1
|
Zhang B, Hu Y, Du H, Han S, Ren L, Cheng H, Wang Y, Gao X, Zheng S, Cui Q, Tian L, Liu T, Sun J, Chai R. Tissue engineering strategies for spiral ganglion neuron protection and regeneration. J Nanobiotechnology 2024; 22:458. [PMID: 39085923 PMCID: PMC11293049 DOI: 10.1186/s12951-024-02742-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024] Open
Abstract
Cochlear implants can directly activate the auditory system's primary sensory neurons, the spiral ganglion neurons (SGNs), via circumvention of defective cochlear hair cells. This bypass restores auditory input to the brainstem. SGN loss etiologies are complex, with limited mammalian regeneration. Protecting and revitalizing SGN is critical. Tissue engineering offers a novel therapeutic strategy, utilizing seed cells, biomolecules, and scaffold materials to create a cellular environment and regulate molecular cues. This review encapsulates the spectrum of both human and animal research, collating the factors contributing to SGN loss, the latest advancements in the utilization of exogenous stem cells for auditory nerve repair and preservation, the taxonomy and mechanism of action of standard biomolecules, and the architectural components of scaffold materials tailored for the inner ear. Furthermore, we delineate the potential and benefits of the biohybrid neural interface, an incipient technology in the realm of implantable devices. Nonetheless, tissue engineering requires refined cell selection and differentiation protocols for consistent SGN quality. In addition, strategies to improve stem cell survival, scaffold biocompatibility, and molecular cue timing are essential for biohybrid neural interface integration.
Collapse
Affiliation(s)
- Bin Zhang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yangnan Hu
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| | - Haoliang Du
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing University, Nanjing, 210008, China
| | - Shanying Han
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Lei Ren
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Hong Cheng
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Yusong Wang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Xin Gao
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Shasha Zheng
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Qingyue Cui
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Lei Tian
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
| | - Tingting Liu
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
| | - Jiaqiang Sun
- Department of Otolaryngology-Head and Neck Surgery, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230001, China.
| | - Renjie Chai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Public Health, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Department of Neurology, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing, China.
- Southeast University Shenzhen Research Institute, Shenzhen, 518063, China.
| |
Collapse
|
2
|
Gross J, Knipper M, Mazurek B. Candidate Key Proteins in Tinnitus: A Bioinformatic Study of Synaptic Transmission in Spiral Ganglion Neurons. Cell Mol Neurobiol 2023; 43:4189-4207. [PMID: 37736859 PMCID: PMC10661836 DOI: 10.1007/s10571-023-01405-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 08/24/2023] [Indexed: 09/23/2023]
Abstract
To study key proteins associated with changes in synaptic transmission in the spiral ganglion in tinnitus, we build three gene lists from the GeneCard database: 1. Perception of sound (PoS), 2. Acoustic stimulation (AcouStim), and 3. Tinnitus (Tin). Enrichment analysis by the DAVID database resulted in similar Gene Ontology (GO) terms for cellular components in all gene lists, reflecting synaptic structures known to be involved in auditory processing. The STRING protein-protein interaction (PPI) network and the Cytoscape data analyzer were used to identify the top two high-degree proteins (HDPs) and their high-score interaction proteins (HSIPs) identified by the combined score (CS) of the corresponding edges. The top two protein pairs (key proteins) for the PoS are BDNF-GDNF and OTOF-CACNA1D and for the AcouStim process BDNF-NTRK2 and TH-CALB1. The Tin process showed BDNF and NGF as HDPs, with high-score interactions with NTRK1 and NGFR at a comparable level. Compared to the PoS and AcouStim process, the number of HSIPs of key proteins (CS > 90. percentile) increases strongly in Tin. In the PoS and AcouStim networks, BDNF receptor signaling is the dominant pathway, and in the Tin network, the NGF-signaling pathway is of similar importance. Key proteins and their HSIPs are good indicators of biological processes and of signaling pathways characteristic for the normal hearing on the one hand and tinnitus on the other.
Collapse
Affiliation(s)
- Johann Gross
- Tinnitus Center, Charité-Universitätsmedizin Berlin, Berlin, Germany.
- Leibniz Society of Science Berlin, Berlin, Germany.
| | - Marlies Knipper
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
- Leibniz Society of Science Berlin, Berlin, Germany
| | - Birgit Mazurek
- Tinnitus Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
3
|
Bertagnoli LE, Seist R, Batts S, Stankovic KM. Potential Ototoxicity of Insulin-like Growth Factor 1 Receptor Signaling Inhibitors: An In Silico Drug Repurposing Study of the Regenerating Cochlear Neuron Transcriptome. J Clin Med 2023; 12:jcm12103485. [PMID: 37240591 DOI: 10.3390/jcm12103485] [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: 02/28/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Spiral ganglion neurons (SGNs) connect cochlear hair cells with higher auditory pathways and their degeneration due to drug toxicity (ototoxicity) contributes to hearing loss. This study aimed to identify drug classes that are negatively correlated with the transcriptome of regenerating SGNs. Human orthologs of differentially expressed genes within the regenerating neonatal mouse SGN transcriptome were entered into CMap and the LINCS unified environment and perturbation-driven gene expression was analyzed. The CMap connectivity scores ranged from 100 (positive correlation) to -100 (negative correlation). Insulin-like growth factor 1/receptor (IGF-1/R) inhibitors were highly negatively correlated with the regenerating SGN transcriptome (connectivity score: -98.87). A systematic literature review of clinical trials and observational studies reporting otologic adverse events (AEs) with IGF-1/R inhibitors identified 108 reports (6141 treated patients). Overall, 16.9% of the treated patients experienced any otologic AE; the rate was highest for teprotumumab (42.9%). In a meta-analysis of two randomized placebo-controlled trials of teprotumumab, there was a significantly higher risk of hearing-related (pooled Peto OR [95% CI]: 7.95 [1.57, 40.17]) and of any otologic AEs (3.56 [1.35, 9.43]) with teprotumumab vs. a placebo, whether or not dizziness/vertigo AEs were included. These results call for close audiological monitoring during IGF-1-targeted treatment, with prompt referral to an otolaryngologist should otologic AEs develop.
Collapse
Affiliation(s)
- Lino E Bertagnoli
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Paracelsus Medical University, 5020 Salzburg, Austria
| | - Richard Seist
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Paracelsus Medical University, 5020 Salzburg, Austria
| | - Shelley Batts
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Konstantina M Stankovic
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Wu Tsai Neuroscience Institute, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
4
|
Wang M, Xu L, Han Y, Wang X, Chen F, Lu J, Wang H, Liu W. Regulation of Spiral Ganglion Neuron Regeneration as a Therapeutic Strategy in Sensorineural Hearing Loss. Front Mol Neurosci 2022; 14:829564. [PMID: 35126054 PMCID: PMC8811300 DOI: 10.3389/fnmol.2021.829564] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 12/27/2021] [Indexed: 12/16/2022] Open
Abstract
In the mammalian cochlea, spiral ganglion neurons (SGNs) are the primary neurons on the auditory conduction pathway that relay sound signals from the inner ear to the brainstem. However, because the SGNs lack the regeneration ability, degeneration and loss of SGNs cause irreversible sensorineural hearing loss (SNHL). Besides, the effectiveness of cochlear implant therapy, which is the major treatment of SNHL currently, relies on healthy and adequate numbers of intact SGNs. Therefore, it is of great clinical significance to explore how to regenerate the SGNs. In recent years, a number of researches have been performed to improve the SGNs regeneration strategy, and some of them have shown promising results, including the progress of SGN regeneration from exogenous stem cells transplantation and endogenous glial cells’ reprogramming. Yet, there are challenges faced in the effectiveness of SGNs regeneration, the maturation and function of newly generated neurons as well as auditory function recovery. In this review, we describe recent advances in researches in SGNs regeneration. In the coming years, regenerating SGNs in the cochleae should become one of the leading biological strategies to recover hearing loss.
Collapse
|
5
|
Jeong M, Bojkovic K, Sagi V, Stankovic KM. Molecular and Clinical Significance of Fibroblast Growth Factor 2 in Development and Regeneration of the Auditory System. Front Mol Neurosci 2022; 14:757441. [PMID: 35002617 PMCID: PMC8733209 DOI: 10.3389/fnmol.2021.757441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/18/2021] [Indexed: 01/25/2023] Open
Abstract
The fibroblast growth factor 2 (FGF2) is a member of the FGF family which is involved in key biological processes including development, cellular proliferation, wound healing, and angiogenesis. Although the utility of the FGF family as therapeutic agents has attracted attention, and FGF2 has been studied in several clinical contexts, there remains an incomplete understanding of the molecular and clinical function of FGF2 in the auditory system. In this review, we highlight the role of FGF2 in inner ear development and hearing protection and present relevant clinical studies for tympanic membrane (TM) repair. We conclude by discussing the future implications of FGF2 as a potential therapeutic agent.
Collapse
Affiliation(s)
- Minjin Jeong
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, United States.,Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear and Harvard Medical School, Boston, MA, United States
| | - Katarina Bojkovic
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear and Harvard Medical School, Boston, MA, United States
| | - Varun Sagi
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, United States.,University of Minnesota Medical School, Minneapolis, MN, United States
| | - Konstantina M Stankovic
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, United States.,Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
6
|
Peng WH, Liao ML, Huang WC, Liu PK, Levi SR, Tseng YJ, Lee CY, Yeh LK, Chen KJ, Chien CL, Wang NK. Conditional Deletion of Activating Rearranged During Transfection Receptor Tyrosine Kinase Leads to Impairment of Photoreceptor Ribbon Synapses and Disrupted Visual Function in Mice. Front Neurosci 2021; 15:728905. [PMID: 34803580 PMCID: PMC8602685 DOI: 10.3389/fnins.2021.728905] [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: 06/30/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022] Open
Abstract
Purpose: The rearranged during transfection (RET) receptor tyrosine kinase plays a key role in transducing signals related to cell growth and differentiation. Ret mutant mice show abnormal retinal activity and abnormal levels and morphology of bipolar cells, yet die on the 21st day after birth as a result of renal underdevelopment. To extend the observation period, we generated the Ret conditional knockout Chx10-Cre;C-Ret lx/lx mouse model and analyzed the retinal function and morphological changes in mature and aging Chx10-Cre;C-Ret lx/lx mice. Methods: Retina-specific depletion of Ret was achieved using mice with floxed alleles of the Ret gene with CHX10-driven Cre recombinase; floxed mice without Cre expression were used as controls. Retinal function was examined using electroretinography (ERG), and 2-, 4-, 12-, and 24-month-old mice were analyzed by hematoxylin staining and immunohistochemistry to evaluate retinal morphological alterations. The ultrastructure of photoreceptor synapses was evaluated using electron microscopy. Results: The results of the ERG testing showed that b-wave amplitudes were reduced in Chx10-Cre;C-Ret lx/lx mice, whereas a-waves were not affected. A histopathological analysis revealed a thinner and disorganized outer plexiform layer at the ages of 12 and 24 months in Chx10-Cre;C-Ret lx/lx mice. Moreover, the data provided by immunohistochemistry showed defects in the synapses of photoreceptor cells. This result was confirmed at the ultrastructural level, thus supporting the participation of Ret in the morphological changes of the synaptic ribbon. Conclusion: Our results provide evidence of the role of Ret in maintaining the function of the retina, which was essential for preserving the structure of the synaptic ribbon and supporting the integrity of the outer plexiform layer.
Collapse
Affiliation(s)
- Wei-Hao Peng
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Meng-Lin Liao
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wan-Chun Huang
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Pei-Kang Liu
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, Columbia University, New York, NY, United States
| | - Sarah R. Levi
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, Columbia University, New York, NY, United States
| | - Yun-Ju Tseng
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, Columbia University, New York, NY, United States
| | - Chia-Ying Lee
- Department of Ophthalmology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Lung-Kun Yeh
- Department of Ophthalmology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kuan-Jen Chen
- Department of Ophthalmology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chung-Liang Chien
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Nan-Kai Wang
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, Columbia University, New York, NY, United States
| |
Collapse
|
7
|
Frick C, Fink S, Schmidbauer D, Rousset F, Eickhoff H, Tropitzsch A, Kramer B, Senn P, Glueckert R, Rask-Andersen H, Wiesmüller KH, Löwenheim H, Müller M. Age-Dependency of Neurite Outgrowth in Postnatal Mouse Cochlear Spiral Ganglion Explants. Brain Sci 2020; 10:E580. [PMID: 32839381 PMCID: PMC7564056 DOI: 10.3390/brainsci10090580] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The spatial gap between cochlear implants (CIs) and the auditory nerve limits frequency selectivity as large populations of spiral ganglion neurons (SGNs) are electrically stimulated synchronously. To improve CI performance, a possible strategy is to promote neurite outgrowth toward the CI, thereby allowing a discrete stimulation of small SGN subpopulations. Brain-derived neurotrophic factor (BDNF) is effective to stimulate neurite outgrowth from SGNs. METHOD TrkB (tropomyosin receptor kinase B) agonists, BDNF, and five known small-molecule BDNF mimetics were tested for their efficacy in stimulating neurite outgrowth in postnatal SGN explants. To modulate Trk receptor-mediated effects, TrkB and TrkC ligands were scavenged by an excess of recombinant receptor proteins. The pan-Trk inhibitor K252a was used to block Trk receptor actions. RESULTS THF (7,8,3'-trihydroxyflavone) partly reproduced the BDNF effect in postnatal day 7 (P7) mouse cochlear spiral ganglion explants (SGEs), but failed to show effectiveness in P4 SGEs. During the same postnatal period, spontaneous and BDNF-stimulated neurite outgrowth increased. The increased neurite outgrowth in P7 SGEs was not caused by the TrkB/TrkC ligands, BDNF and neurotrophin-3 (NT-3). CONCLUSIONS The age-dependency of induction of neurite outgrowth in SGEs was very likely dependent on presently unidentified factors and/or molecular mechanisms which may also be decisive for the age-dependent efficacy of the small-molecule TrkB receptor agonist THF.
Collapse
Affiliation(s)
- Claudia Frick
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen Medical Center, 72076 Tübingen, Germany; (C.F.); (A.T.); (B.K.); (H.L.); (M.M.)
- Department of Microbiome Science, Max Planck Institute for Developmental Biology, 72076 Tübingen, Germany
| | - Stefan Fink
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen Medical Center, 72076 Tübingen, Germany; (C.F.); (A.T.); (B.K.); (H.L.); (M.M.)
| | - Dominik Schmidbauer
- Inner Ear Laboratory Innsbruck, Medical University Innsbruck, 6020 Innsbruck, Austria; (D.S.); (R.G.)
| | - Francis Rousset
- The Inner Ear & Olfaction Lab, Department of Clinical Neurosciences, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland; (F.R.); (P.S.)
| | - Holger Eickhoff
- EMC Microcollections GmbH, 72070 Tübingen, Germany; (H.E.); (K.-H.W.)
| | - Anke Tropitzsch
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen Medical Center, 72076 Tübingen, Germany; (C.F.); (A.T.); (B.K.); (H.L.); (M.M.)
| | - Benedikt Kramer
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen Medical Center, 72076 Tübingen, Germany; (C.F.); (A.T.); (B.K.); (H.L.); (M.M.)
- Department of Otolaryngology, Head and Neck Surgery, University Hospital Mannheim, 68167 Mannheim, Germany
| | - Pascal Senn
- The Inner Ear & Olfaction Lab, Department of Clinical Neurosciences, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland; (F.R.); (P.S.)
| | - Rudolf Glueckert
- Inner Ear Laboratory Innsbruck, Medical University Innsbruck, 6020 Innsbruck, Austria; (D.S.); (R.G.)
- Tirol Kliniken Innsbruck, University Clinic of Otolaryngology, 6020 Innsbruck, Austria
| | - Helge Rask-Andersen
- Department of Surgical Sciences, Otorhinolaryngology and Head and Neck Surgery, University of Uppsala, 751 85 Uppsala, Sweden;
| | | | - Hubert Löwenheim
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen Medical Center, 72076 Tübingen, Germany; (C.F.); (A.T.); (B.K.); (H.L.); (M.M.)
| | - Marcus Müller
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen Medical Center, 72076 Tübingen, Germany; (C.F.); (A.T.); (B.K.); (H.L.); (M.M.)
| |
Collapse
|
8
|
Wong VSC, Meadows M, Goldberg D, Willis DE. Semaphorin 3A induces acute changes in membrane excitability in spiral ganglion neurons in vitro. Eur J Neurosci 2019; 50:1741-1758. [PMID: 30706560 DOI: 10.1111/ejn.14360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 01/10/2019] [Accepted: 01/23/2019] [Indexed: 11/29/2022]
Abstract
The development and survival of spiral ganglion neurons (SGNs) are dependent on multiple trophic factors as well as membrane electrical activity. Semaphorins (Sema) constitute a family of membrane-associated and secreted proteins that have garnered significant attention as a potential SGN "navigator" during cochlea development. Previous studies using mutant mice demonstrated that Sema3A plays a role in the SGN pathfinding. The mechanisms, however, by which Sema3A shapes SGNs firing behavior are not known. In these studies, we found that Sema3A plays a novel role in regulating SGN resting membrane potential and excitability. Using dissociated SGN from pre-hearing (P3-P5) and post-hearing mice (P12-P15), we recorded membrane potentials using whole-cell patch clamp recording techniques in apical and basal SGN populations. Recombinant Sema3A was applied to examine the effects on intrinsic membrane properties and action potentials evoked by current injections. Apical and basal SGNs from newborn mice treated with recombinant Sema3A (100 ng/ml) displayed a higher resting membrane potential, higher threshold, decreased amplitude, and prolonged latency and duration of spikes. Although a similar phenomenon was observed in SGNs from post-hearing mice, the resting membrane potential was essentially indistinguishable before and after Sema3A exposure. Sema3A-mediated changes in membrane excitability were associated with a significant decrease in K+ and Ca2+ currents. Sema3A acts through linopirdine-sensitive K+ channels in apical, but not in the basal SGNs. Therefore, Sema3A induces differential effects in SGN membrane excitability that are dependent on age and location, and constitutes an additional early and novel effect of Sema3A SGNs in vitro.
Collapse
Affiliation(s)
| | - Marc Meadows
- The Vollum Institute, Oregon Health and Science University, Portland, Oregon
| | - David Goldberg
- The Burke Neurological Institute, White Plains, New York
| | - Dianna E Willis
- The Burke Neurological Institute, White Plains, New York.,Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York
| |
Collapse
|
9
|
Akil O, Blits B, Lustig LR, Leake PA. Virally Mediated Overexpression of Glial-Derived Neurotrophic Factor Elicits Age- and Dose-Dependent Neuronal Toxicity and Hearing Loss. Hum Gene Ther 2018; 30:88-105. [PMID: 30183384 DOI: 10.1089/hum.2018.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Contemporary cochlear implants (CI) are generally very effective for remediation of severe to profound sensorineural hearing loss, but outcomes are still highly variable. Auditory nerve survival is likely one of the major factors underlying this variability. Neurotrophin therapy therefore has been proposed for CI recipients, with the goal of improving outcomes by promoting improved survival of cochlear spiral ganglion neurons (SGN) and/or residual hair cells. Previous studies have shown that glial-derived neurotrophic factor (GDNF), brain-derived neurotrophic factor, and neurotrophin-3 can rescue SGNs following insult. The current study was designed to determine whether adeno-associated virus vector serotype 5 (AAV-5) encoding either green fluorescent protein or GDNF can transduce cells in the mouse cochlea to express useful levels of neurotrophin and to approximate the optimum therapeutic dose(s) for transducing hair cells and SGN. The findings demonstrate that AAV-5 is a potentially useful gene therapy vector for the cochlea, resulting in extremely high levels of transgene expression in the cochlear inner hair cells and SGN. However, overexpression of human GDNF in newborn mice caused severe neurological symptoms and hearing loss, likely due to Purkinje cell loss and cochlear nucleus pathology. Thus, extremely high levels of transgene protein expression should be avoided, particularly for proteins that have neurological function in neonatal subjects.
Collapse
Affiliation(s)
- Omar Akil
- 1 Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, California
| | - Bas Blits
- 2 Department of Research and Development, UniQure Biopharma B.V., Amsterdam, The Netherlands
| | - Lawrence R Lustig
- 3 Department of Otolaryngology-Head and Neck Surgery, Columbia University Medical Center, New York, New York
| | - Patricia A Leake
- 1 Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, California
| |
Collapse
|
10
|
Regenerative medicine in hearing recovery. Cytotherapy 2017; 19:909-915. [DOI: 10.1016/j.jcyt.2017.04.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/24/2017] [Accepted: 04/21/2017] [Indexed: 12/20/2022]
|
11
|
Anand SK, Mondal AC. Cellular and molecular attributes of neural stem cell niches in adult zebrafish brain. Dev Neurobiol 2017; 77:1188-1205. [PMID: 28589616 DOI: 10.1002/dneu.22508] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 04/05/2017] [Accepted: 06/02/2017] [Indexed: 12/20/2022]
Abstract
Adult neurogenesis is a complex, presumably conserved phenomenon in vertebrates with a broad range of variations regarding neural progenitor/stem cell niches, cellular composition of these niches, migratory patterns of progenitors and so forth among different species. Current understanding of the reasons underlying the inter-species differences in adult neurogenic potential, the identification and characterization of various neural progenitors, characterization of the permissive environment of neural stem cell niches and other important aspects of adult neurogenesis is insufficient. In the last decade, zebrafish has emerged as a very useful model for addressing these questions. In this review, we have discussed the present knowledge regarding the neural stem cell niches in adult zebrafish brain as well as their cellular and molecular attributes. We have also highlighted their similarities and differences with other vertebrate species. In the end, we shed light on some of the known intrinsic and extrinsic factors that are assumed to regulate the neurogenic process in adult zebrafish brain. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 1188-1205, 2017.
Collapse
Affiliation(s)
- Surendra Kumar Anand
- Cellular and Molecular Neurobiology Lab, School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, India, 110067
| | - Amal Chandra Mondal
- Cellular and Molecular Neurobiology Lab, School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, India, 110067
| |
Collapse
|
12
|
Kwiatkowska M, Reinhard J, Roll L, Kraft N, Dazert S, Faissner A, Volkenstein S. The expression pattern and inhibitory influence of Tenascin-C on the growth of spiral ganglion neurons suggest a regulatory role as boundary formation molecule in the postnatal mouse inner ear. Neuroscience 2016; 319:46-58. [PMID: 26812032 DOI: 10.1016/j.neuroscience.2016.01.039] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 12/08/2015] [Accepted: 01/15/2016] [Indexed: 02/02/2023]
Abstract
Sensorineural hearing loss, as a consequence of acoustic trauma, aging, genetic defects or ototoxic drugs, is highly associated with irreversible damage of cochlear hair cells (HCs) and secondary degeneration of spiral ganglion (SG) cells. Cochlear implants (CIs), which bypass the lost HC function by direct electrical stimulation of the remaining auditory neurons, offer an effective therapy option. Several studies imply that components of the extracellular matrix (ECM) have a great impact on the adhesion and growth of spiral ganglion neurons (SGNs) during development. Based on these findings, ECM proteins might act as bioactive CI substrates to optimize the electrode-nerve interface and to improve efficacy of these implants. In the present study, we focused on the ECM glycoproteins Tenascin-C (TN-C), Laminin (LN), and Fibronectin (FN), which show a prominent expression along the growth route of SGNs and the niche around HCs during murine postnatal development in vivo. We compared their influence on adhesion, neurite length, and neurite number of SGNs in vitro. Moreover, we studied the expression of the chondroitin sulfate proteoglycan (CSPG) dermatan sulfate-dependent proteoglycan-1 (DSD-1-PG), an interaction partner of TN-C. In sum, our in vitro data suggest that TN-C acts as an anti-adhesive and inhibitory factor for the growth of SGNs. The DSD-1 carbohydrate epitope is specifically localized to HC stereocilia and SG fibers. Interestingly, TN-C and the DSD-1-PG exhibit a mutually exclusive expression pattern, with the exception of a very restricted region beneath the habenula perforata, where SG neurites grow through the basilar membrane (BM) toward the HCs. The complementary expression of TN-C, LN, FN, and the DSD-1 epitope suggests that TN-C may act as an important boundary formation molecule in the developing postnatal mouse inner ear, which makes it a promising candidate to regulate neurite outgrowth in the light of CIs.
Collapse
Affiliation(s)
- M Kwiatkowska
- Department of Otorhinolaryngology, Head & Neck Surgery, Ruhr-University Bochum, St. Elisabeth-Hospital, Bleichstrasse 15, 44787 Bochum, Germany
| | - J Reinhard
- Department of Cell Morphology & Molecular Neurobiology, Ruhr-University Bochum, Faculty of Biology & Biotechnology, Universitätsstrasse 150, 44801 Bochum, Germany
| | - L Roll
- Department of Cell Morphology & Molecular Neurobiology, Ruhr-University Bochum, Faculty of Biology & Biotechnology, Universitätsstrasse 150, 44801 Bochum, Germany
| | - N Kraft
- Department of Otorhinolaryngology, Head & Neck Surgery, Ruhr-University Bochum, St. Elisabeth-Hospital, Bleichstrasse 15, 44787 Bochum, Germany
| | - S Dazert
- Department of Otorhinolaryngology, Head & Neck Surgery, Ruhr-University Bochum, St. Elisabeth-Hospital, Bleichstrasse 15, 44787 Bochum, Germany
| | - A Faissner
- Department of Cell Morphology & Molecular Neurobiology, Ruhr-University Bochum, Faculty of Biology & Biotechnology, Universitätsstrasse 150, 44801 Bochum, Germany
| | - S Volkenstein
- Department of Otorhinolaryngology, Head & Neck Surgery, Ruhr-University Bochum, St. Elisabeth-Hospital, Bleichstrasse 15, 44787 Bochum, Germany.
| |
Collapse
|
13
|
The Three-Dimensional Culture System with Matrigel and Neurotrophic Factors Preserves the Structure and Function of Spiral Ganglion Neuron In Vitro. Neural Plast 2016; 2016:4280407. [PMID: 27057364 PMCID: PMC4736769 DOI: 10.1155/2016/4280407] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 11/10/2015] [Indexed: 02/06/2023] Open
Abstract
Whole organ culture of the spiral ganglion region is a resourceful model system facilitating manipulation and analysis of live sprial ganglion neurons (SGNs). Three-dimensional (3D) cultures have been demonstrated to have many biomedical applications, but the effect of 3D culture in maintaining the SGNs structure and function in explant culture remains uninvestigated. In this study, we used the matrigel to encapsulate the spiral ganglion region isolated from neonatal mice. First, we optimized the matrigel concentration for the 3D culture system and found the 3D culture system protected the SGNs against apoptosis, preserved the structure of spiral ganglion region, and promoted the sprouting and outgrowth of SGNs neurites. Next, we found the 3D culture system promoted growth cone growth as evidenced by a higher average number and a longer average length of filopodia and a larger growth cone area. 3D culture system also significantly elevated the synapse density of SGNs. Last, we found that the 3D culture system combined with neurotrophic factors had accumulated effects in promoting the neurites outgrowth compared with 3D culture or NFs treatment only groups. Together, we conclude that the 3D culture system preserves the structure and function of SGN in explant culture.
Collapse
|
14
|
Postnatal expression of neurotrophic factors accessible to spiral ganglion neurons in the auditory system of adult hearing and deafened rats. J Neurosci 2014; 34:13110-26. [PMID: 25253857 DOI: 10.1523/jneurosci.1014-14.2014] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Spiral ganglion neurons (SGNs) receive input from cochlear hair cells and project from the cochlea to the cochlear nucleus. After destruction of hair cells with aminoglycoside antibiotics or noise, SGNs gradually die. It has been assumed that SGN death is attributable to loss of neurotrophic factors (NTFs) derived from hair cells or supporting cells in the organ of Corti (OC). We used quantitative PCR (qPCR) to assay NTF expression-neurotrophin-3 (NT-3), BDNF, GDNF, neurturin, artemin, and CNTF-in the OC and cochlear nucleus at various ages from postnatal day 0 (P0) to P90 in control hearing and neonatally deafened rats. NT-3, neurturin, and CNTF were most abundant in the postnatal hearing OC; CNTF and neurturin most abundant in the cochlear nucleus. In the OC, NT-3 and CNTF showed a postnatal increase in expression approximately concomitant with hearing onset. In rats deafened by daily kanamycin injections (from P8 to P16), surviving inner hair cells were evident at P16 but absent by P19, with most postsynaptic boutons lost before P16. NT-3 and CNTF, which normally increase postnatally, had significantly reduced expression in the OC of deafened rats, although CNTF was expressed throughout the time that SGNs were dying. In contrast, neurturin expression was constant, unaffected by deafening or by age. CNTF and neurturin expression in the cochlear nucleus was unaffected by deafening or age. Thus, NTFs other than NT-3 are available to SGNs even as they are dying after deafening, apparently conflicting with the hypothesis that SGN death is attributable to lack of NTFs.
Collapse
|
15
|
Genetic, cellular, and functional evidence for Ca2+ inflow through Cav1.2 and Cav1.3 channels in murine spiral ganglion neurons. J Neurosci 2014; 34:7383-93. [PMID: 24849370 DOI: 10.1523/jneurosci.5416-13.2014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Spiral ganglion neurons (SGNs) of the eighth nerve serve as the bridge between hair cells and the cochlear nucleus. Hair cells use Cav1.3 as the primary channel for Ca(2+) inflow to mediate transmitter release. In contrast, SGNs are equipped with multiple Ca(2+) channels to mediate Ca(2+)-dependent functions. We examined directly the role of Cav1.3 channels in SGNs using Cav1.3-deficient mice (Cav1.3(-/-)). We revealed a surprising finding that SGNs functionally express the cardiac-specific Cav1.2, as well as neuronal Cav1.3 channels. We show that evoked action potentials recorded from SGNs show a significant decrease in the frequency of firing in Cav1.3(-/-) mice compared with wild-type (Cav1.3(+/+)) littermates. Although Cav1.3 is the designated L-type channel in neurons, whole-cell currents recorded in isolated SGNs from Cav1.3(-/-) mice showed a surprising remnant current with sensitivity toward the dihydropyridine (DHP) agonist and antagonist, and a depolarization shift in the voltage-dependent activation compared with that in the Cav1.3(+/+) mice. Indeed, direct measurement of the elementary properties of Ca(2+) channels, in Cav1.3(+/+) neurons, confirmed the existence of two DHP-sensitive single-channel currents, with distinct open probabilities and conductances. We demonstrate that the DHP-sensitive current in Cav1.3(-/-) mice is derived from Cav1.2 channel activity, providing for the first time, to our knowledge, functional data for the expression of Cav1.2 currents in neurons. Finally, using shRNA gene knockdown methodology, and histological analyses of SGNs from Cav1.2(+/-) and Cav1.3(+/-) mice, we were able to establish the differential roles of Cav1.2 and Cav1.3 in SGNs.
Collapse
|
16
|
Fuentes-Santamaría V, Alvarado JC, Gabaldón-Ull MC, Manuel Juiz J. Upregulation of insulin-like growth factor and interleukin 1β occurs in neurons but not in glial cells in the cochlear nucleus following cochlear ablation. J Comp Neurol 2014; 521:3478-99. [PMID: 23681983 DOI: 10.1002/cne.23362] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2013] [Revised: 04/30/2013] [Accepted: 05/03/2013] [Indexed: 12/30/2022]
Abstract
One of the main mechanisms used by neurons and glial cells to promote repair following brain injury is to upregulate activity-dependent molecules such as insulin-like growth factor 1 (IGF-1) and interleukin-1β (IL-1β). In the auditory system, IGF-1 is crucial for restoring synaptic transmission following hearing loss; however, whether IL-1β is also involved in this process is unknown. In this study, we evaluated the expression of IGF-1 and IL-1β within neurons and glial cells of the ventral cochlear nucleus in adult rats at 1, 7, 15, and 30 days following bilateral cochlear ablation. After the lesion, significant increases in both the overall mean gray levels of IGF-1 immunostaining and the mean gray levels within cells of the cochlear nucleus were observed at 1, 7, and 15 days compared with control animals. The expression and distribution of IL-1β in the ventral cochlear nucleus of ablated animals was temporally and spatially correlated with IGF-1. We also observed a lack of colocalization between IGF-1 and IL-1β with either astrocytes or microglia at any of the time points following ablation. These results suggest that the upregulation of IGF-1 and IL-1β levels within neurons-but not within glial cells-may reflect a plastic mechanism involved in repairing synaptic homeostasis of the overall cellular environment of the cochlear nucleus following bilateral cochlear ablation.
Collapse
Affiliation(s)
- Verónica Fuentes-Santamaría
- Institute for Research on Neurological Disorders (IDINE), Faculty of Medicine, University of Castilla-La Mancha, 02006, Albacete, Spain
| | | | | | | |
Collapse
|
17
|
García-Hernández S, Potashner SJ, Morest DK. Role of fibroblast growth factor 8 in neurite outgrowth from spiral ganglion neurons in vitro. Brain Res 2013; 1529:39-45. [PMID: 23891716 PMCID: PMC5217747 DOI: 10.1016/j.brainres.2013.07.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 07/16/2013] [Accepted: 07/18/2013] [Indexed: 12/12/2022]
Abstract
Many neurons degenerate after injuries resulting from overstimulation, drugs, genetic mutations, and aging. Although several growth factors and neurotrophins delay degeneration and promote regrowth of neural processes, the role of fibroblast growth factor 8 (FGF8) in mammalian spiral ganglion neurons (SGN) neurite outgrowth has not been examined. This study develops and uses SGN cell cultures suitable for experimental analysis, it investigates whether FGF8a and FGF8b isoforms affect the neurite outgrowth from SGN cultured in vitro. We found that both FGF8a and FGF8b promoted the outgrowth of neurites from cultured SGN. This response is mediated by FGF receptors and involves the activation of IκBα-mediated NFκB signaling pathway. These findings suggest that, besides its morphogenetic role during development, FGF8 may have trophic functions in the adult which are relevant to regeneration.
Collapse
Affiliation(s)
- Sofía García-Hernández
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030, USA.
| | | | | |
Collapse
|
18
|
Khalifa SAM, Björk P, Vieider C, Ulfendahl M, Scarfone E. Neuronal polarity mediated by micro-scale protein patterns and Schwann cells in vitro. Tissue Eng Regen Med 2013. [DOI: 10.1007/s13770-013-1084-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
19
|
Posthearing Ca(2+) currents and their roles in shaping the different modes of firing of spiral ganglion neurons. J Neurosci 2013; 32:16314-30. [PMID: 23152615 DOI: 10.1523/jneurosci.2097-12.2012] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Whereas prehearing spiral ganglion neurons (SGNs) rely faithfully on outputs from spontaneously active developing hair cells, the electrical phenotypes of posthearing neurons are shaped by distinct rapid and graded receptor potentials from hair cells. To date, technical difficulties in isolation of fragile posthearing neurons from the rigid bony labyrinth of the inner ear have hindered analyses of the electrical phenotype of SGNs. Therefore, we have recently developed new strategies to isolate posthearing mouse SGNs for functional analyses. Here, we describe the coarse and fine properties of Ca(2+) currents, which sculpt the firing properties of posthearing SGNs. Murine SGNs express multiple Ca(2+) channel currents to enable diverse functions. We have demonstrated that suppression of Ca(2+) currents results in significant hyperpolarization of the resting membrane potential (rmp) of basal SGNs, suggesting that Ca(2+) influx primes rmp for excitation. In contrast, removal of external Ca(2+) has modest effects on rmp of apical SGNs. The blockade of Ca(2+) currents with a mixture of specific blockers attenuates spontaneously active SGNs. Paradoxically, different subtypes of Ca(2+) currents, such as R-type currents, may activate resting outward conductances since blockage of the current results in depolarization of rmp. In keeping with whole-cell current data, single-channel records revealed multiple diverse Ca(2+) channels in SGNs. Additionally, there were differential expressions of distinct Ca(2+) current densities in the apicobasal contour of the adult cochlea. This report provides invaluable insights into Ca(2+)-dependent processes in adult SGNs.
Collapse
|
20
|
Filling the silent void: genetic therapies for hearing impairment. GENETICS RESEARCH INTERNATIONAL 2013; 2012:748698. [PMID: 23304527 PMCID: PMC3529436 DOI: 10.1155/2012/748698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 09/27/2012] [Accepted: 11/04/2012] [Indexed: 12/02/2022]
Abstract
The inner ear cytoarchitecture forms one of the most intricate and delicate organs in the human body and is vulnerable to the effects of genetic disorders, aging, and environmental damage. Owing to the inability of the mammalian cochlea to regenerate sensory hair cells, the loss of hair cells is a leading cause of deafness in humans. Millions of individuals worldwide are affected by the emotionally and financially devastating effects of hearing impairment (HI). This paper provides a brief introduction into the key role of genes regulating inner ear development and function. Potential future therapies that leverage on an improved understanding of these molecular pathways are also described in detail.
Collapse
|
21
|
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
|
22
|
Vemaraju S, Kantarci H, Padanad MS, Riley BB. A spatial and temporal gradient of Fgf differentially regulates distinct stages of neural development in the zebrafish inner ear. PLoS Genet 2012; 8:e1003068. [PMID: 23166517 PMCID: PMC3499369 DOI: 10.1371/journal.pgen.1003068] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 09/21/2012] [Indexed: 01/13/2023] Open
Abstract
Neuroblasts of the statoacoustic ganglion (SAG) initially form in the floor of the otic vesicle during a relatively brief developmental window. They soon delaminate and undergo a protracted phase of proliferation and migration (transit-amplification). Neuroblasts eventually differentiate and extend processes bi-directionally to synapse with hair cells in the inner ear and various targets in the hindbrain. Our studies in zebrafish have shown that Fgf signaling controls multiple phases of this complex developmental process. Moderate levels of Fgf in a gradient emanating from the nascent utricular macula specify SAG neuroblasts in laterally adjacent otic epithelium. At a later stage, differentiating SAG neurons express Fgf5, which serves two functions: First, as SAG neurons accumulate, increasing levels of Fgf exceed an upper threshold that terminates the initial phase of neuroblast specification. Second, elevated Fgf delays differentiation of transit-amplifying cells, balancing the rate of progenitor renewal with neuronal differentiation. Laser-ablation of mature SAG neurons abolishes feedback-inhibition and causes precocious neuronal differentiation. Similar effects are obtained by Fgf5-knockdown or global impairment of Fgf signaling, whereas Fgf misexpression has the opposite effect. Thus Fgf signaling renders SAG development self-regulating, ensuring steady production of an appropriate number of neurons as the larva grows. Neurons of the statoacoustic ganglion (SAG), which innervate the inner ear, are derived from neuroblasts originating from the floor of the otic vesicle. Neuroblasts quickly delaminate from the otic vesicle to form dividing progenitors, which eventually differentiate into mature neurons of the SAG. Fgf has been implicated in initial neuroblast specification in multiple vertebrate species. However, the role of Fgf at later stages remains uncertain, because previous studies have not been able to evaluate the effects of changing levels of Fgf, nor have they been able to clearly distinguish the effects of Fgf at different stages of SAG development. We have combined conditional loss of function, misexpression, and laser-ablation studies in zebrafish to elucidate how graded Fgf coordinates distinct steps in SAG development. Initially moderate Fgf in a spatial gradient specifies neuroblasts within the otic vesicle. Later, mature SAG neurons express Fgf5 and, as additional neurons accumulate outside the otic vesicle, rising levels of Fgf terminate further specification. Elevated Fgf also slows maturation of progenitors, maintaining a stable progenitor pool in which growth and differentiation are evenly balanced. This feedback facilitates steady production of new neurons as the animal grows through larval and adults stages.
Collapse
Affiliation(s)
- Shruti Vemaraju
- Biology Department, Texas A&M University, College Station, Texas, United States of America
| | | | | | | |
Collapse
|
23
|
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
|
24
|
Magariños M, Contreras J, Aburto MR, Varela-Nieto I. Early development of the vertebrate inner ear. Anat Rec (Hoboken) 2012; 295:1775-90. [PMID: 23044927 DOI: 10.1002/ar.22575] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 07/24/2012] [Indexed: 12/12/2022]
Abstract
This is a review of the biological processes and the main signaling pathways required to generate the different otic cell types, with particular emphasis on the actions of insulin-like growth factor I. The sensory organs responsible of hearing and balance have a common embryonic origin in the otic placode. Lineages of neural, sensory, and support cells are generated from common otic neuroepithelial progenitors. The sequential generation of the cell types that will form the adult inner ear requires the coordination of cell proliferation with cell differentiation programs, the strict regulation of cell survival, and the metabolic homeostasis of otic precursors. A network of intracellular signals operates to coordinate the transcriptional response to the extracellular input. Understanding the molecular clues that direct otic development is fundamental for the design of novel treatments for the protection and repair of hearing loss and balance disorders.
Collapse
Affiliation(s)
- Marta Magariños
- Instituto de Investigaciones Biomédicas, Alberto Sols, CSIC-UAM, Madrid, Spain
| | | | | | | |
Collapse
|
25
|
Kizil C, Kaslin J, Kroehne V, Brand M. Adult neurogenesis and brain regeneration in zebrafish. Dev Neurobiol 2012; 72:429-61. [DOI: 10.1002/dneu.20918] [Citation(s) in RCA: 249] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
26
|
Landry TG, Wise AK, Fallon JB, Shepherd RK. Spiral ganglion neuron survival and function in the deafened cochlea following chronic neurotrophic treatment. Hear Res 2011; 282:303-13. [PMID: 21762764 PMCID: PMC3205216 DOI: 10.1016/j.heares.2011.06.007] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 06/28/2011] [Accepted: 06/28/2011] [Indexed: 12/26/2022]
Abstract
Cochlear implants electrically stimulate residual spiral ganglion neurons (SGNs) to provide auditory cues for the severe-profoundly deaf. However, SGNs gradually degenerate following cochlear hair cell loss, leaving fewer neurons available for stimulation. Providing an exogenous supply of neurotrophins (NTs) has been shown to prevent SGN degeneration, and when combined with chronic intracochlear electrical stimulation (ES) following a short period of deafness (5 days), may also promote the formation of new neurons. The present study assessed the histopathological response of guinea pig cochleae treated with NTs (brain-derived neurotrophic factor and neurotrophin-3) with and without ES over a four week period, initiated two weeks after deafening. Results were compared to both NT alone and artificial perilymph (AP) treated animals. AP/ES treated animals exhibited no evidence of SGN rescue compared with untreated deafened controls. In contrast, NT administration showed a significant SGN rescue effect in the lower and middle cochlear turns (two-way ANOVA, p < 0.05) compared with AP-treated control animals. ES in combination with NT did not enhance SGN survival compared with NT alone. SGN function was assessed by measuring electrically-evoked auditory brainstem response (EABR) thresholds. EABR thresholds following NT treatment were significantly lower than animals treated with AP (two-way ANOVA, p = 0.033). Finally, the potential for induced neurogenesis following the combined treatment was investigated using a marker of DNA synthesis. However, no evidence of neurogenesis was observed in the SGN population. The results indicate that chronic NT delivery to the cochlea may be beneficial to cochlear implant patients by increasing the number of viable SGNs and decreasing activation thresholds compared to chronic ES alone.
Collapse
Affiliation(s)
- Thomas G. Landry
- The Bionic Ear Institute, Daly Wing, St. Vincent’s Hospital, Fitzroy, Victoria, 3065, Australia
- The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Andrew K. Wise
- The Bionic Ear Institute, Daly Wing, St. Vincent’s Hospital, Fitzroy, Victoria, 3065, Australia
- The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - James B. Fallon
- The Bionic Ear Institute, Daly Wing, St. Vincent’s Hospital, Fitzroy, Victoria, 3065, Australia
- The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Robert K. Shepherd
- The Bionic Ear Institute, Daly Wing, St. Vincent’s Hospital, Fitzroy, Victoria, 3065, Australia
- The University of Melbourne, Parkville, Victoria, 3052, Australia
| |
Collapse
|
27
|
Lv P, Wei D, Yamoah EN. Kv7-type channel currents in spiral ganglion neurons: involvement in sensorineural hearing loss. J Biol Chem 2010; 285:34699-707. [PMID: 20739290 DOI: 10.1074/jbc.m110.136192] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Alterations in K(v)7-mediated currents in excitable cells result in several diseased conditions. A case in DFNA2, an autosomal dominant version of progressive hearing loss, involves degeneration of hair cells and spiral ganglion neurons (SGNs) from basal to apical cochlea, manifesting as high-to-low frequency hearing loss, and has been ascribed to mutations in K(v)7.4 channels. Analyses of the cellular mechanisms of K(v)7.4 mutations and progressive degeneration of SGNs have been hampered by the paucity of functional data on the role K(v)7 channels play in young and adult neurons. To understand the cellular mechanisms of the disease in SGNs, we examined temporal (young, 0.5 months old, and senescent, 17 months old) and spatial (apical and basal) roles of K(v)7-mediated currents. We report that differential contribution of K(v)7 currents in mice SGNs results in distinct and profound variations of the membrane properties of basal versus apical neurons. The current produces a major impact on the resting membrane potential of basal neurons. Inhibition of the current promotes membrane depolarization, resulting in activation of Ca(2+) currents and a sustained rise in intracellular Ca(2+). Using TUNEL assay, we demonstrate that a sustained increase in intracellular Ca(2+) mediated by inhibition of K(v)7 current results in significant SGN apoptotic death. Thus, this study provides evidence of the cellular etiology and mechanisms of SGN degeneration in DFNA2.
Collapse
Affiliation(s)
- Ping Lv
- Department of Anesthesiology and Pain Medicine, School of Medicine, University of California, Davis, California 95618, USA
| | | | | |
Collapse
|
28
|
Wang SJ, Furusho M, D'Sa C, Kuwada S, Conti L, Morest DK, Bansal R. Inactivation of fibroblast growth factor receptor signaling in myelinating glial cells results in significant loss of adult spiral ganglion neurons accompanied by age-related hearing impairment. J Neurosci Res 2010; 87:3428-37. [PMID: 19598249 DOI: 10.1002/jnr.22164] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Hearing loss has been attributed to many factors, including degeneration of sensory neurons in the auditory pathway and demyelination along the cochlear nerve. Fibroblast growth factors (FGFs), which signal through four receptors (Fgfrs), are produced by auditory neurons and play a key role in embryonic development of the cochlea and in neuroprotection against sound-induced injury. However, the role of FGF signaling in the maintenance of normal auditory function in adult and aging mice remains to be elucidated. Furthermore, the contribution of glial cells, which myelinate the cochlear nerves, is poorly understood. To address these questions, we generated transgenic mice in which Fgfr1 and Fgfr2 were specifically inactivated in Schwann cells and oligodendrocytes but not in neurons. Adult mutant mice exhibited late onset of hearing impairment, which progressed markedly with age. The hearing impairment was accompanied by significant loss of myelinated spiral ganglion neurons. The pathology extended into the cochlear nucleus, without apparent loss of myelin or of the deletion-bearing glial cells themselves. This suggests that perturbation of FGF receptor-mediated glial function leads to the attenuation of glial support of neurons, leading to their loss and impairment of auditory functions. Thus, FGF/FGF receptor signaling provides a potentially novel mechanism of maintaining reciprocal interactions between neurons and glia in adult and aging animals. Dysfunction of glial cells and FGF receptor signaling may therefore be implicated in neurodegenerative hearing loss associated with normal aging.
Collapse
Affiliation(s)
- S J Wang
- Department of Neuroscience, University of Connecticut Medical School, Farmington, Connecticut, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW This review will focus on 'self-repair' of the mammalian inner ear sensory epithelium, including recruiting the in-situ proliferation and differentiation of endogenous cells at the damaged site and the autologous transplantation RECENT FINDINGS Self-repair refers to a favorable structural and functional outcome of damaged inner ear sensory epithelium. Our advanced ability of manipulating the fate of inner ear sensory cells makes in-situ proliferation a possible candidate of hearing restoration. A practical alternative of the unavoidable immune rejection is to introduce autologous cells. Ependymal cells, induced pluripotent stem cells, and olfactory neuroepithelial cells have been recognized as promising sources, which will spur ongoing efforts to evaluate these new cell sources for cell replacement therapy. SUMMARY Further exploration of the innate advantages of in-situ proliferation and use of novel cell sources for autologous transplantation may serve as rehearsals for clinical trials in the near future.
Collapse
|
30
|
Expression of Wnt receptors in adult spiral ganglion neurons: frizzled 9 localization at growth cones of regenerating neurites. Neuroscience 2009; 164:478-87. [PMID: 19716861 DOI: 10.1016/j.neuroscience.2009.08.049] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2009] [Revised: 08/21/2009] [Accepted: 08/22/2009] [Indexed: 01/10/2023]
Abstract
Little is known about signaling pathways, besides those of neurotrophic factors, that are operational in adult spiral ganglion neurons. In patients with sensorineural hearing loss, such pathways could eventually be targeted to stimulate and guide neurite outgrowth from the remnants of the spiral ganglion towards a cochlear implant, thereby improving the fidelity of sound transmission. To systematically identify neuronal receptors for guidance cues in the adult cochlea, we conducted a genome-wide cDNA microarray screen with 2-month-old CBA/CaJ mice. A meta-analysis of our data and those from older mice in two other studies revealed the presence of neuronal transmembrane receptors that represent all four established guidance pathways--ephrin, netrin, semaphorin, and slit--in the mature cochlea as late as 15 months. In addition, we observed the expression of all known receptors for the "wingless-related MMTV integration site" (Wnt) morphogens, whose neuronal guidance function has only recently been recognized. In situ hybridizations located the mRNAs of the Wnt receptors frizzled 1, 4, 6, 9, and 10 specifically in adult spiral ganglion neurons. Finally, frizzled 9 protein was found in the growth cones of adult spiral ganglion neurons that were regenerating neurites in culture. We conclude from our results that adult spiral ganglion neurons are poised to respond to neurite damage, owing to the constitutive expression of a large and diverse collection of guidance receptors. Wnt signaling, in particular, emerges as a candidate pathway for guiding neurite outgrowth towards a cochlear implant after sensorineural hearing loss.
Collapse
|
31
|
Wei D, Levic S, Nie L, Gao WQ, Petit C, Jones EG, Yamoah EN. Cells of adult brain germinal zone have properties akin to hair cells and can be used to replace inner ear sensory cells after damage. Proc Natl Acad Sci U S A 2008; 105:21000-5. [PMID: 19064919 PMCID: PMC2634930 DOI: 10.1073/pnas.0808044105] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2008] [Indexed: 01/27/2023] Open
Abstract
Auditory hair cell defect is a major cause of hearing impairment, often leading to spiral ganglia neuron (SGN) degeneration. The cell loss that follows is irreversible in mammals, because inner ear hair cells (HCs) have a limited capacity to regenerate. Here, we report that in the adult brain of both rodents and humans, the ependymal layer of the lateral ventricle contains cells with proliferative potential, which share morphological and functional characteristics with HCs. In addition, putative neural stem cells (NSCs) from the subventricular zone of the lateral ventricle can differentiate into functional SGNs. Also important, the NSCs can incorporate into the sensory epithelia, demonstrating their therapeutic potential. We assert that NSCs and edendymal cells can undergo an epigenetic functional switch to assume functional characteristics of HCs and SGNs. This study suggests that the functional plasticity of renewable cells and conditions that promote functional reprogramming can be used for cell therapy in the auditory setting.
Collapse
Affiliation(s)
- Dongguang Wei
- Department of Anesthesiology and Pain Medicine, Center for Neuroscience, Program in Communication and Sensory Science, University of California, 1544 Newton Court, Davis, CA 95618
| | - Snezana Levic
- Department of Anesthesiology and Pain Medicine, Center for Neuroscience, Program in Communication and Sensory Science, University of California, 1544 Newton Court, Davis, CA 95618
| | - Liping Nie
- Department of Anesthesiology and Pain Medicine, Center for Neuroscience, Program in Communication and Sensory Science, University of California, 1544 Newton Court, Davis, CA 95618
| | - Wei-qiang Gao
- Department of Molecular Biology, Genentech, Inc., South San Francisco, CA 94080; and
| | - Christine Petit
- Unité de Génétique et Physiologie de l'Audition, Unité Mixte de Recherche S587, Institut National de la Santé et de la Recherche Médicale-Université Paris VI, Collège de France, Institut Pasteur, 25 Rue du Dr Roux, 75724 Paris, Cedex 15, France
| | - Edward G. Jones
- Department of Anesthesiology and Pain Medicine, Center for Neuroscience, Program in Communication and Sensory Science, University of California, 1544 Newton Court, Davis, CA 95618
| | - Ebenezer N. Yamoah
- Department of Anesthesiology and Pain Medicine, Center for Neuroscience, Program in Communication and Sensory Science, University of California, 1544 Newton Court, Davis, CA 95618
| |
Collapse
|
32
|
Grothe C, Jungnickel J, Haastert K. Physiological role of basic FGF in peripheral nerve development and regeneration: potential for reconstruction approaches. FUTURE NEUROLOGY 2008. [DOI: 10.2217/14796708.3.5.605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
According to expression studies and functional analyses in mutant mice and in rats, FGF-2 appears to be specifically involved during development of peripheral nerves and in de-/re-generating processes at the lesion site and in spinal ganglia. In the absence of FGF receptor (FGFR)3, axonal and myelin diameters of peripheral nerves are significantly reduced, suggesting that FGFR3 physiologically regulates axonal development. The normally occurring neuronal cell death in spinal ganglia after peripheral nerve axotomy does not take place in FGF-2 and FGFR3-deleted mice, respectively, suggesting that injury-induced apoptosis is mediated via FGF-2 binding to FGFR3. According to a bimodal function of FGF-2, lesion-induced neuron death in rat spinal ganglia can be prevented by application of FGF-2 to the proximal nerve stump, which could be mediated via FGFR1/2. At the lesion site, FGF-2 appears to be involved in stimulating Schwann cell proliferation, promoting neurite outgrowth, especially of sensory nerve fibers, and regulating remyelination.
Collapse
Affiliation(s)
- Claudia Grothe
- Hannover Medical School, Institute of Neuroanatomy, OE 4140, Carl-Neuberg Str. 1, D-30625, Hannover, Germany
| | - Julia Jungnickel
- Hannover Medical School, Institute of Neuroanatomy, OE 4140, Carl-Neuberg Str. 1, D-30625, Hannover, Germany
| | - Kirsten Haastert
- Hannover Medical School, Institute of Neuroanatomy, OE 4140, Carl-Neuberg Str. 1, D-30625, Hannover, Germany
| |
Collapse
|
33
|
Maruyama J, Miller JM, Ulfendahl M. Glial cell line-derived neurotrophic factor and antioxidants preserve the electrical responsiveness of the spiral ganglion neurons after experimentally induced deafness. Neurobiol Dis 2007; 29:14-21. [PMID: 17870569 PMCID: PMC2680080 DOI: 10.1016/j.nbd.2007.07.026] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2007] [Revised: 07/03/2007] [Accepted: 07/24/2007] [Indexed: 10/23/2022] Open
Abstract
Cochlear implant surgery is currently the therapy of choice for profoundly deaf patients. However, the functionality of cochlear implants depends on the integrity of the auditory spiral ganglion neurons. This study assesses the combined efficacy of two classes of agents found effective in preventing degeneration of the auditory nerve following deafness, neurotrophic factors, and antioxidants. Guinea pigs were deafened and treated for 4 weeks with either local administration of GDNF or a combination of GDNF and systemic injections of the antioxidants ascorbic acid and Trolox. The density of surviving spiral ganglion cells was significantly enhanced and the thresholds for eliciting an electrically evoked brain stem response were significantly reduced in GDNF treated animals compared to deafened-untreated. The addition of antioxidants significantly enhanced the evoked responsiveness over that observed with GDNF alone. The results suggest multiple sites of intervention in the rescue of these cells from deafferentation-induced cell death.
Collapse
Affiliation(s)
- Jun Maruyama
- Center for Hearing and Communication Research and Department of Clinical Neuroscience, Karolinska Institutet
- Department of Otolaryngology, Ehime University School of Medicine, Matsuyama, Japan
| | - Josef M. Miller
- Center for Hearing and Communication Research and Department of Clinical Neuroscience, Karolinska Institutet
- Kresge Hearing Research Institute, University of Michigan, Ann Arbor, USA
| | - Mats Ulfendahl
- Center for Hearing and Communication Research and Department of Clinical Neuroscience, Karolinska Institutet
- Department of Otolaryngology, Karolinska University Hospital – Solna, Stockholm, Sweden
- Corresponding author. Address for correspondence: Mats Ulfendahl, PhD, Center for Hearing and Communication Research, Building M1, Karolinska University Hospital, Solna, SE-171 76 Stockholm, Sweden, Phone: +46 8 51776307 Fax: +46 8 301876,
| |
Collapse
|