboud University Medical Centre, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Henricus P.M. Kunst
- Department of Otorhinolaryngology, Hearing & Genes, Radboud University Medical Centre, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Hannie Kremer
- Department of Otorhinolaryngology, Hearing & Genes, Radboud University Medical Centre, Nijmegen, the Netherlands
- Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Jurgen J. Schelhaas
- Expertise Centre for Epileptology, Sleep Medicine and Neurocognition, Kempenhaege, Heeze, the Netherlands
| | - Ronald J.E. Pennings
- Department of Otorhinolaryngology, Hearing & Genes, Radboud University Medical Centre, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Centre, Nijmegen, the Netherlands
| |
Collapse
|
205
|
Sanchez HA, Bienkowski R, Slavi N, Srinivas M, Verselis VK. Altered inhibition of Cx26 hemichannels by pH and Zn2+ in the A40V mutation associated with keratitis-ichthyosis-deafness syndrome. J Biol Chem 2014; 289:21519-32. [PMID: 24939841 DOI: 10.1074/jbc.m114.578757] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Excessive opening of undocked Cx26 hemichannels in the plasma membrane is associated with disease pathogenesis in keratitis-ichthyosis-deafness (KID) syndrome. Thus far, excessive opening of KID mutant hemichannels has been attributed, almost solely, to aberrant inhibition by extracellular Ca(2+). This study presents two new possible contributing factors, pH and Zn(2+). Plasma pH levels and micromolar concentrations of Zn(2+) inhibit WT Cx26 hemichannels. However, A40V KID mutant hemichannels show substantially reduced inhibition by these factors. Using excised patches, acidification was shown to be effective from either side of the membrane, suggesting a protonation site accessible to H(+) flux through the pore. Sensitivity to pH was not dependent on extracellular aminosulfonate pH buffers. Single channel recordings showed that acidification did not affect unitary conductance or block the hemichannel but rather promoted gating to the closed state with transitions characteristic of the intrinsic loop gating mechanism. Examination of two nearby KID mutants in the E1 domain, G45E and D50N, showed no changes in modulation by pH or Zn(2+). N-bromo-succinimide, but not thiol-specific reagents, attenuated both pH and Zn(2+) responses. Individually mutating each of the five His residues in WT Cx26 did not reveal a key His residue that conferred sensitivity to pH or Zn(2+). From these data and the crystal structure of Cx26 that suggests that Ala-40 contributes to an intrasubunit hydrophobic core, the principal effect of the A40V mutation is probably a perturbation in structure that affects loop gating, thereby affecting multiple factors that act to close Cx26 hemichannels via this gating mechanism.
Collapse
Affiliation(s)
- Helmuth A Sanchez
- From the Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Rick Bienkowski
- the Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia 30322, and
| | - Nefeli Slavi
- the Department of Biological Sciences, SUNY College of Optometry, New York, New York 10036
| | - Miduturu Srinivas
- the Department of Biological Sciences, SUNY College of Optometry, New York, New York 10036
| | - Vytas K Verselis
- From the Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461,
| |
Collapse
|
206
|
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: 1.7] [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
|
207
|
Berger AC, Kelly JJ, Lajoie P, Shao Q, Laird DW. Mutations in Cx30 that are linked to skin disease and non-syndromic hearing loss exhibit several distinct cellular pathologies. J Cell Sci 2014; 127:1751-64. [DOI: 10.1242/jcs.138230] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
ABSTRACT
Connexin 30 (Cx30), a member of the large gap-junction protein family, plays a role in the homeostasis of the epidermis and inner ear through gap junctional intercellular communication (GJIC). Here, we investigate the underlying mechanisms of four autosomal dominant Cx30 gene mutations that are linked to hearing loss and/or various skin diseases. First, the T5M mutant linked to non-syndromic hearing loss formed functional gap junction channels and hemichannels, similar to wild-type Cx30. The loss-of-function V37E mutant associated with Clouston syndrome or keratitis-ichthyosis-deafness syndrome was retained in the endoplasmic reticulum and significantly induced apoptosis. The G59R mutant linked to the Vohwinkel and Bart-Pumphrey syndromes was retained primarily in the Golgi apparatus and exhibited loss of gap junction channel and hemichannel function but did not cause cell death. Lastly, the A88V mutant, which is linked to the development of Clouston syndrome, also significantly induced apoptosis but through an endoplasmic-reticulum-independent mechanism. Collectively, we discovered that four unique Cx30 mutants might cause disease through different mechanisms that also likely include their selective trans-dominant effects on coexpressed connexins, highlighting the overall complexity of connexin-linked diseases and the importance of GJIC in disease prevention.
Collapse
Affiliation(s)
- Amy C. Berger
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON N6A 5C1, Canada
| | - John J. Kelly
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Patrick Lajoie
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Qing Shao
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Dale W. Laird
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON N6A 5C1, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada
| |
Collapse
|
208
|
Chen J, Chen J, Zhu Y, Liang C, Zhao HB. Deafness induced by Connexin 26 (GJB2) deficiency is not determined by endocochlear potential (EP) reduction but is associated with cochlear developmental disorders. Biochem Biophys Res Commun 2014; 448:28-32. [PMID: 24732355 DOI: 10.1016/j.bbrc.2014.04.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 04/06/2014] [Indexed: 10/25/2022]
Abstract
Connexin 26 (Cx26, GJB2) mutations are the major cause of hereditary deafness and are responsible for >50% of nonsyndromic hearing loss. Mouse models show that Cx26 deficiency can cause congenital deafness with cochlear developmental disorders, hair cell degeneration, and the reduction of endocochlear potential (EP) and active cochlear amplification. However, the underlying deafness mechanism still remains undetermined. Our previous studies revealed that hair cell degeneration is not a primary cause of hearing loss. In this study we investigated the role of EP reduction in Cx26 deficiency-induced deafness. We found that the EP reduction is not associated with congenital deafness in Cx26 knockout (KO) mice. The threshold of auditory brainstem response (ABR) in Cx26 KO mice was even greater than 110 dB SPL, demonstrating complete hearing loss. However, the EP in Cx26 KO mice varied and not completely abolished. In some cases, the EP could still remain at higher levels (>70 mV). We further found that the deafness in Cx26 KO mice is associated with cochlear developmental disorders. Deletion of Cx26 in the cochlea before postnatal day 5 (P5) could cause congenital deafness. The cochlea had developmental disorders and the cochlear tunnel was not open. However, no congenital deafness was found when Cx26 was deleted after P5. The cochlea also displayed normal development and the cochlear tunnel was open normally. These data suggest that congenital deafness induced by Cx26 deficiency is not determined by EP reduction and may result from cochlear developmental disorders.
Collapse
Affiliation(s)
- Jin Chen
- Dept. of Otolaryngology, University of Kentucky Medical School, Lexington, KY 40536, USA.; Department of Otolaryngology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Jing Chen
- Dept. of Otolaryngology, University of Kentucky Medical School, Lexington, KY 40536, USA
| | - Yan Zhu
- Dept. of Otolaryngology, University of Kentucky Medical School, Lexington, KY 40536, USA
| | - Chun Liang
- Dept. of Otolaryngology, University of Kentucky Medical School, Lexington, KY 40536, USA
| | - Hong-Bo Zhao
- Dept. of Otolaryngology, University of Kentucky Medical School, Lexington, KY 40536, USA..
| |
Collapse
|
209
|
Bohnenpoll T, Trowe MO, Wojahn I, Taketo MM, Petry M, Kispert A. Canonical Wnt signaling regulates the proliferative expansion and differentiation of fibrocytes in the murine inner ear. Dev Biol 2014; 391:54-65. [PMID: 24727668 DOI: 10.1016/j.ydbio.2014.03.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 03/27/2014] [Accepted: 03/29/2014] [Indexed: 01/18/2023]
Abstract
Otic fibrocytes tether the cochlear duct to the surrounding otic capsule but are also critically involved in maintenance of ion homeostasis in the cochlea, thus, perception of sound. The molecular pathways that regulate the development of this heterogenous group of cells from mesenchymal precursors are poorly understood. Here, we identified epithelial Wnt7a and Wnt7b as possible ligands of Fzd-mediated β-catenin (Ctnnb1)-dependent (canonical) Wnt signaling in the adjacent undifferentiated periotic mesenchyme (POM). Mice with a conditional deletion of Ctnnb1 in the POM exhibited a complete failure of fibrocyte differentiation, a severe reduction of mesenchymal cells surrounding the cochlear duct, loss of pericochlear spaces, a thickening and partial loss of the bony capsule and a secondary disturbance of cochlear duct coiling shortly before birth. Analysis at earlier stages revealed that radial patterning of the POM in two domains with highly condensed cartilaginous precursors and more loosely arranged inner mesenchymal cells occurred normally but that proliferation in the inner domain was reduced and cytodifferentiation failed. Cells with mis/overexpression of a stabilized form of Ctnnb1 in the entire POM mesenchyme sorted to the inner mesenchymal compartment and exhibited increased proliferation. Our analysis suggests that Wnt signals from the cochlear duct epithelium are crucial to induce differentiation and expansion of fibrocyte precursor cells. Our findings emphasize the importance of epithelial-mesenchymal signaling in inner ear development.
Collapse
Affiliation(s)
- Tobias Bohnenpoll
- Institut für Molekularbiologie, OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Mark-Oliver Trowe
- Institut für Molekularbiologie, OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Irina Wojahn
- Institut für Molekularbiologie, OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | | | - Marianne Petry
- Institut für Molekularbiologie, OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Andreas Kispert
- Institut für Molekularbiologie, OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany.
| |
Collapse
|
210
|
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: 29] [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
|
211
|
Slc26a4-insufficiency causes fluctuating hearing loss and stria vascularis dysfunction. Neurobiol Dis 2014; 66:53-65. [PMID: 24561068 DOI: 10.1016/j.nbd.2014.02.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Revised: 02/03/2014] [Accepted: 02/10/2014] [Indexed: 12/13/2022] Open
Abstract
SLC26A4 mutations can cause a distinctive hearing loss phenotype with sudden drops and fluctuation in patients. Existing Slc26a4 mutant mouse lines have a profound loss of hearing and vestibular function, with severe inner ear malformations that do not model this human phenotype. In this study, we generated Slc26a4-insufficient mice by manipulation of doxycycline administration to a transgenic mouse line in which all Slc26a4 expression was under the control of doxycycline. Doxycycline was administered from conception to embryonic day 17.5, and then it was discontinued. Auditory brainstem response thresholds showed significant fluctuation of hearing loss from 1 through 3months of age. The endocochlear potential, which is required for inner ear sensory cell function, correlated with auditory brainstem response thresholds. We observed degeneration of stria vascularis intermediate cells, the cells that generate the endocochlear potential, but no other abnormalities within the cochlea. We conclude that fluctuations of hearing result from fluctuations of the endocochlear potential and stria vascularis dysfunction in Slc26a4-insufficient mouse ears. This model can now be used to test potential interventions to reduce or prevent sudden hearing loss or fluctuation in human patients. Our strategy to generate a hypomorphic mouse model utilizing the tet-on system will be applicable to other diseases in which a hypomorphic allele is needed to model the human phenotype.
Collapse
|
212
|
Zhai J, Wang Q, Tao L. Connexin expression patterns in diseased human corneas. Exp Ther Med 2014; 7:791-798. [PMID: 24669234 PMCID: PMC3961128 DOI: 10.3892/etm.2014.1530] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Accepted: 01/13/2014] [Indexed: 12/22/2022] Open
Abstract
The present study aimed to explore the feasibility of using antisense connexin (Cx) treatment to promote corneal wound healing, and to investigate the changes of Cx gap junction proteins in terms of mRNA, protein expression and distribution in human corneas that were diseased due to various causes. A total of 13 diseased corneas were studied, which were obtained from five eyes injured by chemical burns, five infected eyes and three eyes with Stevens-Johnson syndrome (SJS)-affected corneas. Total RNA was extracted from the corneas and processed by qPCR with isoform primers to detect the expression of eight Cxs. Flow cytometry was adopted to determine the differences in the expression levels of Cx26, Cx31.1 and Cx43. Immunofluorescence was employed to show the localization of the three aforementioned Cxs. The qPCR results indicated that of the eight Cxs, only Cx26, Cx31.1 and Cx43 were upregulated in diseased corneas. Flow cytometry showed that all the diseased corneal tissues, with the exception of the SJS-affected corneas, showed a significantly higher percentage of cells that expressed Cx26 and Cx31.1 compared with the percentage in normal corneas (P<0.05). For Cx43, all three injured corneal groups showed a significantly higher percentage of cells that expressed Cx43 compared with the percentage in normal corneas (P<0.05). Immunohistochemical staining showed that the localization of Cx26, Cx31.1 and Cx43 differed between normal corneas and diseased corneas. This study elucidated the alteration of Cx expression patterns in several corneal diseases. The results indicated that Cx26, Cx31.1 and Cx43 are upregulated in chemically burned and infected corneas at the mRNA and protein levels, whereas only Cx43 is upregulated in SJS-affected corneas.
Collapse
Affiliation(s)
- Jiajie Zhai
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510040, P.R. China
| | - Qin Wang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510040, P.R. China
| | - Liang Tao
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510040, P.R. China
| |
Collapse
|
213
|
Chen J, Zhao HB. The role of an inwardly rectifying K(+) channel (Kir4.1) in the inner ear and hearing loss. Neuroscience 2014; 265:137-46. [PMID: 24480364 DOI: 10.1016/j.neuroscience.2014.01.036] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 01/16/2014] [Accepted: 01/18/2014] [Indexed: 11/18/2022]
Abstract
The KCNJ10 gene which encodes an inwardly rectifying K(+) channel Kir4.1 subunit plays an essential role in the inner ear and hearing. Mutations or deficiency of KCNJ10 can cause hearing loss with EAST or SeSAME syndromes. This review mainly focuses on the expression and function of Kir4.1 potassium channels in the inner ear and hearing. We first introduce general information about inwardly rectifying potassium (Kir) channels. Then, we review the expression and function of Kir4.1 channels in the inner ear, especially in endocochlear potential (EP) generation. Finally, we review KCNJ10 mutation-induced hearing loss and functional impairments. Kir4.1 is strongly expressed on the apical membrane of intermediate cells in the stria vascularis and in the satellite cells of cochlear ganglia. Functionally, Kir4.1 has critical roles in cochlear development and hearing through two distinct aspects of extracellular K(+) homeostasis: First, it participates in the generation and maintenance of EP and high K(+) concentration in the endolymph inside the scala media. Second, Kir4.1 is the major K(+) channel in satellite glial cells surrounding spiral ganglion neurons to sink K(+) ions expelled by the ganglion neurons during excitation. Kir4.1 deficiency leads to hearing loss with the absence of EP and spiral ganglion neuron degeneration. Deafness mutants show loss-of-function and reduced channel membrane-targeting and currents, which can be rescued upon by co-expression with wild-type Kir4.1. This review provides insights for further understanding Kir potassium channel function in the inner ear and the pathogenesis of deafness due to KCNJ10 deficiency, and also provides insights for developing therapeutic strategies targeting this deafness.
Collapse
Affiliation(s)
- J Chen
- Department of Morphology, Medical College of China Three Gorges University, Yichang, Hubei 443002, PR China; Department of Otolaryngology, University of Kentucky Medical Center, Lexington, KY 40536-0293, USA
| | - H-B Zhao
- Department of Otolaryngology, University of Kentucky Medical Center, Lexington, KY 40536-0293, USA.
| |
Collapse
|
214
|
Ceschi P, Bohl A, Sternberg K, Neumeister A, Senz V, Schmitz K, Kietzmann M, Scheper V, Lenarz T, Stöver T, Paasche G. Biodegradable polymeric coatings on cochlear implant surfaces and their influence on spiral ganglion cell survival. J Biomed Mater Res B Appl Biomater 2014; 102:1255-67. [DOI: 10.1002/jbm.b.33110] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 11/08/2013] [Accepted: 01/04/2014] [Indexed: 12/31/2022]
Affiliation(s)
- P. Ceschi
- Hannover Medical School; Department of Otolaryngology; Hannover Germany
- School of Veterinary Medicine Hannover Foundation; Department of Pharmacology; Toxicology and Pharmacy Hannover Germany
| | - A. Bohl
- University of Rostock, Institute for Biomedical Engineering; Rostock Germany
| | - K. Sternberg
- University of Rostock, Institute for Biomedical Engineering; Rostock Germany
| | | | - V. Senz
- University of Rostock, Institute for Biomedical Engineering; Rostock Germany
| | - K.P. Schmitz
- University of Rostock, Institute for Biomedical Engineering; Rostock Germany
| | - M. Kietzmann
- School of Veterinary Medicine Hannover Foundation; Department of Pharmacology; Toxicology and Pharmacy Hannover Germany
| | - V. Scheper
- Hannover Medical School; Department of Otolaryngology; Hannover Germany
| | - T. Lenarz
- Hannover Medical School; Department of Otolaryngology; Hannover Germany
| | - T. Stöver
- Hannover Medical School; Department of Otolaryngology; Hannover Germany
- KGU; Department of Otolaryngology; Frankfurt Germany
| | - G. Paasche
- Hannover Medical School; Department of Otolaryngology; Hannover Germany
| |
Collapse
|
215
|
Abstract
Fuchs' corneal dystrophy (FCD) is a common late-onset genetic disorder of the corneal endothelium. It causes loss of endothelial cell density and excrescences in the Descemet membrane, eventually progressing to corneal edema, necessitating corneal transplantation. The genetic basis of FCD is complex and heterogeneous, demonstrating variable expressivity and incomplete penetrance. To date, three causal genes, ZEB1, SLC4A11 and LOXHD1, have been identified, representing a small proportion of the total genetic load of FCD. An additional four loci have been localized, including a region on chromosome 18 that is potentially responsible for a large proportion of all FCD cases. The elucidation of the causal genes underlying these loci will begin to clarify the pathogenesis of FCD and pave the way for the emergence of nonsurgical treatments.
Collapse
Affiliation(s)
- Benjamin W Iliff
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | |
Collapse
|
216
|
Wangemann P. Mouse models for pendrin-associated loss of cochlear and vestibular function. Cell Physiol Biochem 2013; 32:157-65. [PMID: 24429822 PMCID: PMC4415819 DOI: 10.1159/000356635] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2013] [Indexed: 01/17/2023] Open
Abstract
The human gene SLC26A4 and the mouse ortholog Slc26a4 code for the protein pendrin, which is an anion exchanger expressed in apical membranes of selected epithelia. In the inner ear, pendrin is expressed in the cochlea, the vestibular labyrinth and the endolymphatic sac. Loss-of-function and hypo-functional mutations cause an enlargement of the vestibular aqueduct (EVA) and sensorineural hearing loss. The relatively high prevalence of SLC26A4 mutations provides a strong imperative to develop rational interventions that delay, ameliorate or prevent pendrin-associated loss of cochlear and vestibular function. This review summarizes recent studies in mouse models that have been developed to delineate the role of pendrin in the physiology of hearing and balance and that have brought forward the concept that a temporally and spatially limited therapy may be sufficient to secure a life-time of normal hearing in children bearing mutations of SLC26A4.
Collapse
Affiliation(s)
- Philine Wangemann
- Anatomy & Physiology Department, Kansas State University, Manhattan, Kansas, USA
| |
Collapse
|
217
|
Precise toxigenic ablation of intermediate cells abolishes the "battery" of the cochlear duct. J Neurosci 2013; 33:14601-6. [PMID: 24005310 DOI: 10.1523/jneurosci.2147-13.2013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The extracellular potential of excitable and nonexcitable cells with respect to ground is ∼0 mV. One of the known exceptions in mammals is the cochlear duct, where the potential is ∼80-100 mV, called the endocochlear potential (EP). The EP serves as the "battery" for transduction of sound, contributing toward the sensitivity of the auditory system. The stria vascularis (StV) of the cochlear duct is the station where the EP is generated, but the cell-specific roles in the StV are ill defined. Using the intermediate cell (IC)-specific tyrosinase promoter, under the control of diphtheria toxin (DT), we eliminated and/or halted differentiation of neural crest melanocytes after migration to the StV. The ensuing adult transgenic mice are profoundly deaf. Additionally, the EP was abolished. Expression of melanocyte early marker and Kir4.1 in ICs precedes the onset of pigment synthesis. Activation of DT leads to loss of ICs. Finally, in accord with the distinct embryology of retinal pigmented cells, transgenic mice with toxigenic ablation of neural crest-derived melanocytes have intact visual responses. We assert that the tyrosinase promoter is the distinct target for genetic manipulation of IC-specific genes.
Collapse
|
218
|
Treatment with 17-allylamino-17-demethoxygeldanamycin ameliorated symptoms of Bartter syndrome type IV caused by mutated Bsnd in mice. Biochem Biophys Res Commun 2013; 441:544-9. [PMID: 24189473 DOI: 10.1016/j.bbrc.2013.10.129] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Accepted: 10/24/2013] [Indexed: 11/21/2022]
Abstract
Mutations of BSND, which encodes barttin, cause Bartter syndrome type IV. This disease is characterized by salt and fluid loss, hypokalemia, metabolic alkalosis, and sensorineural hearing impairment. Barttin is the β-subunit of the ClC-K chloride channel, which recruits it to the plasma membranes, and the ClC-K/barttin complex contributes to transepithelial chloride transport in the kidney and inner ear. The retention of mutant forms of barttin in the endoplasmic reticulum (ER) is etiologically linked to Bartter syndrome type IV. Here, we report that treatment with 17-allylamino-17-demethoxygeldanamycin (17-AAG), an Hsp90 inhibitor, enhanced the plasma membrane expression of mutant barttins (R8L and G47R) in Madin-Darby canine kidney cells. Administration of 17-AAG to Bsnd(R8L/R8L) knock-in mice elevated the plasma membrane expression of R8L in the kidney and inner ear, thereby mitigating hypokalemia, metabolic alkalosis, and hearing loss. These results suggest that drugs that rescue ER-retained mutant barttin may be useful for treating patients with Bartter syndrome type IV.
Collapse
|
219
|
Efficient siRNA transfection to the inner ear through the intact round window by a novel proteidic delivery technology in the chinchilla. Gene Ther 2013; 21:10-8. [PMID: 24108151 PMCID: PMC3881030 DOI: 10.1038/gt.2013.49] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Revised: 07/21/2013] [Accepted: 08/27/2013] [Indexed: 12/27/2022]
Abstract
The use of small-interfering RNA (siRNA) has great potential for the development of drugs designed to knock down the expression of damage- or disease-causing genes. However, because of the high molecular weight and negative charge of siRNA, it is restricted from crossing the blood-cochlear barrier, which limits the concentration and size of molecules that are able to gain access to cells of the inner ear. Intratympanic approaches, which deliver siRNA to the middle ear, rely on permeation through the round window for access to the structures of the inner ear. We developed an innovative siRNA delivery recombination protein, TAT double-stranded RNA-binding domains (TAT-DRBDs), which can transfect Cy3-labeled siRNA into cells of the inner ear, including the inner and outer hair cells, crista ampullaris, macula utriculi and macula sacculi, through intact round-window permeation in the chinchilla in vivo, and there were no apparent morphological damages for the time of observation. We also found that Cy3-labeled siRNA could directly enter spiral ganglion neurons and the epithelium of the stria vascularis independently; however, the mechanism is unknown. Therefore, as a non-viral vector, TAT-DRBD is a good candidate for the delivery of double-stranded siRNAs for treating various inner ear ailments and preservation of hearing function.
Collapse
|
220
|
Abstract
Congenital long QT syndrome (LQTS) is a genetically heterogeneous group of heritable disorders of myocardial repolarization linked by the shared clinical phenotype of QT prolongation on electrocardiogram and an increased risk of potentially life-threatening cardiac arrhythmias. At the molecular level, mutations in 15 distinct LQTS-susceptibility genes that encode ion channel pore-forming α-subunits and accessory β-subunits central to the electromechanical function of the heart have been implicated in its pathogenesis. Over the past 2 decades, our evolving understanding of the electrophysiological mechanisms by which specific genetic substrates perturb the cardiac action potential has translated into vastly improved approaches to the diagnosis, risk stratification, and treatment of patients with LQTS. In this review, we describe how our understanding of the molecular underpinnings of LQTS has yielded numerous clinically meaningful genotype-phenotype correlations and how these insights have translated into genotype- and phenotype-guided approaches to the clinical management of LQTS.
Collapse
Affiliation(s)
| | - Michael J. Ackerman
- Departments of Medicine (Division of Cardiovascular Diseases), Pediatrics (Division of Pediatric Cardiology), and Molecular Pharmacology & Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN
| |
Collapse
|
221
|
Yang H, Xiong H, Huang Q, Pang J, Zheng X, Chen L, Yu R, Zheng Y. Compromised potassium recycling in the cochlea contributes to conservation of endocochlear potential in a mouse model of age-related hearing loss. Neurosci Lett 2013; 555:97-101. [PMID: 24055606 DOI: 10.1016/j.neulet.2013.09.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Revised: 05/10/2013] [Accepted: 09/10/2013] [Indexed: 12/13/2022]
Abstract
The C57BL/6 strain is considered an excellent model to study age-related hearing loss (AHL). Aging C57BL/6 mice are characterized by profound hearing loss but conservation of the endocochlear potential (EP). Here we show 12-month-old C57BL/6 mice display a notable hearing loss at 4, 8, 16 and 32kHz while the EP is maintained at normal level. Morphological examination shows significant outer hair cells loss in the cochlear basal turn and atrophy of the stria vascularis (SV). Fluorescence immunohistochemical studies reveal that potassium channel KCNJ10 and KCNQ1 expression dramatically decreased in the SV. Concomitant with this, mRNA levels of KCNJ10 and KCNQ1 are also reduced. In addition, three other potassium transporters, including α1-Na,K-ATPase, α2-Na,K-ATPase and NKCC1, reduce their expression at mRNA levels as well. These observations suggest that conservation of the EP in aging C57BL/6 mice is attributable to the SV generating a new balance for potassium influx and efflux at a relatively lower level.
Collapse
Affiliation(s)
- Haidi Yang
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, China; Institute of Hearing and Speech-Language Science, Sun Yat-sen University, China
| | | | | | | | | | | | | | | |
Collapse
|
222
|
Adachi N, Yoshida T, Nin F, Ogata G, Yamaguchi S, Suzuki T, Komune S, Hisa Y, Hibino H, Kurachi Y. The mechanism underlying maintenance of the endocochlear potential by the K+ transport system in fibrocytes of the inner ear. J Physiol 2013; 591:4459-72. [PMID: 23836687 PMCID: PMC3784193 DOI: 10.1113/jphysiol.2013.258046] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 07/03/2013] [Indexed: 11/08/2022] Open
Abstract
The endocochlear potential (EP) of +80 mV in the scala media, which is indispensable for audition, is controlled by K+ transport across the lateral cochlear wall. This wall includes two epithelial barriers, the syncytium and the marginal cells. The former contains multiple cell types, such as fibrocytes, which are exposed to perilymph on their basolateral surfaces. The apical surfaces of the marginal cells face endolymph. Between the two barriers lies the intrastrial space (IS), an extracellular space with a low K+ concentration ([K+]) and a potential similar to the EP. This intrastrial potential (ISP) dominates the EP and represents the sum of the diffusion potential elicited by a large K+ gradient across the apical surface of the syncytium and the syncytium's potential, which is slightly positive relative to perilymph. Although a K+ transport system in fibrocytes seems to contribute to the EP, the mechanism remains uncertain. We examined the electrochemical properties of the lateral wall of guinea pigs with electrodes sensitive to potential and K+ while perfusing into the perilymph of the scala tympani blockers of Na+,K+-ATPase, the K+ pump thought to be essential to the system. Inhibiting Na+,K+-ATPase barely affected [K+] in the IS but greatly decreased [K+] within the syncytium, reducing the K+ gradient across its apical surface. The treatment hyperpolarized the syncytium only moderately. Consequently, both the ISP and the EP declined. Fibrocytes evidently use the Na+,K+-ATPase to achieve local K+ transport, maintaining the syncytium's high [K+] that is crucial for the K+ diffusion underlying the positive ISP.
Collapse
Affiliation(s)
- Naoko Adachi
- H. Hibino: Department of Molecular Physiology, Niigata University School of Medicine, 1-757 Asahimachi-dori, Chuo-ku, Niigata, Niigata 951-8510, Japan. Y. Kurachi: Division of Molecular and Cellular Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
223
|
Tsai EA, Berman MA, Conlin LK, Rehm HL, Francey LJ, Deardorff MA, Holst J, Kaur M, Gallant E, Clark DM, Glessner JT, Jensen ST, Grant SFA, Gruber PJ, Hakonarson H, Spinner NB, Krantz ID. PECONPI: a novel software for uncovering pathogenic copy number variations in non-syndromic sensorineural hearing loss and other genetically heterogeneous disorders. Am J Med Genet A 2013; 161A:2134-47. [PMID: 23897863 DOI: 10.1002/ajmg.a.36038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 04/11/2013] [Indexed: 11/10/2022]
Abstract
This report describes an algorithm developed to predict the pathogenicity of copy number variants (CNVs) in large sample cohorts. CNVs (genomic deletions and duplications) are found in healthy individuals and in individuals with genetic diagnoses, and differentiation of these two classes of CNVs can be challenging and usually requires extensive manual curation. We have developed PECONPI, an algorithm to assess the pathogenicity of CNVs based on gene content and CNV frequency. This software was applied to a large cohort of patients with genetically heterogeneous non-syndromic hearing loss to score and rank each CNV based on its relative pathogenicity. Of 636 individuals tested, we identified the likely underlying etiology of the hearing loss in 14 (2%) of the patients (1 with a homozygous deletion, 7 with a deletion of a known hearing loss gene and a point mutation on the trans allele and 6 with a deletion larger than 1 Mb). We also identified two probands with smaller deletions encompassing genes that may be functionally related to their hearing loss. The ability of PECONPI to determine the pathogenicity of CNVs was tested on a second genetically heterogeneous cohort with congenital heart defects (CHDs). It successfully identified a likely etiology in 6 of 355 individuals (2%). We believe this tool is useful for researchers with large genetically heterogeneous cohorts to help identify known pathogenic causes and novel disease genes.
Collapse
Affiliation(s)
- Ellen A Tsai
- Genomics and Computational Biology Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
224
|
Ahmed S, Vorasubin N, Lopez IA, Hosokawa S, Ishiyama G, Ishiyama A. The expression of glutamate aspartate transporter (GLAST) within the human cochlea and its distribution in various patient populations. Brain Res 2013; 1529:134-42. [PMID: 23850643 DOI: 10.1016/j.brainres.2013.06.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 06/14/2013] [Accepted: 06/26/2013] [Indexed: 10/26/2022]
Abstract
Glutamate plays an important role in the central nervous system as an excitatory neurotransmitter. However, its abundance can lead to excitotoxicity which necessitates the proper function of active glutamate transporters. The glutamate-aspartate transporter (GLAST) has been shown to exist and function within non-human cochlear specimens regulating the inner ear glutamate concentration. In this study, we examined human cochleas from formalin-fixed celloidin-embedded temporal bone specimens of three different types of patients (Meniere's disease, normal controls, and other otopathologic conditions) and examined the differential expression of GLAST in the spiral ligament of the basal, middle, and apical turns of the cochlea. Immunohistochemical staining was performed with polyclonal antibodies against GLAST and image analysis was carried out with the Image J analysis software. In contrast to other studies with non-human specimens, GLAST was expressed in the spiral ligament fibrocytes but was not detected in the satellite cells of the spiral ganglia or supporting cells of the Organ of Corti in the human cochlea. Our data also showed that GLAST expression significantly differs in the basal and apical turns of the cochlea. Lastly, post-hoc analysis showed a difference in the GLAST immunoreactive area of patients with Meniere's disease when compared to that of patients with other otopathologic conditions-such as presbycusis or ototoxicity. These results may potentially lead to further understanding of different disease states that affect hearing.
Collapse
Affiliation(s)
- Sameer Ahmed
- Department of Head and Neck Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1624, USA
| | | | | | | | | | | |
Collapse
|
225
|
Kim AH, Nahm E, Sollas A, Mattiace L, Rozental R. Connexin 43 and hearing: possible implications for retrocochlear auditory processing. Laryngoscope 2013; 123:3185-93. [PMID: 23817980 DOI: 10.1002/lary.24249] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Revised: 05/10/2013] [Accepted: 05/22/2013] [Indexed: 11/09/2022]
Abstract
OBJECTIVES/HYPOTHESIS To examine the relationship between hearing and connexin 43, a dominant gap junctional protein in the central nervous system. STUDY DESIGN Original research. METHODS Connexin 43 heterozygous mice are used to assess its mutational effect on hearing. Results are compared to controls consisting of connexin 43, wild type and CBA/J mice. Hearing is assessed using auditory brainstem response and distortion product otoacoustic emissions tests. Distribution of connexin 43 in the organ of Corti and the retrocochlear auditory centers (eight nerve, cochlear nucleus, olivary complex, lateral lemniscus, inferior colliculus, respectively) is examined. Fluorescent markers are used to elucidate cell types. RESULTS Mean click auditory brainstem response threshold for the young connexin 43 heterozygous mice (3-4 months) was 36.7 ± 12.6 dB compared to 25 ± 0 dB for control mice (P < 0.05). Mean threshold difference became more pronounced (68 ± 7.5 dB vs. 31 ± 2.2 dB) at 10 months (P < 0.05). Tonal auditory brainstem response testing showed elevated thresholds (>60 dB) at all frequencies (4-32 kHz) compared to the controls. Distortion product otoacoustic emissions (DPOAE) were present in all the mice, although the older connexin 43 heterozygous mice responded at higher thresholds. The pattern of connexin 43 immunoreactivity was distinctive from connexin 26 and 30, showing minimal presence in the organ of Corti but robustly present in the retrocochlear centers. CONCLUSION Connexin 43 heterozygous mice demonstrated greater degree of hearing loss compared to age-matched controls. It is abundantly found in the retrocochlear auditory centers. The mechanism of hearing loss in these mice does not appear to be related to hair cell loss.
Collapse
Affiliation(s)
- Ana H Kim
- Department of Otolaryngology, New York Eye and Ear Infirmary, New York; New York Medical College, Valhalla, New York
| | | | | | | | | |
Collapse
|
226
|
Liang C, Hong Q, Jiang TT, Gao Y, Yao XF, Luo XX, Zhuo XH, Shinn JB, Jones RO, Zhao HB, Lu GJ. The effects and outcomes of electrolyte disturbances and asphyxia on newborns hearing. Int J Pediatr Otorhinolaryngol 2013; 77:1072-6. [PMID: 23648318 PMCID: PMC3738180 DOI: 10.1016/j.ijporl.2013.03.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 03/25/2013] [Accepted: 03/28/2013] [Indexed: 10/26/2022]
Abstract
OBJECTIVE To determine the effect of electrolyte disturbances (ED) and asphyxia on infant hearing and hearing outcomes. STUDY DESIGN We conducted newborn hearing screening with transient evoked otoacoustic emission (TEOAE) test on a large scale (>5000 infants). The effects of ED and asphyxia on infant hearing and hearing outcomes were evaluated. RESULT The pass rate of TEOAE test was significantly reduced in preterm infants with ED (83.1%, multiple logistic regression analysis: P<0.01) but not in full-term infants with ED (93.6%, P=0.41). However, there was no significant reduction in the pass rate in infants with asphyxia (P=0.85). We further found that hypocalcaemia significantly reduced the pass rate of TEOAE test (86.8%, P<0.01). In the follow-up recheck at 3 months of age, the pass rate remained low (44.4%, P<0.01). CONCLUSION ED is a high-risk factor for preterm infant hearing. Hypocalcaemia can produce more significant impairment with a low recovery rate.
Collapse
Affiliation(s)
- Chun Liang
- Maternal and Child Health Care Hospital of Baoan, Shenzhen, P.R. of China, 518133,Dept. of Otolaryngology, University of Kentucky Medical Center, Lexington, KY, USA 40536
| | - Qi Hong
- Maternal and Child Health Care Hospital of Baoan, Shenzhen, P.R. of China, 518133
| | - Tao-Tao Jiang
- Maternal and Child Health Care Hospital of Baoan, Shenzhen, P.R. of China, 518133
| | - Yan Gao
- Maternal and Child Health Care Hospital of Baoan, Shenzhen, P.R. of China, 518133
| | - Xiao-Fang Yao
- Maternal and Child Health Care Hospital of Baoan, Shenzhen, P.R. of China, 518133
| | - Xiao-Xing Luo
- Maternal and Child Health Care Hospital of Baoan, Shenzhen, P.R. of China, 518133
| | - Xiu-Hui Zhuo
- Maternal and Child Health Care Hospital of Baoan, Shenzhen, P.R. of China, 518133
| | - Jennifer B. Shinn
- Dept. of Otolaryngology, University of Kentucky Medical Center, Lexington, KY, USA 40536
| | - Raleigh O. Jones
- Dept. of Otolaryngology, University of Kentucky Medical Center, Lexington, KY, USA 40536
| | - Hong-Bo Zhao
- Dept. of Otolaryngology, University of Kentucky Medical Center, Lexington, KY, USA 40536
| | - Guang-Jin Lu
- Maternal and Child Health Care Hospital of Baoan, Shenzhen, P.R. of China, 518133
| |
Collapse
|
227
|
Inner ear tissue remodeling and ion homeostasis gene alteration in murine chronic otitis media. Otol Neurotol 2013; 34:338-46. [PMID: 23269288 DOI: 10.1097/mao.0b013e31827b4d0a] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
HYPOTHESIS Studies were designed to ascertain the impact of chronic middle ear infection on the numerous ion and water channels, transporters, and tissue remodeling genes in the inner and middle ear. BACKGROUND Permanent sensorineural hearing loss is a significant problem resulting from chronic middle ear disease, although the inner ear processes involved are poorly defined. Maintaining a balanced ionic composition of endolymph in the inner ear is crucial for hearing; thus, it was hypothesized that this may be at risk with inflammation. METHODS Inner and middle ear RNA collected separately from 6-month-old C3H/HeJ mice with prolonged middle ear disease were subjected to qRT-PCR for 8 common inflammatory cytokine genes, 24 genes for channels controlling ion (sodium, potassium, and chloride) and water (aquaporin) transport, tight junction claudins, and gap junction connexins, and 32 tissue remodeling genes. Uninfected Balb/c mice were used as controls. RESULTS Significant increase in inner ear inflammatory and ion homeostasis (claudin, aquaporin, and gap junction) gene expression, and both upregulation and downregulation of tissue remodeling gene expression occurred. Alteration in middle ear ion homeostasis and tissue remodeling gene expression was noted in the setting of uniform upregulation of cytokine genes. CONCLUSION Chronic inflammatory middle ear disease can impact inner ear ion and water transport functions and induce tissue remodeling. Recognizing these inner ear mechanisms at risk may identify potential therapeutic targets to maintain hearing during prolonged otitis media.
Collapse
|
228
|
Gao Y, Yechikov S, Vázquez AE, Chen D, Nie L. Impaired surface expression and conductance of the KCNQ4 channel lead to sensorineural hearing loss. J Cell Mol Med 2013; 17:889-900. [PMID: 23750663 PMCID: PMC3729637 DOI: 10.1111/jcmm.12080] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 03/29/2013] [Indexed: 01/21/2023] Open
Abstract
KCNQ4, a voltage-gated potassium channel, plays an important role in maintaining cochlear ion homoeostasis and regulating hair cell membrane potential, both essential for normal auditory function. Mutations in the KCNQ4 gene lead to DFNA2, a subtype of autosomal dominant non-syndromic deafness that is characterized by progressive sensorineural hearing loss across all frequencies. Despite recent advances in the identification of pathogenic KCNQ4 mutations, the molecular aetiology of DFNA2 remains unknown. We report here that decreased cell surface expression and impaired conductance of the KCNQ4 channel are two mechanisms underlying hearing loss in DFNA2. In HEK293T cells, a dramatic decrease in cell surface expression was detected by immunofluorescent microscopy and confirmed by Western blot for the pathogenic KCNQ4 mutants L274H, W276S, L281S, G285C, G285S, G296S and G321S, while their overall cellular levels remained normal. In addition, none of these mutations affected tetrameric assembly of KCNQ4 channels. Consistent with these results, all mutants showed strong dominant-negative effects on the wild-type (WT) channel function. Most importantly, overexpression of HSP90β, a key component of the molecular chaperone network that controls the KCNQ4 biogenesis, significantly increased cell surface expression of the KCNQ4 mutants L281S, G296S and G321S. KCNQ4 surface expression was restored or considerably improved in HEK293T cells mimicking the heterozygous condition of these mutations in DFNA2 patients. Finally, our electrophysiological studies demonstrated that these mutations directly compromise the conductance of the KCNQ4 channel, since no significant change in KCNQ4 current was observed after KCNQ4 surface expression was restored or improved.
Collapse
Affiliation(s)
- Yanhong Gao
- Department of Otolaryngology, University of California Davis, Davis, CA, USA
| | | | | | | | | |
Collapse
|
229
|
Li X, Zhou F, Marcus DC, Wangemann P. Endolymphatic Na⁺ and K⁺ concentrations during cochlear growth and enlargement in mice lacking Slc26a4/pendrin. PLoS One 2013; 8:e65977. [PMID: 23741519 PMCID: PMC3669272 DOI: 10.1371/journal.pone.0065977] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 05/01/2013] [Indexed: 11/30/2022] Open
Abstract
Slc26a4 (Δ/Δ) mice are deaf, develop an enlarged membranous labyrinth, and thereby largely resemble the human phenotype where mutations of SLC26A4 cause an enlarged vestibular aqueduct and sensorineural hearing loss. The enlargement is likely caused by abnormal ion and fluid transport during the time of embryonic development, however, neither the mechanisms of ion transport nor the ionic composition of the luminal fluid during this time of development are known. Here we determine the ionic composition of inner ear fluids at the time at which the enlargement develops and the onset of expression of selected ion transporters. Concentrations of Na(+) and K(+) were measured with double-barreled ion-selective electrodes in the cochlea and the endolymphatic sac of Slc26a4 (Δ/+), which develop normal hearing, and of Slc26a4 (Δ/Δ) mice, which fail to develop hearing. The expression of specific ion transporters was examined by quantitative RT-PCR and immunohistochemistry. High Na(+) (∼141 mM) and low K(+) concentrations (∼11 mM) were found at embryonic day (E) 16.5 in cochlear endolymph of Slc26a4 (Δ/+) and Slc26a4 (Δ/Δ) mice. Shortly before birth the K(+) concentration began to rise. Immediately after birth (postnatal day 0), the Na(+) and K(+) concentrations in cochlear endolymph were each ∼80 mM. In Slc26a4 (Δ/Δ) mice, the rise in the K(+) concentration occurred with a ∼3 day delay. K(+) concentrations were also found to be low (∼15 mM) in the embryonic endolymphatic sac. The onset of expression of the K(+) channel KCNQ1 and the Na(+)/2Cl(-)/K(+) cotransporter SLC12A2 occurred in the cochlea at E19.5 in Slc26a4 (Δ/+) and Slc26a4 (Δ/Δ) mice. These data demonstrate that endolymph, at the time at which the enlargement develops, is a Na(+)-rich fluid, which transitions into a K(+)-rich fluid before birth. The data suggest that the endolymphatic enlargement caused by a loss of Slc26a4 is a consequence of disrupted Na(+) transport.
Collapse
Affiliation(s)
- Xiangming Li
- Anatomy and Physiology Department, Kansas State University, Manhattan, Kansas, United States of America
| | - Fei Zhou
- Anatomy and Physiology Department, Kansas State University, Manhattan, Kansas, United States of America
| | - Daniel C. Marcus
- Anatomy and Physiology Department, Kansas State University, Manhattan, Kansas, United States of America
| | - Philine Wangemann
- Anatomy and Physiology Department, Kansas State University, Manhattan, Kansas, United States of America
| |
Collapse
|
230
|
Hou J. A connected tale of claudins from the renal duct to the sensory system. Tissue Barriers 2013; 1:e24968. [PMID: 24533254 PMCID: PMC3867513 DOI: 10.4161/tisb.24968] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 05/06/2013] [Accepted: 05/08/2013] [Indexed: 12/17/2022] Open
Abstract
Claudins are tight junction membrane proteins that regulate paracellular permeability to ions and solutes in many physiological systems. The electric property of claudin is the most interesting and pertains to two important organ functions: the renal and sensorineural functions. The kidney comprises of three major segments of epithelial tubules with different paracellular permeabilities: the proximal tubule (PT), the thick acending limb of Henle’s loop (TALH) and the collecting duct (CD). Claudins act as ion channels allowing selective permeation of Na+ in the PT, Ca2+ and Mg2+ in the TALH and Cl− in the CD. The inner ear, on the other hand, expresses claudins as a barrier to block K+ permeation between endolymph and perilymph. The permeability properties of claudins in different organs can be attributed to claudin interaction within the cell membrane and between neighboring cells. The first extracellular loop of claudins contains determinants of paracellular ionic permeability. While analogous to transmembrane ion channels in many ways, the biophysical and biochemical properties of claudin based paracellular channels remain to be fully characterized.
Collapse
Affiliation(s)
- Jianghui Hou
- Washington University Renal Division; St. Louis, MO USA
| |
Collapse
|
231
|
Inner ear supporting cells: rethinking the silent majority. Semin Cell Dev Biol 2013; 24:448-59. [PMID: 23545368 DOI: 10.1016/j.semcdb.2013.03.009] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 03/21/2013] [Indexed: 11/21/2022]
Abstract
Sensory epithelia of the inner ear contain two major cell types: hair cells and supporting cells. It has been clear for a long time that hair cells play critical roles in mechanoreception and synaptic transmission. In contrast, until recently the more abundant supporting cells were viewed as serving primarily structural and homeostatic functions. In this review, we discuss the growing information about the roles that supporting cells play in the development, function and maintenance of the inner ear, their activities in pathological states, their potential for hair cell regeneration, and the mechanisms underlying these processes.
Collapse
|
232
|
Vele O, Schrijver I. Inherited hearing loss: molecular genetics and diagnostic testing. ACTA ACUST UNITED AC 2013; 2:231-48. [PMID: 23495655 DOI: 10.1517/17530059.2.3.231] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Hearing loss is a clinically and genetically heterogeneous condition with major medical and social consequences. It affects up to 8% of the general population. OBJECTIVE This review recapitulates the principles of auditory physiology and the molecular basis of hearing loss, outlines the main types of non-syndromic and syndromic deafness by mode of inheritance, and provides an overview of current clinically available genetic testing. METHODS This paper reviews the literature on auditory physiology and on genes, associated with hearing loss, for which genetic testing is presently offered. RESULTS/CONCLUSION The advent of molecular diagnostic assays for hereditary hearing loss permits earlier detection of the underlying causes, facilitates appropriate interventions, and is expected to generate the data necessary for more specific genotype-phenotype correlations.
Collapse
Affiliation(s)
- Oana Vele
- Stanford University School of Medicine, Department of Pathology and Pediatrics, L235, 300 Pasteur Drive, Stanford, CA 94305, USA +1 650 724 2403 ; +1 650 724 1567 ;
| | | |
Collapse
|
233
|
Gao Y, Yechikov S, Vazquez AE, Chen D, Nie L. Distinct roles of molecular chaperones HSP90α and HSP90β in the biogenesis of KCNQ4 channels. PLoS One 2013; 8:e57282. [PMID: 23431407 PMCID: PMC3576372 DOI: 10.1371/journal.pone.0057282] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 01/21/2013] [Indexed: 01/30/2023] Open
Abstract
Loss-of-function mutations in the KCNQ4 channel cause DFNA2, a subtype of autosomal dominant non-syndromic deafness that is characterized by progressive sensorineural hearing loss. Previous studies have demonstrated that the majority of the pathogenic KCNQ4 mutations lead to trafficking deficiency and loss of KCNQ4 currents. Over the last two decades, various strategies have been developed to rescue trafficking deficiency of pathogenic mutants; the most exciting advances have been made by manipulating activities of molecular chaperones involved in the biogenesis and quality control of the target protein. However, such strategies have not been established for KCNQ4 mutants and little is known about the molecular chaperones governing the KCNQ4 biogenesis. To identify KCNQ4-associated molecular chaperones, a proteomic approach was used in this study. As a result, two major molecular chaperones, HSP70 and HSP90, were identified and then confirmed by reciprocal co-immunoprecipitation assays, suggesting that the HSP90 chaperone pathway might be involved in the KCNQ4 biogenesis. Manipulating chaperone expression further revealed that two different isoforms of HSP90, the inducible HSP90α and the constitutive HSP90β, had opposite effects on the cellular level of the KCNQ4 channel; that HSP40, HSP70, and HOP, three key components of the HSP90 chaperone pathway, were crucial in facilitating KCNQ4 biogenesis. In contrast, CHIP, a major E3 ubiquitin ligase, had an opposite effect. Collectively, our data suggest that HSP90α and HSP90β play key roles in controlling KCNQ4 homeostasis via the HSP40-HSP70-HOP-HSP90 chaperone pathway and the ubiquitin-proteasome pathway. Most importantly, we found that over-expression of HSP90β significantly improved cell surface expression of the trafficking-deficient, pathogenic KCNQ4 mutants L274H and W276S. KCNQ4 surface expression was restored by HSP90β in cells mimicking heterozygous conditions of the DFNA2 patients, even though it was not sufficient to rescue the function of KCNQ4 channels.
Collapse
Affiliation(s)
- Yanhong Gao
- Department of Otolaryngology, University of California Davis, Davis, California, United States of America
| | - Sergey Yechikov
- Department of Otolaryngology, University of California Davis, Davis, California, United States of America
| | - Ana E. Vazquez
- Department of Otolaryngology, University of California Davis, Davis, California, United States of America
- * E-mail: (AEV); (LN)
| | - Dongyang Chen
- Department of Otolaryngology, University of California Davis, Davis, California, United States of America
| | - Liping Nie
- Department of Otolaryngology, University of California Davis, Davis, California, United States of America
- * E-mail: (AEV); (LN)
| |
Collapse
|
234
|
Giudicessi JR, Ackerman MJ. Prevalence and potential genetic determinants of sensorineural deafness in KCNQ1 homozygosity and compound heterozygosity. ACTA ACUST UNITED AC 2013; 6:193-200. [PMID: 23392653 DOI: 10.1161/circgenetics.112.964684] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND- Homozygous or compound heterozygous mutations in KCNQ1 cause Jervell and Lange-Nielsen syndrome, a rare, autosomal-recessive form of long-QT syndrome characterized by deafness, marked QT prolongation, and a high risk of sudden death. However, it is not understood why some individuals with mutations on both KCNQ1 alleles present without deafness. In this study, we sought to determine the prevalence and genetic determinants of this phenomenon in a large referral population of patients with long-QT syndrome. METHODS AND RESULTS- A retrospective analysis of all patients with long-QT syndrome evaluated from July 1998 to April 2012 was used to identify those with ≥1 KCNQ1 mutation. Of the 249 KCNQ1-positive patients identified, 15 (6.0%) harbored a rare putative pathogenic mutation on both KCNQ1 alleles. Surprisingly, 11 of these patients (73%) presented without the sensorineural deafness associated with Jervell and Lange-Nielsen syndrome. The degree of QT-interval prolongation and the number of breakthrough cardiac events were similar between patients with and without deafness. Interestingly, truncating mutations were more prevalent in patients with Jervell and Lange-Nielsen syndrome (79%) than in nondeaf patients (36%; P<0.001) derived from this study and those in the literature. CONCLUSIONS- In this study, we provide evidence that the recessive inheritance of a severe long-QT syndrome type 1 phenotype in the absence of an auditory phenotype may represent a more common pattern of long-QT syndrome inheritance than previously anticipated and that these cases should be treated as a higher-risk long-QT syndrome subset similar to their Jervell and Lange-Nielsen syndrome counterparts. Furthermore, mutation type may serve as a genetic determinant of deafness, but not cardiac expressivity, in individuals harboring ≥1 KCNQ1 mutation on each allele.
Collapse
|
235
|
Monzack EL, Cunningham LL. Lead roles for supporting actors: critical functions of inner ear supporting cells. Hear Res 2013; 303:20-9. [PMID: 23347917 DOI: 10.1016/j.heares.2013.01.008] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 12/20/2012] [Accepted: 01/10/2013] [Indexed: 12/27/2022]
Abstract
Many studies that aim to investigate the underlying mechanisms of hearing loss or balance disorders focus on the hair cells and spiral ganglion neurons of the inner ear. Fewer studies have examined the supporting cells that contact both of these cell types in the cochlea and vestibular end organs. While the roles of supporting cells are still being elucidated, emerging evidence indicates that they serve many functions vital to maintaining healthy populations of hair cells and spiral ganglion neurons. Here we review recent studies that highlight the critical roles supporting cells play in the development, function, survival, death, phagocytosis, and regeneration of other cell types within the inner ear. Many of these roles have also been described for glial cells in other parts of the nervous system, and lessons from these other systems continue to inform our understanding of supporting cell functions. This article is part of a Special Issue entitled "Annual Reviews 2013".
Collapse
Affiliation(s)
- Elyssa L Monzack
- National Institute on Deafness and Other Communication Disorders, 5 Research Court, Rockville, MD 20850, USA.
| | | |
Collapse
|
236
|
Reimann K, Krishnamoorthy G, Wangemann P. NOS inhibition enhances myogenic tone by increasing rho-kinase mediated Ca2+ sensitivity in the male but not the female gerbil spiral modiolar artery. PLoS One 2013; 8:e53655. [PMID: 23301097 PMCID: PMC3536759 DOI: 10.1371/journal.pone.0053655] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 12/04/2012] [Indexed: 11/22/2022] Open
Abstract
Cochlear blood flow regulation is important to prevent hearing loss caused by ischemia and oxidative stress. Cochlear blood supply is provided by the spiral modiolar artery (SMA). The myogenic tone of the SMA is enhanced by the nitric oxide synthase (NOS) blocker L-NG-Nitro-Arginine (LNNA) in males, but not in females. Here, we investigated whether this gender difference is based on differences in the cytosolic Ca2+ concentration and/or the Ca2+ sensitivity of the myofilaments. Vascular diameter, myogenic tone, cytosolic Ca2+, and Ca2+ sensitivity were evaluated in pressurized SMA segments isolated from male and female gerbils using laser-scanning microscopy and microfluorometry. The gender difference of the LNNA-induced tone was compared, in the same vessel segments, to tone induced by 150 mM K+ and endothelin-1, neither of which showed an apparent gender-difference. Interestingly, LNNA-induced tone in male SMAs was observed in protocols that included changes in intramural pressure, but not when the intramural pressure was held constant. LNNA in male SMAs did not increase the global Ca2+ concentration in smooth muscle cells but increased the Ca2+ sensitivity. This increase in the Ca2+ sensitivity was abolished in the presence of the guanylyl cyclase inhibitor ODQ or by extrinsic application of either the nitric oxide (NO)-donor DEA-NONOate or the cGMP analog 8-pCPT-cGMP. The rho-kinase blocker Y27632 decreased the basal Ca2+ sensitivity and abolished the LNNA-induced increase in Ca2+ sensitivity in male SMAs. Neither LNNA nor Y27632 changed the Ca2+ sensitivity in female SMAs. The data suggest that the gender difference in LNNA-induced tone is based on a gender difference in the regulation of rho-kinase mediated Ca2+ sensitivity. Rho-kinase and NO thus emerge as critical factors in the regulation of cochlear blood flow. The larger role of NO-dependent mechanisms in male SMAs predicts greater restrictions on cochlear blood flow under conditions of impaired endothelial cell function.
Collapse
Affiliation(s)
- Katrin Reimann
- Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas, United States of America
- Universitätsklinik für Hals-, Nasen- und Ohrenheilkunde, Eberhard-Karls Universität, Tübingen, Germany
| | - Gayathri Krishnamoorthy
- Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas, United States of America
| | - Philine Wangemann
- Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas, United States of America
- * E-mail:
| |
Collapse
|
237
|
Anatomy and physiology of the external, middle and inner ear. DISORDERS OF PERIPHERAL AND CENTRAL AUDITORY PROCESSING 2013. [DOI: 10.1016/b978-0-7020-5310-8.00001-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
238
|
Cell-type-specific roles of Na+/K+ ATPase subunits in Drosophila auditory mechanosensation. Proc Natl Acad Sci U S A 2012; 110:181-6. [PMID: 23248276 DOI: 10.1073/pnas.1208866110] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Ion homeostasis is a fundamental cellular process particularly important in excitable cell activities such as hearing. It relies on the Na(+)/K(+) ATPase (also referred to as the Na pump), which is composed of a catalytic α subunit and a β subunit required for its transport to the plasma membrane and for regulating its activity. We show that α and β subunits are expressed in Johnston's organ (JO), the Drosophila auditory organ. We knocked down expression of α subunits (ATPα and α-like) and β subunits (nrv1, nrv2, and nrv3) individually in JO with UAS/Gal4-mediated RNAi. ATPα shows elevated expression in the ablumenal membrane of scolopale cells, which enwrap JO neuronal dendrites in endolymph-like compartments. Knocking down ATPα, but not α-like, in the entire JO or only in scolopale cells using specific drivers, resulted in complete deafness. Among β subunits, nrv2 is expressed in scolopale cells and nrv3 in JO neurons. Knocking down nrv2 in scolopale cells blocked Nrv2 expression, reduced ATPα expression in the scolopale cells, and caused almost complete deafness. Furthermore, knockdown of either nrv2 or ATPα specifically in scolopale cells causes abnormal, electron-dense material accumulation in the scolopale space. Similarly, nrv3 functions in JO but not in scolopale cells, suggesting neuron specificity that parallels nrv2 scolopale cell-specific support of the catalytic ATPα. Our studies provide an amenable model to investigate generation of endolymph-like extracellular compartments.
Collapse
|
239
|
Bashir ZEH, Latief N, Belyantseva IA, Iqbal F, Riazuddin SA, Amer Riazuddin S, Khan SN, Friedman TB, Riazuddin S, Riazuddin S. Phenotypic variability of CLDN14 mutations causing DFNB29 hearing loss in the Pakistani population. J Hum Genet 2012; 58:102-8. [PMID: 23235333 PMCID: PMC3596117 DOI: 10.1038/jhg.2012.143] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Human hereditary deafness at the DFNB29 autosomal locus on chromosome 21q22.1 is caused by recessive mutations of CLDN14, encoding claudin 14. This tight junction protein is tetra-membrane spanning that localizes to the apical tight junctions of organ of Corti hair cells and in many other tissues. Typically, the DFNB29 phenotype is characterized by pre-lingual, bi-lateral, sensorineural hearing loss. The goal of this study was to define the identity and frequency of CLDN14 mutations and associated inner ear phenotypes in a cohort of 800 Pakistani families segregating deafness. Hearing loss in 15 multi-generational families was found to co-segregate with CLDN14-linked STR markers. The sequence of the six exons and regions flanking the introns of CLDN14 in these 15 families revealed five likely pathogenic alleles. Two are novel missense substitutions (p.Ser87Ile and p.Ala94Val) while p.Arg81His, p.Val85Asp and p.Met133ArgfsX23 have been reported previously. Haplotype analyses indicate that p.Val85Asp and p.Met133ArgfsX23 are founder mutations. The p.Val85Asp accounts for approximately 67% of the mutant alleles of CLDN14 in our cohort. Combined with previously reported data, CLDN14 mutations were identified in 18 of 800 Pakistani families (2.25%; 95% CI, 1.4-3.5%). Hearing loss in the affected individuals homozygous for CLDN14 mutations varied from moderate to profound. This phenotypic variability may be due to environmental factors (e.g. drug and noise exposure) and/or genetic modifiers.
Collapse
Affiliation(s)
- Zil-e-Huma Bashir
- National Center of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
240
|
Affiliation(s)
- Kenneth Shepard
- Department of Electrical Engineering and Biomedical Engineering, Columbia University, New York, New York, USA.
| | | | | |
Collapse
|
241
|
Rabconnectin3α promotes stable activity of the H+ pump on synaptic vesicles in hair cells. J Neurosci 2012; 32:11144-56. [PMID: 22875945 DOI: 10.1523/jneurosci.1705-12.2012] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Acidification of synaptic vesicles relies on the vacuolar-type ATPase (V-ATPase) and provides the electrochemical driving force for neurotransmitter exchange. The regulatory mechanisms that ensure assembly of the V-ATPase holoenzyme on synaptic vesicles are unknown. Rabconnectin3α (Rbc3α) is a potential candidate for regulation of V-ATPase activity because of its association with synaptic vesicles and its requirement for acidification of intracellular compartments. Here, we provide the first evidence for a role of Rbc3α in synaptic vesicle acidification and neurotransmission. In this study, we characterized mutant alleles of rbc3α isolated from a large-scale screen for zebrafish with auditory/vestibular defects. We show that Rbc3α is localized to basal regions of hair cells in which synaptic vesicles are present. To determine whether Rbc3α regulates V-ATPase activity, we examined the acidification of synaptic vesicles and localization of the V-ATPase in hair cells. In contrast to wild-type hair cells, we observed that synaptic vesicles had elevated pH, and a cytosolic subunit of the V-ATPase was no longer enriched in synaptic regions of mutant hair cells. As a consequence of defective acidification of synaptic vesicles, afferent neurons in rbc3α mutants had reduced firing rates and reduced accuracy of phase-locked action potentials in response to mechanical stimulation of hair cells. Collectively, our data suggest that Rbc3α modulates synaptic transmission in hair cells by promoting V-ATPase activity in synaptic vesicles.
Collapse
|
242
|
Magariños M, Contreras J, Aburto MR, Varela-Nieto I. Early development of the vertebrate inner ear. Anat Rec (Hoboken) 2012; 295:1775-90. [PMID: 23044927 DOI: 10.1002/ar.22575] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 07/24/2012] [Indexed: 12/12/2022]
Abstract
This is a review of the biological processes and the main signaling pathways required to generate the different otic cell types, with particular emphasis on the actions of insulin-like growth factor I. The sensory organs responsible of hearing and balance have a common embryonic origin in the otic placode. Lineages of neural, sensory, and support cells are generated from common otic neuroepithelial progenitors. The sequential generation of the cell types that will form the adult inner ear requires the coordination of cell proliferation with cell differentiation programs, the strict regulation of cell survival, and the metabolic homeostasis of otic precursors. A network of intracellular signals operates to coordinate the transcriptional response to the extracellular input. Understanding the molecular clues that direct otic development is fundamental for the design of novel treatments for the protection and repair of hearing loss and balance disorders.
Collapse
Affiliation(s)
- Marta Magariños
- Instituto de Investigaciones Biomédicas, Alberto Sols, CSIC-UAM, Madrid, Spain
| | | | | | | |
Collapse
|
243
|
Water channel proteins in the inner ear and their link to hearing impairment and deafness. Mol Aspects Med 2012; 33:612-37. [DOI: 10.1016/j.mam.2012.06.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 06/11/2012] [Accepted: 06/17/2012] [Indexed: 11/24/2022]
|
244
|
Mammano F. ATP-dependent intercellular Ca2+ signaling in the developing cochlea: facts, fantasies and perspectives. Semin Cell Dev Biol 2012; 24:31-9. [PMID: 23022499 DOI: 10.1016/j.semcdb.2012.09.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 09/14/2012] [Indexed: 10/27/2022]
Abstract
Hearing relies on a sensitive mechanoelectrical transduction process in the cochlea of the inner ear. The cochlea contains sensory, secretory, neural, supporting and epithelial cells which are all essential to the sound transduction process. It is well known that a complex extracellular purinergic signaling system contributes to cochlear homeostasis, altering cochlear sensitivity and neural output via ATP-gated ion channels (P2X receptors) and G protein-coupled P2Y receptors. This review focuses on the emerging roles of ATP that are currently under investigation in the developing sensory epithelium, with particular emphasis on the link between ATP release, Ca(2+) signaling, the expression and function of gap junction proteins connexin26 and connexin30, and the acquisition of hearing.
Collapse
Affiliation(s)
- Fabio Mammano
- Dipartimento di Fisica e Astronomia G. Galilei, Università di Padova, 35131 Padova, Italy.
| |
Collapse
|
245
|
Curhan SG, Shargorodsky J, Eavey R, Curhan GC. Analgesic use and the risk of hearing loss in women. Am J Epidemiol 2012; 176:544-54. [PMID: 22933387 DOI: 10.1093/aje/kws146] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Use of analgesics is common and is associated with increased risk of hearing loss in men; however, the relation has not been examined prospectively in women. The authors prospectively examined the relation between frequency of aspirin, ibuprofen, and acetaminophen use and risk of hearing loss among 62,261 women aged 31-48 years at baseline (1995) in Nurses' Health Study II. The outcome was self-reported hearing loss (n = 10,012), and the follow-up period was 1995-2009. Cox proportional hazards regression was used to adjust for potential confounders. During 764,247 person-years of follow-up, ibuprofen use and acetaminophen use were independently associated with increased risk of hearing loss, but aspirin use was not. For ibuprofen, the multivariate-adjusted relative risk of hearing loss was 1.13 (95% confidence interval (CI): 1.06, 1.19) for use 2-3 days/week, 1.21 (95% CI: 1.11, 1.32) for use 4-5 days/week, and 1.24 (95% CI: 1.14, 1.35) for use ≥6 days/week (P-trend < 0.0001), compared with use less than once per week. For acetaminophen, the corresponding relative risks were 1.11 (95% CI: 1.02, 1.19), 1.21 (95% CI: 1.07, 1.37), and 1.08 (95% CI: 0.95, 1.22), respectively (P-trend = 0.0007). In this study, use of ibuprofen or acetaminophen (but not aspirin) 2 or more days per week was associated with an increased risk of hearing loss in women.
Collapse
Affiliation(s)
- Sharon G Curhan
- Channing Laboratory, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts 02115, USA.
| | | | | | | |
Collapse
|
246
|
Mistrík P, Daudet N, Morandell K, Ashmore JF. Mammalian prestin is a weak Cl⁻/HCO₃⁻ electrogenic antiporter. J Physiol 2012; 590:5597-610. [PMID: 22890707 DOI: 10.1113/jphysiol.2012.241448] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The lateral membrane of mammalian cochlear outer hair cells contains prestin, a protein which can act as a fast voltage-driven actuator responsible for electromotility and enhanced sensitivity to sound. The protein belongs to the SLC26 family of transporters whose members are characterised as able to exchange halides for SO(4)(2-) or HCO(3)(-) yet previous analyses of mammalian prestin have suggested that such exchange functions were minimal. Here anion transport is investigated both in guinea-pig outer hair cells (OHCs) and in an expression system where we employ a sensitive intracellular pH (pH(i)) probe, pHluorin, to report HCO(3)(-) transport and to monitor the small pH(i) changes observable in the cells. In the presence of extracellular HCO(3)(-), pH(i) recovered from an acid load 4 times faster in prestin-transfected cells. The acceleration required a chloride gradient established by reducing extracellular chloride to 2 mm. Similar results were also shown using BCECF as an alternative pH(i) sensor, but with recovery only found in those cells expressing prestin. Simultaneous electrophysiological recording of the transfected cells during bicarbonate exposure produced a shift in the reversal potential to more negative potentials, consistent with electrogenic transport. These data therefore suggest that prestin can act as a weak Cl(-)/HCO(3)(-) antiporter and it is proposed that, in addition to participating in wide band cochlear sound amplification, prestin may also be involved in the slow time scale (>10 s) phenomena where changes in cell stiffness and internal pressure have been implicated. The results show the importance of considering the effects of the endogenous bicarbonate buffering system in evaluating the function of prestin in cochlear outer hair cells.
Collapse
Affiliation(s)
- P Mistrík
- UCL Ear Institute, 332 Gray’s Inn Road, London WC1E 6BT, UK
| | | | | | | |
Collapse
|
247
|
Eckhard A, Gleiser C, Rask-Andersen H, Arnold H, Liu W, Mack A, Müller M, Löwenheim H, Hirt B. Co-localisation of Kir4.1 and AQP4 in rat and human cochleae reveals a gap in water channel expression at the transduction sites of endocochlear K+ recycling routes. Cell Tissue Res 2012; 350:27-43. [DOI: 10.1007/s00441-012-1456-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 05/22/2012] [Indexed: 11/30/2022]
|
248
|
Powers TR, Virk SM, Trujillo-Provencio C, Serrano EE. Probing the Xenopus laevis inner ear transcriptome for biological function. BMC Genomics 2012; 13:225. [PMID: 22676585 PMCID: PMC3532188 DOI: 10.1186/1471-2164-13-225] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 05/21/2012] [Indexed: 01/27/2023] Open
Abstract
Background The senses of hearing and balance depend upon mechanoreception, a process that originates in the inner ear and shares features across species. Amphibians have been widely used for physiological studies of mechanotransduction by sensory hair cells. In contrast, much less is known of the genetic basis of auditory and vestibular function in this class of animals. Among amphibians, the genus Xenopus is a well-characterized genetic and developmental model that offers unique opportunities for inner ear research because of the amphibian capacity for tissue and organ regeneration. For these reasons, we implemented a functional genomics approach as a means to undertake a large-scale analysis of the Xenopus laevis inner ear transcriptome through microarray analysis. Results Microarray analysis uncovered genes within the X. laevis inner ear transcriptome associated with inner ear function and impairment in other organisms, thereby supporting the inclusion of Xenopus in cross-species genetic studies of the inner ear. The use of gene categories (inner ear tissue; deafness; ion channels; ion transporters; transcription factors) facilitated the assignment of functional significance to probe set identifiers. We enhanced the biological relevance of our microarray data by using a variety of curation approaches to increase the annotation of the Affymetrix GeneChip® Xenopus laevis Genome array. In addition, annotation analysis revealed the prevalence of inner ear transcripts represented by probe set identifiers that lack functional characterization. Conclusions We identified an abundance of targets for genetic analysis of auditory and vestibular function. The orthologues to human genes with known inner ear function and the highly expressed transcripts that lack annotation are particularly interesting candidates for future analyses. We used informatics approaches to impart biologically relevant information to the Xenopus inner ear transcriptome, thereby addressing the impediment imposed by insufficient gene annotation. These findings heighten the relevance of Xenopus as a model organism for genetic investigations of inner ear organogenesis, morphogenesis, and regeneration.
Collapse
Affiliation(s)
- TuShun R Powers
- Biology Department, New Mexico State University, Las Cruces, USA
| | | | | | | |
Collapse
|
249
|
Heinrich UR, Helling K. Nitric oxide--a versatile key player in cochlear function and hearing disorders. Nitric Oxide 2012; 27:106-16. [PMID: 22659013 DOI: 10.1016/j.niox.2012.05.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 05/10/2012] [Accepted: 05/24/2012] [Indexed: 12/13/2022]
Abstract
Nitric oxide (NO) is a signaling molecule which can generally be formed by three nitric oxide synthases (NOS). Two of them, the endothelial nitric oxide synthase (eNOS) and the neural nitric oxide synthase (nNOS), are calcium/calmodulin-dependent and constitutively expressed in many cell types. Both isoforms are found in the vertebrate cochlea. The inducible nitric oxide synthase (iNOS) is independent of calcium and normally not detectable in the un-stimulated cochlea. In the inner ear, as in other tissues, NO was identified as a multitask molecule involved in various processes such as neurotransmission and neuromodulation. In addition, increasing evidence demonstrates that the NO-dependent processes of cell protection or, alternatively, cell destruction seem to depend, among other things, on changes in the local cochlear NO-concentration. These alterations can occur at the cellular level or within a distinct cell population both leading to an NO-imbalance within the hearing organ. This dysfunction can result in hearing loss or even in deafness. In cases of cochlear malfunction, regulatory systems such as the gap junction system, the blood vessels or the synaptic region might be affected temporarily or permanently by an altered NO-level. This review discusses potential cellular mechanisms how NO might contribute to different forms of hearing disorders. Approaches of NO-reduction are evaluated and the transfer of results obtained from experimental animal models to human medication is discussed.
Collapse
Affiliation(s)
- Ulf-Rüdiger Heinrich
- Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center of The Johannes Gutenberg-University Mainz, Germany.
| | | |
Collapse
|
250
|
Computational model of a circulation current that controls electrochemical properties in the mammalian cochlea. Proc Natl Acad Sci U S A 2012; 109:9191-6. [PMID: 22619324 DOI: 10.1073/pnas.1120067109] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Sound-evoked mechanical stimuli permit endolymphatic K(+) to enter sensory hair cells. This transduction is sensitized by an endocochlear potential (EP) of +80 mV in endolymph. After depolarizing the cells, K(+) leaves hair cells in perilymph, and it is then circulated back to endolymph across the lateral cochlear wall. In theory, this process entails a continuous and unidirectional current carried by apical K(+) channels and basolateral K(+) uptake transporters in both the marginal cell and syncytial layers of the lateral wall. The transporters regulate intracellular and extracellular [K(+)], allowing the channels to form K(+) diffusion potentials across each of the two layers. These diffusion potentials govern the EP. What remains uncertain is whether these transport mechanisms accumulating across diverse cell layers make up a continuous circulation current in the lateral wall and how this current might affect the characteristics of the endolymph. To address this question, we developed an electrophysiological model that incorporates channels and transporters of the lateral wall and channels of hair cells that derive a circulation current. The simulation replicated normal experimental EP values and reproduced experimentally measured changes in the EP and intra- and extracellular [K(+)] in the lateral wall when different transporters and channels were blocked. The model predicts that, under these different conditions, the circulation current's contribution to the EP arises from different sources. Finally, our model also accurately simulated EP loss in a mouse model of a chloride channelopathy associated with deafness.
Collapse
|