1
|
Kostrikov S, Hjortkjaer J, Dau T, Corfas G, Liberman LD, Liberman MC. A modiolar-pillar gradient in auditory-nerve dendritic length: A novel post-synaptic contribution to dynamic range? Hear Res 2025; 456:109172. [PMID: 39708764 PMCID: PMC11772111 DOI: 10.1016/j.heares.2024.109172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/06/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Auditory-nerve fibers (ANFs) from a given cochlear region can vary in threshold sensitivity by up to 60 dB, corresponding to a 1000-fold difference in stimulus level, although each fiber innervates a single inner hair cell (IHC) via a single synapse. ANFs with high-thresholds also have low spontaneous rates (SRs) and synapse on the side of the IHC closer to the modiolus, whereas the low-threshold, high-SR fibers synapse on the side closer to the pillar cells. Prior biophysical work has identified modiolar-pillar differences in both pre- and post-synaptic properties, but a comprehensive explanation for the wide range of sensitivities remains elusive. Here, in guinea pigs, we used immunostaining for several neuronal markers, including Caspr, a key protein in nodes of Ranvier, to reveal a novel modiolar-pillar gradient in the location of the first ANF heminodes, presumed to be the site of the spike generator, just outside the sensory epithelium. Along the cochlea, from apex to base, the unmyelinated terminal dendrites of modiolar ANFs were 2-4 times longer than those of pillar ANFs. This modiolar-pillar gradient in dendritic length, coupled with the 2-4 fold smaller caliber of modiolar dendrites seen in prior single-fiber labeling studies, suggests there could be a large difference in the number of length constants between the synapse and the spike initiation zone for low- vs high-SR fibers. The resultant differences in attenuation of post-synaptic potentials propagating along these unmyelinated dendrites could be a key contributor to the observed range of threshold sensitivities among ANFs.
Collapse
Affiliation(s)
- Serhii Kostrikov
- Department of Health Technology, Centre for Auditory Neuroscience, Hearing Systems, Technical University of Denmark, Lyngby, Denmark
| | - Jens Hjortkjaer
- Department of Health Technology, Centre for Auditory Neuroscience, Hearing Systems, Technical University of Denmark, Lyngby, Denmark
| | - Torsten Dau
- Department of Health Technology, Centre for Auditory Neuroscience, Hearing Systems, Technical University of Denmark, Lyngby, Denmark
| | - Gabriel Corfas
- Department of Otolaryngology-Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI, United States
| | - Leslie D Liberman
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA, United States
| | - M Charles Liberman
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA, United States; Department of Otolaryngology-Head & Neck Surgery, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
2
|
Siebald C, Vincent PFY, Bottom RT, Sun S, Reijntjes DOJ, Manca M, Glowatzki E, Müller U. Molecular signatures define subtypes of auditory afferents with distinct peripheral projection patterns and physiological properties. Proc Natl Acad Sci U S A 2023; 120:e2217033120. [PMID: 37487063 PMCID: PMC10400978 DOI: 10.1073/pnas.2217033120] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 06/06/2023] [Indexed: 07/26/2023] Open
Abstract
Type I spiral ganglion neurons (SGNs) are the auditory afferents that transmit sound information from cochlear inner hair cells (IHCs) to the brainstem. These afferents consist of physiological subtypes that differ in their spontaneous firing rate (SR), activation threshold, and dynamic range and have been described as low, medium, and high SR fibers. Lately, single-cell RNA sequencing experiments have revealed three molecularly defined type I SGN subtypes. The extent to which physiological type I SGN subtypes correspond to molecularly defined subtypes is unclear. To address this question, we have generated mouse lines expressing CreERT2 in SGN subtypes that allow for a physiological assessment of molecular subtypes. We show that Lypd1-CreERT2 expressing SGNs represent a well-defined group of neurons that preferentially innervate the IHC modiolar side and exhibit a narrow range of low SRs. In contrast, Calb2-CreERT2 expressing SGNs preferentially innervate the IHC pillar side and exhibit a wider range of SRs, thus suggesting that a strict stratification of all SGNs into three molecular subclasses is not obvious, at least not with the CreERT2 tools used here. Genetically marked neuronal subtypes refine their innervation specificity onto IHCs postnatally during the time when activity is required to refine their molecular phenotype. Type I SGNs thus consist of genetically defined subtypes with distinct physiological properties and innervation patterns. The molecular subtype-specific lines characterized here will provide important tools for investigating the role of the physiologically distinct type I SGNs in encoding sound signals.
Collapse
Affiliation(s)
- Caroline Siebald
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Philippe F. Y. Vincent
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Riley T. Bottom
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Shuohao Sun
- National Institute of Biological Science, Beijing102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing100084, China
| | - Daniel O. J. Reijntjes
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Marco Manca
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Elisabeth Glowatzki
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Ulrich Müller
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
| |
Collapse
|
3
|
Liu W, Johansson Å, Rask-Andersen H, Rask-Andersen M. A combined genome-wide association and molecular study of age-related hearing loss in H. sapiens. BMC Med 2021; 19:302. [PMID: 34847940 PMCID: PMC8638543 DOI: 10.1186/s12916-021-02169-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 10/21/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Sensorineural hearing loss is one of the most common sensory deficiencies. However, the molecular contribution to age-related hearing loss is not fully elucidated. METHODS We performed genome-wide association studies (GWAS) for hearing loss-related traits in the UK Biobank (N = 362,396) and selected a high confidence set of ten hearing-associated gene products for staining in human cochlear samples: EYA4, LMX1A, PTK2/FAK, UBE3B, MMP2, SYNJ2, GRM5, TRIOBP, LMO-7, and NOX4. RESULTS All proteins were found to be expressed in human cochlear structures. Our findings illustrate cochlear structures that mediate mechano-electric transduction of auditory stimuli, neuronal conductance, and neuronal plasticity to be involved in age-related hearing loss. CONCLUSIONS Our results suggest common genetic variation to influence structural resilience to damage as well as cochlear recovery after trauma, which protect against accumulated damage to cochlear structures and the development of hearing loss over time.
Collapse
Affiliation(s)
- Wei Liu
- Department of Surgical Sciences, Section of Otorhinolaryngology and Head & Neck Surgery, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Åsa Johansson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Helge Rask-Andersen
- Department of Surgical Sciences, Section of Otorhinolaryngology and Head & Neck Surgery, Uppsala University, SE-751 85, Uppsala, Sweden.
| | - Mathias Rask-Andersen
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
4
|
Grandi FC, De Tomasi L, Mustapha M. Single-Cell RNA Analysis of Type I Spiral Ganglion Neurons Reveals a Lmx1a Population in the Cochlea. Front Mol Neurosci 2020; 13:83. [PMID: 32523514 PMCID: PMC7261882 DOI: 10.3389/fnmol.2020.00083] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 04/24/2020] [Indexed: 12/13/2022] Open
Abstract
In the mature cochlea, each inner hair cell (IHC) is innervated by multiple spiral ganglion neurons of type I (SGNI). SGNIs are morphologically and electro-physiologically diverse. Also, they differ in their susceptibility to noise insult. However, the molecular underpinnings of their identity and physiological differences remain poorly understood. In this study, we developed a novel triple transgenic mouse, which enabled the isolation of pure populations of SGNIs and the analysis of a 96-gene panel via single-cell qPCR. We found three distinct populations of Type I SGNs, which were marked by their exclusive expression of Lmx1a, Slc4a4, or Mfap4/Fzd2, respectively, at postnatal days P3, P8, and P12. Our data suggest that afferent SGN subtypes are established genetically before the onset of hearing and that the expression of key physiological markers, such as ion channels, is heterogeneous and may be underlying the heterogeneous firing proprieties of SGNIs.
Collapse
Affiliation(s)
| | - Lara De Tomasi
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Mirna Mustapha
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom.,Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
5
|
Johnson SL, Safieddine S, Mustapha M, Marcotti W. Hair Cell Afferent Synapses: Function and Dysfunction. Cold Spring Harb Perspect Med 2019; 9:a033175. [PMID: 30617058 PMCID: PMC6886459 DOI: 10.1101/cshperspect.a033175] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
To provide a meaningful representation of the auditory landscape, mammalian cochlear hair cells are optimized to detect sounds over an incredibly broad range of frequencies and intensities with unparalleled accuracy. This ability is largely conferred by specialized ribbon synapses that continuously transmit acoustic information with high fidelity and sub-millisecond precision to the afferent dendrites of the spiral ganglion neurons. To achieve this extraordinary task, ribbon synapses employ a unique combination of molecules and mechanisms that are tailored to sounds of different frequencies. Here we review the current understanding of how the hair cell's presynaptic machinery and its postsynaptic afferent connections are formed, how they mature, and how their function is adapted for an accurate perception of sound.
Collapse
Affiliation(s)
- Stuart L Johnson
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - Saaid Safieddine
- UMRS 1120, Institut Pasteur, Paris, France
- Sorbonne Universités, UPMC Université Paris 06, Complexité du Vivant, Paris, France
| | - Mirna Mustapha
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, United Kingdom
- Department of Otolaryngology-Head & Neck Surgery, Stanford University, Stanford, California 94035
| | - Walter Marcotti
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, United Kingdom
| |
Collapse
|
6
|
Yu Y, Hu B, Bao J, Mulvany J, Bielefeld E, Harrison RT, Neton SA, Thirumala P, Chen Y, Lei D, Qiu Z, Zheng Q, Ren J, Perez-Flores MC, Yamoah EN, Salehi P. Otoprotective Effects of Stephania tetrandra S. Moore Herb Isolate against Acoustic Trauma. J Assoc Res Otolaryngol 2018; 19:653-668. [PMID: 30187298 PMCID: PMC6249158 DOI: 10.1007/s10162-018-00690-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 07/08/2018] [Indexed: 01/10/2023] Open
Abstract
Noise is the most common occupational and environmental hazard, and noise-induced hearing loss (NIHL) is the second most common form of sensorineural hearing deficit. Although therapeutics that target the free-radical pathway have shown promise, none of these compounds is currently approved against NIHL by the United States Food and Drug Administration. The present study has demonstrated that tetrandrine (TET), a traditional Chinese medicinal alkaloid and the main chemical isolate of the Stephania tetrandra S. Moore herb, significantly attenuated NIHL in CBA/CaJ mice. TET is known to exert antihypertensive and antiarrhythmic effects through the blocking of calcium channels. Whole-cell patch-clamp recording from adult spiral ganglion neurons showed that TET blocked the transient Ca2+ current in a dose-dependent manner and the half-blocking concentration was 0.6 + 0.1 μM. Consistent with previous findings that modulations of calcium-based signaling pathways have both prophylactic and therapeutic effects against neural trauma, NIHL was significantly diminished by TET administration. Importantly, TET has a long-lasting protective effect after noise exposure (48 weeks) in comparison to 2 weeks after noise exposure. The otoprotective effects of TET were achieved mainly by preventing outer hair cell damage and synapse loss between inner hair cells and spiral ganglion neurons. Thus, our data indicate that TET has great potential in the prevention and treatment of NIHL.
Collapse
Affiliation(s)
- Yan Yu
- The First People’s Hospital of Zhangjiagang, 68 W Jiyang Road, Zhangjiagang City, 215600 Jiangsu China
- Translational Research Center, Northeast Ohio Medical University, Rootstown, OH 44272 USA
| | - Bing Hu
- Translational Research Center, Northeast Ohio Medical University, Rootstown, OH 44272 USA
- Department of Otolaryngology-Head and Neck Surgery, Case Western Reserve University School of Medicine, Cleveland, OH 44106 USA
- Department of Otolaryngology Head and Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, 440011 Hunan China
| | - Jianxin Bao
- Translational Research Center, Northeast Ohio Medical University, Rootstown, OH 44272 USA
- Department of Research and Development, Gateway Biotechnology Inc., Rootstown, OH 44272 USA
| | - Jessica Mulvany
- Translational Research Center, Northeast Ohio Medical University, Rootstown, OH 44272 USA
- Department of Research and Development, Gateway Biotechnology Inc., Rootstown, OH 44272 USA
| | - Eric Bielefeld
- Department of Speech and Hearing Science, Ohio State University, Columbus, OH 43210 USA
| | - Ryan T. Harrison
- Department of Speech and Hearing Science, Ohio State University, Columbus, OH 43210 USA
| | - Sarah A. Neton
- Department of Speech and Hearing Science, Ohio State University, Columbus, OH 43210 USA
| | - Partha Thirumala
- The University of Pittsburgh Medical Center, Suite B-400, 200 Lothrop Street, Pittsburgh, PA 15213 USA
| | - Yingying Chen
- Translational Research Center, Northeast Ohio Medical University, Rootstown, OH 44272 USA
| | - Debin Lei
- Translational Research Center, Northeast Ohio Medical University, Rootstown, OH 44272 USA
| | - Ziyu Qiu
- Department of Research and Development, Gateway Biotechnology Inc., Rootstown, OH 44272 USA
| | - Qingyin Zheng
- Department of Otolaryngology-Head and Neck Surgery, Case Western Reserve University School of Medicine, Cleveland, OH 44106 USA
| | - Jihao Ren
- Department of Otolaryngology Head and Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, 440011 Hunan China
| | - Maria Cristina Perez-Flores
- Department of Physiology and Cell Biology, University of Nevada Reno, 1664 North Virginia St, Reno, NV 89557 USA
| | - Ebenezer N. Yamoah
- Department of Physiology and Cell Biology, University of Nevada Reno, 1664 North Virginia St, Reno, NV 89557 USA
| | - Pezhman Salehi
- Translational Research Center, Northeast Ohio Medical University, Rootstown, OH 44272 USA
| |
Collapse
|
7
|
Ramamurthy P, White JB, Yull Park J, Hume RI, Ebisu F, Mendez F, Takayama S, Barald KF. Concomitant differentiation of a population of mouse embryonic stem cells into neuron-like cells and schwann cell-like cells in a slow-flow microfluidic device. Dev Dyn 2017; 246:7-27. [PMID: 27761977 PMCID: PMC5159187 DOI: 10.1002/dvdy.24466] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 09/16/2016] [Accepted: 09/30/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND To send meaningful information to the brain, an inner ear cochlear implant (CI) must become closely coupled to as large and healthy a population of remaining spiral ganglion neurons (SGN) as possible. Inner ear gangliogenesis depends on macrophage migration inhibitory factor (MIF), a directionally attractant neurotrophic cytokine made by both Schwann and supporting cells (Bank et al., 2012). MIF-induced mouse embryonic stem cell (mESC)-derived "neurons" could potentially substitute for lost or damaged SGN. mESC-derived "Schwann cells" produce MIF, as do all Schwann cells (Huang et al., a; Roth et al., 2007; Roth et al., 2008) and could attract SGN to a "cell-coated" implant. RESULTS Neuron- and Schwann cell-like cells were produced from a common population of mESCs in an ultra-slow-flow microfluidic device. As the populations interacted, "neurons" grew over the "Schwann cell" lawn, and early events in myelination were documented. Blocking MIF on the Schwann cell side greatly reduced directional neurite outgrowth. MIF-expressing "Schwann cells" were used to coat a CI: Mouse SGN and MIF-induced "neurons" grew directionally to the CI and to a wild-type but not MIF-knockout organ of Corti explant. CONCLUSIONS Two novel stem cell-based approaches for treating the problem of sensorineural hearing loss are described. Developmental Dynamics 246:7-27, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Poornapriya Ramamurthy
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Joshua B White
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan
| | - Joong Yull Park
- School of Mechanical Engineering, College of Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Richard I Hume
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan
| | - Fumi Ebisu
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Flor Mendez
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Shuichi Takayama
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan
| | - Kate F Barald
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
8
|
Pan C, Chu H, Lai Y, Liu Y, Sun Y, Du Z, Chen J, Tong T, Chen Q, Zhou L, Bing D, Tao Y. Down-regulation of the large conductance Ca(2+)-activated K(+) channel expression in C57BL/6J cochlea. Acta Otolaryngol 2016; 136:875-8. [PMID: 27093472 DOI: 10.3109/00016489.2016.1168941] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONCLUSION The large conductance Ca(2+)-activated K(+ )channels (BK) expression is decreased in the cochleae of age-related hearing loss (AHL) mice. BK channel may be associated with AHL. OBJECTIVE AHL is the most common among elderly persons. BK channels act as sensors for membrane voltage and intracellular Ca(2+ )and are essential for hearing. To investigate the distribution of BK channel in the cochleae of C57BL/6J mice, and the relationship between the expression of BK channel and the etiology of AHL. METHODS BK expression was studied in the cochleae of C57BL/6J mice at various ages (4, 12, 26, 52 weeks). The expressions of BK at the protein and mRNA levels were detected by immunofluorescence technique, western blot and quantitative real time PCR. RESULTS In comparison with 4-week-old mice, BK expressions in the cochleae at 12, 26 and 52 weeks of age were significantly and gradually decreased at both the protein and the mRNA levels. The immunofluorescence technique showed the BK channel was located in the hair cells and cells of the spiral ganglion, spiral ligament and stria vascularis and its expression also decreased with aging.
Collapse
Affiliation(s)
- Chunchen Pan
- a Department of Otolaryngology-Head and Neck Surgery , Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , PR China
| | - Hanqi Chu
- a Department of Otolaryngology-Head and Neck Surgery , Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , PR China
| | - Yanbing Lai
- b Department of Otolaryngology-Head and Neck Surgery , the First People's Hospital of Foshan , Foshan , Guangdong , PR China
| | - Yun Liu
- a Department of Otolaryngology-Head and Neck Surgery , Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , PR China
| | - Yanbo Sun
- a Department of Otolaryngology-Head and Neck Surgery , Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , PR China
| | - Zhihui Du
- a Department of Otolaryngology-Head and Neck Surgery , Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , PR China
| | - Jin Chen
- a Department of Otolaryngology-Head and Neck Surgery , Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , PR China
| | - Ting Tong
- a Department of Otolaryngology-Head and Neck Surgery , Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , PR China
| | - Qingguo Chen
- a Department of Otolaryngology-Head and Neck Surgery , Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , PR China
| | - Liangqiang Zhou
- a Department of Otolaryngology-Head and Neck Surgery , Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , PR China
| | - Dan Bing
- a Department of Otolaryngology-Head and Neck Surgery , Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , PR China
| | - Yanling Tao
- a Department of Otolaryngology-Head and Neck Surgery , Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , PR China
| |
Collapse
|
9
|
Kwan KY. Single-Cell Transcriptome Analysis of Developing and Regenerating Spiral Ganglion Neurons. ACTA ACUST UNITED AC 2016; 2:211-220. [PMID: 28758056 DOI: 10.1007/s40495-016-0064-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The spiral ganglion neurons (SGNs) of the cochlea are essential for our ability to hear. SGN loss after exposure to ototoxic drugs or loud noise results in hearing loss. Pluripotent stem cell-derived and endogenous progenitor cell types have the potential to become SGNs and are cellular foundations for replacement therapies. Repurposing transcriptional regulatory networks to promote SGN differentiation from progenitor cells is a strategy for regeneration. Advances in the Fludigm C1 workflow or Drop-seq allow sequencing of single cell transcriptomes to reveal variability between cells. During differentiation, the individual transcriptomes obtained from single-cell RNA-seq can be exploited to identify different cellular states. Pseudotemporal ordering of transcriptomes describes the differentiation trajectory, allows monitoring of transcriptional changes and determines molecular barriers that prevent the progression of progenitors into SGNs. Analysis of single cell transcriptomes will help develop novel strategies for guiding efficient SGN regeneration.
Collapse
Affiliation(s)
- Kelvin Y Kwan
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway, NJ 08854, USA.,Stem Cell Research Center and Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
10
|
Gilles A, Schlee W, Rabau S, Wouters K, Fransen E, Van de Heyning P. Decreased Speech-In-Noise Understanding in Young Adults with Tinnitus. Front Neurosci 2016; 10:288. [PMID: 27445661 PMCID: PMC4923253 DOI: 10.3389/fnins.2016.00288] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 06/09/2016] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Young people are often exposed to high music levels which make them more at risk to develop noise-induced symptoms such as hearing loss, hyperacusis, and tinnitus of which the latter is the symptom perceived the most by young adults. Although, subclinical neural damage was demonstrated in animal experiments, the human correlate remains under debate. Controversy exists on the underlying condition of young adults with normal hearing thresholds and noise-induced tinnitus (NIT) due to leisure noise. The present study aimed to assess differences in audiological characteristics between noise-exposed adolescents with and without NIT. METHODS A group of 87 young adults with a history of recreational noise exposure was investigated by use of the following tests: otoscopy, impedance measurements, pure-tone audiometry including high-frequencies, transient and distortion product otoacoustic emissions, speech-in-noise testing with continuous and modulated noise (amplitude-modulated by 15 Hz), auditory brainstem responses (ABR) and questionnaires.Nineteen students reported NIT due to recreational noise exposure, and their measures were compared to the non-tinnitus subjects. RESULTS No significant differences between tinnitus and non-tinnitus subjects could be found for hearing thresholds, otoacoustic emissions, and ABR results.Tinnitus subjects had significantly worse speech reception in noise compared to non-tinnitus subjects for sentences embedded in steady-state noise (mean speech reception threshold (SRT) scores, respectively -5.77 and -6.90 dB SNR; p = 0.025) as well as for sentences embedded in 15 Hz AM-noise (mean SRT scores, respectively -13.04 and -15.17 dB SNR; p = 0.013). In both groups speech reception was significantly improved during AM-15 Hz noise compared to the steady-state noise condition (p < 0.001). However, the modulation masking release was not affected by the presence of NIT. CONCLUSIONS Young adults with and without NIT did not differ regarding audiometry, OAE, and ABR.However, tinnitus patients showed decreased speech-in-noise reception. The results are discussed in the light of previous findings suggestion NIT may occur in the absence of measurable peripheral damage as reflected in speech-in-noise deficits in tinnitus subjects.
Collapse
Affiliation(s)
- Annick Gilles
- University Department of Otorhinolaryngology and Head and Neck Surgery, Antwerp University HospitalEdegem, Belgium; Department of Translational Neurosciences, Faculty of Medicine and Health Sciences, University of AntwerpWilrijk, Belgium; Department of Human and Social Welfare, University College GhentGhent, Belgium
| | - Winny Schlee
- University Department of Psychology, University of Konstanz Konstanz, Germany
| | - Sarah Rabau
- University Department of Otorhinolaryngology and Head and Neck Surgery, Antwerp University HospitalEdegem, Belgium; Department of Translational Neurosciences, Faculty of Medicine and Health Sciences, University of AntwerpWilrijk, Belgium
| | - Kristien Wouters
- Department of Translational Neurosciences, Faculty of Medicine and Health Sciences, University of AntwerpWilrijk, Belgium; University Department of Scientific Coordination and Biostatistics, Antwerp University HospitalEdegem, Belgium
| | - Erik Fransen
- Department of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp Wilrijk, Belgium
| | - Paul Van de Heyning
- University Department of Otorhinolaryngology and Head and Neck Surgery, Antwerp University HospitalEdegem, Belgium; Department of Translational Neurosciences, Faculty of Medicine and Health Sciences, University of AntwerpWilrijk, Belgium
| |
Collapse
|
11
|
Reijntjes DO, Pyott SJ. The afferent signaling complex: Regulation of type I spiral ganglion neuron responses in the auditory periphery. Hear Res 2016; 336:1-16. [DOI: 10.1016/j.heares.2016.03.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 02/12/2016] [Accepted: 03/07/2016] [Indexed: 12/19/2022]
|
12
|
Goodrich LV. Early Development of the Spiral Ganglion. THE PRIMARY AUDITORY NEURONS OF THE MAMMALIAN COCHLEA 2016. [DOI: 10.1007/978-1-4939-3031-9_2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
13
|
Quantitative analysis linking inner hair cell voltage changes and postsynaptic conductance change: a modelling study. BIOMED RESEARCH INTERNATIONAL 2015; 2015:626971. [PMID: 25654117 PMCID: PMC4299359 DOI: 10.1155/2015/626971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 08/20/2014] [Accepted: 09/04/2014] [Indexed: 01/11/2023]
Abstract
This paper presents a computational model which estimates the postsynaptic conductance change of mammalian Type I afferent peripheral process when airborne acoustic waves impact on the tympanic membrane. A model of the human auditory periphery is used to estimate the inner hair cell potential change in response to airborne sound. A generic and tunable topology of the mammalian synaptic ribbon is generated and the voltage dependence of its substructures is used to calculate discrete and probabilistic neurotransmitter vesicle release. Results suggest an almost linear relationship between increasing sound level (in dB SPL) and the postsynaptic conductance for frequencies considered too high for neurons to phase lock with (i.e., a few kHz). Furthermore coordinated vesicle release is shown for up to 300–400 Hz and a mechanism of phase shifting the subharmonic content of a stimulating signal is suggested. Model outputs suggest that strong onset response and highly synchronised multivesicular release rely on compound fusion of ribbon tethered vesicles.
Collapse
|
14
|
Liu Q, Lee E, Davis RL. Heterogeneous intrinsic excitability of murine spiral ganglion neurons is determined by Kv1 and HCN channels. Neuroscience 2013; 257:96-110. [PMID: 24200924 DOI: 10.1016/j.neuroscience.2013.10.065] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Revised: 10/02/2013] [Accepted: 10/26/2013] [Indexed: 02/03/2023]
Abstract
The spiral ganglion conveys afferent auditory information predominantly through a single class of type I neurons that receive signals from inner hair cell sensory receptors. These auditory primary afferents, like in other systems (Puopolo and Belluzzi, 1998; Gascon and Moqrich, 2010; Leao et al., 2012) possess a marked diversity in their electrophysiological features (Taberner and Liberman, 2005). Consistent with these observations, when the auditory primary afferents were assessed in neuronal explants separated from their peripheral and central targets it was found that individual neurons were markedly heterogeneous in their endogenous electrophysiological features. One aspect of this heterogeneity, obvious throughout the ganglion, was their wide range of excitability as assessed by voltage threshold measurements (Liu and Davis, 2007). Thus, while neurons in the base differed significantly from apical and middle neurons in their voltage thresholds, each region showed distinctly wide ranges of values. To determine whether the resting membrane potentials (RMPs) of these neurons correlate with the threshold distribution and to identify the ion channel regulatory elements underlying heterogeneous neuronal excitability in the ganglion, patch-clamp recordings were made from postnatal day (P5-8) murine spiral ganglion neurons in vitro. We found that RMP mirrored the tonotopic threshold distribution, and contributed an additional level of heterogeneity in each cochlear location. Pharmacological experiments further indicated that threshold and RMP was coupled through the Kv1 current, which had a dual impact on both electrophysiological parameters. Whereas, hyperpolarization-activated cationic channels decoupled these two processes by primarily affecting RMP without altering threshold level. Thus, beyond mechanical and synaptic specializations, ion channel regulation of intrinsic membrane properties imbues spiral ganglion neurons with different excitability levels, a feature that contributes to primary auditory afferent diversity.
Collapse
Affiliation(s)
- Q Liu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - E Lee
- Rutgers University, New Jersey Medical School, Newark, NJ 07746, USA
| | - R L Davis
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
15
|
Wang W, Kim HJ, Lv P, Tempel B, Yamoah EN. Association of the Kv1 family of K+ channels and their functional blueprint in the properties of auditory neurons as revealed by genetic and functional analyses. J Neurophysiol 2013; 110:1751-64. [PMID: 23864368 DOI: 10.1152/jn.00290.2013] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Developmental plasticity in spiral ganglion neurons (SGNs) ensues from profound alterations in the functional properties of the developing hair cell (HC). For example, prehearing HCs are spontaneously active. However, at the posthearing stage, HC membrane properties transition to graded receptor potentials. The dendrotoxin (DTX)-sensitive Kv1 channel subunits (Kv1.1, 1.2, and 1.6) shape the firing properties and membrane potential of SGNs, and the expression of the channel undergoes developmental changes. Because of the stochastic nature of Kv subunit heteromultimerization, it has been difficult to determine physiologically relevant subunit-specific interactions and their functions in the underlying mechanisms of Kv1 channel plasticity in SGNs. Using Kcna2 null mutant mice, we demonstrate a surprising paradox in changes in the membrane properties of SGNs. The resting membrane potential of Kcna2(-/-) SGNs was significantly hyperpolarized compared with that of age-matched wild-type (WT) SGNs. Analyses of outward currents in the mutant SGNs suggest an apparent approximately twofold increase in outward K(+) currents. We show that in vivo and in vitro heteromultimerization of Kv1.2 and Kv1.4 α-subunits underlies the striking and unexpected alterations in the properties of SGNs. The results suggest that heteromeric interactions of Kv1.2 and Kv1.4 dominate the defining features of Kv1 channels in SGNs.
Collapse
Affiliation(s)
- Wenying Wang
- Program in Communication Science, Center for Neuroscience, University of California, Davis, School of Medicine, Davis, California
| | | | | | | | | |
Collapse
|
16
|
van Loon MC, Ramekers D, Agterberg MJ, de Groot JC, Grolman W, Klis SF, Versnel H. Spiral ganglion cell morphology in guinea pigs after deafening and neurotrophic treatment. Hear Res 2013; 298:17-26. [DOI: 10.1016/j.heares.2013.01.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 12/21/2012] [Accepted: 01/17/2013] [Indexed: 12/17/2022]
|
17
|
Glial cell line-derived neurotrophic factor (GDNF) induces neuritogenesis in the cochlear spiral ganglion via neural cell adhesion molecule (NCAM). Mol Cell Neurosci 2012; 54:30-43. [PMID: 23262364 DOI: 10.1016/j.mcn.2012.12.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 11/24/2012] [Accepted: 12/12/2012] [Indexed: 11/21/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) increases survival and neurite extension of spiral ganglion neurons (SGNs), the primary neurons of the auditory system, via yet unknown signaling mechanisms. In other cell types, signaling is achieved by the GPI-linked GDNF family receptor α1 (GFRα1) via recruitment of transmembrane receptors: Ret (re-arranged during transformation) and/or NCAM (neural cell adhesion molecule). Here we show that GDNF enhances neuritogenesis in organotypic cultures of spiral ganglia from 5-day-old rats and mice. Addition of GFRα1-Fc increases this effect. GDNF/GFRα1-Fc stimulation activates intracellular PI3K/Akt and MEK/Erk signaling cascades as detected by Western blot analysis of cultures prepared from rats at postnatal days 5 (P5, before the onset of hearing) and 20 (P20, after the onset of hearing). Both cascades mediate GDNF stimulation of neuritogenesis, since application of the Akt inhibitor Wortmannin or the Erk inhibitor U0126 abolished GDNF/GFRα1-Fc stimulated neuritogenesis in P5 rats. Since cultures of P5 NCAM-deficient mice failed to respond by neuritogenesis to GDNF/GFRα1-Fc, we conclude that NCAM serves as a receptor for GDNF signaling responsible for neuritogenesis in early postnatal spiral ganglion.
Collapse
|
18
|
Jin Y, Kondo K, Ushio M, Kaga K, Ryan AF, Yamasoba T. Developmental changes in the responsiveness of rat spiral ganglion neurons to neurotrophic factors in dissociated culture: differential responses for survival, neuritogenesis and neuronal morphology. Cell Tissue Res 2012; 351:15-27. [PMID: 23149719 DOI: 10.1007/s00441-012-1526-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Accepted: 10/24/2012] [Indexed: 12/26/2022]
Abstract
The way that the development of the inner ear innervation is regulated by various neurotrophic factors and/or their combinations at different postnatal developmental stages remains largely unclear. Moreover, survival and neuritogenesis in deafferented adult neurons is important for cochlear implant function. To address these issues, developmental changes in the responsiveness of postnatal rat spiral ganglion neurons (SGNs) to neurotrophin-3 (NT-3), brain-derived neurotrophic factor (BDNF) and leukemia inhibitory factor (LIF) were examined by using a dissociated cell culture system. SGNs at postnatal day (P) 0, P5 and P20 (young adult) were cultured with the addition of NT-3, BDNF, or LIF or of a combination of NT-3 and BDNF (N + B) or of NT-3, BDNF and LIF (ALL factors). SGNs were analyzed for three parameters: survival, longest neurite length (LNL) and neuronal morphology. At P0, SGNs required exposure to N + B or ALL factors for enhanced survival and the ALL factors combination showed a synergistic effect much greater than the sum of the individual factors. At P5, SGNs responded to a wider range of treatment conditions for enhanced survival and combinations showed only an additive improvement over individual factors. The survival percentage of untreated SGNs was highest at P20 but combinations of neurotrophic factors were no more effective than individual factors. LNL of each SGN was enhanced by LIF alone or ALL factors at P0 and P5 but was suppressed by NT-3, BDNF and N + B at P5 in a dose-dependent manner. The LNL at P20 was enhanced by ALL factors and suppressed by N + B. Treatment with ALL factors increased the proportion of SGNs that had two or more primary neurites in all age groups. These findings suggest that NT-3, BDNF, LIF and their combinations predominantly support different ontogenetic events at different developmental stages in the innervation of the inner ear.
Collapse
Affiliation(s)
- Yulian Jin
- Department of Otolaryngology and Head and Neck Surgery, Graduate School of Medicine, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
19
|
Budenz CL, Pfingst BE, Raphael Y. The use of neurotrophin therapy in the inner ear to augment cochlear implantation outcomes. Anat Rec (Hoboken) 2012; 295:1896-908. [PMID: 23044834 DOI: 10.1002/ar.22586] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 07/24/2012] [Indexed: 12/12/2022]
Abstract
Severe to profound deafness is most often secondary to a loss of or injury to cochlear mechanosensory cells, and there is often an associated loss of the peripheral auditory neural structures, specifically the spiral ganglion neurons and peripheral auditory fibers. Cochlear implantation is currently our best hearing rehabilitation strategy for severe to profound deafness. These implants work by directly electrically stimulating the remnant auditory neural structures within the deafened cochlea. When administered to the deafened cochlea in animal models, neurotrophins, specifically brain derived neurotrophic factor and neurotrophin-3, have been shown to dramatically improve spiral ganglion neuron survival and stimulate peripheral auditory fiber regrowth. In animal models, neurotrophins administered in combination with cochlear implantation has resulted in significant improvements in the electrophysiological and psychophysical measures of outcome. While further research must be done before these therapies can be applied clinically, neurotrophin therapies for the inner ear show great promise in enhancing CI outcomes and the treatment of hearing loss.
Collapse
Affiliation(s)
- Cameron L Budenz
- Kresge Hearing Research Institute, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | |
Collapse
|
20
|
Green SH, Bailey E, Wang Q, Davis RL. The Trk A, B, C's of Neurotrophins in the Cochlea. Anat Rec (Hoboken) 2012; 295:1877-95. [DOI: 10.1002/ar.22587] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 07/24/2012] [Indexed: 12/20/2022]
|
21
|
Needham K, Nayagam BA, Minter RL, O'Leary SJ. Combined application of brain-derived neurotrophic factor and neurotrophin-3 and its impact on spiral ganglion neuron firing properties and hyperpolarization-activated currents. Hear Res 2012; 291:1-14. [PMID: 22796476 DOI: 10.1016/j.heares.2012.07.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 06/29/2012] [Accepted: 07/03/2012] [Indexed: 01/11/2023]
Abstract
Neurotrophins provide an effective tool for the rescue and regeneration of spiral ganglion neurons (SGNs) following sensorineural hearing loss. However, these nerve growth factors are also potent modulators of ion channel activity and expression, and in the peripheral auditory system brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT3) have previously been shown to alter the firing properties of auditory neurons and differentially regulate the expression of some potassium channels in vitro. In this study we examined the activity of the hyperpolarization-mediated mixed-cation current (I(h)) in early post-natal cultured rat SGNs following exposure to combined BDNF and NT3. Whole-cell patch-clamp recordings made after 1 or 2 days in vitro revealed no change in the firing adaptation of neurons in the presence of BDNF and NT3. Resting membrane potentials were also maintained, but spike latency and firing threshold was subject to regulation by both neurotrophins and time in vitro. Current clamp recordings revealed an activity profile consistent with activation of the hyperpolarization-activated current. Rapid membrane hyperpolarization was followed by a voltage- and time-dependent depolarizing voltage sag. In voltage clamp, membrane hyperpolarization evoked a slowly-activating inward current that was reversibly blocked with cesium and inhibited by ZD7288. The amplitude and current density of I(h) was significantly larger in BDNF and NT3 supplemented cultures, but this did not translate to a significant alteration in voltage sag magnitude. Neurotrophins provided at 50 ng/ml produced a hyperpolarizing shift in the voltage-dependence and slower time course of I(h) activation compared to SGNs in control groups or cultured with 10 ng/ml BDNF and NT3. Our results indicate that combined BDNF and NT3 increase the activity of hyperpolarization-activated currents and that the voltage-dependence and activation kinetics of I(h) in SGNs are sensitive to changes in neurotrophin concentration. In addition, BDNF and NT3 applied together induce a decrease in firing threshold, but does not generate a shift in firing adaptation.
Collapse
Affiliation(s)
- Karina Needham
- Department of Otolaryngology, University of Melbourne, Royal Victorian Eye & Ear Hospital, Level 2, 32 Gisborne St., East Melbourne, Victoria 3002, Australia.
| | | | | | | |
Collapse
|
22
|
Son EJ, Wu L, Yoon H, Kim S, Choi JY, Bok J. Developmental gene expression profiling along the tonotopic axis of the mouse cochlea. PLoS One 2012; 7:e40735. [PMID: 22808246 PMCID: PMC3395647 DOI: 10.1371/journal.pone.0040735] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 06/12/2012] [Indexed: 12/13/2022] Open
Abstract
The mammalian cochlear duct is tonotopically organized such that the basal cochlea is tuned to high frequency sounds and the apical cochlea to low frequency sounds. In an effort to understand how this tonotopic organization is established, we searched for genes that are differentially expressed along the tonotopic axis during neonatal development. Cochlear tissues dissected from P0 and P8 mice were divided into three equal pieces, representing the base, middle and apex, and gene expression profiles were determined using the microarray technique. The gene expression profiles were grouped according to changes in expression levels along the tonotopic axis as well as changes during neonatal development. The classified groups were further analyzed by functional annotation clustering analysis to determine whether genes associated with specific biological function or processes are particularly enriched in each group. These analyses identified several candidate genes that may be involved in cochlear development and acquisition of tonotopy. We examined the expression domains for a few candidate genes in the developing mouse cochlea. Tnc (tenacin C) and Nov (nephroblastoma overexpressed gene) are expressed in the basilar membrane, with increased expression toward the apex, which may contribute to graded changes in the structure of the basilar membrane along the tonotopic axis. In addition, Fst (Follistatin), an antagonist of TGF-β/BMP signaling, is expressed in the lesser epithelial ridge and at gradually higher levels towards the apex. The graded expression pattern of Fst is established at the time of cochlear specification and maintained throughout embryonic and postnatal development, suggesting its possible role in the organization of tonotopy. Our data will provide a good resource for investigating the developmental mechanisms of the mammalian cochlea including the acquisition of tonotopy.
Collapse
Affiliation(s)
- Eun Jin Son
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
- * E-mail: (JB); (EJS)
| | - Ling Wu
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
- BK21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Heejei Yoon
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
| | - Sunhee Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Young Choi
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
- BK21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Jinwoong Bok
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
- BK21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
- * E-mail: (JB); (EJS)
| |
Collapse
|
23
|
Complex distribution patterns of voltage-gated calcium channel α-subunits in the spiral ganglion. Hear Res 2011; 278:52-68. [PMID: 21281707 DOI: 10.1016/j.heares.2011.01.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Revised: 01/21/2011] [Accepted: 01/21/2011] [Indexed: 01/10/2023]
Abstract
As with other elements of the peripheral auditory system, spiral ganglion neurons display specializations that vary as a function of location along the tonotopic axis. Previous work has shown that voltage-gated K(+) channels and synaptic proteins show graded changes in their density that confers rapid responsiveness to neurons in the high frequency, basal region of the cochlea and slower, more maintained responsiveness to neurons in the low frequency, apical region of the cochlea. In order to understand how voltage-gated calcium channels (VGCCs) may contribute to these diverse phenotypes, we identified the VGCC α-subunits expressed in the ganglion, investigated aspects of Ca(2+)-dependent neuronal firing patterns, and mapped the intracellular and intercellular distributions of seven VGCC α-subunits in the spiral ganglion in vitro. Initial experiments with qRT-PCR showed that eight of the ten known VGCC α-subunits were expressed in the ganglion and electrophysiological analysis revealed firing patterns that were consistent with the presence of both LVA and HVA Ca(2+) channels. Moreover, we were able to study seven of the α-subunits with immunocytochemistry, and we found that all were present in spiral ganglion neurons, three of which were neuron-specific (Ca(V)1.3, Ca(V)2.2, and Ca(V)3.3). Further characterization of neuron-specific α-subunits showed that Ca(V)1.3 and Ca(V)3.3 were tonotopically-distributed, whereas Ca(V)2.2 was uniformly distributed in apical and basal neurons. Multiple VGCC α-subunits were also immunolocalized to Schwann cells, having distinct intracellular localizations, and, significantly, appearing to distinguish putative compact (Ca(V)2.3, Ca(V)3.1) from loose (Ca(V)1.2) myelin. Electrophysiological evaluation of spiral ganglion neurons in the presence of TEA revealed Ca(2+) plateau potentials with slopes that varied proportionately with the cochlear region from which neurons were isolated. Because afterhyperpolarizations were minimal or absent under these conditions, we hypothesize that differential density and/or kinetics of one or more of the VGCC α-subunits could account for observed tonotopic differences. These experiments have set the stage for defining the clear multiplicity of functional control in neurons and Schwann cells of the spiral ganglion.
Collapse
|