1
|
Rose KP, Manilla G, Milon B, Zalzman O, Song Y, Coate TM, Hertzano R. Spatially distinct otic mesenchyme cells show molecular and functional heterogeneity patterns before hearing onset. iScience 2023; 26:107769. [PMID: 37720106 PMCID: PMC10502415 DOI: 10.1016/j.isci.2023.107769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 06/29/2023] [Accepted: 08/25/2023] [Indexed: 09/19/2023] Open
Abstract
The cochlea consists of diverse cellular populations working in harmony to convert mechanical stimuli into electrical signals for the perception of sound. Otic mesenchyme cells (OMCs), often considered a homogeneous cell type, are essential for normal cochlear development and hearing. Despite being the most numerous cell type in the developing cochlea, OMCs are poorly understood. OMCs are known to differentiate into spatially and functionally distinct cell types, including fibrocytes of the lateral wall and spiral limbus, modiolar osteoblasts, and specialized tympanic border cells of the basilar membrane. Here, we show that OMCs are transcriptionally and functionally heterogeneous and can be divided into four distinct populations that spatially correspond to OMC-derived cochlear structures. We also show that this heterogeneity and complexity of OMCs commences during early phases of cochlear development. Finally, we describe the cell-cell communication network of the developing cochlea, inferring a major role for OMC in outgoing signaling.
Collapse
Affiliation(s)
- Kevin P. Rose
- Neurotology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gabriella Manilla
- Neurotology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Beatrice Milon
- Neurotology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ori Zalzman
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yang Song
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Thomas M. Coate
- Department of Biology, Georgetown University, Washington, DC 20007, USA
| | - Ronna Hertzano
- Neurotology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Otorhinolaryngology Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
2
|
Bernardinelli E, Huber F, Roesch S, Dossena S. Clinical and Molecular Aspects Associated with Defects in the Transcription Factor POU3F4: A Review. Biomedicines 2023; 11:1695. [PMID: 37371790 DOI: 10.3390/biomedicines11061695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
X-linked deafness (DFNX) is estimated to account for up to 2% of cases of hereditary hearing loss and occurs in both syndromic and non-syndromic forms. POU3F4 is the gene most commonly associated with X-linked deafness (DFNX2, DFN3) and accounts for about 50% of the cases of X-linked non-syndromic hearing loss. This gene codes for a transcription factor of the POU family that plays a major role in the development of the middle and inner ear. The clinical features of POU3F4-related hearing loss include a pathognomonic malformation of the inner ear defined as incomplete partition of the cochlea type 3 (IP-III). Often, a perilymphatic gusher is observed upon stapedectomy during surgery, possibly as a consequence of an incomplete separation of the cochlea from the internal auditory canal. Here we present an overview of the pathogenic gene variants of POU3F4 reported in the literature and discuss the associated clinical features, including hearing loss combined with additional phenotypes such as cognitive and motor developmental delays. Research on the transcriptional targets of POU3F4 in the ear and brain is in its early stages and is expected to greatly advance our understanding of the pathophysiology of POU3F4-linked hearing loss.
Collapse
Affiliation(s)
- Emanuele Bernardinelli
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Florian Huber
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Sebastian Roesch
- Department of Otorhinolaryngology, Head and Neck Surgery, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Silvia Dossena
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, 5020 Salzburg, Austria
| |
Collapse
|
3
|
Bernardinelli E, Roesch S, Simoni E, Marino A, Rasp G, Astolfi L, Sarikas A, Dossena S. Novel POU3F4 variants identified in patients with inner ear malformations exhibit aberrant cellular distribution and lack of SLC6A20 transcriptional upregulation. Front Mol Neurosci 2022; 15:999833. [PMID: 36245926 PMCID: PMC9558712 DOI: 10.3389/fnmol.2022.999833] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/13/2022] [Indexed: 11/14/2022] Open
Abstract
Hearing loss (HL) is the most common sensory defect and affects 450 million people worldwide in a disabling form. Pathogenic sequence alterations in the POU3F4 gene, which encodes a transcription factor, are causative of the most common type of X-linked deafness (X-linked deafness type 3, DFN3, DFNX2). POU3F4-related deafness is characterized by a typical inner ear malformation, namely an incomplete partition of the cochlea type 3 (IP3), with or without an enlargement of the vestibular aqueduct (EVA). The pathomechanism underlying POU3F4-related deafness and the corresponding transcriptional targets are largely uncharacterized. Two male patients belonging to a Caucasian cohort with HL and EVA who presented with an IP3 were submitted to genetic analysis. Two novel sequence variants in POU3F4 were identified by Sanger sequencing. In cell-based assays, the corresponding protein variants (p.S74Afs*8 and p.C327*) showed an aberrant expression and subcellular distribution and lack of transcriptional activity. These two protein variants failed to upregulate the transcript levels of the amino acid transporter gene SLC6A20, which was identified as a novel transcriptional target of POU3F4 by RNA sequencing and RT-qPCR. Accordingly, POU3F4 silencing by siRNA resulted in downregulation of SLC6A20 in mouse embryonic fibroblasts. Moreover, we showed for the first time that SLC6A20 is expressed in the mouse cochlea, and co-localized with POU3F4 in the spiral ligament. The findings presented here point to a novel role of amino acid transporters in the inner ear and pave the way for mechanistic studies of POU3F4-related HL.
Collapse
Affiliation(s)
- Emanuele Bernardinelli
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, Salzburg, Austria
| | - Sebastian Roesch
- Department of Otorhinolaryngology, Head and Neck Surgery, Paracelsus Medical University, Salzburg, Austria
| | - Edi Simoni
- Bioacoustic Research Laboratory, Department of Neuroscience, Biomedical Campus Pietro d’Abano, University of Padua, Padua, Italy
| | - Angela Marino
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, Messina, Italy
| | - Gerd Rasp
- Department of Otorhinolaryngology, Head and Neck Surgery, Paracelsus Medical University, Salzburg, Austria
| | - Laura Astolfi
- Bioacoustic Research Laboratory, Department of Neuroscience, Biomedical Campus Pietro d’Abano, University of Padua, Padua, Italy
- Interdepartmental Research Center of International Auditory Processing Project in Venice (I-APPROVE), Department of Neurosciences, University of Padova, Santi Giovanni e Paolo Hospital, ULSS3, Venice, Italy
| | - Antonio Sarikas
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, Salzburg, Austria
- *Correspondence: Antonio Sarikas,
| | - Silvia Dossena
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, Salzburg, Austria
- Silvia Dossena,
| |
Collapse
|
4
|
Defourny J. Considering gene therapy to protect from X-linked deafness DFNX2 and associated neurodevelopmental disorders. IBRAIN 2022; 8:431-441. [PMID: 37786584 PMCID: PMC10529175 DOI: 10.1002/ibra.12068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 08/10/2022] [Accepted: 09/13/2022] [Indexed: 10/04/2023]
Abstract
Mutations and deletions in the gene or upstream of the gene encoding the POU3F4 transcription factor cause X-linked progressive deafness DFNX2 and additional neurodevelopmental disorders in humans. Hearing loss can be purely sensorineural or mixed, that is, with both conductive and sensorineural components. Affected males show anatomical abnormalities of the inner ear, which are jointly defined as incomplete partition type III. Current approaches to improve hearing and speech skills of DFNX2 patients do not seem to be fully effective. Owing to inner ear malformations, cochlear implantation is surgically difficult and may predispose towards severe complications. Even in cases where implantation is safely performed, hearing and speech outcomes remain highly variable among patients. Mouse models for DFNX2 deafness revealed that sensorineural loss could arise from a dysfunction of spiral ligament fibrocytes in the lateral wall of the cochlea, which leads to reduced endocochlear potential. Highly positive endocochlear potential is critical for sensory hair cell mechanotransduction and hearing. In this context, here, we propose to develop a therapeutic approach in male Pou3f4 -/y mice based on an adeno-associated viral (AAV) vector-mediated gene transfer in cochlear spiral ligament fibrocytes. Among a broad range of AAV vectors, AAV7 was found to show a strong tropism for the spiral ligament. Thus, we suggest that an AAV7-mediated delivery of Pou3f4 complementary DNA in the spiral ligament of Pou3f4 -/y mice could represent an attractive strategy to prevent fibrocyte degeneration and to restore normal cochlear functions and properties, including a positive endocochlear potential, before hearing loss progresses to profound deafness.
Collapse
Affiliation(s)
- Jean Defourny
- GIGA‐Neurosciences, Unit of Cell and Tissue BiologyUniversity of Liège, C.H.U. B36LiègeBelgium
| |
Collapse
|
5
|
Genetic Load of Alternations of Transcription Factor Genes in Non-Syndromic Deafness and the Associated Clinical Phenotypes: Experience from Two Tertiary Referral Centers. Biomedicines 2022; 10:biomedicines10092125. [PMID: 36140227 PMCID: PMC9495667 DOI: 10.3390/biomedicines10092125] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/18/2022] [Accepted: 08/25/2022] [Indexed: 11/30/2022] Open
Abstract
Sensorineural hearing loss is one of the most common inherited sensory disorders. Functional classifications of deafness genes have shed light on genotype- and mechanism-based pharmacological approaches and on gene therapy strategies. In this study, we characterized the clinical phenotypes and genotypes of non-syndromic deafness caused by transcription factor (TF) gene variants, one of the functional classifications of genetic hearing loss. Of 1280 probands whose genomic DNA was subjected to molecular genetic testing, TF genes were responsible for hearing loss in 2.6%. Thirty-three pathogenic variants, including nine novel variants, accounting for non-syndromic deafness were clustered in only four TF genes (POU3F4, POU4F3, LMX1A, and EYA4), which is indicative of a narrow molecular etiologic spectrum of TF genes, and the functional redundancy of many other TF genes, in the context of non-syndromic deafness. The audiological and radiological characteristics associated with the four TF genes differed significantly, with a wide phenotypic spectrum. The results of this study reveal the genetic load of TF gene alterations among a cohort with non-syndromic hearing loss. Additionally, we have further refined the clinical profiles associated with TF gene variants as a basis for a personalized, genetically tailored approach to audiological rehabilitation.
Collapse
|
6
|
Early S, Du E, Boussaty E, Friedman R. Genetics of noise-induced hearing loss in the mouse model. Hear Res 2022; 425:108505. [DOI: 10.1016/j.heares.2022.108505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 03/28/2022] [Accepted: 04/07/2022] [Indexed: 12/01/2022]
|
7
|
Zhang L, Chen S, Sun Y. Mechanism and Prevention of Spiral Ganglion Neuron Degeneration in the Cochlea. Front Cell Neurosci 2022; 15:814891. [PMID: 35069120 PMCID: PMC8766678 DOI: 10.3389/fncel.2021.814891] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 12/09/2021] [Indexed: 12/14/2022] Open
Abstract
Sensorineural hearing loss (SNHL) is one of the most prevalent sensory deficits in humans, and approximately 360 million people worldwide are affected. The current treatment option for severe to profound hearing loss is cochlear implantation (CI), but its treatment efficacy is related to the survival of spiral ganglion neurons (SGNs). SGNs are the primary sensory neurons, transmitting complex acoustic information from hair cells to second-order sensory neurons in the cochlear nucleus. In mammals, SGNs have very limited regeneration ability, and SGN loss causes irreversible hearing loss. In most cases of SNHL, SGN damage is the dominant pathogenesis, and it could be caused by noise exposure, ototoxic drugs, hereditary defects, presbycusis, etc. Tremendous efforts have been made to identify novel treatments to prevent or reverse the damage to SGNs, including gene therapy and stem cell therapy. This review summarizes the major causes and the corresponding mechanisms of SGN loss and the current protection strategies, especially gene therapy and stem cell therapy, to promote the development of new therapeutic methods.
Collapse
Affiliation(s)
- Li Zhang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sen Chen
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Sun
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Otorhinolaryngology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
The effects of substrate composition and topography on the characteristics and growth of cell cultures of cochlear fibrocytes. Hear Res 2021; 415:108427. [PMID: 34999290 DOI: 10.1016/j.heares.2021.108427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 12/02/2021] [Accepted: 12/23/2021] [Indexed: 11/22/2022]
Abstract
Spiral ligament fibrocytes of the cochlea play homoeostatic roles in hearing and their degeneration contributes to hearing loss. Culturing fibrocytes in vitro provides a way to evaluate their functional characteristics and study possible therapies for hearing loss. We investigated whether in vivo characteristics of fibrocytes could be recapitulated in vitro by modifying the culture substrates and carried out proof of concept studies for potential transplantation of culture cells into the inner ear. Fibrocytes cultured from 4 to 5-week old CD/1 mice were grown on 2D substrates coated with collagen I, II, V or IX and, after harvesting, onto or into 3D substrates (hydrogels) of collagen I alone or mixed collagen I and II at a 1:1 ratio. We also assessed magnetic nanoparticle (MNP) uptake. Cell counts, immunohistochemical and ultrastructural studies showed that fibrocytes grown on 2D substrates proliferated, formed both small spindle-shaped and large flat cells that avidly took up MNPs. Of the different collagen coatings, only collagen II had an effect, causing a reduced size of the larger cells. On hydrogels, the cells were plump/rounded with extended processes, resembling native cells. They formed networks over the surface and became incorporated into the gel. In all culture formats, the majority co-expressed caldesmon, aquaporin 1, S-100 and sodium potassium ATPase, indicating a mixed or uncharacterised phenotype. Time-course experiments showed a decrease to ∼50% of the starting population by 4d after seeding on collagen I hydrogels, but better survival (∼60%) was found on collagen I + II gels, whilst TEM revealed the presence of apoptotic cells. Cells grown within gels additionally showed necrosis. These results demonstrate that fibrocytes grown in 3D recapitulate in vivo morphology of native fibrocytes, but have poorer survival, compared with 2D. Therefore hydrogel cultures could be used to study fibrocyte function and might also offer avenues for cell-replacement therapies, but need more optimization for therapeutic use. Fibrocyte function could be modified using MNPs in combination, for example, with gene transfection.
Collapse
|
9
|
Roesch S, Rasp G, Sarikas A, Dossena S. Genetic Determinants of Non-Syndromic Enlarged Vestibular Aqueduct: A Review. Audiol Res 2021; 11:423-442. [PMID: 34562878 PMCID: PMC8482117 DOI: 10.3390/audiolres11030040] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/02/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022] Open
Abstract
Hearing loss is the most common sensorial deficit in humans and one of the most common birth defects. In developed countries, at least 60% of cases of hearing loss are of genetic origin and may arise from pathogenic sequence alterations in one of more than 300 genes known to be involved in the hearing function. Hearing loss of genetic origin is frequently associated with inner ear malformations; of these, the most commonly detected is the enlarged vestibular aqueduct (EVA). EVA may be associated to other cochleovestibular malformations, such as cochlear incomplete partitions, and can be found in syndromic as well as non-syndromic forms of hearing loss. Genes that have been linked to non-syndromic EVA are SLC26A4, GJB2, FOXI1, KCNJ10, and POU3F4. SLC26A4 and FOXI1 are also involved in determining syndromic forms of hearing loss with EVA, which are Pendred syndrome and distal renal tubular acidosis with deafness, respectively. In Caucasian cohorts, approximately 50% of cases of non-syndromic EVA are linked to SLC26A4 and a large fraction of patients remain undiagnosed, thus providing a strong imperative to further explore the etiology of this condition.
Collapse
Affiliation(s)
- Sebastian Roesch
- Department of Otorhinolaryngology, Head and Neck Surgery, Paracelsus Medical University, 5020 Salzburg, Austria; (S.R.); (G.R.)
| | - Gerd Rasp
- Department of Otorhinolaryngology, Head and Neck Surgery, Paracelsus Medical University, 5020 Salzburg, Austria; (S.R.); (G.R.)
| | - Antonio Sarikas
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, 5020 Salzburg, Austria;
| | - Silvia Dossena
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, 5020 Salzburg, Austria;
- Correspondence: ; Tel.: +43-(0)662-2420-80564
| |
Collapse
|
10
|
X-linked Malformation Deafness: Neurodevelopmental Symptoms Are Common in Children With IP3 Malformation and Mutation in POU3F4. Ear Hear 2021; 43:53-69. [PMID: 34133399 PMCID: PMC8694264 DOI: 10.1097/aud.0000000000001073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Supplemental Digital Content is available in the text. Incomplete partition type 3 (IP3) malformation deafness is a rare hereditary cause of congenital or rapid progressive hearing loss. The children present with a severe to profound mixed hearing loss and temporal bone imaging show a typical inner ear malformation classified as IP3. Cochlear implantation is one option of hearing restoration in severe cases. Little is known about other specific difficulties these children might exhibit, for instance possible neurodevelopmental symptoms.
Collapse
|
11
|
Eckert MA, Harris KC, Lang H, Lewis MA, Schmiedt RA, Schulte BA, Steel KP, Vaden KI, Dubno JR. Translational and interdisciplinary insights into presbyacusis: A multidimensional disease. Hear Res 2021; 402:108109. [PMID: 33189490 PMCID: PMC7927149 DOI: 10.1016/j.heares.2020.108109] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 10/19/2020] [Accepted: 10/25/2020] [Indexed: 12/18/2022]
Abstract
There are multiple etiologies and phenotypes of age-related hearing loss or presbyacusis. In this review we summarize findings from animal and human studies of presbyacusis, including those that provide the theoretical framework for distinct metabolic, sensory, and neural presbyacusis phenotypes. A key finding in quiet-aged animals is a decline in the endocochlear potential (EP) that results in elevated pure-tone thresholds across frequencies with greater losses at higher frequencies. In contrast, sensory presbyacusis appears to derive, in part, from acute and cumulative effects on hair cells of a lifetime of environmental exposures (e.g., noise), which often result in pronounced high frequency hearing loss. These patterns of hearing loss in animals are recognizable in the human audiogram and can be classified into metabolic and sensory presbyacusis phenotypes, as well as a mixed metabolic+sensory phenotype. However, the audiogram does not fully characterize age-related changes in auditory function. Along with the effects of peripheral auditory system declines on the auditory nerve, primary degeneration in the spiral ganglion also appears to contribute to central auditory system aging. These inner ear alterations often correlate with structural and functional changes throughout the central nervous system and may explain suprathreshold speech communication difficulties in older adults with hearing loss. Throughout this review we highlight potential methods and research directions, with the goal of advancing our understanding, prevention, diagnosis, and treatment of presbyacusis.
Collapse
Affiliation(s)
- Mark A Eckert
- Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA.
| | - Kelly C Harris
- Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA
| | - Hainan Lang
- Medical University of South Carolina, Department of Pathology and Laboratory Medicine, Charleston, SC 29425, USA
| | - Morag A Lewis
- King's College London, Wolfson Centre for Age-Related Diseases, London SE1 1UL, United Kingdom
| | - Richard A Schmiedt
- Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA
| | - Bradley A Schulte
- Medical University of South Carolina, Department of Pathology and Laboratory Medicine, Charleston, SC 29425, USA; Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA
| | - Karen P Steel
- King's College London, Wolfson Centre for Age-Related Diseases, London SE1 1UL, United Kingdom
| | - Kenneth I Vaden
- Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA
| | - Judy R Dubno
- Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA; Medical University of South Carolina, Department of Pathology and Laboratory Medicine, Charleston, SC 29425, USA
| |
Collapse
|
12
|
Peeleman N, Verdoodt D, Ponsaerts P, Van Rompaey V. On the Role of Fibrocytes and the Extracellular Matrix in the Physiology and Pathophysiology of the Spiral Ligament. Front Neurol 2020; 11:580639. [PMID: 33193034 PMCID: PMC7653186 DOI: 10.3389/fneur.2020.580639] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/10/2020] [Indexed: 12/19/2022] Open
Abstract
The spiral ligament in the cochlea has been suggested to play a significant role in the pathophysiology of different etiologies of strial hearing loss. Spiral ligament fibrocytes (SLFs), the main cell type in the lateral wall, are crucial in maintaining the endocochlear potential and regulating blood flow. SLF dysfunction can therefore cause cochlear dysfunction and thus hearing impairment. Recent studies have highlighted the role of SLFs in the immune response of the cochlea. In contrast to sensory cells in the inner ear, SLFs (more specifically type III fibrocytes) have also demonstrated the ability to regenerate after different types of trauma such as drug toxicity and noise. SLFs are responsible for producing proteins, such as collagen and cochlin, that create an adequate extracellular matrix to thrive in. Any dysfunction of SLFs or structural changes to the extracellular matrix can significantly impact hearing function. However, SLFs may prove useful in restoring hearing by their potential to regenerate cells in the spiral ligament.
Collapse
Affiliation(s)
- Noa Peeleman
- Department of Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Dorien Verdoodt
- Department of Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Vincent Van Rompaey
- Department of Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Department of Otorhinolaryngology and Head and Neck Surgery, Antwerp University Hospital, Edegem, Belgium
| |
Collapse
|
13
|
Petrina NE, Marakhonov AV, Zinchenko RA. [Presentation of a rare case of hereditary hearing loss with X-linked recessive inheritance associated with the POU3F4 gene]. Vestn Otorinolaringol 2020; 85:65-69. [PMID: 32885640 DOI: 10.17116/otorino20208504165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Congenital hearing loss is one of the most frequent inherited human pathologies, occurring in 1-2 out of 1000 newborns. X-linked hearing loss occurs in 1-5% of all congenital hearing impairments. The proband (a man) and his affected brother have profound prelingual non-syndromic neurosensory hearing loss. Their parents are healthy. The aim of the study was to determine the cause of hearing loss in a given family and to assess the population frequency of the revealed pathogenic genetic variant. NGS analysis identified a pathogenic variant c.907C>T (p.Pro303Ser) in the POU3F4 gene mapped to the Xq21.1 locus. This is the second case of X-linked hearing loss (DFNX2, OMIM 304400) in Europe, caused by the c.907C>T variant in the POU3F4 gene. DFNX2-hearing loss is manifested with abnormalities of the inner ear, predisposing to the "gusher effect" - otoliquorrhea during stapedoplasty. The brother was diagnosed with a c.907C>T variant in the POU3F4 gene in the hemizygous state while in their mother - in the heterozygous state. Their father had no variant c.907C>T. Molecular genetic analysis showed that the genetic variant c.907C>T was not detected in the control sample of healthy female from the Nogai population, which suggests its low frequency in the population.
Collapse
Affiliation(s)
- N E Petrina
- Research Center for Medical Genetics, Moscow, Russia
| | | | - R A Zinchenko
- Research Center for Medical Genetics, Moscow, Russia.,N.A. Semashko National Research Institute of Public Health, Moscow, Russia
| |
Collapse
|
14
|
Brooks PM, Rose KP, MacRae ML, Rangoussis KM, Gurjar M, Hertzano R, Coate TM. Pou3f4-expressing otic mesenchyme cells promote spiral ganglion neuron survival in the postnatal mouse cochlea. J Comp Neurol 2020; 528:1967-1985. [PMID: 31994726 DOI: 10.1002/cne.24867] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/13/2020] [Accepted: 01/16/2020] [Indexed: 12/20/2022]
Abstract
During inner ear development, primary auditory neurons named spiral ganglion neurons (SGNs) are surrounded by otic mesenchyme cells, which express the transcription factor Pou3f4. Mutations in Pou3f4 are associated with DFNX2, the most common form of X-linked deafness and typically include developmental malformations of the middle ear and inner ear. It is known that interactions between Pou3f4-expressing mesenchyme cells and SGNs are important for proper axon bundling during development. However, Pou3f4 continues to be expressed through later phases of development, and potential interactions between Pou3f4 and SGNs during this period had not been explored. To address this, we documented Pou3f4 protein expression in the early postnatal mouse cochlea and compared SGNs in Pou3f4 knockout mice and littermate controls. In Pou3f4y/- mice, SGN density begins to decline by the end of the first postnatal week, with approximately 25% of SGNs ultimately lost. This period of SGN loss in Pou3f4y/- cochleae coincides with significant elevations in SGN apoptosis. Interestingly, this period also coincides with the presence of a transient population of Pou3f4-expressing cells around and within the spiral ganglion. To determine if Pou3f4 is normally required for SGN peripheral axon extension into the sensory domain, we used a genetic sparse labeling approach to track SGNs and found no differences compared with controls. We also found that Pou3f4 loss did not lead to changes in the proportions of Type I SGN subtypes. Overall, these data suggest that otic mesenchyme cells may play a role in maintaining SGN populations during the early postnatal period.
Collapse
Affiliation(s)
- Paige M Brooks
- Department of Biology, Georgetown University, Washington, District of Columbia
| | - Kevin P Rose
- Department of Otorhinolaryngology Head and Neck Surgery, University of Maryland School of Medicine, University of Maryland, Baltimore, Maryland
| | - Meaghan L MacRae
- Department of Biology, Georgetown University, Washington, District of Columbia
| | | | - Mansa Gurjar
- Department of Biology, Georgetown University, Washington, District of Columbia
| | - Ronna Hertzano
- Department of Otorhinolaryngology Head and Neck Surgery, University of Maryland School of Medicine, University of Maryland, Baltimore, Maryland.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, University of Maryland, Baltimore, Maryland.,Institute for Genome Sciences, University of Maryland School of Medicine, University of Maryland, Baltimore, Maryland
| | - Thomas M Coate
- Department of Biology, Georgetown University, Washington, District of Columbia
| |
Collapse
|
15
|
Giannantonio S, Agolini E, Scorpecci A, Anzivino R, Bellacchio E, Cocciadiferro D, Novelli A, Digilio MC, Marsella P. Genetic identification and molecular modeling characterization of a novel POU3F4 variant in two Italian deaf brothers. Int J Pediatr Otorhinolaryngol 2020; 129:109790. [PMID: 31786483 DOI: 10.1016/j.ijporl.2019.109790] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 11/18/2019] [Accepted: 11/19/2019] [Indexed: 11/27/2022]
Abstract
In this report, we describe a novel, probably pathogenic hemizygous variant c.870G > T (p.Lys290Asn) in the POU3F4 gene in two deaf brothers from one Italian family with identical inner ear abnormalities specific to X-linked deafness-2 (DFNX2). In addition, we performed homology modeling to predict the effect of the missense variant on the protein structure showing a possible disruption of the normal folding. The identification of pathogenic variants causing X-linked recessive deafness will improve molecular diagnosis, genetic counseling, and knowledge of the molecular epidemiology of hearing loss among Italian individuals. Taken together, we recommend preoperative gene mutation analysis in patients who have DFNX2 diagnosed on the basis of characteristic radiological findings, in order to provide with better prognostic information, the risk of recurrence, and improved rehabilitation options. Finally, the present work strengthens the hypothesis that DFNX-2 could be considered as a syndromic deafness, since mixed hearing loss is associated with other dysfunctions of the neuropsychological profile of the patients.
Collapse
Affiliation(s)
- Sara Giannantonio
- Audiology and Otosurgery Unit, "Bambino Gesù" Pediatric Hospital, Rome, Italy.
| | | | | | - Roberta Anzivino
- Audiology and Otosurgery Unit, "Bambino Gesù" Pediatric Hospital, Rome, Italy
| | | | | | - Antonio Novelli
- Genetics Unit, "Bambino Gesù" Pediatric Hospital, Rome, Italy
| | | | - Pasquale Marsella
- Audiology and Otosurgery Unit, "Bambino Gesù" Pediatric Hospital, Rome, Italy
| |
Collapse
|
16
|
Baanannou A, Rastegar S, Bouzid A, Takamiya M, Gerber V, Souissi A, Beil T, Jrad O, Strähle U, Masmoudi S. Gene duplication and functional divergence of the zebrafish otospiralin genes. Dev Genes Evol 2019; 230:27-36. [PMID: 31838648 DOI: 10.1007/s00427-019-00642-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 11/24/2019] [Indexed: 11/29/2022]
Abstract
Otospiralin (OTOSP) is a small protein of unknown function, expressed in fibrocytes of the inner ear and required for normal cochlear auditory function. Despite its conservation from fish to mammals, expression of otospiralin was only investigated in mammals. Here, we report for the first time the expression profile of OTOS orthologous genes in zebrafish (Danio rerio): otospiralin and si:ch73-23l24.1 (designated otospiralin-like). In situ hybridization analyses in zebrafish embryos showed a specific expression of otospiralin-like in notochord (from 14 to 48 hpf) and similar expression patterns for otospiralin and otospiralin-like in gut (from 72 to 120 hpf), swim bladder (from 96 to 120 hpf) and inner ear (at 120 hpf). Morpholino knockdown of otospiralin and otospiralin-like showed no strong change of the body structure of the embryos at 5 dpf and the inner ear was normally formed. Nevertheless, knockdown embryos showed a reduced number of kinocilia in the lateral crista, indicating that these genes play an important role in kinocilium formation. RT-qPCR revealed that otospiralin is highly expressed in adult zebrafish inner ear comparing to the others analyzed tissues as previously shown for mice. Interestingly, otospiralin-like was not detected in the inner ear which suggests that otospiralin have a more important function in hearing than otospiralin-like. Phylogenetic analysis of otospiralin proteins in vertebrates indicated the presence of two subgroups and supported the functional divergence observed in zebrafish for otospiralin and otospiralin-like genes. This study offers the first insight into the expression of otospiralin and otospiralin-like in zebrafish. Expression data point to an important role for otospiralin in zebrafish hearing and a specific role for otospiralin-like in notochord vacuolization.
Collapse
Affiliation(s)
- Aissette Baanannou
- Laboratory of Molecular and Cellular Screening Processes, Center of Biotechnology of Sfax, Sfax, Tunisia.
| | - Sepand Rastegar
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, 76021, Karlsruhe, Germany
| | - Amal Bouzid
- Laboratory of Molecular and Cellular Screening Processes, Center of Biotechnology of Sfax, Sfax, Tunisia
| | - Masanari Takamiya
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, 76021, Karlsruhe, Germany
| | - Vanessa Gerber
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, 76021, Karlsruhe, Germany
| | - Amal Souissi
- Laboratory of Molecular and Cellular Screening Processes, Center of Biotechnology of Sfax, Sfax, Tunisia
| | - Tanja Beil
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, 76021, Karlsruhe, Germany
| | - Olfa Jrad
- Laboratory of Molecular and Cellular Screening Processes, Center of Biotechnology of Sfax, Sfax, Tunisia
| | - Uwe Strähle
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, 76021, Karlsruhe, Germany
| | - Saber Masmoudi
- Laboratory of Molecular and Cellular Screening Processes, Center of Biotechnology of Sfax, Sfax, Tunisia
| |
Collapse
|
17
|
Furness DN. Forgotten Fibrocytes: A Neglected, Supporting Cell Type of the Cochlea With the Potential to be an Alternative Therapeutic Target in Hearing Loss. Front Cell Neurosci 2019; 13:532. [PMID: 31866825 PMCID: PMC6908467 DOI: 10.3389/fncel.2019.00532] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 11/15/2019] [Indexed: 12/31/2022] Open
Abstract
Cochlear fibrocytes are a homeostatic supporting cell type embedded in the vascularized extracellular matrix of the spiral ligament, within the lateral wall. Here, they participate in the connective tissue syncytium that enables potassium recirculation into the scala media to take place and ensures development of the endolymphatic potential that helps drive current into hair cells during acoustic stimulation. They have also been implicated in inflammatory responses in the cochlea. Some fibrocytes interact closely with the capillaries of the vasculature in a way which suggests potential involvement, together with the stria vascularis, also in the blood-labyrinth barrier. Several lines of evidence suggests that pathology of the fibrocytes, along with other degenerative changes in this region, contribute to metabolic hearing loss (MHL) during aging that is becoming recognized as distinct from, and potentially a precursor for, sensorineural hearing loss (SNHL). This pathology may underlie a significant proportion of cases of presbycusis. Some evidence points also to an association between fibrocyte degeneration and Ménière’s disease (MD). Fibrocytes are mesenchymal; this characteristic, and their location, make them amenable to potential cell therapy in the form of cell replacement or genetic modification to arrest the process of degeneration that leads to MHL. This review explores the properties and roles of this neglected cell type and suggests potential therapeutic approaches, such as cell transplantation or genetic engineering of fibrocytes, which could be used to prevent this form of presbycusis or provide a therapeutic avenue for MD.
Collapse
Affiliation(s)
- David N Furness
- School of Life Sciences, Keele University, Keele, United Kingdom
| |
Collapse
|
18
|
Shin SA, Lyu AR, Jeong SH, Kim TH, Park MJ, Park YH. Acoustic Trauma Modulates Cochlear Blood Flow and Vasoactive Factors in a Rodent Model of Noise-Induced Hearing Loss. Int J Mol Sci 2019; 20:ijms20215316. [PMID: 31731459 PMCID: PMC6862585 DOI: 10.3390/ijms20215316] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 12/12/2022] Open
Abstract
Noise exposure affects the organ of Corti and the lateral wall of the cochlea, including the stria vascularis and spiral ligament. Although the inner ear vasculature and spiral ligament fibrocytes in the lateral wall consist of a significant proportion of cells in the cochlea, relatively little is known regarding their functional significance. In this study, 6-week-old male C57BL/6 mice were exposed to noise trauma to induce transient hearing threshold shift (TTS) or permanent hearing threshold shift (PTS). Compared to mice with TTS, mice with PTS exhibited lower cochlear blood flow and lower vessel diameter in the stria vascularis, accompanied by reduced expression levels of genes involved in vasodilation and increased expression levels of genes related to vasoconstriction. Ultrastructural analyses by transmission electron microscopy revealed that the stria vascularis and spiral ligament fibrocytes were more damaged by PTS than by TTS. Moreover, mice with PTS expressed significantly higher levels of proinflammatory cytokines in the cochlea (e.g., IL-1β, IL-6, and TNF-α). Overall, our findings suggest that cochlear microcirculation and lateral wall pathologies are differentially modulated by the severity of acoustic trauma and are associated with changes in vasoactive factors and inflammatory responses in the cochlea.
Collapse
Affiliation(s)
- Sun-Ae Shin
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (S.-A.S.); (A.-R.L.)
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea;
| | - Ah-Ra Lyu
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (S.-A.S.); (A.-R.L.)
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea;
| | - Seong-Hun Jeong
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea;
| | - Tae Hwan Kim
- Biomedical Convergence Research Center, Chungnam National University Hospital, Daejeon 35015, Korea;
| | - Min Jung Park
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (S.-A.S.); (A.-R.L.)
- Brain Research Institute, College of Medicine, Chungnam National University, Daejeon 35015, Korea
- Correspondence: (M.J.P.); (Y.-H.P.); Tel.: +82-42-280-7697(M.J.P.); Fax: +82-42-253-4059 (M.J.P.)
| | - Yong-Ho Park
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (S.-A.S.); (A.-R.L.)
- Brain Research Institute, College of Medicine, Chungnam National University, Daejeon 35015, Korea
- Correspondence: (M.J.P.); (Y.-H.P.); Tel.: +82-42-280-7697(M.J.P.); Fax: +82-42-253-4059 (M.J.P.)
| |
Collapse
|
19
|
Miwa T, Minoda R, Ishikawa Y, Kajii T, Orita Y, Ohyama T. Role of Dach1 revealed using a novel inner ear-specific Dach1-knockdown mouse model. Biol Open 2019; 8:bio.043612. [PMID: 31405829 PMCID: PMC6737983 DOI: 10.1242/bio.043612] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Dach1 gene is expressed in the inner ear of normal mouse embryos in the area that differentiates into the cochlear stria vascularis (SV). We hypothesised that Dach1 downregulation in the inner ear would lead to SV dysplasia. However, because Dach1 knockout is embryonic lethal in mice, the role of Dach1 in the inner ear is unclear. Here, we established inner ear-specific Dach1-knockdown mice and showed that Dach1 downregulation resulted in hearing loss, reduced endocochlear potential and secondary outer hair cell loss. There were no abnormalities in marginal cells and basal cells in the SV or spiral ligament in inner ear-specific Dach1-knockdown mature mice. However, intermediate cell dysplasia and thinning of the SV were observed. Moreover, dynamic changes in the expression of key genes related to the epithelial-mesenchymal transition were observed in the lateral wall of the cochlear epithelium, which differentiated into the SV in inner ear-specific Dach1-knockdown mice at embryonic stages. In summary, suppression of Dach1 expression in the inner ear caused the epithelial-mesenchymal transition in the lateral wall of cochlear epithelium, resulting in loss of intermediate cells in the SV and SV dysplasia. This article has an associated First Person interview with the first author of the paper. Summary: Inner ear-specific downregulation of Dach1 reveals that the epithelial–mesenchymal transition is crucial for the generation of the stria vascularis.
Collapse
Affiliation(s)
- Toru Miwa
- Department of Otolaryngology and Head and Neck Surgery, Graduate of School of Medicine, Kumamoto University, Kumamoto 8608556, Japan .,Otolaryngology-Head and Neck Surgery, JCHO Kumamoto General Hospital, Kumamoto 8668660, Japan
| | - Ryosei Minoda
- Otolaryngology-Head and Neck Surgery, JCHO Kumamoto General Hospital, Kumamoto 8668660, Japan
| | - Yoshihide Ishikawa
- Department of Otolaryngology and Head and Neck Surgery, Graduate of School of Medicine, Kumamoto University, Kumamoto 8608556, Japan
| | - Tomohito Kajii
- Department of Otolaryngology and Head and Neck Surgery, Graduate of School of Medicine, Kumamoto University, Kumamoto 8608556, Japan
| | - Yorihisa Orita
- Department of Otolaryngology and Head and Neck Surgery, Graduate of School of Medicine, Kumamoto University, Kumamoto 8608556, Japan
| | - Takahiro Ohyama
- USC-Tina and Rick Caruso Department of Otolaryngology-Head & Neck Surgery, Zilkha Neurogenetic Institute, USC Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
20
|
Kada S, Hamaguchi K, Ito J, Omori K, Nakagawa T. Bone Marrow Stromal Cells Accelerate Hearing Recovery via Regeneration or Maintenance of Cochlear Fibrocytes in Mouse Spiral Ligaments. Anat Rec (Hoboken) 2019; 303:478-486. [DOI: 10.1002/ar.24063] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 04/14/2018] [Accepted: 04/26/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Shinpei Kada
- Department of Otolaryngology, Head and Neck SurgeryGraduate School of Medicine, Kyoto University Kyoto Japan
- Department of Otolaryngology, Head and Neck SurgeryNational Hospital Organization Kyoto Medical Center Kyoto Japan
| | - Kiyomi Hamaguchi
- Department of Otolaryngology, Head and Neck SurgeryGraduate School of Medicine, Kyoto University Kyoto Japan
- Department of Otolaryngology, Head and Neck SurgeryShizuoka City Shizuoka Hospital Shizuoka Japan
| | - Juichi Ito
- Department of Otolaryngology, Head and Neck SurgeryGraduate School of Medicine, Kyoto University Kyoto Japan
- Shiga Medical Center Research Institute Moriyama Japan
| | - Koichi Omori
- Department of Otolaryngology, Head and Neck SurgeryGraduate School of Medicine, Kyoto University Kyoto Japan
| | - Takayuki Nakagawa
- Department of Otolaryngology, Head and Neck SurgeryGraduate School of Medicine, Kyoto University Kyoto Japan
| |
Collapse
|
21
|
Defourny J. Eph/ephrin signalling in the development and function of the mammalian cochlea. Dev Biol 2019; 449:35-40. [PMID: 30771305 DOI: 10.1016/j.ydbio.2019.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/10/2019] [Accepted: 02/11/2019] [Indexed: 12/12/2022]
Abstract
In mammals, the functional development of the cochlea requires the tight regulation of multiple molecules and signalling pathways including fibroblast growth factors, bone morphogenetic proteins, Wnt and Notch signalling pathways. Over the last decade, the Eph/ephrin system also emerged as a key player of the development and function of the mammalian cochlea. In this review, we discuss the recent advances on the role of Eph/ephrin signalling in patterning the cochlear sensory epithelium and the complex innervation of mechanosensory hair cells by spiral ganglion neurons. Finally, we address the issue of a syndromic form of hearing loss caused by a deficient member of the Eph/ephrin family.
Collapse
Affiliation(s)
- Jean Defourny
- GIGA-Neurosciences, Unit of Cell and Tissue Biology, University of Liège, C.H.U. B36, B-4000, Liège, Belgium.
| |
Collapse
|
22
|
Nonomura Y, Sawamura S, Hanzawa K, Nishikaze T, Sekiya S, Higuchi T, Nin F, Uetsuka S, Inohara H, Okuda S, Miyoshi E, Horii A, Takahashi S, Natsuka S, Hibino H. Characterisation of N-glycans in the epithelial-like tissue of the rat cochlea. Sci Rep 2019; 9:1551. [PMID: 30733536 PMCID: PMC6367448 DOI: 10.1038/s41598-018-38079-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 12/17/2018] [Indexed: 01/08/2023] Open
Abstract
Membrane proteins (such as ion channels, transporters, and receptors) and secreted proteins are essential for cellular activities. N-linked glycosylation is involved in stability and function of these proteins and occurs at Asn residues. In several organs, profiles of N-glycans have been determined by comprehensive analyses. Nevertheless, the cochlea of the mammalian inner ear, a tiny organ mediating hearing, has yet to be examined. Here, we focused on the stria vascularis, an epithelial-like tissue in the cochlea, and characterised N-glycans by liquid chromatography with mass spectrometry. This hypervascular tissue not only expresses several ion transporters and channels to control the electrochemical balance in the cochlea but also harbours different transporters and receptors that maintain structure and activity of the organ. Seventy-nine N-linked glycans were identified in the rat stria vascularis. Among these, in 55 glycans, the complete structures were determined; in the other 24 species, partial glycosidic linkage patterns and full profiles of the monosaccharide composition were identified. In the process of characterisation, several sialylated glycans were subjected sequentially to two different alkylamidation reactions; this derivatisation helped to distinguish α2,3-linkage and α2,6-linkage sialyl isomers with mass spectrometry. These data should accelerate elucidation of the molecular architecture of the cochlea.
Collapse
Affiliation(s)
- Yoriko Nonomura
- Department of Molecular Physiology, Niigata University School of Medicine, Niigata, Japan
- Department of Otorhinolaryngology-Head and Neck Surgery, Niigata University School of Medicine, Niigata, Japan
| | - Seishiro Sawamura
- Department of Molecular Physiology, Niigata University School of Medicine, Niigata, Japan
| | - Ken Hanzawa
- Department of Biology, Faculty of Science, Niigata University, Niigata, Japan
| | - Takashi Nishikaze
- Koichi Tanaka Mass Spectrometry Research Laboratory, Shimadzu Corporation, Kyoto, Japan
| | - Sadanori Sekiya
- Koichi Tanaka Mass Spectrometry Research Laboratory, Shimadzu Corporation, Kyoto, Japan
| | - Taiga Higuchi
- Department of Molecular Physiology, Niigata University School of Medicine, Niigata, Japan
| | - Fumiaki Nin
- Department of Molecular Physiology, Niigata University School of Medicine, Niigata, Japan
- Center for Transdisciplinary Research, Niigata University, Niigata, Japan
| | - Satoru Uetsuka
- Department of Otorhinolaryngology-Head and Neck Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hidenori Inohara
- Department of Otorhinolaryngology-Head and Neck Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shujiro Okuda
- Bioinformatics Laboratory, Niigata University School of Medicine, Niigata, Japan
| | - Eiji Miyoshi
- Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Arata Horii
- Department of Otorhinolaryngology-Head and Neck Surgery, Niigata University School of Medicine, Niigata, Japan
| | - Sugata Takahashi
- Department of Otorhinolaryngology-Head and Neck Surgery, Niigata University School of Medicine, Niigata, Japan
| | - Shunji Natsuka
- Department of Biology, Faculty of Science, Niigata University, Niigata, Japan
| | - Hiroshi Hibino
- Department of Molecular Physiology, Niigata University School of Medicine, Niigata, Japan.
- Center for Transdisciplinary Research, Niigata University, Niigata, Japan.
- AMED-CREST, AMED, Niigata, Japan.
| |
Collapse
|
23
|
Yang T, Guo L, Wang L, Yu X. Diagnosis, Intervention, and Prevention of Genetic Hearing Loss. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1130:73-92. [PMID: 30915702 DOI: 10.1007/978-981-13-6123-4_5] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
It is estimated that at least 50% of congenital or childhood hearing loss is attributable to genetic causes. In non-syndromic hearing loss, which accounts for 70% of genetic hearing loss, approximately 80% of cases are autosomal recessive, 15% autosomal dominant, and 1-2% mitochondrial or X-linked. In addition, 30% of genetic hearing loss is syndromic. The genetic causes of hearing loss are highly heterogeneous. So far, more than 140 deafness-related genes have been discovered. Studies on those genes tremendously increased our understanding of the inner ear functions at the molecular level. It also offers important information for the patients and allows personalized and accurate genetic counseling. In many cases, genetic diagnosis of hearing loss can help to avoid unnecessary and costly clinical testing, offer prognostic information, and guide future medical management. On the other hand, a variety of gene therapeutic approaches have been developed aiming to relieve or converse the hearing loss due to genetic causes. Prevention of genetic hearing loss is feasible through prepregnancy and prenatal genetic diagnosis and counseling.
Collapse
Affiliation(s)
- Tao Yang
- Department of Otorhinolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China. .,Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China. .,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China.
| | - Luo Guo
- Key Laboratory of Hearing Medicine of NHFPC, ENT Institute and Otorhinolaryngology Department, Shanghai Engineering Research Centre of Cochlear Implant, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Longhao Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Xiaoyu Yu
- Department of Otorhinolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| |
Collapse
|
24
|
Espino Guarch M, Font-Llitjós M, Murillo-Cuesta S, Errasti-Murugarren E, Celaya AM, Girotto G, Vuckovic D, Mezzavilla M, Vilches C, Bodoy S, Sahún I, González L, Prat E, Zorzano A, Dierssen M, Varela-Nieto I, Gasparini P, Palacín M, Nunes V. Mutations in L-type amino acid transporter-2 support SLC7A8 as a novel gene involved in age-related hearing loss. eLife 2018; 7:31511. [PMID: 29355479 PMCID: PMC5811215 DOI: 10.7554/elife.31511] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 01/18/2018] [Indexed: 12/19/2022] Open
Abstract
Age-related hearing loss (ARHL) is the most common sensory deficit in the elderly. The disease has a multifactorial etiology with both environmental and genetic factors involved being largely unknown. SLC7A8/SLC3A2 heterodimer is a neutral amino acid exchanger. Here, we demonstrated that SLC7A8 is expressed in the mouse inner ear and that its ablation resulted in ARHL, due to the damage of different cochlear structures. These findings make SLC7A8 transporter a strong candidate for ARHL in humans. Thus, a screening of a cohort of ARHL patients and controls was carried out revealing several variants in SLC7A8, whose role was further investigated by in vitro functional studies. Significant decreases in SLC7A8 transport activity was detected for patient’s variants (p.Val302Ile, p.Arg418His, p.Thr402Met and p.Val460Glu) further supporting a causative role for SLC7A8 in ARHL. Moreover, our preliminary data suggest that a relevant proportion of ARHL cases could be explained by SLC7A8 mutations. Age-related hearing loss affects about one in three individuals between the ages of 65 and 74. The first symptom is difficulty hearing high-pitched sounds like children’s voices. The disease starts gradually and worsens over time. Changes in the ear, the nerve that connects it to the brain, or the brain itself can cause hearing loss. Sometimes all three play a role. Genetics, exposure to noise, disease, and aging may all contribute. The condition is so complex it is difficult for scientists to pinpoint a primary suspect or develop treatments. Now, Guarch, Font-Llitjós et al. show that errors in a protein called SLC7A8 cause age-related hearing loss in mice and humans. The SLC7A8 protein acts like a door that allows amino acids – the building blocks of proteins – to enter or leave a cell. This door is blocked in mice lacking SLC7A8 and damage occurs in the part of their inner ear responsible for hearing. As a result, the animals lose their hearing. Next, Guarch, Font-Llitjós et al. scanned the genomes of 147 people from isolated villages in Italy for mutations in the gene for SLC7A8. The people also underwent hearing tests. Mutations in the gene for SLC7A8 that partially block the door and prevent the flow of amino acids were found in people with hearing loss. Some mutations in SLC7A8 that allow the door to stay open where found in people who could hear. The experiments suggest that certain mutations in the gene for SLC7A8 are likely an inherited cause of age-related hearing loss. It is possible that other proteins that control the flow of amino acids into or out of cells also may play a role in hearing. More studies are needed to see if it is possible to fix errors in the SLC7A8 protein to delay or restore the hearing loss.
Collapse
Affiliation(s)
- Meritxell Espino Guarch
- Experimental Genetics, Sidra Medical and Research Center, Doha, Qatar.,Genes, Disease and Therapy Program, Molecular Genetics Laboratory - IDIBELL, Barcelona, Spain.,Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mariona Font-Llitjós
- Genes, Disease and Therapy Program, Molecular Genetics Laboratory - IDIBELL, Barcelona, Spain.,Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain
| | - Silvia Murillo-Cuesta
- Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Alberto Sols Biomedical Research Institute (CSIC/UAM), Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Ekaitz Errasti-Murugarren
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain
| | - Adelaida M Celaya
- Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Alberto Sols Biomedical Research Institute (CSIC/UAM), Madrid, Spain
| | - Giorgia Girotto
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy.,Medical Genetics, Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - Dragana Vuckovic
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy.,Medical Genetics, Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | | | - Clara Vilches
- Genes, Disease and Therapy Program, Molecular Genetics Laboratory - IDIBELL, Barcelona, Spain
| | - Susanna Bodoy
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain
| | - Ignasi Sahún
- Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Laura González
- Genes, Disease and Therapy Program, Molecular Genetics Laboratory - IDIBELL, Barcelona, Spain.,Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain
| | - Esther Prat
- Genes, Disease and Therapy Program, Molecular Genetics Laboratory - IDIBELL, Barcelona, Spain.,Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Genetics Section, Physiological Sciences Department, Health Sciences and Medicine Faculty, University of Barcelona, Barcelona, Spain
| | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Biochemistry and Molecular Biomedicine Department, Faculty of Biology, University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre on Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - Mara Dierssen
- Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Isabel Varela-Nieto
- Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Alberto Sols Biomedical Research Institute (CSIC/UAM), Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Paolo Gasparini
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy.,Medical Genetics, Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - Manuel Palacín
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Biochemistry and Molecular Biomedicine Department, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Virginia Nunes
- Genes, Disease and Therapy Program, Molecular Genetics Laboratory - IDIBELL, Barcelona, Spain.,Biomedical Research Networking Centre on Rare Diseases (CIBERER), Institute of Health Carlos III, Barcelona, Spain.,Genetics Section, Physiological Sciences Department, Health Sciences and Medicine Faculty, University of Barcelona, Barcelona, Spain
| |
Collapse
|
25
|
Frejo L, Requena T, Okawa S, Gallego-Martinez A, Martinez-Bueno M, Aran I, Batuecas-Caletrio A, Benitez-Rosario J, Espinosa-Sanchez JM, Fraile-Rodrigo JJ, García-Arumi AM, González-Aguado R, Marques P, Martin-Sanz E, Perez-Fernandez N, Pérez-Vázquez P, Perez-Garrigues H, Santos-Perez S, Soto-Varela A, Tapia MC, Trinidad-Ruiz G, Del Sol A, Alarcon Riquelme ME, Lopez-Escamez JA. Regulation of Fn14 Receptor and NF-κB Underlies Inflammation in Meniere's Disease. Front Immunol 2017; 8:1739. [PMID: 29326686 PMCID: PMC5733484 DOI: 10.3389/fimmu.2017.01739] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 11/23/2017] [Indexed: 12/17/2022] Open
Abstract
Meniere’s disease (MD) is a rare disorder characterized by episodic vertigo, sensorineural hearing loss, tinnitus, and aural fullness. It is associated with a fluid imbalance between the secretion of endolymph in the cochlear duct and its reabsorption into the subarachnoid space, leading to an accumulation of endolymph in the inner ear. Epidemiological evidence, including familial aggregation, indicates a genetic contribution and a consistent association with autoimmune diseases (AD). We conducted a case–control study in two phases using an immune genotyping array in a total of 420 patients with bilateral MD and 1,630 controls. We have identified the first locus, at 6p21.33, suggesting an association with bilateral MD [meta-analysis leading signal rs4947296, OR = 2.089 (1.661–2.627); p = 1.39 × 10−09]. Gene expression profiles of homozygous genotype-selected peripheral blood mononuclear cells (PBMCs) demonstrated that this region is a trans-expression quantitative trait locus (eQTL) in PBMCs. Signaling analysis predicted several tumor necrosis factor-related pathways, the TWEAK/Fn14 pathway being the top candidate (p = 2.42 × 10−11). This pathway is involved in the modulation of inflammation in several human AD, including multiple sclerosis, systemic lupus erythematosus, or rheumatoid arthritis. In vitro studies with genotype-selected lymphoblastoid cells from patients with MD suggest that this trans-eQTL may regulate cellular proliferation in lymphoid cells through the TWEAK/Fn14 pathway by increasing the translation of NF-κB. Taken together; these findings suggest that the carriers of the risk genotype may develop an NF-κB-mediated inflammatory response in MD.
Collapse
Affiliation(s)
- Lidia Frejo
- Otology and Neurotology Group CTS495, Department of Genomic Medicine - Centre for Genomics and Oncological Research - Pfizer/Universidad de Granada/Junta de Andalucía (GENYO), Granada, Spain
| | - Teresa Requena
- Otology and Neurotology Group CTS495, Department of Genomic Medicine - Centre for Genomics and Oncological Research - Pfizer/Universidad de Granada/Junta de Andalucía (GENYO), Granada, Spain
| | - Satoshi Okawa
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine (LCSB), Universite du Luxembourg, Belval, Luxembourg
| | - Alvaro Gallego-Martinez
- Otology and Neurotology Group CTS495, Department of Genomic Medicine - Centre for Genomics and Oncological Research - Pfizer/Universidad de Granada/Junta de Andalucía (GENYO), Granada, Spain
| | - Manuel Martinez-Bueno
- Group of Genetics of Complex Diseases, Department of Genomic Medicine - Centre for Genomics and Oncological Research - Pfizer/Universidad de Granada/Junta de Andalucía (GENYO), Granada, Spain
| | - Ismael Aran
- Department of Otolaryngology, Complexo Hospitalario de Pontevedra, Pontevedra, Spain
| | | | - Jesus Benitez-Rosario
- Department of Otolaryngology, Hospital Universitario de Gran Canaria Dr Negrin, Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Juan M Espinosa-Sanchez
- Otology and Neurotology Group CTS495, Department of Genomic Medicine - Centre for Genomics and Oncological Research - Pfizer/Universidad de Granada/Junta de Andalucía (GENYO), Granada, Spain.,Department of Otolaryngology, Instituto de Investigación Biosanitaria ibs.GRANADA, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | | | - Ana María García-Arumi
- Department of Otorhinolaryngology, Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | - Rocío González-Aguado
- Department of Otorhinolaryngology, Hospital Universitario Marqués de Valdecilla, Santander, Cantabria, Spain
| | - Pedro Marques
- Department of Otorhinolaryngology, Centro Hospitalar de S.João, EPE, University of Porto Medical School, Porto, Portugal
| | - Eduardo Martin-Sanz
- Department of Otolaryngology, Hospital Universitario de Getafe, Getafe, Madrid, Spain
| | | | - Paz Pérez-Vázquez
- Department of Otorhinolaryngology, Hospital Universitario de Cabueñes, Gijón, Asturias, Spain
| | | | - Sofía Santos-Perez
- Division of Otoneurology, Department of Otorhinolaryngology, Complexo Hospitalario Universitario, Santiago de Compostela, Spain
| | - Andres Soto-Varela
- Division of Otoneurology, Department of Otorhinolaryngology, Complexo Hospitalario Universitario, Santiago de Compostela, Spain
| | - Maria C Tapia
- Department of Otorhinolaryngology, Instituto Antolí Candela, Madrid, Spain
| | - Gabriel Trinidad-Ruiz
- Division of Otoneurology, Department of Otorhinolaryngology, Complejo Hospitalario Badajoz, Badajoz, Spain
| | - Antonio Del Sol
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine (LCSB), Universite du Luxembourg, Belval, Luxembourg
| | - Marta E Alarcon Riquelme
- Group of Genetics of Complex Diseases, Department of Genomic Medicine - Centre for Genomics and Oncological Research - Pfizer/Universidad de Granada/Junta de Andalucía (GENYO), Granada, Spain.,Unit of Chronic Inflammatory Diseases, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jose A Lopez-Escamez
- Otology and Neurotology Group CTS495, Department of Genomic Medicine - Centre for Genomics and Oncological Research - Pfizer/Universidad de Granada/Junta de Andalucía (GENYO), Granada, Spain.,Department of Otolaryngology, Instituto de Investigación Biosanitaria ibs.GRANADA, Hospital Universitario Virgen de las Nieves, Granada, Spain.,Luxembourg Centre for System Biomedicine (LCSB), Universite du Luxembourg, Belval, Luxembourg
| |
Collapse
|
26
|
HPN-07, a free radical spin trapping agent, protects against functional, cellular and electrophysiological changes in the cochlea induced by acute acoustic trauma. PLoS One 2017; 12:e0183089. [PMID: 28832600 PMCID: PMC5568441 DOI: 10.1371/journal.pone.0183089] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 07/28/2017] [Indexed: 11/29/2022] Open
Abstract
Oxidative stress is considered a major cause of the structural and functional changes associated with auditory pathologies induced by exposure to acute acoustic trauma AAT). In the present study, we examined the otoprotective effects of 2,4-disulfophenyl-N-tert-butylnitrone (HPN-07), a nitrone-based free radical trap, on the physiological and cellular changes in the auditory system of chinchilla following a six-hour exposure to 4 kHz octave band noise at 105 dB SPL. HPN-07 has been shown to suppress oxidative stress in biological models of a variety of disorders. Our results show that administration of HPN-07 beginning four hours after acoustic trauma accelerated and enhanced auditory/cochlear functional recovery, as measured by auditory brainstem responses (ABR), distortion product otoacoustic emissions (DPOAE), compound action potentials (CAP), and cochlear microphonics (CM). The normally tight correlation between the endocochlear potential (EP) and evoked potentials of CAP and CM were persistently disrupted after noise trauma in untreated animals but returned to homeostatic conditions in HPN-07 treated animals. Histological analyses revealed several therapeutic advantages associated with HPN-07 treatment following AAT, including reductions in inner and outer hair cell loss; reductions in AAT-induced loss of calretinin-positive afferent nerve fibers in the spiral lamina; and reductions in fibrocyte loss within the spiral ligament. These findings support the conclusion that early intervention with HPN-07 following an AAT efficiently blocks the propagative ototoxic effects of oxidative stress, thereby preserving the homeostatic and functional integrity of the cochlea.
Collapse
|
27
|
Liu W, Schrott-Fischer A, Glueckert R, Benav H, Rask-Andersen H. The Human "Cochlear Battery" - Claudin-11 Barrier and Ion Transport Proteins in the Lateral Wall of the Cochlea. Front Mol Neurosci 2017; 10:239. [PMID: 28848383 PMCID: PMC5554435 DOI: 10.3389/fnmol.2017.00239] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 07/14/2017] [Indexed: 11/21/2022] Open
Abstract
Background: The cochlea produces an electric field potential essential for hair cell transduction and hearing. This biological “battery” is situated in the lateral wall of the cochlea and contains molecular machinery that secretes and recycles K+ ions. Its functioning depends on junctional proteins that restrict the para-cellular escape of ions. The tight junction protein Claudin-11 has been found to be one of the major constituents of this barrier that maintains ion gradients (Gow et al., 2004; Kitajiri et al., 2004a). We are the first to elucidate the human Claudin-11 framework and the associated ion transport machinery using super-resolution fluorescence illumination microscopy (SR-SIM). Methods: Archival cochleae obtained during meningioma surgery were used for SR-SIM together with transmission electron microscopy after ethical consent. Results: Claudin-11-expressing cells formed parallel tight junction lamellae that insulated the epithelial syncytium of the stria vascularis and extended to the suprastrial region. Intercellular gap junctions were found between the barrier cells and fibrocytes. Conclusion: Transmission electron microscopy, confocal microscopy and SR-SIM revealed exclusive cell specialization in the various subdomains of the lateral wall of the human cochlea. The Claudin-11-expressing cells exhibited both conductor and isolator characteristics, and these micro-porous separators may selectively mediate the movement of charged units to the intrastrial space in a manner that is analogous to a conventional electrochemical “battery.” The function and relevance of this battery for the development of inner ear disease are discussed.
Collapse
Affiliation(s)
- Wei Liu
- Department of Surgical Sciences, Section of Otolaryngology, Uppsala University HospitalUppsala, Sweden
| | | | - Rudolf Glueckert
- Department of Otolaryngology, Medical University of InnsbruckInnsbruck, Austria
| | | | - Helge Rask-Andersen
- Department of Surgical Sciences, Head and Neck Surgery, Section of Otolaryngology, Uppsala University HospitalUppsala, Sweden
| |
Collapse
|
28
|
Abstract
Cochlear spiral ligament fibrocytes (SLFs) play essential roles in the physiology of hearing including ion recycling and the generation of endocochlear potential. In adult animals, SLFs can repopulate after damages, yet little is known about the characteristics of proliferating cells that support SLFs' self-renewal. Here we report in detail about the characteristics of cycling cells in the spiral ligament (SL). Fifteen P6 mice and six noise-exposed P28 mice were injected with 5-bromo-2'-deoxyuridine (BrdU) for 7 days and we chased BrdU retaining cells for as long as 60 days. Immunohistochemistry revealed that the BrdU positive IB4 (an endotherial marker) negative cells expressed an early SLF marker Pou3f4 but negative for cleaved-Caspase 3. Marker studies revealed that type 3 SLFs displayed significantly higher percentage of BrdU+ cells compared to other subtypes. Notably, the cells retained BrdU until P72, demonstrating they were dividing slowly. In the noise-damaged mice, in contrast to the loss of the other types, the number of type 3 SLFs did not altered and the BrdU incorporating- phosphorylated Histone H3 positive type 3 cells were increased from day 1 to 14 after noise exposure. Furthermore, the cells repopulating type 1 area, where the cells diminished profoundly after damage, were positive for the type 3 SLF markers. Collectively, in the latral wall of the cochlea, type 3 SLFs have the stem cell capacity and may contribute to the endogenous regeneration of lateral wall spiral ligament. Manipulating type 3 cells may be employed for potential regenerative therapies.
Collapse
Affiliation(s)
- Yang Li
- Department of Otorhinolaryngology, School of Medicine, Keio University,35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital, Xi'an Jiaotong University, Xi Wu Lu, Xi'an, China
| | - Kotaro Watanabe
- Department of Otorhinolaryngology, School of Medicine, Keio University,35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Masato Fujioka
- Department of Otorhinolaryngology, School of Medicine, Keio University,35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
- * E-mail:
| | - Kaoru Ogawa
- Department of Otorhinolaryngology, School of Medicine, Keio University,35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
29
|
Kanno A, Mutai H, Namba K, Morita N, Nakano A, Ogahara N, Sugiuchi T, Ogawa K, Matsunaga T. Frequency and specific characteristics of the incomplete partition type III anomaly in children. Laryngoscope 2016; 127:1663-1669. [DOI: 10.1002/lary.26245] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 07/11/2016] [Accepted: 07/14/2016] [Indexed: 11/05/2022]
Affiliation(s)
- Ayako Kanno
- Division of Hearing and Balance Research; National Institute of Sensory Organs; National Hospital Organization Tokyo Medical Center; Tokyo Japan
- Department of Otolaryngology; Inagi Municipal Hospital; Tokyo Japan
- Department of Otolaryngology; Keio University School of Medicine; Tokyo Japan
| | - Hideki Mutai
- Division of Hearing and Balance Research; National Institute of Sensory Organs; National Hospital Organization Tokyo Medical Center; Tokyo Japan
| | - Kazunori Namba
- Division of Hearing and Balance Research; National Institute of Sensory Organs; National Hospital Organization Tokyo Medical Center; Tokyo Japan
| | - Noriko Morita
- Department of Otolaryngology; Kobari General Hospital; Chiba Japan
| | - Atsuko Nakano
- Division of Otolaryngology; Chiba Children's Hospital; Chiba Japan
| | - Noboru Ogahara
- Department of Otorhinolaryngology; Kanagawa Children's Medical Center; Kanagawa Japan
| | - Tomoko Sugiuchi
- Department of Otolaryngology; Kanto Rosai Hospital; Kanagawa Japan
| | - Kaoru Ogawa
- Department of Otolaryngology; Keio University School of Medicine; Tokyo Japan
| | - Tatsuo Matsunaga
- Division of Hearing and Balance Research; National Institute of Sensory Organs; National Hospital Organization Tokyo Medical Center; Tokyo Japan
- Medical Genetics Center; National Hospital Organization Tokyo Medical Center; Tokyo Japan
| |
Collapse
|
30
|
Deletion of Tricellulin Causes Progressive Hearing Loss Associated with Degeneration of Cochlear Hair Cells. Sci Rep 2015; 5:18402. [PMID: 26677943 PMCID: PMC4683410 DOI: 10.1038/srep18402] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 11/17/2015] [Indexed: 12/20/2022] Open
Abstract
Tricellulin (also known as MARVELD2) is considered as a central component of tricellular tight junctions and is distributed among various epithelial tissues. Although mutations in the gene encoding tricellulin are known to cause deafness in humans (DFNB49) and mice, the influence of its systemic deletion in vivo remains unknown. When we generated tricellulin-knockout mice (Tric−/−), we found an early-onset rapidly progressive hearing loss associated with the degeneration of hair cells (HCs); however, their body size and overall appearance were normal. Tric−/− mice did not show any morphological change pertaining to other organs such as the gastrointestinal tract, liver, kidney, thyroid gland and heart. The endocochlear potential (EP) was normal in Tric−/− mice, suggesting that the tight junction barrier is maintained in the stria vascularis, where EP is generated. The degeneration of HCs, which occurred after the maturation of EP, was prevented in the culture medium with an ion concentration similar to that of the perilymph. These data demonstrate the specific requirement of tricellulin for maintaining ion homeostasis around cochlear HCs to ensure their survival. The Tric−/− mouse provides a new model for understanding the distinct roles of tricellulin in different epithelial systems as well as in the pathogenesis of DFNB49.
Collapse
|
31
|
Cochlear afferent innervation development. Hear Res 2015; 330:157-69. [DOI: 10.1016/j.heares.2015.07.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 06/02/2015] [Accepted: 07/21/2015] [Indexed: 01/11/2023]
|
32
|
Choi BY, An YH, Song JJ, Koo JW, Lee JH, Oh SH, Chang SO, Kim CS, Park JH. Clinical observations and molecular variables of patients with hearing loss and incomplete partition type III. Laryngoscope 2015; 126:E123-8. [DOI: 10.1002/lary.25573] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 05/27/2015] [Accepted: 07/23/2015] [Indexed: 11/08/2022]
Affiliation(s)
- Byung Yoon Choi
- Department of Otorhinolaryngology-Head and Neck Surgery; Seoul National University Bundang Hospital; Seongnam South Korea
| | - Yong-Hwi An
- Department of Otorhinolaryngology-Head and Neck Surgery; Eulji University School of Medicine; Seoul South Korea
| | - Jae-Jin Song
- Department of Otorhinolaryngology-Head and Neck Surgery; Seoul National University Bundang Hospital; Seongnam South Korea
| | - Ja-Won Koo
- Department of Otorhinolaryngology-Head and Neck Surgery; Seoul National University Bundang Hospital; Seongnam South Korea
| | - Jun Ho Lee
- Department of Otorhinolaryngology-Head and Neck Surgery; Seoul National University Hospital; Seoul South Korea
| | - Seung Ha Oh
- Department of Otorhinolaryngology-Head and Neck Surgery; Seoul National University Hospital; Seoul South Korea
| | - Sun O. Chang
- Department of Otorhinolaryngology-Head and Neck Surgery; Seoul National University Hospital; Seoul South Korea
| | - Chong Sun Kim
- Department of Otorhinolaryngology-Head and Neck Surgery; Seoul National University Hospital; Seoul South Korea
| | - Joo Hyun Park
- Department of Otorhinolaryngology-Head and Neck Surgery; Dongguk University Ilsan Hospital; Goyang South Korea
| |
Collapse
|
33
|
Uetsuka S, Ogata G, Nagamori S, Isozumi N, Nin F, Yoshida T, Komune S, Kitahara T, Kikkawa Y, Inohara H, Kanai Y, Hibino H. Molecular architecture of the stria vascularis membrane transport system, which is essential for physiological functions of the mammalian cochlea. Eur J Neurosci 2015; 42:1984-2002. [PMID: 26060893 DOI: 10.1111/ejn.12973] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 06/02/2015] [Accepted: 06/02/2015] [Indexed: 11/29/2022]
Abstract
Stria vascularis of the mammalian cochlea transports K(+) to establish the electrochemical property in the endolymph crucial for hearing. This epithelial tissue also transports various small molecules. To clarify the profile of proteins participating in the transport system in the stria vascularis, membrane components purified from the stria of adult rats were analysed by liquid chromatography tandem mass spectrometry. Of the 3236 proteins detected in the analysis, 1807 were membrane proteins. Ingenuity Knowledge Base and literature data identified 513 proteins as being expressed on the 'plasma membrane', these included 25 ion channels and 79 transporters. Sixteen of the former and 62 of the latter had not yet been identified in the stria. Unexpectedly, many Cl(-) and Ca(2+) transport systems were found, suggesting that the dynamics of these ions play multiple roles. Several transporters for organic substances were also detected. Network analysis demonstrated that a few kinases, including protein kinase A, and Ca(2+) were key regulators for the strial transports. In the library of channels and transporters, 19 new candidates for uncloned deafness-related genes were identified. These resources provide a platform for understanding the molecular mechanisms underlying the epithelial transport essential for cochlear function and the pathophysiological processes involved in hearing disorders.
Collapse
Affiliation(s)
- Satoru Uetsuka
- Department of Molecular Physiology, Niigata University School of Medicine, 1-757 Asahimachi-dori, 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, Osaka, Japan
| | - Genki Ogata
- Department of Molecular Physiology, Niigata University School of Medicine, 1-757 Asahimachi-dori, Niigata, 951-8510, Japan.,Center for Transdisciplinary Research, Niigata University, Niigata, Japan
| | - Shushi Nagamori
- Division of Bio-system Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Noriyoshi Isozumi
- Division of Bio-system Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Fumiaki Nin
- Department of Molecular Physiology, Niigata University School of Medicine, 1-757 Asahimachi-dori, Niigata, 951-8510, Japan.,Center for Transdisciplinary Research, Niigata University, Niigata, Japan
| | - Takamasa Yoshida
- Department of Molecular Physiology, Niigata University School of Medicine, 1-757 Asahimachi-dori, Niigata, 951-8510, Japan.,Center for Transdisciplinary Research, Niigata University, Niigata, Japan.,Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shizuo Komune
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tadashi Kitahara
- Department of Otorhinolaryngology - Head and Neck Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Otorhinolaryngology - Head and Neck Surgery, Nara Medical University, Nara, Japan
| | - Yoshiaki Kikkawa
- Mammalian Genetics Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Hidenori Inohara
- Department of Otorhinolaryngology - Head and Neck Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yoshikatsu Kanai
- Division of Bio-system Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hiroshi Hibino
- Department of Molecular Physiology, Niigata University School of Medicine, 1-757 Asahimachi-dori, Niigata, 951-8510, Japan.,Center for Transdisciplinary Research, Niigata University, Niigata, Japan
| |
Collapse
|
34
|
Wu CC, Lin YH, Liu TC, Lin KN, Yang WS, Hsu CJ, Chen PL, Wu CM. Identifying Children With Poor Cochlear Implantation Outcomes Using Massively Parallel Sequencing. Medicine (Baltimore) 2015; 94:e1073. [PMID: 26166082 PMCID: PMC4504554 DOI: 10.1097/md.0000000000001073] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cochlear implantation is currently the treatment of choice for children with severe to profound hearing impairment. However, the outcomes with cochlear implants (CIs) vary significantly among recipients. The purpose of the present study is to identify the genetic determinants of poor CI outcomes. Twelve children with poor CI outcomes (the "cases") and 30 "matched controls" with good CI outcomes were subjected to comprehensive genetic analyses using massively parallel sequencing, which targeted 129 known deafness genes. Audiological features, imaging findings, and auditory/speech performance with CIs were then correlated to the genetic diagnoses. We identified genetic variants which are associated with poor CI outcomes in 7 (58%) of the 12 cases; 4 cases had bi-allelic PCDH15 pathogenic mutations and 3 cases were homozygous for the DFNB59 p.G292R variant. Mutations in the WFS1, GJB3, ESRRB, LRTOMT, MYO3A, and POU3F4 genes were detected in 7 (23%) of the 30 matched controls. The allele frequencies of PCDH15 and DFNB59 variants were significantly higher in the cases than in the matched controls (both P < 0.001). In the 7 CI recipients with PCDH15 or DFNB59 variants, otoacoustic emissions were absent in both ears, and imaging findings were normal in all 7 implanted ears. PCDH15 or DFNB59 variants are associated with poor CI performance, yet children with PCDH15 or DFNB59 variants might show clinical features indistinguishable from those of other typical pediatric CI recipients. Accordingly, genetic examination is indicated in all CI candidates before operation.
Collapse
Affiliation(s)
- Chen-Chi Wu
- From Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan (C-CW, Y-HL, T-CL, K-NL, C-JH); Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan (C-CW, W-SY, P-LC); Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, Taiwan (Y-HL, P-LC); Department of Otolaryngology, Cardinal Tien Hospital, New Taipei, Taiwan (K-NL); Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan (W-SY, P-LC); Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan (W-SY, P-LC); Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan (W-SY, P-LC); Department of Otolaryngology, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan (C-JH); and Department of Otolaryngology, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan (C-MW)
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Deleyrolle L, Sabourin JC, Rothhut B, Fujita H, Guichet PO, Teigell M, Ripoll C, Chauvet N, Perrin F, Mamaeva D, Noda T, Mori K, Yoshihara Y, Hugnot JP. OCAM regulates embryonic spinal cord stem cell proliferation by modulating ErbB2 receptor. PLoS One 2015; 10:e0122337. [PMID: 25875008 PMCID: PMC4395419 DOI: 10.1371/journal.pone.0122337] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 02/10/2015] [Indexed: 01/07/2023] Open
Abstract
The proliferation and differentiation of neural stem cells are tightly controlled by intrinsic and extrinsic cues. Cell adhesion molecules are increasingly recognized as regulators of these processes. Here we report the expression of the olfactory cell adhesion molecule (OCAM/NCAM2/RNCAM) during mouse spinal cord development and in neural stem cells cultured as neurospheres. OCAM is also weakly expressed in the dormant adult stem cell niche around the central canal and is overexpressed after spinal cord injury. Both transmembrane (TM) and glycosylphosphatidylinositol (GPI)-linked isoforms are present in neurospheres. Electron microscopy and internalisation experiments revealed a dynamic trafficking of OCAM between the membrane and intracellular compartments. After differentiation, OCAM remains in neurons and oligodendrocytes whereas no expression is detected in astrocytes. Using OCAM knockout (KO) mice, we found that mutant spinal cord stem cells showed an increased proliferation and self-renewal rates although no effect on differentiation was observed. This effect was reversed by lentivirus-mediated re-introduction of OCAM. Mechanistically, we identified the ErbB2/Neu/HER2 protein as being implicated in the enhanced proliferation of mutant cells. ErbB2 protein expression and phosphorylation level were significantly increased in KO cells whereas no difference was observed at the mRNA level. Overexpression of ErbB2 in wild-type and mutant cells also increased their growth while reintroduction of OCAM in mutant cells reduced the level of phosphorylated ErbB2. These results indicate that OCAM exerts a posttranscriptional control on the ErbB2 signalling in spinal cord stem cells. This study adds further support for considering cell adhesion molecules as regulators of the ErbB signalling.
Collapse
Affiliation(s)
- Loïc Deleyrolle
- Department of Neurosurgery, College of Medicine, University of Florida Gainesville, Gainesville, Florida, United States of America
| | | | - Bernard Rothhut
- INSERM U1051, Institute for Neuroscience, Hôpital Saint Eloi, Montpellier, France
- * E-mail:
| | | | | | - Marisa Teigell
- INSERM U1051, Institute for Neuroscience, Hôpital Saint Eloi, Montpellier, France
| | - Chantal Ripoll
- INSERM U1051, Institute for Neuroscience, Hôpital Saint Eloi, Montpellier, France
| | - Norbert Chauvet
- INSERM U661, Department of Endocrinology, Institute of Functional Genomics, Montpellier, France
- University of Montpellier 2, Montpellier, France
| | - Florence Perrin
- INSERM U1051, Institute for Neuroscience, Hôpital Saint Eloi, Montpellier, France
| | - Daria Mamaeva
- INSERM U1051, Institute for Neuroscience, Hôpital Saint Eloi, Montpellier, France
| | - Tetsuo Noda
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kensaku Mori
- Department of Physiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Jean-Philippe Hugnot
- INSERM U1051, Institute for Neuroscience, Hôpital Saint Eloi, Montpellier, France
- University of Montpellier 2, Montpellier, France
| |
Collapse
|
36
|
Nishio SY, Hattori M, Moteki H, Tsukada K, Miyagawa M, Naito T, Yoshimura H, Iwasa YI, Mori K, Shima Y, Sakuma N, Usami SI. Gene expression profiles of the cochlea and vestibular endorgans: localization and function of genes causing deafness. Ann Otol Rhinol Laryngol 2015; 124 Suppl 1:6S-48S. [PMID: 25814645 DOI: 10.1177/0003489415575549] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVES We sought to elucidate the gene expression profiles of the causative genes as well as the localization of the encoded proteins involved in hereditary hearing loss. METHODS Relevant articles (as of September 2014) were searched in PubMed databases, and the gene symbols of the genes reported to be associated with deafness were located on the Hereditary Hearing Loss Homepage using localization, expression, and distribution as keywords. RESULTS Our review of the literature allowed us to systematize the gene expression profiles for genetic deafness in the inner ear, clarifying the unique functions and specific expression patterns of these genes in the cochlea and vestibular endorgans. CONCLUSIONS The coordinated actions of various encoded molecules are essential for the normal development and maintenance of auditory and vestibular function.
Collapse
Affiliation(s)
- Shin-Ya Nishio
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto, Japan
| | - Mitsuru Hattori
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hideaki Moteki
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Keita Tsukada
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Maiko Miyagawa
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takehiko Naito
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hidekane Yoshimura
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yoh-Ichiro Iwasa
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Kentaro Mori
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yutaka Shima
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Naoko Sakuma
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan Department of Otorhinolaryngology and Head and Neck Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Shin-Ichi Usami
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|
37
|
Iizuka T, Kamiya K, Gotoh S, Sugitani Y, Suzuki M, Noda T, Minowa O, Ikeda K. Perinatal Gjb2 gene transfer rescues hearing in a mouse model of hereditary deafness. Hum Mol Genet 2015; 24:3651-61. [PMID: 25801282 DOI: 10.1093/hmg/ddv109] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 03/17/2015] [Indexed: 11/12/2022] Open
Abstract
Hearing loss is the most widespread sensory disorder, with an incidence of congenital genetic deafness of 1 in 1600 children. For many ethnic populations, the most prevalent form of genetic deafness is caused by recessive mutations in the gene gap junction protein, beta 2, 26 kDa (GJB2), which is also known as connexin 26 (Cx26). Despite this knowledge, existing treatment strategies do not completely recover speech perception. Here we used a gene delivery system to rescue hearing in a mouse model of Gjb2 deletion. Mice lacking Cx26 are characterized by profound deafness from birth and improper development of cochlear cells. Cochlear delivery of Gjb2 using an adeno-associated virus significantly improved the auditory responses and development of the cochlear structure. Using gene replacement to restore hearing in a new mouse model of Gjb2-related deafness may lead to the development of therapies for human hereditary deafness.
Collapse
Affiliation(s)
- Takashi Iizuka
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo 113-8421, Japan
| | - Kazusaku Kamiya
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo 113-8421, Japan
| | - Satoru Gotoh
- Department of Cell Biology, Japanese Foundation for Cancer Research, Cancer Institute, Tokyo 135-8550, Japan
| | - Yoshinobu Sugitani
- Department of Cell Biology, Japanese Foundation for Cancer Research, Cancer Institute, Tokyo 135-8550, Japan
| | - Masaaki Suzuki
- Department of Otolaryngology, Teikyo University Chiba Medical Center, Ichihara 299-0111, Japan and
| | - Tetsuo Noda
- Department of Cell Biology, Japanese Foundation for Cancer Research, Cancer Institute, Tokyo 135-8550, Japan, Team for Advanced Development and Evaluation of Human Disease Models, RIKEN BioResource Center, Tsukuba 305-0074, Japan
| | - Osamu Minowa
- Department of Cell Biology, Japanese Foundation for Cancer Research, Cancer Institute, Tokyo 135-8550, Japan, Team for Advanced Development and Evaluation of Human Disease Models, RIKEN BioResource Center, Tsukuba 305-0074, Japan
| | - Katsuhisa Ikeda
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo 113-8421, Japan,
| |
Collapse
|
38
|
Bowl MR, Dawson SJ. The mouse as a model for age-related hearing loss - a mini-review. Gerontology 2014; 61:149-57. [PMID: 25471225 DOI: 10.1159/000368399] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 09/17/2014] [Indexed: 11/19/2022] Open
Abstract
The most common form of sensory disability is age-related hearing loss (ARHL), also referred to as presbycusis. ARHL is a complex disorder with a mixture of genetic and environmental components, a combination that leads to a progressive decline in hearing function with increased age. In the last 15 years, there has been a vast increase in our knowledge of the genes that underlie congenital deafness and the critical components of hearing. In contrast, knowledge of the pathological processes involved in ARHL remains very limited. The mouse has proved an essential tool in the identification of early-onset deafness genes and in revealing the basic mechanisms of hearing. As focus is now turning toward elucidating the most common form of hearing loss, ARHL, the mouse will again play a fundamental role in this research. Here, we review the need for an animal model and discuss the suitability of the mouse as an ARHL model. Finally, we outline the ways in which hearing researchers are utilising the mouse in the investigation of ARHL and provide perspectives on the need for these data to be integrated with the results of human genetic studies.
Collapse
Affiliation(s)
- Michael R Bowl
- Mammalian Genetics Unit, MRC Harwell, Harwell Oxford, UK
| | | |
Collapse
|
39
|
Chen J, Ingham N, Kelly J, Jadeja S, Goulding D, Pass J, Mahajan VB, Tsang SH, Nijnik A, Jackson IJ, White JK, Forge A, Jagger D, Steel KP. Spinster homolog 2 (spns2) deficiency causes early onset progressive hearing loss. PLoS Genet 2014; 10:e1004688. [PMID: 25356849 PMCID: PMC4214598 DOI: 10.1371/journal.pgen.1004688] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 08/19/2014] [Indexed: 12/13/2022] Open
Abstract
Spinster homolog 2 (Spns2) acts as a Sphingosine-1-phosphate (S1P) transporter in zebrafish and mice, regulating heart development and lymphocyte trafficking respectively. S1P is a biologically active lysophospholipid with multiple roles in signalling. The mechanism of action of Spns2 is still elusive in mammals. Here, we report that Spns2-deficient mice rapidly lost auditory sensitivity and endocochlear potential (EP) from 2 to 3 weeks old. We found progressive degeneration of sensory hair cells in the organ of Corti, but the earliest defect was a decline in the EP, suggesting that dysfunction of the lateral wall was the primary lesion. In the lateral wall of adult mutants, we observed structural changes of marginal cell boundaries and of strial capillaries, and reduced expression of several key proteins involved in the generation of the EP (Kcnj10, Kcnq1, Gjb2 and Gjb6), but these changes were likely to be secondary. Permeability of the boundaries of the stria vascularis and of the strial capillaries appeared normal. We also found focal retinal degeneration and anomalies of retinal capillaries together with anterior eye defects in Spns2 mutant mice. Targeted inactivation of Spns2 in red blood cells, platelets, or lymphatic or vascular endothelial cells did not affect hearing, but targeted ablation of Spns2 in the cochlea using a Sox10-Cre allele produced a similar auditory phenotype to the original mutation, suggesting that local Spns2 expression is critical for hearing in mammals. These findings indicate that Spns2 is required for normal maintenance of the EP and hence for normal auditory function, and support a role for S1P signalling in hearing. Progressive hearing loss is common in the human population but we know very little about the molecular mechanisms involved. Mutant mice are useful for investigating these mechanisms and have revealed a wide range of different abnormalities that can all lead to the same outcome: deafness. We report here our findings of a new mouse line with a mutation in the Spns2 gene, affecting the release of a lipid called sphingosine-1-phosphate, which has an important role in several processes in the body. For the first time, we report that this molecular pathway is required for normal hearing through a role in generating a voltage difference that acts like a battery, allowing the sensory hair cells of the cochlea to detect sounds at extremely low levels. Without the normal function of the Spns2 gene and release of sphingosine-1-phosphate locally in the inner ear, the voltage in the cochlea declines, leading to rapid loss of sensitivity to sound and ultimately to complete deafness. The human version of this gene, SPNS2, may be involved in human deafness, and understanding the underlying mechanism presents an opportunity to develop potential treatments for this form of hearing loss.
Collapse
Affiliation(s)
- Jing Chen
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | - Neil Ingham
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | - John Kelly
- Centre for Auditory Research, UCL Ear Institute, London, United Kingdom
| | - Shalini Jadeja
- MRC Human Genetics Unit, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom, and Roslin Institute, University of Edinburgh, Easter Bush, United Kingdom
| | - David Goulding
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
| | - Johanna Pass
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | - Vinit B. Mahajan
- Omics Laboratory, University of Iowa, Iowa City, Iowa, United States of America
| | - Stephen H. Tsang
- Edward S. Harkness Eye Institute, Columbia University, New York, New York, United States of America
| | - Anastasia Nijnik
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
- Department of Physiology, Complex Traits Group, McGill University, Montreal, Quebec, Canada
| | - Ian J. Jackson
- MRC Human Genetics Unit, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom, and Roslin Institute, University of Edinburgh, Easter Bush, United Kingdom
| | | | - Andrew Forge
- Centre for Auditory Research, UCL Ear Institute, London, United Kingdom
| | - Daniel Jagger
- Centre for Auditory Research, UCL Ear Institute, London, United Kingdom
| | - Karen P. Steel
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
40
|
Raft S, Coate TM, Kelley MW, Crenshaw EB, Wu DK. Pou3f4-mediated regulation of ephrin-b2 controls temporal bone development in the mouse. PLoS One 2014; 9:e109043. [PMID: 25299585 PMCID: PMC4192298 DOI: 10.1371/journal.pone.0109043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 09/01/2014] [Indexed: 12/25/2022] Open
Abstract
The temporal bone encases conductive and sensorineural elements of the ear. Mutations of POU3F4 are associated with unique temporal bone abnormalities and X-linked mixed deafness (DFNX2/DFN3). However, the target genes and developmental processes controlled by POU3F4 transcription factor activity have remained largely uncharacterized. Ephrin-B2 (Efnb2) is a signaling molecule with well-documented effects on cell adhesion, proliferation, and migration. Our analyses of targeted mouse mutants revealed that Efnb2 loss-of-function phenocopies temporal bone abnormalities of Pou3f4 hemizygous null neonates: qualitatively identical malformations of the stapes, styloid process, internal auditory canal, and cochlear capsule were present in both mutants. Using failed/insufficient separation of the stapes and styloid process as a quantitative trait, we found that single gene Efnb2 loss-of-function and compound Pou3f4/Efnb2 loss-of-function caused a more severe phenotype than single gene Pou3f4 loss-of-function. Pou3f4 and Efnb2 gene expression domains overlapped at the site of impending stapes-styloid process separation and at subcapsular mesenchyme surrounding the cochlea; at both these sites, Efnb2 expression was attenuated in Pou3f4 hemizygous null mutants relative to control. Results of immunoprecipitation experiments using chromatin isolated from nascent middle ear mesenchyme supported the hypothesis of a physical association between Pou3f4 and specific non-coding sequence of Efnb2. We propose that Efnb2 is a target of Pou3f4 transcription factor activity and an effector of mesenchymal patterning during temporal bone development.
Collapse
Affiliation(s)
- Steven Raft
- Section on Sensory Cell Regeneration and Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Thomas M. Coate
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Matthew W. Kelley
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, United States of America
| | - E. Bryan Crenshaw
- Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Doris K. Wu
- Section on Sensory Cell Regeneration and Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
41
|
Deficiency of transcription factor Brn4 disrupts cochlear gap junction plaques in a model of DFN3 non-syndromic deafness. PLoS One 2014; 9:e108216. [PMID: 25259580 PMCID: PMC4178122 DOI: 10.1371/journal.pone.0108216] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 08/26/2014] [Indexed: 11/19/2022] Open
Abstract
Brn4, which encodes a POU transcription factor, is the gene responsible for DFN3, an X chromosome-linked, non-syndromic type of hearing loss. Brn4-deficient mice have a low endocochlear potential (EP), hearing loss, and ultrastructural alterations in spiral ligament fibrocytes, however the molecular pathology through which Brn4 deficiency causes low EP is still unclear. Mutations in the Gjb2 and Gjb6 genes encoding the gap junction proteins connexin26 (Cx26) and connexin30 (Cx30) genes, respectively, which encode gap junction proteins and are expressed in cochlear fibrocytes and non-sensory epithelial cells (i.e., cochlear supporting cells) to maintain the proper EP, are responsible for hereditary sensorineural deafness. It has been hypothesized that the gap junction in the cochlea provides an intercellular passage by which K+ is transported to maintain the EP at the high level necessary for sensory hair cell excitation. Here we analyzed the formation of gap junction plaques in cochlear supporting cells of Brn4-deficient mice at different stages by confocal microscopy and three-dimensional graphic reconstructions. Gap junctions from control mice, which are composed mainly of Cx26 and Cx30, formed linear plaques along the cell-cell junction sites with adjacent cells. These plaques formed pentagonal or hexagonal outlines of the normal inner sulcus cells and border cells. Gap junction plaques in Brn4-deficient mice did not, however, show the normal linear structure but instead formed small spots around the cell-cell junction sites. Gap junction lengths were significantly shorter, and the level of Cx26 and Cx30 was significantly reduced in Brn4-deficient mice compared with littermate controls. Thus the Brn4 mutation affected the assembly and localization of gap junction proteins at the cell borders of cochlear supporting cells, suggesting that Brn4 substantially contributes to cochlear gap junction properties to maintain the proper EP in cochleae, similar to connexin-related deafness.
Collapse
|
42
|
Mizutari K. Spontaneous recovery of cochlear fibrocytes after severe degeneration caused by acute energy failure. Front Pharmacol 2014; 5:198. [PMID: 25206337 PMCID: PMC4143613 DOI: 10.3389/fphar.2014.00198] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 08/11/2014] [Indexed: 12/03/2022] Open
Abstract
Cochlear fibrocytes in the lateral wall region play a critical role in the regulation of inner ear ion and fluid homeostasis, although these are non-sensory cells. Along with other non-sensory cells, fibrocytes in the spiral ligament have been reported to repopulate themselves after damage. However, the studies of regeneration of cochlear fibrocytes have been difficult because a suitable fibrocyte-specific degeneration model did not exist. Therefore, we analyzed cochlear fibrocytes using a rat model of acute cochlear energy failure induced by a mitochondrial toxin. This model is unique because hearing loss is caused by apoptosis of fibrocytes in the cochlear lateral wall not by damage to sensory cells. Although this model involves severe damage to the cochlear lateral wall, delayed spontaneous regeneration occurs without any treatment. Moreover, partial hearing recovery is accompanied by morphological remodeling of the cochlear lateral wall. Two hypotheses are conceivable regarding this spontaneous recovery of cochlear fibrocytes. One is that residual cochlear fibrocytes proliferate spontaneously, followed by remodeling of the functional region of the lateral wall. Another is that some foreign cells such as bone marrow-derived cells promote morphological and functional recovery of the lateral wall. Acceleration of the lateral wall recovery promoted by these mechanisms may be a new therapeutic strategy against hearing loss.
Collapse
Affiliation(s)
- Kunio Mizutari
- Department of Otolaryngology, National Defense Medical College, Saitama Japan
| |
Collapse
|
43
|
Li X, Shi X, Qiao Y, Xu K, Zeng L, Wang C, Xu Z, Niu H. Observation of permeability of blood-labyrinth barrier during cytomegalovirus-induced hearing loss. Int J Pediatr Otorhinolaryngol 2014; 78:995-9. [PMID: 24814236 DOI: 10.1016/j.ijporl.2014.03.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 03/17/2014] [Accepted: 03/18/2014] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Congenital cytomegalovirus (CMV) infection is the most common infectious cause of sensorineural hearing loss in children. This study aims to investigate the pathogenesis CMV-induced hearing loss from the view of integrity of blood-labyrinth-barrier (BLB). METHODS Newborn BALB/c mice were randomly divided into three groups (n=22, respectively): CMV group, control group and normal group. The CMV group and control group were intracerebrally injected with equal volume (15 μl) of murine CMV (MCMV; 10(4)IU/0.1 ml) and PBS, respectively, and normal group did not receive any treatment. After three weeks, auditory-evoked brainstem response was assessed, and permeability of BLB was evaluated by Evans blue method. Means between groups were compared using t-test. RESULTS We observed that mice injected with MCMV had a hearing loss and it was connected with the permeability changes of BLB. Besides, using hematoxylin-eosin staining, we noticed hyperaemia in stria vascularis and spiral ligament and bleeding in scala vestibule and scala tympani in CMV group. CONCLUSION All these data indicated the possible association between CMV-induced hearing loss and BLB dysfunction with the characteristics of inflammation. Our data provide a possible path to investigate the mechanism of CMV-induced hearing damage.
Collapse
Affiliation(s)
- Xuanyi Li
- Department of Otolaryngology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou 221006, China
| | - Xi Shi
- Institute of Audiology and Speech Science of Xuzhou Medical College, Xuzhou 221006, China
| | - Yuehua Qiao
- Department of Otolaryngology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou 221006, China; Institute of Audiology and Speech Science of Xuzhou Medical College, Xuzhou 221006, China.
| | - Kailin Xu
- Laboratory of Transplantation Immunity of Xuzhou Medical College, Xuzhou 221006, China
| | - Lingyu Zeng
- Laboratory of Transplantation Immunity of Xuzhou Medical College, Xuzhou 221006, China
| | - Caiji Wang
- Department of Otolaryngology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou 221006, China
| | - Zhou Xu
- School of Basic Education Science of Xuzhou Medical College, Xuzhou 221006, China
| | - Haichen Niu
- Institute of Audiology and Speech Science of Xuzhou Medical College, Xuzhou 221006, China
| |
Collapse
|
44
|
Hao X, Xing Y, Moore MW, Zhang J, Han D, Schulte BA, Dubno JR, Lang H. Sox10 expressing cells in the lateral wall of the aged mouse and human cochlea. PLoS One 2014; 9:e97389. [PMID: 24887110 PMCID: PMC4041576 DOI: 10.1371/journal.pone.0097389] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 04/17/2014] [Indexed: 12/20/2022] Open
Abstract
Age-related hearing loss (presbycusis) is a common human disorder, affecting one in three Americans aged 60 and over. Previous studies have shown that presbyacusis is associated with a loss of non-sensory cells in the cochlear lateral wall. Sox10 is a transcription factor crucial to the development and maintenance of neural crest-derived cells including some non-sensory cell types in the cochlea. Mutations of the Sox10 gene are known to cause various combinations of hearing loss and pigmentation defects in humans. This study investigated the potential relationship between Sox10 gene expression and pathological changes in the cochlear lateral wall of aged CBA/CaJ mice and human temporal bones from older donors. Cochlear tissues prepared from young adult (1–3 month-old) and aged (2–2.5 year-old) mice, and human temporal bone donors were examined using quantitative immunohistochemical analysis and transmission electron microscopy. Cells expressing Sox10 were present in the stria vascularis, outer sulcus and spiral prominence in mouse and human cochleas. The Sox10+ cell types included marginal and intermediate cells and outer sulcus cells, including those that border the scala media and those extending into root processes (root cells) in the spiral ligament. Quantitative analysis of immunostaining revealed a significant decrease in the number of Sox10+ marginal cells and outer sulcus cells in aged mice. Electron microscopic evaluation revealed degenerative alterations in the surviving Sox10+ cells in aged mice. Strial marginal cells in human cochleas from donors aged 87 and older showed only weak immunostaining for Sox10. Decreases in Sox10 expression levels and a loss of Sox10+ cells in both mouse and human aged ears suggests an important role of Sox10 in the maintenance of structural and functional integrity of the lateral wall. A loss of Sox10+ cells may also be associated with a decline in the repair capabilities of non-sensory cells in the aged ear.
Collapse
Affiliation(s)
- Xinping Hao
- Department of Otolaryngology – Head & Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Yazhi Xing
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Michael W. Moore
- Department of Otolaryngology – Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Jianning Zhang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Department of Otolaryngology, Shanghai Yueyang Integrated Medicine Hospital, Shanghai, China
| | - Demin Han
- Department of Otolaryngology – Head & Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- * E-mail: (HL); (DH)
| | - Bradley A. Schulte
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Department of Otolaryngology – Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Judy R. Dubno
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Department of Otolaryngology – Head & Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Hainan Lang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- * E-mail: (HL); (DH)
| |
Collapse
|
45
|
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: 2.1] [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
|
46
|
A modular gain-of-function approach to generate cortical interneuron subtypes from ES cells. Neuron 2014; 80:1145-58. [PMID: 24314726 DOI: 10.1016/j.neuron.2013.09.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2013] [Indexed: 01/18/2023]
Abstract
Whereas past work indicates that cortical interneurons (cINs) can be generically produced from stem cells, generating large numbers of specific subtypes of this population has remained elusive. This reflects an information gap in our understanding of the transcriptional programs required for different interneuron subtypes. Here, we have utilized the directed differentiation of stem cells into specific subpopulations of cortical interneurons as a means to identify some of these missing factors. To establish this approach, we utilized two factors known to be required for the generation of cINs, Nkx2-1 and Dlx2. As predicted, their regulated transient expression greatly improved the differentiation efficiency and specificity over baseline. We extended upon this "cIN-primed" model in order to establish a modular system whereby a third transcription factor could be systematically introduced. Using this approach, we identified Lmo3 and Pou3f4 as genes that can augment the differentiation and/or subtype specificity of cINs in vitro.
Collapse
|
47
|
The expression of PTEN in the development of mouse cochlear lateral wall. Neuroscience 2014; 258:263-9. [DOI: 10.1016/j.neuroscience.2013.11.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 11/07/2013] [Accepted: 11/07/2013] [Indexed: 12/20/2022]
|
48
|
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: 3.1] [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
|
49
|
Nayak G, Lee SI, Yousaf R, Edelmann SE, Trincot C, Van Itallie CM, Sinha GP, Rafeeq M, Jones SM, Belyantseva IA, Anderson JM, Forge A, Frolenkov GI, Riazuddin S. Tricellulin deficiency affects tight junction architecture and cochlear hair cells. J Clin Invest 2013; 123:4036-49. [PMID: 23979167 DOI: 10.1172/jci69031] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 05/30/2013] [Indexed: 01/06/2023] Open
Abstract
The two compositionally distinct extracellular cochlear fluids, endolymph and perilymph, are separated by tight junctions that outline the scala media and reticular lamina. Mutations in TRIC (also known as MARVELD2), which encodes a tricellular tight junction protein known as tricellulin, lead to nonsyndromic hearing loss (DFNB49). We generated a knockin mouse that carries a mutation orthologous to the TRIC coding mutation linked to DFNB49 hearing loss in humans. Tricellulin was absent from the tricellular junctions in the inner ear epithelia of the mutant animals, which developed rapidly progressing hearing loss accompanied by loss of mechanosensory cochlear hair cells, while the endocochlear potential and paracellular permeability of a biotin-based tracer in the stria vascularis were unaltered. Freeze-fracture electron microscopy revealed disruption of the strands of intramembrane particles connecting bicellular and tricellular junctions in the inner ear epithelia of tricellulin-deficient mice. These ultrastructural changes may selectively affect the paracellular permeability of ions or small molecules, resulting in a toxic microenvironment for cochlear hair cells. Consistent with this hypothesis, hair cell loss was rescued in tricellulin-deficient mice when generation of normal endolymph was inhibited by a concomitant deletion of the transcription factor, Pou3f4. Finally, comprehensive phenotypic screening showed a broader pathological phenotype in the mutant mice, which highlights the non-redundant roles played by tricellulin.
Collapse
Affiliation(s)
- Gowri Nayak
- Laboratory of Molecular Genetics, Division of Pediatric Otolaryngology / Head and Neck Surgery, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio 45229, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
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: 1.2] [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
|