1
|
Manickam V, Maity S, Murali SV, Gawande DY, Stothert AR, Batalkina L, Cardona AE, Kaur T. Local delivery of soluble fractalkine (CX3CL1) peptide restores ribbon synapses after noise-induced cochlear synaptopathy. Front Cell Neurosci 2024; 18:1486740. [PMID: 39539341 PMCID: PMC11557324 DOI: 10.3389/fncel.2024.1486740] [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: 08/26/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Cochlear ribbon synapses between sensory inner hair cells (IHCs) and spiral ganglion neurons (SGNs) are vulnerable to rapid and primary damage and/or loss due to noise overexposure. Such damaged ribbon synapses can repair spontaneously in mouse and guinea pig. However, the mechanisms for synaptic repair are unclear. Previously, we have demonstrated a critical role for the fractalkine signaling axis (CX3CL1-CX3CR1) in synaptic repair, wherein noise-damaged ribbon synapses are spontaneously repaired in the presence of fractalkine receptor (CX3CR1) expressed by cochlear macrophages. Here, we examined whether local administration of chemokine fractalkine ligand (CX3CL1 or FKN) in the form of a peptide is effective in restoring synapses and hearing loss after noise-induced cochlear synaptopathy (NICS). Specifically, the efficacy of different isoforms of FKN was evaluated for restoration of loss of IHC ribbon synapses and hearing after NICS. A single transtympanic injection of soluble isoform of FKN (sFKN) peptide at 1 day after synaptopathic noise trauma for 2 hours at 93 decibel sound pressure level led to significant recovery of auditory brainstem response (ABR) thresholds, ABR peak I amplitudes and ribbon synapses in FKN knockout mice when compared to mice injected with membrane-bound FKN peptide (mFKN). Likewise, local treatment with sFKN peptide in FKN wild type mice restored synaptopathic noise-damaged ribbon synapses and ABR peak I amplitudes. Mechanistically, FKN regulates macrophage numbers in the damaged cochlea and in the absence of macrophages, sFKN failed to restore loss of synapses and hearing after NICS. Furthermore, sFKN treatment attenuated cochlear inflammation after NICS without altering the expression of CX3CR1. Finally, injected sFKN peptide was detectable inside the cochlea for 24 h localized to the basilar membrane and spiral lamina near the sensory epithelium. These data provide a proof-of-principle that local delivery of an immune factor, sFKN is effective in restoring ribbon synapses and hearing loss after NICS in a macrophage-dependent manner and highlights the potential of sFKN as an immunotherapy for cochlear synaptopathy due to noise.
Collapse
Affiliation(s)
| | - Sibaprasad Maity
- Department of Biomedical Sciences, Creighton University, Omaha, NE, United States
| | - Sree Varshini Murali
- Department of Biomedical Sciences, Creighton University, Omaha, NE, United States
- Department of Otolaryngology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, United States
| | - Dinesh Y. Gawande
- Department of Biomedical Sciences, Creighton University, Omaha, NE, United States
- Department of Otolaryngology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, United States
| | - Andrew R. Stothert
- Department of Biomedical Sciences, Creighton University, Omaha, NE, United States
| | - Lyudamila Batalkina
- Department of Biomedical Sciences, Creighton University, Omaha, NE, United States
| | - Astrid E. Cardona
- Department of Molecular Microbiology and Immunology, University of Texas, San Antonio, TX, United States
| | - Tejbeer Kaur
- Department of Biomedical Sciences, Creighton University, Omaha, NE, United States
- Department of Otolaryngology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, United States
| |
Collapse
|
2
|
Blum K, Schepsky P, Derleder P, Schätzle P, Nasri F, Fischer P, Engel J, Kurt S. Noise-induced cochlear synaptopathy in C57BL/6 N mice as a function of trauma strength: ribbons are more vulnerable than postsynapses. Front Cell Neurosci 2024; 18:1465216. [PMID: 39411002 PMCID: PMC11473312 DOI: 10.3389/fncel.2024.1465216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
Noise-induced cochlear synaptopathy is characterized by irreversible loss of synapses between inner hair cells (IHCs) and spiral ganglion neurons (SGNs) despite normal hearing thresholds. We analyzed hearing performance and cochlear structure in C57BL/6 N mice exposed to 100, 106, or 112 dB SPL broadband noise (8-16 kHz) for 2 h. Auditory brainstem responses (ABRs) were assessed before, directly after, and up to 28 days post-trauma. Finally, the number, size, and pairing of IHC presynaptic (CtBP2-positive) ribbons and postsynaptic AMPA receptor scaffold (Homer1-positive) clusters were analyzed along the cochlea. Four weeks after the 100 dB SPL trauma, a permanent threshold shift (PTS) was observed at 45 kHz, which after the higher traumata extended toward middle to low frequencies. Loss in ABR wave I amplitudes scaled with trauma strength indicating loss of functional IHC synaptic connections. Latencies of wave I mostly increased with trauma strength. No trauma-related OHC loss was found. The number of synaptic pairs was reduced in the midbasal and basal cochlear region in all trauma conditions, with ribbon loss amounting up to 46% of control. Ribbons surviving the trauma were paired, whereas 4-6 unpaired postsynapses/IHC were found in the medial, midbasal, and basal regions irrespective of trauma strength, contrasting findings in CBA/CaJ mice. Our data confirm the susceptibility of ribbon synapses and ABR wave I amplitudes to a noise trauma of 100 dB SPL or larger. Notably, peripheral dendrites bearing IHC postsynapses were less vulnerable than presynaptic ribbons in C57BL/6 N mice.
Collapse
Affiliation(s)
- Kerstin Blum
- Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Department of Biophysics, Saarland University, Homburg, Germany
- Center for Gender-specific Biology and Medicine (CGBM), School of Medicine, Saarland University, Homburg, Germany
| | - Pauline Schepsky
- Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Department of Biophysics, Saarland University, Homburg, Germany
| | - Philip Derleder
- Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Department of Biophysics, Saarland University, Homburg, Germany
| | - Philipp Schätzle
- Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Department of Biophysics, Saarland University, Homburg, Germany
| | - Fahmi Nasri
- Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Department of Biophysics, Saarland University, Homburg, Germany
| | - Philipp Fischer
- Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Department of Biophysics, Saarland University, Homburg, Germany
| | - Jutta Engel
- Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Department of Biophysics, Saarland University, Homburg, Germany
- Center for Gender-specific Biology and Medicine (CGBM), School of Medicine, Saarland University, Homburg, Germany
| | - Simone Kurt
- Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Department of Biophysics, Saarland University, Homburg, Germany
- Center for Gender-specific Biology and Medicine (CGBM), School of Medicine, Saarland University, Homburg, Germany
| |
Collapse
|
3
|
Wang L, Xu M, Zhang Q, Li GL. Amitriptyline protects afferent synapses in the cochlea against excitotoxic trauma in vitro. FEBS J 2024; 291:4111-4124. [PMID: 39128014 DOI: 10.1111/febs.17233] [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: 11/09/2023] [Revised: 03/20/2024] [Accepted: 07/19/2024] [Indexed: 08/13/2024]
Abstract
Afferent synapses between inner hair cells (IHCs) and the type I spiral ganglion neurons (SGNs) in the cochlea provide over 95% of sensory signals for auditory perception in the brain. However, these afferent synapses are particularly vulnerable to damage, for example from excitotoxicity, and exposure to noise in the environment which often leads to noise-induced cochlear synaptopathy (NICS). In this study, we simulated excitotoxic trauma by incubating kainic acid, a non-desensitizing agonist for AMPA type glutamate receptors on cultured cochleae. The possible protective effects of amitriptyline against NICS were examined. We found that, in IHCs, amitriptyline reversed the decrease of Ca2+ current and exocytosis caused by excitotoxic trauma. In SGNs, amitriptyline promoted the recovery of neurite loss caused by excitotoxic trauma. Furthermore, we found that the protective effects of amitriptyline are likely mediated by suppressing apoptosis factors that were upregulated during excitotoxic trauma. In conclusion, our results suggest that amitriptyline could protect afferent synapses in the cochlea from NICS, making it a potential drug candidate for hearing protection.
Collapse
Affiliation(s)
- Liqin Wang
- Department of Otorhinolaryngology, ENT Institute, and NHC Key Laboratory of Hearing Medicine, Eye & ENT Hospital, Fudan University, Shanghai, China
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Mengfan Xu
- School of Life Sciences, Anhui University, Hefei, China
- Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China
| | - Qing Zhang
- Department of Otorhinolaryngology-Head & Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, China
| | - Geng-Lin Li
- Department of Otorhinolaryngology, ENT Institute, and NHC Key Laboratory of Hearing Medicine, Eye & ENT Hospital, Fudan University, Shanghai, China
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Vincent PFY, Young ED, Edge ASB, Glowatzki E. Auditory hair cells and spiral ganglion neurons regenerate synapses with refined release properties in vitro. Proc Natl Acad Sci U S A 2024; 121:e2315599121. [PMID: 39058581 PMCID: PMC11294990 DOI: 10.1073/pnas.2315599121] [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: 09/10/2023] [Accepted: 06/12/2024] [Indexed: 07/28/2024] Open
Abstract
Ribbon synapses between inner hair cells (IHCs) and type I spiral ganglion neurons (SGNs) in the inner ear are damaged by noise trauma and with aging, causing "synaptopathy" and hearing loss. Cocultures of neonatal denervated organs of Corti and newly introduced SGNs have been developed to find strategies for improving IHC synapse regeneration, but evidence of the physiological normality of regenerated synapses is missing. This study utilizes IHC optogenetic stimulation and SGN recordings, showing that, when P3-5 denervated organs of Corti are cocultured with SGNs, newly formed IHC/SGN synapses are indeed functional, exhibiting glutamatergic excitatory postsynaptic currents. When using older organs of Corti at P10-11, synaptic activity probed by deconvolution showed more mature release properties, closer to the specialized mode of IHC synaptic transmission crucial for coding the sound signal. This functional assessment of newly formed IHC synapses developed here, provides a powerful tool for testing approaches to improve synapse regeneration.
Collapse
Affiliation(s)
- Philippe F. Y. Vincent
- The Center for Hearing and Balance, The Johns Hopkins School of Medicine, Baltimore, MD21205
- Department of Otolaryngology Head and Neck Surgery, The Johns Hopkins School of Medicine, Baltimore, MD21205
| | - Eric D. Young
- The Center for Hearing and Balance, The Johns Hopkins School of Medicine, Baltimore, MD21205
- Department of Otolaryngology Head and Neck Surgery, The Johns Hopkins School of Medicine, Baltimore, MD21205
- Department of Neuroscience, The Johns Hopkins School of Medicine, Baltimore, MD21205
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD21205
| | - Albert S. B. Edge
- Department of Otolaryngology, Harvard Medical School, Boston, MA02115
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, MA02114
- Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, MA02115
- Harvard Stem Cell Institute, Cambridge, MA02139
| | - Elisabeth Glowatzki
- The Center for Hearing and Balance, The Johns Hopkins School of Medicine, Baltimore, MD21205
- Department of Otolaryngology Head and Neck Surgery, The Johns Hopkins School of Medicine, Baltimore, MD21205
- Department of Neuroscience, The Johns Hopkins School of Medicine, Baltimore, MD21205
| |
Collapse
|
5
|
Vijayakumar S, DiGuiseppi JA, Dabestani PJ, Ryan WG, Quevedo RV, Li Y, Diers J, Tu S, Fleegel J, Nguyen C, Rhoda LM, Imami AS, Hamoud ARA, Lovas S, McCullumsmith RE, Zallocchi M, Zuo J. In silico transcriptome screens identify epidermal growth factor receptor inhibitors as therapeutics for noise-induced hearing loss. SCIENCE ADVANCES 2024; 10:eadk2299. [PMID: 38896614 PMCID: PMC11186505 DOI: 10.1126/sciadv.adk2299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 05/14/2024] [Indexed: 06/21/2024]
Abstract
Noise-induced hearing loss (NIHL) is a common sensorineural hearing impairment that lacks U.S. Food and Drug Administration-approved drugs. To fill the gap in effective screening models, we used an in silico transcriptome-based drug screening approach, identifying 22 biological pathways and 64 potential small molecule treatments for NIHL. Two of these, afatinib and zorifertinib [epidermal growth factor receptor (EGFR) inhibitors], showed efficacy in zebrafish and mouse models. Further tests with EGFR knockout mice and EGF-morpholino zebrafish confirmed their protective role against NIHL. Molecular studies in mice highlighted EGFR's crucial involvement in NIHL and the protective effect of zorifertinib. When given orally, zorifertinib was found in the perilymph with favorable pharmacokinetics. In addition, zorifertinib combined with AZD5438 (a cyclin-dependent kinase 2 inhibitor) synergistically prevented NIHL in zebrafish. Our results underscore the potential for in silico transcriptome-based drug screening in diseases lacking efficient models and suggest EGFR inhibitors as potential treatments for NIHL, meriting clinical trials.
Collapse
Affiliation(s)
- Sarath Vijayakumar
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Joseph A. DiGuiseppi
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Parinaz Jila Dabestani
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - William G. Ryan
- Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA.
| | - Rene Vielman Quevedo
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Yuju Li
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Jack Diers
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Shu Tu
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Jonathan Fleegel
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Cassidy Nguyen
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Lauren M. Rhoda
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Ali Sajid Imami
- Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA.
| | | | - Sándor Lovas
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Robert E. McCullumsmith
- Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA.
- Neurosciences Institute, ProMedica, Toledo, OH 43606, USA
| | - Marisa Zallocchi
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Jian Zuo
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE 68178, USA
- Ting Therapeutics, University of California San Diego, 9310 Athena Circle, San Diego, CA 92037, USA
| |
Collapse
|
6
|
Hemachandran S, Hu N, Kane CJ, Green SH. Cyclic AMP signaling promotes regeneration of cochlear synapses after excitotoxic or noise trauma. Front Cell Neurosci 2024; 18:1363219. [PMID: 38694536 PMCID: PMC11061447 DOI: 10.3389/fncel.2024.1363219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 03/27/2024] [Indexed: 05/04/2024] Open
Abstract
Introduction Cochlear afferent synapses connecting inner hair cells to spiral ganglion neurons are susceptible to excitotoxic trauma on exposure to loud sound, resulting in a noise-induced cochlear synaptopathy (NICS). Here we assessed the ability of cyclic AMP-dependent protein kinase (PKA) signaling to promote cochlear synapse regeneration, inferred from its ability to promote axon regeneration in axotomized CNS neurons, another system refractory to regeneration. Methods We mimicked NICS in vitro by applying a glutamate receptor agonist, kainic acid (KA) to organotypic cochlear explant cultures and experimentally manipulated cAMP signaling to determine whether PKA could promote synapse regeneration. We then delivered the cAMP phosphodiesterase inhibitor rolipram via implanted subcutaneous minipumps in noise-exposed CBA/CaJ mice to test the hypothesis that cAMP signaling could promote cochlear synapse regeneration in vivo. Results We showed that the application of the cell membrane-permeable cAMP agonist 8-cpt-cAMP or the cAMP phosphodiesterase inhibitor rolipram promotes significant regeneration of synapses in vitro within twelve hours after their destruction by KA. This is independent of neurotrophin-3, which also promotes synapse regeneration. Moreover, of the two independent signaling effectors activated by cAMP - the cAMP Exchange Protein Activated by cAMP and the cAMP-dependent protein kinase - it is the latter that mediates synapse regeneration. Finally, we showed that systemic delivery of rolipram promotes synapse regeneration in vivo following NICS. Discussion In vitro experiments show that cAMP signaling promotes synapse regeneration after excitotoxic destruction of cochlear synapses and does so via PKA signaling. The cAMP phosphodiesterase inhibitor rolipram promotes synapse regeneration in vivo in noise-exposed mice. Systemic administration of rolipram or similar compounds appears to provide a minimally invasive therapeutic approach to reversing synaptopathy post-noise.
Collapse
Affiliation(s)
| | | | | | - Steven H. Green
- Department of Biology, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
7
|
Ismail Mohamad N, Santra P, Park Y, Matthews IR, Taketa E, Chan DK. Synaptic ribbon dynamics after noise exposure in the hearing cochlea. Commun Biol 2024; 7:421. [PMID: 38582813 PMCID: PMC10998851 DOI: 10.1038/s42003-024-06067-y] [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: 09/21/2023] [Accepted: 03/18/2024] [Indexed: 04/08/2024] Open
Abstract
Moderate noise exposure induces cochlear synaptopathy, the loss of afferent ribbon synapses between cochlear hair cells and spiral ganglion neurons, which is associated with functional hearing decline. Prior studies have demonstrated noise-induced changes in the distribution and number of synaptic components, but the dynamic changes that occur after noise exposure have not been directly visualized. Here, we describe a live imaging model using RIBEYE-tagRFP to enable direct observation of pre-synaptic ribbons in mature hearing mouse cochleae after synaptopathic noise exposure. Ribbon number does not change, but noise induces an increase in ribbon volume as well as movement suggesting unanchoring from synaptic tethers. A subgroup of basal ribbons displays concerted motion towards the cochlear nucleus with subsequent migration back to the cell membrane after noise cessation. Understanding the immediate dynamics of synaptic damage after noise exposure may facilitate identification of specific target pathways to treat cochlear synaptopathy.
Collapse
Affiliation(s)
- Noura Ismail Mohamad
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Peu Santra
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Yesai Park
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Ian R Matthews
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Emily Taketa
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Dylan K Chan
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
8
|
Wu PZ, Liberman LD, Liberman MC. Noise-induced synaptic loss and its post-exposure recovery in CBA/CaJ vs. C57BL/6J mice. Hear Res 2024; 445:108996. [PMID: 38547565 PMCID: PMC11024800 DOI: 10.1016/j.heares.2024.108996] [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: 01/23/2024] [Revised: 03/11/2024] [Accepted: 03/22/2024] [Indexed: 04/07/2024]
Abstract
Acute noise-induced loss of synapses between inner hair cells (IHCs) and auditory nerve fibers (ANFs) has been documented in several strains of mice, but the extent of post-exposure recovery reportedly varies dramatically. If such inter-strain heterogeneity is real, it could be exploited to probe molecular pathways mediating neural remodeling in the adult cochlea. Here, we compared synaptopathy repair in CBA/CaJ vs. C57BL/6J, which are at opposite ends of the reported recovery spectrum. We evaluated C57BL/6J mice 0 h, 24 h, 2 wks or 8 wks after exposure for 2 h to octave-band noise (8-16 kHz) at either 90, 94 or 98 dB SPL, to compare with analogous post-exposure results in CBA/CaJ at 98 or 101 dB. We counted pre- and post-synaptic puncta in immunostained cochleas, using machine learning to classify paired (GluA2 and CtBP2) vs. orphan (CtBP2 only) puncta, and batch-processing to quantify immunostaining intensity. At 98 dB, both strains show ongoing loss of ribbons and synapses between 0 and 24 h, followed by partial recovery, however the extent and degree of these changes were greater in C57BL/6J. Much of the synaptic recovery is due to transient reduction in GluA2 intensity in synaptopathic regions. In contrast, CtBP2 intensity showed only transient increases (at 2 wks). Neurofilament staining revealed transient extension of ANF terminals in C57BL/6J, but not in CBA/CaJ, peaking at 24 h and reverting by 2 wks. Thus, although interstrain differences in synapse recovery are dominated by reversible changes in GluA2 receptor levels, the neurite extension seen in C57BL/6J suggests a qualitative difference in regenerative capacity.
Collapse
Affiliation(s)
- Pei-Zhe Wu
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA 02114, USA; Department of Otolaryngology, Harvard Medical School, Boston, MA 02115, USA
| | - Leslie D Liberman
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - M Charles Liberman
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA 02114, USA; Department of Otolaryngology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
9
|
Feng B, Dong T, Song X, Zheng X, Jin C, Cheng Z, Liu Y, Zhang W, Wang X, Tao Y, Wu H. Personalized Porous Gelatin Methacryloyl Sustained-Release Nicotinamide Protects Against Noise-Induced Hearing Loss. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305682. [PMID: 38225752 DOI: 10.1002/advs.202305682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/11/2023] [Indexed: 01/17/2024]
Abstract
There are no Food and Drug Administration-approved drugs for treating noise-induced hearing loss (NIHL), reflecting the absence of clear specific therapeutic targets and effective delivery strategies. Noise trauma is demonstrated results in nicotinamide adenine dinucleotide (NAD+) downregulation and mitochondrial dysfunction in cochlear hair cells (HCs) and spiral ganglion neurons (SGNs) in mice, and NAD+ boosted by nicotinamide (NAM) supplementation maintains cochlear mitochondrial homeostasis and prevents neuroexcitatory toxic injury in vitro and ex vivo, also significantly ameliorated NIHL in vivo. To tackle the limited drug delivery efficiency due to sophisticated anatomical barriers and unique clearance pathway in ear, personalized NAM-encapsulated porous gelatin methacryloyl (PGMA@NAM) are developed based on anatomy topography of murine temporal bone by micro-computed tomography and reconstruction of round window (RW) niche, realizing hydrogel in situ implantation completely, NAM sustained-release and long-term auditory preservation in mice. This study strongly supports personalized PGMA@NAM as NIHL protection drug with effective inner ear delivery, providing new inspiration for drug-based treatment of NIHL.
Collapse
Affiliation(s)
- Baoyi Feng
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639, Zhizaoju Road, Shanghai, 200011, P. R. China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, No.115, Jinzun Road, Shanghai, 200125, P. R. China
- Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200125, P. R. China
| | - Tingting Dong
- Ear Institute, Shanghai Jiao Tong University School of Medicine, No.115, Jinzun Road, Shanghai, 200125, P. R. China
- Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200125, P. R. China
- Biobank of Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.115, Jinzun Road, Shanghai, 200125, P. R. China
| | - Xinyu Song
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Xiaofei Zheng
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639, Zhizaoju Road, Shanghai, 200011, P. R. China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, No.115, Jinzun Road, Shanghai, 200125, P. R. China
- Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200125, P. R. China
| | - Chenxi Jin
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639, Zhizaoju Road, Shanghai, 200011, P. R. China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, No.115, Jinzun Road, Shanghai, 200125, P. R. China
- Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200125, P. R. China
| | - Zhenzhe Cheng
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639, Zhizaoju Road, Shanghai, 200011, P. R. China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, No.115, Jinzun Road, Shanghai, 200125, P. R. China
- Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200125, P. R. China
| | - Yiqing Liu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639, Zhizaoju Road, Shanghai, 200011, P. R. China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, No.115, Jinzun Road, Shanghai, 200125, P. R. China
- Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200125, P. R. China
| | - Wenjie Zhang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Xueling Wang
- Ear Institute, Shanghai Jiao Tong University School of Medicine, No.115, Jinzun Road, Shanghai, 200125, P. R. China
- Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200125, P. R. China
- Biobank of Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.115, Jinzun Road, Shanghai, 200125, P. R. China
| | - Yong Tao
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639, Zhizaoju Road, Shanghai, 200011, P. R. China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, No.115, Jinzun Road, Shanghai, 200125, P. R. China
- Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200125, P. R. China
| | - Hao Wu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639, Zhizaoju Road, Shanghai, 200011, P. R. China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, No.115, Jinzun Road, Shanghai, 200125, P. R. China
- Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200125, P. R. China
| |
Collapse
|
10
|
Yu Q, Liu S, Guo R, Chen K, Li Y, Jiang D, Gong S, Yin L, Liu K. Complete Restoration of Hearing Loss and Cochlear Synaptopathy via Minimally Invasive, Single-Dose, and Controllable Middle Ear Delivery of Brain-Derived Neurotrophic Factor-Poly(dl-lactic acid- co-glycolic acid)-Loaded Hydrogel. ACS NANO 2024; 18:6298-6313. [PMID: 38345574 DOI: 10.1021/acsnano.3c11049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Noise-induced hearing loss (NIHL) often accompanies cochlear synaptopathy, which can be potentially reversed to restore hearing. However, there has been little success in achieving complete recovery of sensorineural deafness using nearly noninvasive middle ear drug delivery before. Here, we present a study demonstrating the efficacy of a middle ear delivery system employing brain-derived neurotrophic factor (BDNF)-poly-(dl-lactic acid-co-glycolic acid) (PLGA)-loaded hydrogel in reversing synaptopathy and restoring hearing function in a mouse model with NIHL. The mouse model achieved using the single noise exposure (NE, 115 dBL, 4 h) exhibited an average 20 dBL elevation of hearing thresholds with intact cochlear hair cells but a loss of ribbon synapses as the primary cause of hearing impairment. We developed a BDNF-PLGA-loaded thermosensitive hydrogel, which was administered via a single controllable injection into the tympanic cavity of noise-exposed mice, allowing its presence in the middle ear for a duration of 2 weeks. This intervention resulted in complete restoration of NIHL at frequencies of click, 4, 8, 16, and 32 kHz. Moreover, the cochlear ribbon synapses exhibited significant recovery, whereas other cochlear components (hair cells and auditory nerves) remained unchanged. Additionally, the cochlea of NE treated mice revealed activation of tropomyosin receptor kinase B (TRKB) signaling upon exposure to BDNF. These findings demonstrate a controllable and minimally invasive therapeutic approach that utilizes a BDNF-PLGA-loaded hydrogel to restore NIHL by specifically repairing cochlear synaptopathy. This tailored middle ear delivery system holds great promise for achieving ideal clinical outcomes in the treatment of NIHL and cochlear synaptopathy.
Collapse
Affiliation(s)
- Qianru Yu
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Shengnan Liu
- School of Materials Science and Engineering,Tsinghua University, Beijing 100084, China
| | - Rui Guo
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Kuntao Chen
- School of Materials Science and Engineering,Tsinghua University, Beijing 100084, China
| | - Yang Li
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Dan Jiang
- Hearing Implant Centre, Guy's and St. Thomas NHS Foundation Trust, London SE1 7EH, United Kingdom
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, United Kingdom
| | - Shusheng Gong
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
- Clinical Center for Hearing Loss, Capital Medical University, Beijing 100050, China
| | - Lan Yin
- School of Materials Science and Engineering,Tsinghua University, Beijing 100084, China
| | - Ke Liu
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
- Clinical Center for Hearing Loss, Capital Medical University, Beijing 100050, China
| |
Collapse
|
11
|
Saidia AR, François F, Casas F, Mechaly I, Venteo S, Veechi JT, Ruel J, Puel JL, Wang J. Oxidative Stress Plays an Important Role in Glutamatergic Excitotoxicity-Induced Cochlear Synaptopathy: Implication for Therapeutic Molecules Screening. Antioxidants (Basel) 2024; 13:149. [PMID: 38397748 PMCID: PMC10886292 DOI: 10.3390/antiox13020149] [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: 12/12/2023] [Revised: 01/12/2024] [Accepted: 01/20/2024] [Indexed: 02/25/2024] Open
Abstract
The disruption of the synaptic connection between the sensory inner hair cells (IHCs) and the auditory nerve fiber terminals of the type I spiral ganglion neurons (SGN) has been observed early in several auditory pathologies (e.g., noise-induced or ototoxic drug-induced or age-related hearing loss). It has been suggested that glutamate excitotoxicity may be an inciting element in the degenerative cascade observed in these pathological cochlear conditions. Moreover, oxidative damage induced by free hydroxyl radicals and nitric oxide may dramatically enhance cochlear damage induced by glutamate excitotoxicity. To investigate the underlying molecular mechanisms involved in cochlear excitotoxicity, we examined the molecular basis responsible for kainic acid (KA, a full agonist of AMPA/KA-preferring glutamate receptors)-induced IHC synapse loss and degeneration of the terminals of the type I spiral ganglion afferent neurons using a cochlear explant culture from P3 mouse pups. Our results demonstrated that disruption of the synaptic connection between IHCs and SGNs induced increased levels of oxidative stress, as well as altered both mitochondrial function and neurotrophin signaling pathways. Additionally, the application of exogenous antioxidants and neurotrophins (NT3, BDNF, and small molecule TrkB agonists) clearly increases synaptogenesis. These results suggest that understanding the molecular pathways involved in cochlear excitotoxicity is of crucial importance for the future clinical trials of drug interventions for auditory synaptopathies.
Collapse
Affiliation(s)
- Anissa Rym Saidia
- Institute for Neurosciences of Montpellier (INM), INSERM U1298, University Montpellier, 34295 Montpellier, France; (F.F.); (I.M.); (S.V.); (J.T.V.); (J.-L.P.)
| | - Florence François
- Institute for Neurosciences of Montpellier (INM), INSERM U1298, University Montpellier, 34295 Montpellier, France; (F.F.); (I.M.); (S.V.); (J.T.V.); (J.-L.P.)
| | - François Casas
- INRA, UMR 866 Dynamique Musculaire et Métabolisme, 34060 Montpellier, France;
| | - Ilana Mechaly
- Institute for Neurosciences of Montpellier (INM), INSERM U1298, University Montpellier, 34295 Montpellier, France; (F.F.); (I.M.); (S.V.); (J.T.V.); (J.-L.P.)
| | - Stéphanie Venteo
- Institute for Neurosciences of Montpellier (INM), INSERM U1298, University Montpellier, 34295 Montpellier, France; (F.F.); (I.M.); (S.V.); (J.T.V.); (J.-L.P.)
| | - Joseph T. Veechi
- Institute for Neurosciences of Montpellier (INM), INSERM U1298, University Montpellier, 34295 Montpellier, France; (F.F.); (I.M.); (S.V.); (J.T.V.); (J.-L.P.)
| | - Jérôme Ruel
- Centre de Recherche en CardioVasculaire et Nutrition, Aix-Marseille Université-INSERM, 1263-INRAE 1260, 13385 Marseille, France;
| | - Jean-Luc Puel
- Institute for Neurosciences of Montpellier (INM), INSERM U1298, University Montpellier, 34295 Montpellier, France; (F.F.); (I.M.); (S.V.); (J.T.V.); (J.-L.P.)
| | - Jing Wang
- Institute for Neurosciences of Montpellier (INM), INSERM U1298, University Montpellier, 34295 Montpellier, France; (F.F.); (I.M.); (S.V.); (J.T.V.); (J.-L.P.)
| |
Collapse
|
12
|
Vincent PF, Young ED, Edge AS, Glowatzki E. Auditory Hair Cells and Spiral Ganglion Neurons Regenerate Synapses with Refined Release Properties In Vitro. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.05.561095. [PMID: 38076928 PMCID: PMC10705289 DOI: 10.1101/2023.10.05.561095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Ribbon synapses between inner hair cells (IHCs) and type I spiral ganglion neurons (SGNs) in the inner ear are damaged by noise trauma and with aging, causing 'synaptopathy 'and hearing loss. Co-cultures of neonatal denervated organs of Corti and newly introduced SGNs have been developed to find strategies for improving IHC synapse regeneration, but evidence of the physiological normality of regenerated synapses is missing. This study utilizes IHC optogenetic stimulation and SGN recordings, showing that newly formed IHC synapses are indeed functional, exhibiting glutamatergic excitatory postsynaptic currents. When older organs of Corti were plated, synaptic activity probed by deconvolution, showed more mature release properties, closer to the highly specialized mode of IHC synaptic transmission that is crucial for coding the sound signal. This newly developed functional assessment of regenerated IHC synapses provides a powerful tool for testing approaches to improve synapse regeneration.
Collapse
Affiliation(s)
- Philippe F.Y. Vincent
- The Center for Hearing and Balance, The Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Otolaryngology Head and Neck Surgery, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Eric D. Young
- The Center for Hearing and Balance, The Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Otolaryngology Head and Neck Surgery, The Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Neuroscience, The Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Albert S.B. Edge
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts, USA
- Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Elisabeth Glowatzki
- The Center for Hearing and Balance, The Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Otolaryngology Head and Neck Surgery, The Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Neuroscience, The Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
13
|
Moverman DJ, Liberman LD, Kraemer S, Corfas G, Liberman MC. Ultrastructure of noise-induced cochlear synaptopathy. Sci Rep 2023; 13:19456. [PMID: 37945811 PMCID: PMC10636047 DOI: 10.1038/s41598-023-46859-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023] Open
Abstract
Acoustic overexposure can eliminate synapses between inner hair cells (IHCs) and auditory nerve fibers (ANFs), even if hair-cell function recovers. This synaptopathy has been extensively studied by confocal microscopy, however, understanding the nature and sequence of damage requires ultrastructural analysis. Here, we used focused ion-beam scanning electron microscopy to mill, image, segment and reconstruct ANF terminals in mice, 1 day and 1 week after synaptopathic exposure (8-16 kHz, 98 dB SPL). At both survivals, ANF terminals were normal in number, but 62% and 53%, respectively, lacked normal synaptic specializations. Most non-synapsing fibers (57% and 48% at 1 day and 1 week) remained in contact with an IHC and contained healthy-looking organelles. ANFs showed a transient increase in mitochondrial content (51%) and efferent innervation (34%) at 1 day. Fibers maintaining synaptic connections showed hypertrophy of pre-synaptic ribbons at both 1 day and 1 week. Non-synaptic fibers were lower in mitochondrial content and typically on the modiolar side of the IHC, where ANFs with high-thresholds and low spontaneous rates are normally found. Even 1 week post-exposure, many ANF terminals remained in IHC contact despite loss of synaptic specializations, thus, regeneration efforts at early post-exposure times should concentrate on synaptogenesis rather than neurite extension.
Collapse
Affiliation(s)
- Daniel J Moverman
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, 243 Charles St., Boston, MA, 02114-3096, USA
| | - Leslie D Liberman
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, 243 Charles St., Boston, MA, 02114-3096, USA
| | - Stephan Kraemer
- Center for Nanoscale Systems, Harvard College, Cambridge, MA, 02138, USA
| | - Gabriel Corfas
- Department of Otolaryngology-Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI, USA
| | - M Charles Liberman
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, 243 Charles St., Boston, MA, 02114-3096, USA.
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
14
|
Wang Y, Abrams KS, Youngman M, Henry KS. Histological Correlates of Auditory Nerve Injury from Kainic Acid in the Budgerigar (Melopsittacus undulatus). J Assoc Res Otolaryngol 2023; 24:473-485. [PMID: 37798548 PMCID: PMC10695905 DOI: 10.1007/s10162-023-00910-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 09/11/2023] [Indexed: 10/07/2023] Open
Abstract
PURPOSE Loss of auditory nerve afferent synapses with cochlear hair cells, called cochlear synaptopathy, is a common pathology in humans caused by aging and noise overexposure. The perceptual consequences of synaptopathy in isolation from other cochlear pathologies are still unclear. Animal models provide an effective approach to resolve uncertainty regarding the physiological and perceptual consequences of auditory nerve loss, because neural lesions can be induced and readily quantified. The budgerigar, a parakeet species, has recently emerged as an animal model for synaptopathy studies based on its capacity for vocal learning and ability to behaviorally discriminate simple and complex sounds with acuity similar to humans. Kainic acid infusions in the budgerigar produce a profound reduction of compound auditory nerve responses, including wave I of the auditory brainstem response, without impacting physiological hair cell measures. These results suggest selective auditory nerve damage. However, histological correlates of neural injury from kainic acid are still lacking. METHODS We quantified the histological effects caused by intracochlear infusion of kainic acid (1 mM; 2.5 µL), and evaluated correlations between the histological and physiological assessments of auditory nerve status. RESULTS Kainic acid infusion in budgerigars produced pronounced loss of neural auditory nerve soma (60% on average) in the cochlear ganglion, and of peripheral axons, at time points 2 or more months following injury. The hair cell epithelium was unaffected by kainic acid. Neural loss was significantly correlated with reduction of compound auditory nerve responses and auditory brainstem response wave I. CONCLUSION Compound auditory nerve responses and wave I provide a useful index of cochlear synaptopathy in this animal model.
Collapse
Affiliation(s)
- Yingxuan Wang
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA
| | - Kristina S Abrams
- Department of Neuroscience, University of Rochester, Rochester, NY 14642, USA
| | - Margaret Youngman
- Department of Otolaryngology, University of Rochester, Rochester, NY 14642, USA
| | - Kenneth S Henry
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA.
- Department of Neuroscience, University of Rochester, Rochester, NY 14642, USA.
- Department of Otolaryngology, University of Rochester, Rochester, NY 14642, USA.
| |
Collapse
|
15
|
Kempfle JS, Jung DH. Experimental drugs for the prevention or treatment of sensorineural hearing loss. Expert Opin Investig Drugs 2023; 32:643-654. [PMID: 37598357 DOI: 10.1080/13543784.2023.2242253] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 07/26/2023] [Indexed: 08/22/2023]
Abstract
INTRODUCTION Sensorineural hearing loss results in irreversible loss of inner ear hair cells and spiral ganglion neurons. Reduced sound detection and speech discrimination can span all ages, and sensorineural hearing rehabilitation is limited to amplification with hearing aids or cochlear implants. Recent insights into experimental drug treatments for inner ear regeneration and otoprotection have paved the way for clinical trials in order to restore a more physiological hearing experience. Paired with the development of innovative minimally invasive approaches for drug delivery to the inner ear, new, emerging treatments for hearing protection and restoration are within reach. AREAS COVERED This expert opinion provides an overview of the latest experimental drug therapies to protect from and to restore sensorineural hearing loss. EXPERT OPINION The degree and type of cellular damage to the cochlea, the responsiveness of remaining, endogenous cells to regenerative treatments, and the duration of drug availability within cochlear fluids will determine the success of hearing protection or restoration.
Collapse
Affiliation(s)
- Judith S Kempfle
- Department of Otolaryngology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Otolaryngology, Head & Neck Surgery, Harvard Medical School, Boston, MA, USA
- Department of Otolaryngology, UMass Memorial Medical Center, Worcester, MA, USA
- Department of Otolaryngology, Head & Neck Surgery, University of Massachusetts Medical School, Worcester, MA, USA
| | - David H Jung
- Department of Otolaryngology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Otolaryngology, Head & Neck Surgery, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
16
|
Vijayakumar S, DiGuiseppi JA, Dabestani J, Ryan WG, Vielman Quevedo R, Li Y, Diers J, Tu S, Fleegel J, Nguyen C, Rhoda LM, Imami AS, Hamoud AAR, Lovas S, McCullumsmith R, Zallocchi M, Zuo J. In Silico Transcriptome-based Screens Identify Epidermal Growth Factor Receptor Inhibitors as Therapeutics for Noise-induced Hearing Loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.07.544128. [PMID: 37333346 PMCID: PMC10274759 DOI: 10.1101/2023.06.07.544128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Noise-Induced Hearing Loss (NIHL) represents a widespread disease for which no therapeutics have been approved by the Food and Drug Administration (FDA). Addressing the conspicuous void of efficacious in vitro or animal models for high throughput pharmacological screening, we utilized an in silico transcriptome-oriented drug screening strategy, unveiling 22 biological pathways and 64 promising small molecule candidates for NIHL protection. Afatinib and zorifertinib, both inhibitors of the Epidermal Growth Factor Receptor (EGFR), were validated for their protective efficacy against NIHL in experimental zebrafish and murine models. This protective effect was further confirmed with EGFR conditional knockout mice and EGF knockdown zebrafish, both demonstrating protection against NIHL. Molecular analysis using Western blot and kinome signaling arrays on adult mouse cochlear lysates unveiled the intricate involvement of several signaling pathways, with particular emphasis on EGFR and its downstream pathways being modulated by noise exposure and Zorifertinib treatment. Administered orally, Zorifertinib was successfully detected in the perilymph fluid of the inner ear in mice with favorable pharmacokinetic attributes. Zorifertinib, in conjunction with AZD5438 - a potent inhibitor of cyclin dependent kinase 2 - produced synergistic protection against NIHL in the zebrafish model. Collectively, our findings underscore the potential application of in silico transcriptome-based drug screening for diseases bereft of efficient screening models and posit EGFR inhibitors as promising therapeutic agents warranting clinical exploration for combatting NIHL. Highlights In silico transcriptome-based drug screens identify pathways and drugs against NIHL.EGFR signaling is activated by noise but reduced by zorifertinib in mouse cochleae.Afatinib, zorifertinib and EGFR knockout protect against NIHL in mice and zebrafish.Orally delivered zorifertinib has inner ear PK and synergizes with a CDK2 inhibitor.
Collapse
|
17
|
Choi JE, Carpena NT, Lee JH, Chang SY, Lee MY, Jung JY, Chung WH. Round-window delivery of lithium chloride regenerates cochlear synapses damaged by noise-induced excitotoxic trauma via inhibition of the NMDA receptor in the rat. PLoS One 2023; 18:e0284626. [PMID: 37216352 DOI: 10.1371/journal.pone.0284626] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 04/04/2023] [Indexed: 05/24/2023] Open
Abstract
Noise exposure can destroy the synaptic connections between hair cells and auditory nerve fibers without damaging the hair cells, and this synaptic loss could contribute to difficult hearing in noisy environments. In this study, we investigated whether delivering lithium chloride to the round-window can regenerate synaptic loss of cochlea after acoustic overexposure. Our rat animal model of noise-induced cochlear synaptopathy caused about 50% loss of synapses in the cochlear basal region without damaging hair cells. We locally delivered a single treatment of poloxamer 407 (vehicle) containing lithium chloride (either 1 mM or 2 mM) to the round-window niche 24 hours after noise exposure. Controls included animals exposed to noise who received only the vehicle. Auditory brainstem responses were measured 3 days, 1 week, and 2 weeks post-exposure treatment, and cochleas were harvested 1 week and 2 weeks post-exposure treatment for histological analysis. As documented by confocal microscopy of immunostained ribbon synapses, local delivery of 2 mM lithium chloride produced synaptic regeneration coupled with corresponding functional recovery, as seen in the suprathreshold amplitude of auditory brainstem response wave 1. Western blot analyses revealed that 2 mM lithium chloride suppressed N-methyl-D-aspartate (NMDA) receptor expression 7 days after noise-exposure. Thus, round-window delivery of lithium chloride using poloxamer 407 reduces cochlear synaptic loss after acoustic overexposure by inhibiting NMDA receptor activity in rat model.
Collapse
Affiliation(s)
- Ji Eun Choi
- Department of Otolaryngology Head and Neck Surgery, Dankook University Hospital, College of Medicine, Dankook University, Cheonan, South Korea
- Multi-modality Treatment Research Center for Auditory/Vestibular Disease, College of Medicine, Dankook University, Cheonan, South Korea
| | - Nathaniel T Carpena
- Multi-modality Treatment Research Center for Auditory/Vestibular Disease, College of Medicine, Dankook University, Cheonan, South Korea
| | - Jae-Hun Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - So-Young Chang
- Multi-modality Treatment Research Center for Auditory/Vestibular Disease, College of Medicine, Dankook University, Cheonan, South Korea
| | - Min Young Lee
- Department of Otolaryngology Head and Neck Surgery, Dankook University Hospital, College of Medicine, Dankook University, Cheonan, South Korea
- Multi-modality Treatment Research Center for Auditory/Vestibular Disease, College of Medicine, Dankook University, Cheonan, South Korea
| | - Jae Yun Jung
- Department of Otolaryngology Head and Neck Surgery, Dankook University Hospital, College of Medicine, Dankook University, Cheonan, South Korea
- Multi-modality Treatment Research Center for Auditory/Vestibular Disease, College of Medicine, Dankook University, Cheonan, South Korea
| | - Won-Ho Chung
- Department of Otorhinolaryngology-Head and Neck Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
18
|
Al Aameri RFH, Alanisi EMA, Oluwatosin A, Al Sallami D, Sheth S, Alberts I, Patel S, Rybak LP, Ramkumar V. Targeting CXCL1 chemokine signaling for treating cisplatin ototoxicity. Front Immunol 2023; 14:1125948. [PMID: 37063917 PMCID: PMC10102581 DOI: 10.3389/fimmu.2023.1125948] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 03/17/2023] [Indexed: 04/03/2023] Open
Abstract
Cisplatin is chemotherapy used for solid tumor treatment like lung, bladder, head and neck, ovarian and testicular cancers. However, cisplatin-induced ototoxicity limits the utility of this agent in cancer patients, especially when dose escalations are needed. Ototoxicity is associated with cochlear cell death through DNA damage, the generation of reactive oxygen species (ROS) and the consequent activation of caspase, glutamate excitotoxicity, inflammation, apoptosis and/or necrosis. Previous studies have demonstrated a role of CXC chemokines in cisplatin ototoxicity. In this study, we investigated the role of CXCL1, a cytokine which increased in the serum and cochlea by 24 h following cisplatin administration. Adult male Wistar rats treated with cisplatin demonstrated significant hearing loss, assessed by auditory brainstem responses (ABRs), hair cell loss and loss of ribbon synapse. Immunohistochemical studies evaluated the levels of CXCL1 along with increased presence of CD68 and CD45-positive immune cells in cochlea. Increases in CXCL1 was time-dependent in the spiral ganglion neurons and organ of Corti and was associated with progressive increases in CD45, CD68 and IBA1-positive immune cells. Trans-tympanic administration of SB225002, a chemical inhibitor of CXCR2 (receptor target for CXCL1) reduced immune cell migration, protected against cisplatin-induced hearing loss and preserved hair cell integrity. We show that SB225002 reduced the expression of CXCL1, NOX3, iNOS, TNF-α, IL-6 and COX-2. Similarly, knockdown of CXCR2 by trans-tympanic administration of CXCR2 siRNA protected against hearing loss and loss of outer hair cells and reduced ribbon synapses. In addition, SB225002 reduced the expression of inflammatory mediators induced by cisplatin. These results implicate the CXCL1 chemokine as an early player in cisplatin ototoxicity, possibly by initiating the immune cascade, and indicate that CXCR2 is a relevant target for treating cisplatin ototoxicity.
Collapse
Affiliation(s)
- Raheem F. H. Al Aameri
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Entkhab M. A. Alanisi
- Department of Pharmaceutical Sciences, Larkin University College of Pharmacy, Miami, FL, United States
| | - Adu Oluwatosin
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Dheyaa Al Sallami
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Sandeep Sheth
- Department of Pharmaceutical Sciences, Larkin University College of Pharmacy, Miami, FL, United States
| | - Ian Alberts
- Medical Microbiology, Immunology and Cell Biology (MMICB), Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Shree Patel
- Medical Microbiology, Immunology and Cell Biology (MMICB), Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Leonard P. Rybak
- Department of Surgery, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Vickram Ramkumar
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
- *Correspondence: Vickram Ramkumar,
| |
Collapse
|
19
|
Malfeld K, Armbrecht N, Pich A, Volk HA, Lenarz T, Scheper V. Prevention of Noise-Induced Hearing Loss In Vivo: Continuous Application of Insulin-like Growth Factor 1 and Its Effect on Inner Ear Synapses, Auditory Function and Perilymph Proteins. Int J Mol Sci 2022; 24:ijms24010291. [PMID: 36613734 PMCID: PMC9820558 DOI: 10.3390/ijms24010291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/19/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
As noise-induced hearing loss (NIHL) is a leading cause of occupational diseases, there is an urgent need for the development of preventive and therapeutic interventions. To avoid user-compliance-based problems occurring with conventional protection devices, the pharmacological prevention is currently in the focus of hearing research. Noise exposure leads to an increase in reactive oxygen species (ROS) in the cochlea. This way antioxidant agents are a promising option for pharmacological interventions. Previous animal studies reported preventive as well as therapeutic effects of Insulin-like growth factor 1 (IGF-1) in the context of NIHL. Unfortunately, in patients the time point of the noise trauma cannot always be predicted, and additive effects may occur. Therefore, continuous prevention seems to be beneficial. The present study aimed to investigate the preventive potential of continuous administration of low concentrations of IGF-1 to the inner ear in an animal model of NIHL. Guinea pigs were unilaterally implanted with an osmotic minipump. One week after surgery they received noise trauma, inducing a temporary threshold shift. Continuous IGF-1 delivery lasted for seven more days. It did not lead to significantly improved hearing thresholds compared to control animals. Quite the contrary, there is a hint for a higher noise susceptibility. Nevertheless, changes in the perilymph proteome indicate a reduced damage and better repair mechanisms through the IGF-1 treatment. Thus, future studies should investigate delivery methods enabling continuous prevention but reducing the risk of an overdosage.
Collapse
Affiliation(s)
- Kathrin Malfeld
- Department of Otolaryngology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany
| | - Nina Armbrecht
- Department of Otolaryngology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Andreas Pich
- Core Facility Proteomics, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Holger A. Volk
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany
| | - Thomas Lenarz
- Department of Otolaryngology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Cluster of Excellence “Hearing4all”, German Research Foundation (DFG; “Deutsche Forschungsgemeinschaft”), Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Verena Scheper
- Department of Otolaryngology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Cluster of Excellence “Hearing4all”, German Research Foundation (DFG; “Deutsche Forschungsgemeinschaft”), Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Correspondence:
| |
Collapse
|
20
|
Schwieger J, Frisch AS, Rau TS, Lenarz T, Hügl S, Scheper V. 3D Printed Cell Culture Chamber for Testing the Effect of Pump-Based Chronic Drug Delivery on Inner Ear Tissue. Biomolecules 2022; 12:biom12040589. [PMID: 35454178 PMCID: PMC9032916 DOI: 10.3390/biom12040589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/11/2022] [Accepted: 04/15/2022] [Indexed: 11/16/2022] Open
Abstract
Cochlear hair cell damage and spiral ganglion neuron (SGN) degeneration are the main causes of sensory neural hearing loss. Cochlear implants (CIs) can replace the function of the hair cells and stimulate the SGNs electrically. The condition of the SGNs and their spatial distance to the CI are key factors for CI-functionality. For a better performance, a high number of neurons and a closer contact to the electrode are intended. Neurotrophic factors are able to enhance SGN survival and neurite outgrowth, and thereby might optimize the electrode-nerve interaction. This would require chronic factor treatment, which is not yet established for the inner ear. Investigations on chronic drug delivery to SGNs could benefit from an appropriate in vitro model. Thus, an inner ear inspired Neurite Outgrowth Chamber (NOC), which allows the incorporation of a mini-osmotic pump for long-term drug delivery, was designed and three-dimensionally printed. The NOC’s function was validated using spiral ganglion explants treated with ciliary neurotrophic factor, neurotrophin-3, or control fluid released via pumps over two weeks. The NOC proved to be suitable for explant cultivation and observation of pump-based drug delivery over the examined period, with neurotrophin-3 significantly increasing neurite outgrowth compared to the other groups.
Collapse
Affiliation(s)
- Jana Schwieger
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, 30625 Hannover, Germany; (A.S.F.); (T.S.R.); (T.L.); (S.H.); (V.S.)
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), Stadtfelddamm 34, 30625 Hannover, Germany
- Cluster of Excellence “Hearing4all” EXC 1077/2, 30625 Hannover, Germany
- Correspondence: ; Tel.: +49-5115327262
| | - Anna Sophie Frisch
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, 30625 Hannover, Germany; (A.S.F.); (T.S.R.); (T.L.); (S.H.); (V.S.)
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), Stadtfelddamm 34, 30625 Hannover, Germany
| | - Thomas S. Rau
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, 30625 Hannover, Germany; (A.S.F.); (T.S.R.); (T.L.); (S.H.); (V.S.)
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), Stadtfelddamm 34, 30625 Hannover, Germany
- Cluster of Excellence “Hearing4all” EXC 1077/2, 30625 Hannover, Germany
| | - Thomas Lenarz
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, 30625 Hannover, Germany; (A.S.F.); (T.S.R.); (T.L.); (S.H.); (V.S.)
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), Stadtfelddamm 34, 30625 Hannover, Germany
- Cluster of Excellence “Hearing4all” EXC 1077/2, 30625 Hannover, Germany
| | - Silke Hügl
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, 30625 Hannover, Germany; (A.S.F.); (T.S.R.); (T.L.); (S.H.); (V.S.)
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), Stadtfelddamm 34, 30625 Hannover, Germany
- Cluster of Excellence “Hearing4all” EXC 1077/2, 30625 Hannover, Germany
| | - Verena Scheper
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, 30625 Hannover, Germany; (A.S.F.); (T.S.R.); (T.L.); (S.H.); (V.S.)
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), Stadtfelddamm 34, 30625 Hannover, Germany
- Cluster of Excellence “Hearing4all” EXC 1077/2, 30625 Hannover, Germany
| |
Collapse
|
21
|
Guo R, Xu Y, Xiong W, Wei W, Qi Y, Du Z, Gong S, Tao Z, Liu K. Autophagy-Mediated Synaptic Refinement and Auditory Neural Pruning Contribute to Ribbon Synaptic Maturity in the Developing Cochlea. Front Mol Neurosci 2022; 15:850035. [PMID: 35310883 PMCID: PMC8931412 DOI: 10.3389/fnmol.2022.850035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 01/28/2022] [Indexed: 11/13/2022] Open
Abstract
In rodents, massive initial synapses are formed in the auditory peripheral nervous system at the early postnatal stage, and one of the major phenomena is that the number of afferent synapses in the cochlea is significantly reduced in the duration of development. This raises the hypothesis that the number of cochlear ribbon synapses are dramatically changed with hearing development and maturation. In this study, several tracers identifying activities of autophagy were applied to estimate the level of autophagy activity in the process of ribbon synapse development in mice; further, changes in the synaptic number and spiral ganglion nerve (SGN) fibers were quantitatively measured. We found robust expression of LC3B and lysosomal-associated membrane protein 1 as well as LysoTracker in or near inner hair cells and cochlear ribbon synapses in the early stage of postnatal development. Moreover, we found a significant loss in the intensity of SGN fibers at ribbon synaptic development and hearing onset. Thus, this study demonstrates that ribbon synaptic refinement and SGN fibers pruning are closely associated with the morphological and functional maturation of ribbon synapses and that synaptic refinement and SGN fiber pruning are regulated by the robust activities of autophagy in the earlier stages of auditory development.
Collapse
Affiliation(s)
- Rui Guo
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yice Xu
- Department of Otolaryngology Head and Neck Surgery, Xiaogan Central Hospital, Wuhan University of Science and Technology, Xiaogan, China
| | - Wei Xiong
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wei Wei
- Department of Otology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Yue Qi
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhengde Du
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Shusheng Gong
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- *Correspondence: Shusheng Gong,
| | - Zezhang Tao
- Department of Otolaryngology Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ke Liu
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Ke Liu,
| |
Collapse
|
22
|
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
|
23
|
Chen F, Zhao F, Mahafza N, Lu W. Detecting Noise-Induced Cochlear Synaptopathy by Auditory Brainstem Response in Tinnitus Patients With Normal Hearing Thresholds: A Meta-Analysis. Front Neurosci 2021; 15:778197. [PMID: 34987358 PMCID: PMC8721093 DOI: 10.3389/fnins.2021.778197] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/15/2021] [Indexed: 01/10/2023] Open
Abstract
Noise-induced cochlear synaptopathy (CS) is defined as a permanent loss of synapses in the auditory nerve pathway following noise exposure. Several studies using auditory brainstem response (ABR) have indicated the presence of CS and increased central gain in tinnitus patients with normal hearing thresholds (TNHT), but the results were inconsistent. This meta-analysis aimed to review the evidence of CS and its pathological changes in the central auditory system in TNHT. Published studies using ABR to study TNHT were reviewed. PubMed, EMBASE, and Scopus databases were selected to search for relevant literature. Studies (489) were retrieved, and 11 were included for meta-analysis. The results supported significantly reduced wave I amplitude in TNHT, whereas the alternations in wave V amplitude were inconsistent among the studies. Consistently increased V/I ratio indicated noise-induced central gain enhancement. The results indicated the evidence of noise-induced cochlear synaptopathy in tinnitus patients with normal hearing. However, inconsistent changes in wave V amplitude may be explained by that the failure of central gain that triggers the pathological neural changes in the central auditory system and/or that increased central gain may be necessary to generate tinnitus but not to maintain tinnitus.
Collapse
Affiliation(s)
- Feifan Chen
- Centre for Speech and Language Therapy and Hearing Science, Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, United Kingdom
| | - Fei Zhao
- Centre for Speech and Language Therapy and Hearing Science, Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, United Kingdom
- Department of Hearing and Speech Science, Guangzhou Xinhua College, Guangzhou, China
| | - Nadeem Mahafza
- Centre for Speech and Language Therapy and Hearing Science, Cardiff School of Sport and Health Sciences, Cardiff Metropolitan University, Cardiff, United Kingdom
| | - Wei Lu
- Department of Otolaryngology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
24
|
Ding D, Qi W, Jiang H, Salvi R. Excitotoxic damage to auditory nerve afferents and spiral ganglion neurons is correlated with developmental upregulation of AMPA and KA receptors. Hear Res 2021; 411:108358. [PMID: 34607211 DOI: 10.1016/j.heares.2021.108358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/29/2021] [Accepted: 09/21/2021] [Indexed: 12/31/2022]
Abstract
Excess release of glutamate at the inner hair cell-type I auditory nerve synapse results in excitotoxicity characterized by rapid swelling and disintegration of the afferent synapses, but in some cases, the damage expands to the spiral ganglion soma. Cochlear excitotoxic damage is largely mediated by α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPAR) and kainate receptor (KAR) and potentially N-methyl-D-aspartate receptors (NMDAR). Because these receptors are developmentally regulated, the pattern of excitotoxic damage could change during development. To test this hypothesis, we compared AMPAR, NMDAR and KAR immunolabeling and excitotoxic damage patterns in rat postnatal day 3 (P3) and adult cochlear cultures. At P3, AMPAR and KAR immunolabeling, but not NMDAR, was abundantly expressed on peripheral nerve terminals adjacent to IHCs. In contrast, AMPAR, KAR and NMDAR immunolabeling was minimal or undetectable on the SGN soma. In adult rats, however, AMPAR, KAR and NMDAR immunolabeling occurred on both peripheral nerve terminals near IHCs as well as the soma of SGNs. High doses of Glu and KA only damaged peripheral nerve terminals near IHCs, but not SGNs, at P3, consistent with selective expression of AMPAR and KAR expression on the terminals. However, in adults, Glu and KA damaged both peripheral nerve terminals near IHCs and SGNs both of which expressed AMPAR and KAR. These results indicate that cochlear excitotoxic damage is closely correlated with structures that express AMPAR and KAR.
Collapse
Affiliation(s)
- Dalian Ding
- Center for Hearing and Deafness, University at Buffalo, 137 Cary Hall, Buffalo, NY 14214, USA
| | - Weidong Qi
- Department of Otolaryngology, Huashan Hospital Fudan University, Shanghai 200040, China
| | - Haiyan Jiang
- Center for Hearing and Deafness, University at Buffalo, 137 Cary Hall, Buffalo, NY 14214, USA
| | - Richard Salvi
- Center for Hearing and Deafness, University at Buffalo, 137 Cary Hall, Buffalo, NY 14214, USA.
| |
Collapse
|
25
|
Kempfle JS, Duro MV, Zhang A, Amador CD, Kuang R, Lu R, Kashemirov BA, Edge AS, McKenna CE, Jung DH. A Novel Small Molecule Neurotrophin-3 Analogue Promotes Inner Ear Neurite Outgrowth and Synaptogenesis In vitro. Front Cell Neurosci 2021; 15:666706. [PMID: 34335184 PMCID: PMC8319950 DOI: 10.3389/fncel.2021.666706] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 06/18/2021] [Indexed: 11/15/2022] Open
Abstract
Sensorineural hearing loss is irreversible and is associated with the loss of spiral ganglion neurons (SGNs) and sensory hair cells within the inner ear. Improving spiral ganglion neuron (SGN) survival, neurite outgrowth, and synaptogenesis could lead to significant gains for hearing-impaired patients. There has therefore been intense interest in the use of neurotrophic factors in the inner ear to promote both survival of SGNs and re-wiring of sensory hair cells by surviving SGNs. Neurotrophin-3 (NT-3) and brain-derived neurotrophic factor (BDNF) represent the primary neurotrophins in the inner ear during development and throughout adulthood, and have demonstrated potential for SGN survival and neurite outgrowth. We have pioneered a hybrid molecule approach to maximize SGN stimulation in vivo, in which small molecule analogues of neurotrophins are linked to bisphosphonates, which in turn bind to cochlear bone. We have previously shown that a small molecule BDNF analogue coupled to risedronate binds to bone matrix and promotes SGN neurite outgrowth and synaptogenesis in vitro. Because NT-3 has been shown in a variety of contexts to have a greater regenerative capacity in the cochlea than BDNF, we sought to develop a similar approach for NT-3. 1Aa is a small molecule analogue of NT-3 that has been shown to activate cells through TrkC, the NT-3 receptor, although its activity on SGNs has not previously been described. Herein we describe the design and synthesis of 1Aa and a covalent conjugate of 1Aa with risedronate, Ris-1Aa. We demonstrate that both 1Aa and Ris-1Aa stimulate neurite outgrowth in SGN cultures at a significantly higher level compared to controls. Ris-1Aa maintained its neurotrophic activity when bound to hydroxyapatite, the primary mineral component of bone. Both 1Aa and Ris-1Aa promote significant synaptic regeneration in cochlear explant cultures, and both 1Aa and Ris-1Aa appear to act at least partly through TrkC. Our results provide the first evidence that a small molecule analogue of NT-3 can stimulate SGNs and promote regeneration of synapses between SGNs and inner hair cells. Our findings support the promise of hydroxyapatite-targeting bisphosphonate conjugation as a novel strategy to deliver neurotrophic agents to SGNs encased within cochlear bone.
Collapse
Affiliation(s)
- Judith S Kempfle
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, United States.,Department of Otolaryngology, University Medical Center Tübingen, Tübingen, Germany
| | - Marlon V Duro
- Department of Chemistry, University of Southern California, Los Angeles, CA, United States
| | - Andrea Zhang
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, United States
| | - Carolina D Amador
- Department of Chemistry, University of Southern California, Los Angeles, CA, United States
| | - Richard Kuang
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, United States
| | - Ryan Lu
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, United States
| | - Boris A Kashemirov
- Department of Chemistry, University of Southern California, Los Angeles, CA, United States
| | - Albert S Edge
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, United States
| | - Charles E McKenna
- Department of Chemistry, University of Southern California, Los Angeles, CA, United States
| | - David H Jung
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
26
|
Zhang C, Li Q, Chen M, Lu T, Min S, Li S. The role of oxidative stress in the susceptibility of noise-impaired cochleae to synaptic loss induced by intracochlear electrical stimulation. Neuropharmacology 2021; 196:108707. [PMID: 34246683 DOI: 10.1016/j.neuropharm.2021.108707] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/15/2021] [Accepted: 07/06/2021] [Indexed: 11/20/2022]
Abstract
Intracochlear electrical stimulation (ES) generated by cochlear implants (CIs) is used to activate auditory nerves to restore hearing perception in deaf subjects and those with residual hearing who use electroacoustic stimulation (EAS) technology. Approximately 1/3 of EAS recipients experience loss of residual hearing a few months after ES activation, but the underlying mechanism is unknown. Clinical evidence indicates that the loss is related to the previous history of noise-induced hearing loss (NIHL). In this report, we investigated the impact of intracochlear ES on oxidative stress levels and synaptic counts in inner hair cells (IHCs) of the apical, middle and basal regions of guinea pigs with normal hearing (NH) and NIHL. Our results demonstrated that intracochlear ES with an intensity of 6 dB above the thresholds of electrically evoked compound action potentials (ECAPs) could induce the elevation of oxidative stress levels, resulting in a loss of IHC synapses near the electrodes in the basal and middle regions of the NH cochleae. Furthermore, the apical region of cochleae with NIHL were more susceptible to synaptic loss induced by relatively low-intensity ES than that of NH cochleae, resulting from the additional elevation of oxidative stress levels and the reduced antioxidant capability throughout the whole cochlea.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Otolaryngology - Head and Neck Surgery, Eye & ENT Hospital of Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Hearing Medicine, Shanghai, China
| | - Qiang Li
- Department of Otolaryngology - Head and Neck Surgery, Eye & ENT Hospital of Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Hearing Medicine, Shanghai, China
| | - Min Chen
- Department of Otolaryngology - Head and Neck Surgery, Eye & ENT Hospital of Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Hearing Medicine, Shanghai, China
| | - Tianhao Lu
- Department of Otolaryngology - Head and Neck Surgery, Eye & ENT Hospital of Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Hearing Medicine, Shanghai, China
| | - Shiyao Min
- Department of Otolaryngology - Head and Neck Surgery, Eye & ENT Hospital of Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Hearing Medicine, Shanghai, China
| | - Shufeng Li
- Department of Otolaryngology - Head and Neck Surgery, Eye & ENT Hospital of Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Hearing Medicine, Shanghai, China.
| |
Collapse
|
27
|
Koizumi Y, Mizutari K, Kawauchi S, Sato S, Shiotani A, Kakehata S. Y-27632, a ROCK inhibitor, improved laser-induced shock wave (LISW)-induced cochlear synaptopathy in mice. Mol Brain 2021; 14:105. [PMID: 34217338 PMCID: PMC8254252 DOI: 10.1186/s13041-021-00819-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/28/2021] [Indexed: 11/10/2022] Open
Abstract
Recently, a pathological condition called cochlear synaptopathy has been clarified, and as a disorder of the auditory nerve synapses that occurs prior to failure of hair cells, it has been recognized as a major cause of sensorineural hearing loss. However, cochlear synaptopathy is untreatable. Inhibition of rho-associated coiled-coil containing protein kinase (ROCK), a serine-threonine protein kinase, has been reported to have neuroprotective and regenerative effects on synaptic pathways in the nervous system, including those in the inner ear. We previously demonstrated the regenerative effect of the ROCK inhibitor, Y-27632, on an excitotoxic cochlear nerve damage model in vitro. In this study, we aimed to validate the effect of ROCK inhibition on mice with cochlear synaptopathy induced by laser-induced shock wave (LISW) in vivo. After the elevation of ROCK1/2 expression in the damaged cochlea was confirmed, we administered Y-27632 locally via the middle ear. The amplitude of wave I in the auditory brainstem response and the number of synapses in the Y-27632-treated cochlea increased significantly. These results clearly demonstrate that ROCK inhibition has a promising clinical application in the treatment of cochlear synaptopathy, which is the major pathology of sensorineural hearing loss.
Collapse
Affiliation(s)
- Yutaka Koizumi
- Department of Otolaryngology-Head and Neck Surgery, Yamagata University Faculty of Medicine, 2-2-2 Iida-Nishi, Yamagata, 990-9585, Japan
| | - Kunio Mizutari
- Department of Otolaryngology-Head and Neck Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan.
| | - Satoko Kawauchi
- Division of Bioinformation and Therapeutic Systems, National Defense Medical College Research Institute, Saitama, 359-8513, Japan
| | - Shunichi Sato
- Division of Bioinformation and Therapeutic Systems, National Defense Medical College Research Institute, Saitama, 359-8513, Japan
| | - Akihiro Shiotani
- Department of Otolaryngology-Head and Neck Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Seiji Kakehata
- Department of Otolaryngology-Head and Neck Surgery, Yamagata University Faculty of Medicine, 2-2-2 Iida-Nishi, Yamagata, 990-9585, Japan.
| |
Collapse
|
28
|
In Situ 3D-Imaging of the Inner Ear Synapses with a Cochlear Implant. Life (Basel) 2021; 11:life11040301. [PMID: 33915846 PMCID: PMC8066088 DOI: 10.3390/life11040301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/27/2021] [Accepted: 03/28/2021] [Indexed: 11/17/2022] Open
Abstract
In recent years sensorineural hearing loss was found to affect not exclusively, nor at first, the sensory cells of the inner ear. The sensory cells' synapses and subsequent neurites are initially damaged. Auditory synaptopathies also play an important role in cochlear implant (CI) care, as they can lead to a loss of physiological hearing in patients with residual hearing. These auditory synaptopathies and in general the cascades of hearing pathologies have been in the focus of research in recent years with the aim to develop more targeted and individually tailored therapeutics. In the current study, a method to examine implanted inner ears of guinea pigs was developed to examine the synapse level. For this purpose, the cochlea is made transparent and scanned with the implant in situ using confocal laser scanning microscopy. Three different preparation methods were compared to enable both an overview image of the cochlea for assessing the CI position and images of the synapses on the same specimen. The best results were achieved by dissection of the bony capsule of the cochlea.
Collapse
|
29
|
Fernandez KA, Watabe T, Tong M, Meng X, Tani K, Kujawa SG, Edge AS. Trk agonist drugs rescue noise-induced hidden hearing loss. JCI Insight 2021; 6:142572. [PMID: 33373328 PMCID: PMC7934864 DOI: 10.1172/jci.insight.142572] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 12/16/2020] [Indexed: 12/21/2022] Open
Abstract
TrkB agonist drugs are shown here to have a significant effect on the regeneration of afferent cochlear synapses after noise-induced synaptopathy. The effects were consistent with regeneration of cochlear synapses that we observed in vitro after synaptic loss due to kainic acid-induced glutamate toxicity and were elicited by administration of TrkB agonists, amitriptyline, and 7,8-dihydroxyflavone, directly into the cochlea via the posterior semicircular canal 48 hours after exposure to noise. Synaptic counts at the inner hair cell and wave 1 amplitudes in the auditory brainstem response (ABR) were partially restored 2 weeks after drug treatment. Effects of amitriptyline on wave 1 amplitude and afferent auditory synapse numbers in noise-exposed ears after systemic (as opposed to local) delivery were profound and long-lasting; synapses in the treated animals remained intact 1 year after the treatment. However, the effect of systemically delivered amitriptyline on synaptic rescue was dependent on dose and the time window of administration: it was only effective when given before noise exposure at the highest injected dose. The long-lasting effect and the efficacy of postexposure treatment indicate a potential broad application for the treatment of synaptopathy, which often goes undetected until well after the original damaging exposures.
Collapse
Affiliation(s)
- Katharine A Fernandez
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA.,Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, Massachusetts, USA
| | - Takahisa Watabe
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA.,Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, Massachusetts, USA
| | - Mingjie Tong
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA.,Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, Massachusetts, USA
| | - Xiankai Meng
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA.,Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, Massachusetts, USA
| | - Kohsuke Tani
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA.,Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, Massachusetts, USA
| | - Sharon G Kujawa
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA.,Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, Massachusetts, USA
| | - Albert Sb Edge
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA.,Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
30
|
Nevoux J, Alexandru M, Bellocq T, Tanaka L, Hayashi Y, Watabe T, Lahlou H, Tani K, Edge ASB. An antibody to RGMa promotes regeneration of cochlear synapses after noise exposure. Sci Rep 2021; 11:2937. [PMID: 33536466 PMCID: PMC7859405 DOI: 10.1038/s41598-021-81294-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 01/05/2021] [Indexed: 11/09/2022] Open
Abstract
Auditory neuropathy is caused by the loss of afferent input to the brainstem via the components of the neural pathway comprising inner hair cells and the first order neurons of the spiral ganglion. Recent work has identified the synapse between cochlear primary afferent neurons and sensory hair cells as a particularly vulnerable component of this pathway. Loss of these synapses due to noise exposure or aging results in the pathology identified as hidden hearing loss, an initial stage of cochlear dysfunction that goes undetected in standard hearing tests. We show here that repulsive axonal guidance molecule a (RGMa) acts to prevent regrowth and synaptogenesis of peripheral auditory nerve fibers with inner hair cells. Treatment of noise-exposed animals with an anti-RGMa blocking antibody regenerated inner hair cell synapses and resulted in recovery of wave-I amplitude of the auditory brainstem response, indicating effective reversal of synaptopathy.
Collapse
Affiliation(s)
- Jerome Nevoux
- Department of Otololaryngology, Harvard Medical School, Boston, MA, 02115, USA.,Eaton-Peabody Laboratories, Massachusetts Eye and Ear, 243 Charles St., Boston, MA, 02114, USA
| | - Mihaela Alexandru
- Department of Otololaryngology, Harvard Medical School, Boston, MA, 02115, USA.,Eaton-Peabody Laboratories, Massachusetts Eye and Ear, 243 Charles St., Boston, MA, 02114, USA
| | - Thomas Bellocq
- Department of Otololaryngology, Harvard Medical School, Boston, MA, 02115, USA.,Eaton-Peabody Laboratories, Massachusetts Eye and Ear, 243 Charles St., Boston, MA, 02114, USA
| | - Lei Tanaka
- Department of Otololaryngology, Harvard Medical School, Boston, MA, 02115, USA.,Eaton-Peabody Laboratories, Massachusetts Eye and Ear, 243 Charles St., Boston, MA, 02114, USA
| | - Yushi Hayashi
- Department of Otololaryngology, Harvard Medical School, Boston, MA, 02115, USA.,Eaton-Peabody Laboratories, Massachusetts Eye and Ear, 243 Charles St., Boston, MA, 02114, USA
| | - Takahisa Watabe
- Department of Otololaryngology, Harvard Medical School, Boston, MA, 02115, USA.,Eaton-Peabody Laboratories, Massachusetts Eye and Ear, 243 Charles St., Boston, MA, 02114, USA
| | - Hanae Lahlou
- Department of Otololaryngology, Harvard Medical School, Boston, MA, 02115, USA.,Eaton-Peabody Laboratories, Massachusetts Eye and Ear, 243 Charles St., Boston, MA, 02114, USA
| | - Kohsuke Tani
- Department of Otololaryngology, Harvard Medical School, Boston, MA, 02115, USA.,Eaton-Peabody Laboratories, Massachusetts Eye and Ear, 243 Charles St., Boston, MA, 02114, USA
| | - Albert S B Edge
- Department of Otololaryngology, Harvard Medical School, Boston, MA, 02115, USA. .,Eaton-Peabody Laboratories, Massachusetts Eye and Ear, 243 Charles St., Boston, MA, 02114, USA. .,Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, MA, 02115, USA. .,Harvard Stem Cell Institute, Cambridge, MA, 02138, USA.
| |
Collapse
|
31
|
Koizumi Y, Ito T, Mizutari K, Kakehata S. Regenerative Effect of a ROCK Inhibitor, Y-27632, on Excitotoxic Trauma in an Organotypic Culture of the Cochlea. Front Cell Neurosci 2020; 14:572434. [PMID: 33328888 PMCID: PMC7717995 DOI: 10.3389/fncel.2020.572434] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 10/26/2020] [Indexed: 11/28/2022] Open
Abstract
In the past, most inner ear diseases were thought to start with the impairment of the sensory epithelium of the cochlea before subsequently progressing to secondary neural degeneration. However, recent studies show that loss of primary synapses accompanied by excitotoxic degeneration of peripheral axons is likely to be the underlying pathology in sensorineural hearing loss. Rho-associated coiled-coil containing protein kinase (ROCK) inhibition has been reported to have neuroprotective and regenerative effects on synaptic pathways. Therefore, we analyzed the effect of ROCK inhibition using Y-27632 in a model of peripheral axonal damage in the spiral ganglion neurons created using the glutamate agonists, N-methyl-D-aspartate (NMDA) and kainic acid, to induce excitotoxic trauma in the explanted cochlea. The number of axons projecting to hair cells in the cochlea treated with Y-27632 was significantly greater than those in the cochlea treated only with NMDA + kainic acid. Furthermore, there was a significant increase in synapses between the spiral ganglion and the inner hair cells in the cochlea treated with Y-27632. The findings of this study suggest that ROCK inhibition could be a potential strategy for the regeneration of peripheral axons in the spiral ganglion and synapse formation in the inner hair cells of a cochlea that has sustained excitotoxic injury, which is one of the primary etiologies of inner ear disease.
Collapse
Affiliation(s)
- Yutaka Koizumi
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Tsukasa Ito
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Kunio Mizutari
- Department of Otolaryngology-Head and Neck Surgery, National Defense Medical College, Saitama, Japan
| | - Seiji Kakehata
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine, Yamagata University, Yamagata, Japan
| |
Collapse
|
32
|
Gao L, Kita T, Katsuno T, Yamamoto N, Omori K, Nakagawa T. Insulin-Like Growth Factor 1 on the Maintenance of Ribbon Synapses in Mouse Cochlear Explant Cultures. Front Cell Neurosci 2020; 14:571155. [PMID: 33132846 PMCID: PMC7579230 DOI: 10.3389/fncel.2020.571155] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/27/2020] [Indexed: 01/31/2023] Open
Abstract
Hearing loss has become one of the most common disabilities worldwide. The synaptic connections between inner hair cells (IHCs) and spiral ganglion neurons have specialized synaptic constructions, termed ribbon synapses, which are important for auditory function. The ribbon synapses in the cochlea are quite vulnerable to various insults. As such, the maintenance of ribbon synapses is important for ensuring hearing function. Insulin-like growth factor 1 (IGF1) plays a critical role in the development and maintenance of the cochlea and has the potential to protect cochlear hair cells from various insults. In this study, we examined the role of IGF1 in the maintenance of ribbon synapses in cochlear explants of postnatal day four mice. We cultured cochlear explants with an IGF1 receptor antagonist, JB1, which is an IGF1 peptide analog. Results showed that exposure to JB1 for 24 h resulted in the loss of ribbon synapses. After an additional 24-h culture without JB1, the number of ribbon synapses spontaneously recovered. The application of exogenous IGF1 showed two different aspects of ribbon synapses. Low doses of exogenous IGF1 promoted the recovery of ribbon synapses, while it compromised the spontaneous recovery of ribbon synapses at high doses. Altogether, these results indicate that the paracrine or autocrine release of IGF1 in the cochlea plays a crucial role in the maintenance of cochlear ribbon synapses.
Collapse
Affiliation(s)
- Li Gao
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoko Kita
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tatsuya Katsuno
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Norio Yamamoto
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koichi Omori
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takayuki Nakagawa
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
33
|
Wu CC, Brugeaud A, Seist R, Lin HC, Yeh WH, Petrillo M, Coppola G, Edge ASB, Stankovic KM. Altered expression of genes regulating inflammation and synaptogenesis during regrowth of afferent neurons to cochlear hair cells. PLoS One 2020; 15:e0238578. [PMID: 33001981 PMCID: PMC7529247 DOI: 10.1371/journal.pone.0238578] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 08/19/2020] [Indexed: 12/20/2022] Open
Abstract
The spiral ganglion neurons constitute the primary connection between auditory hair cells and the brain. The spiral ganglion afferent fibers and their synapse with hair cells do not regenerate to any significant degree in adult mammalian ears after damage. We have investigated gene expression changes after kainate-induced disruption of the synapses in a neonatal cochlear explant model in which peripheral fibers and the afferent synapse do regenerate. We compared gene expression early after damage, during regeneration of the fibers and synapses, and after completion of in vitro regeneration. These analyses revealed a total of 2.5% differentially regulated transcripts (588 out of 24,000) based on a threshold of p<0.005. Inflammatory response genes as well as genes involved in regeneration of neural circuits were upregulated in the spiral ganglion neurons and organ of Corti, where the hair cells reside. Prominent genes upregulated at several time points included genes with roles in neurogenesis (Elavl4 and Sox21), neural outgrowth (Ntrk3 and Ppp1r1c), axonal guidance (Rgmb and Sema7a), synaptogenesis (Nlgn2 and Psd2), and synaptic vesicular function (Syt8 and Syn1). Immunohistochemical and in situ hybridization analysis of genes that had not previously been described in the cochlea confirmed their cochlear expression. The time course of expression of these genes suggests that kainate treatment resulted in a two-phase response in spiral ganglion neurons: an acute response consistent with inflammation, followed by an upregulation of neural regeneration genes. Identification of the genes activated during regeneration of these fibers suggests candidates that could be targeted to enhance regeneration in adult ears.
Collapse
Affiliation(s)
- Chen-Chi Wu
- Eaton Peabody Laboratories and Department of Otolaryngology—Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
- Department of Otolaryngology—Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Aurore Brugeaud
- Eaton Peabody Laboratories and Department of Otolaryngology—Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
- Department of Otolaryngology—Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Richard Seist
- Eaton Peabody Laboratories and Department of Otolaryngology—Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
- Department of Otolaryngology—Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Otorhinolaryngology-Head and Neck Surgery, Paracelsus Medical University, Salzburg, Austria
| | - Hsiao-Chun Lin
- Eaton Peabody Laboratories and Department of Otolaryngology—Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
- Department of Otolaryngology—Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Wei-Hsi Yeh
- Eaton Peabody Laboratories and Department of Otolaryngology—Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
- Department of Otolaryngology—Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
- Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Marco Petrillo
- Eaton Peabody Laboratories and Department of Otolaryngology—Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
- Department of Otolaryngology—Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Giovanni Coppola
- Program in Neurogenetics, Department of Neurology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Albert S. B. Edge
- Eaton Peabody Laboratories and Department of Otolaryngology—Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
- Department of Otolaryngology—Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
- Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Konstantina M. Stankovic
- Eaton Peabody Laboratories and Department of Otolaryngology—Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
- Department of Otolaryngology—Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
- Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, Massachusetts, United States of America
- Harvard Program in Therapeutic Science, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
34
|
Shen N, Zhou L, Lai B, Li S. The Influence of Cochlear Implant-Based Electric Stimulation on the Electrophysiological Characteristics of Cultured Spiral Ganglion Neurons. Neural Plast 2020; 2020:3108490. [PMID: 32963515 PMCID: PMC7490630 DOI: 10.1155/2020/3108490] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/22/2020] [Accepted: 08/17/2020] [Indexed: 11/17/2022] Open
Abstract
Background Cochlear implant-based electrical stimulation may be an important reason to induce the residual hearing loss after cochlear implantation. In our previous study, we found that charge-balanced biphasic electrical stimulation inhibited the neurite growth of spiral ganglion neurons (SGNs) and decreased Schwann cell density in vitro. In this study, we want to know whether cochlear implant-based electrical stimulation can induce the change of electrical activity in cultured SGNs. Methods Spiral ganglion neuron electrical stimulation in vitro model is established using the devices delivering cochlear implant-based electrical stimulation. After 48 h treatment by 50 μA or 100 μA electrical stimulation, the action potential (AP) and voltage depended calcium current (I Ca) of SGNs are recorded using whole-cell electrophysiological method. Results The results show that the I Ca of SGNs is decreased significantly in 50 μA and 100 μA electrical stimulation groups. The reversal potential of I Ca is nearly +80 mV in control SGN, but the reversal potential decreases to +50 mV in 50 μA and 100 μA electrical stimulation groups. Interestingly, the AP amplitude, the AP latency, and the AP duration of SGNs have no statistically significant differences in all three groups. Conclusion Our study suggests cochlear implant-based electrical stimulation only significantly inhibit the I Ca of cultured SGNs but has no effect on the firing of AP, and the relation of I Ca inhibition and SGN damage induced by electrical stimulation and its mechanism needs to be further studied.
Collapse
Affiliation(s)
- Na Shen
- Department of Otolaryngology, Zhongshan Hospital, Fudan University, Shanghai, China
- ENT Institute and Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Lei Zhou
- Department of Otolaryngology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bin Lai
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Shufeng Li
- ENT Institute and Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, China
| |
Collapse
|
35
|
Li Q, Chen M, Zhang C, Lu T, Min S, Li S. Opposite Roles of NT-3 and BDNF in Synaptic Remodeling of the Inner Ear Induced by Electrical Stimulation. Cell Mol Neurobiol 2020; 41:1665-1682. [PMID: 32770528 DOI: 10.1007/s10571-020-00935-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 08/01/2020] [Indexed: 12/11/2022]
Abstract
With the development of neural prostheses, neural plasticity including synaptic remodeling under electrical stimulation is drawing more and more attention. Indeed, intracochlear electrical stimulation used to restore hearing in deaf can induce the loss of residual hearing and synapses of the inner hair cells (IHCs). However, the mechanism under this process is largely unknown. Considering that the guinea pig is always a suitable and convenient choice for the animal model of cochlea implant (CI), in the present study, normal-hearing guinea pigs were implanted with CIs. Four-hour electrical stimulation with the intensity of 6 dB above electrically evoked compound action potential (ECAP) threshold (which can decrease the quantity of IHC synapses and the excitability of the auditory nerve) resulted in the upregulation of Bdnf (p < 0.0001) and downregulation of Nt-3 (p < 0.05). Intracochlear perfusion of exogenous NT-3 or TrkC/Fc (which blocks NT-3) can, respectively, resist or aggravate the synaptic loss induced by electrical stimulation. In contrast, local delivery of exogenous BDNF or TrkB/Fc (which blocks BDNF) to the cochlea, respectively, exacerbated or protected against the synaptic loss caused by electrical stimulation. Notably, the synaptic changes were only observed in the basal and middle halves of the cochlea. All the findings above suggested that NT-3 and BDNF may play opposite roles in the remodeling of IHC synapses induced by intracochlear electrical stimulation, i.e. NT-3 and BDNF promoted the regeneration and degeneration of IHC synapses, respectively.
Collapse
Affiliation(s)
- Qiang Li
- ENT Institute and Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, China
| | - Min Chen
- ENT Institute and Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, China
| | - Chen Zhang
- ENT Institute and Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, China
| | - Tianhao Lu
- ENT Institute and Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, China
| | - Shiyao Min
- ENT Institute and Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, China
| | - Shufeng Li
- ENT Institute and Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China. .,NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, China.
| |
Collapse
|
36
|
Swiderski DL, Colesa DJ, Hughes AP, Raphael Y, Pfingst BE. Relationships between Intrascalar Tissue, Neuron Survival, and Cochlear Implant Function. J Assoc Res Otolaryngol 2020; 21:337-352. [PMID: 32691251 PMCID: PMC7445211 DOI: 10.1007/s10162-020-00761-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 07/12/2020] [Indexed: 12/15/2022] Open
Abstract
Fibrous tissue and/or new bone are often found surrounding a cochlear implant in the cochlear scalae. This new intrascalar tissue could potentially limit cochlear implant function by increasing impedance and altering signaling pathways between the implant and the auditory nerve. In this study, we investigated the relationship between intrascalar tissue and 5 measures of implant function in guinea pigs. Variation in both spiral ganglion neuron (SGN) survival and intrascalar tissue was produced by implanting hearing ears, ears deafened with neomycin, and neomycin-deafened ears treated with a neurotrophin. We found significant effects of SGN density on 4 functional measures but adding intrascalar tissue level to the analysis did not explain more variation in any measure than was explained by SGN density alone. These results suggest that effects of intrascalar tissue on electrical hearing are relatively unimportant in comparison to degeneration of the auditory nerve, although additional studies in human implant recipients are still needed to assess the effects of this tissue on complex hearing tasks like speech perception. The results also suggest that efforts to minimize the trauma that aggravates both tissue development and SGN loss could be beneficial.
Collapse
Affiliation(s)
- Donald L Swiderski
- Kresge Hearing Research Institute, Department of Otolaryngology-Head and Neck Surgery, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Deborah J Colesa
- Kresge Hearing Research Institute, Department of Otolaryngology-Head and Neck Surgery, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Aaron P Hughes
- Kresge Hearing Research Institute, Department of Otolaryngology-Head and Neck Surgery, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Yehoash Raphael
- Kresge Hearing Research Institute, Department of Otolaryngology-Head and Neck Surgery, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Bryan E Pfingst
- Kresge Hearing Research Institute, Department of Otolaryngology-Head and Neck Surgery, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
37
|
Feng S, Yang L, Hui L, Luo Y, Du Z, Xiong W, Liu K, Jiang X. Long-term exposure to low-intensity environmental noise aggravates age-related hearing loss via disruption of cochlear ribbon synapses. Am J Transl Res 2020; 12:3674-3687. [PMID: 32774726 PMCID: PMC7407738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 06/27/2020] [Indexed: 06/11/2023]
Abstract
Noise pollution is a major public hazard. Previous studies have shown that environmental noise affects the reorganization of the auditory cortex and leads to behavioral abnormality; however, the effects of long-term environmental noise exposure on the inner ear and hearing remain to be elucidated. In this study, we simulated environmental noise with a long-term 70 dB sound pressure level "white" noise, observed its effect on the inner ears of C57BL/6J mice, and developed an in vitro model for mechanistic studies. We found that environmental noise increased the hearing threshold, decreased the auditory response amplitude, and aggravated the range and extent of age-related hearing loss (ARHL), especially in the intermediate frequency band in mice. Cochlear ribbon synapse is the primary site of inner ear injury caused by environmental noise. We also verified, through an in vitro simulation of the excitatory toxicity of glutamate and aging effects, that the activation of NLRP3 inflammasome plays a vital role in the cochlear ribbon synaptic damage. Our results show that long-term exposure to low-intensity environmental noise can lead to hearing loss via the disruption of ribbon synapses, which is caused by an inflammatory reaction. Additionally, environmental noise can further aggravate the progression of ARHL. This study expounded the pathogenesis of the inner ear damage caused by environmental noise exposure and provides a new direction for the prevention and treatment of hearing loss.
Collapse
Affiliation(s)
- Shuai Feng
- Department of Otolaryngology, The First Affiliated Hospital of China Medical UniversityShenyang 110001, China
| | - Le Yang
- Department of Otolaryngology-Head and Neck Surgery, Guangdong No. 2 Provincial People’s Hospital, Southern Medical UniversityGuangzhou 510000, China
| | - Lian Hui
- Department of Otolaryngology, The First Affiliated Hospital of China Medical UniversityShenyang 110001, China
| | - Yangtuo Luo
- Department of Otolaryngology, The First Affiliated Hospital of China Medical UniversityShenyang 110001, China
| | - Zhengde Du
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical UniversityBeijing 100050, China
| | - Wei Xiong
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical UniversityBeijing 100050, China
| | - Ke Liu
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical UniversityBeijing 100050, China
| | - Xuejun Jiang
- Department of Otolaryngology, The First Affiliated Hospital of China Medical UniversityShenyang 110001, China
| |
Collapse
|
38
|
Seist R, Tong M, Landegger LD, Vasilijic S, Hyakusoku H, Katsumi S, McKenna CE, Edge ASB, Stankovic KM. Regeneration of Cochlear Synapses by Systemic Administration of a Bisphosphonate. Front Mol Neurosci 2020; 13:87. [PMID: 32765216 PMCID: PMC7381223 DOI: 10.3389/fnmol.2020.00087] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 04/28/2020] [Indexed: 12/12/2022] Open
Abstract
Sensorineural hearing loss (SNHL) caused by noise exposure and attendant loss of glutamatergic synapses between cochlear spiral ganglion neurons (SGNs) and hair cells is the most common sensory deficit worldwide. We show here that systemic administration of a bisphosphonate to mice 24 h after synaptopathic noise exposure regenerated synapses between inner hair cells and SGNs and restored cochlear function. We further demonstrate that this effect is mediated by inhibition of the mevalonate pathway. These results are highly significant because they suggest that bisphosphonates could reverse cochlear synaptopathy for the treatment of SNHL.
Collapse
Affiliation(s)
- Richard Seist
- Eaton-Peabody Laboratories, Department of Otolaryngology – Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, United States
- Department of Otolaryngology – Head and Neck Surgery, Harvard Medical School, Boston, MA, United States
- Department of Otorhinolaryngology-Head and Neck Surgery, Paracelsus Medical University, Salzburg, Austria
| | - Mingjie Tong
- Eaton-Peabody Laboratories, Department of Otolaryngology – Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, United States
- Department of Otolaryngology – Head and Neck Surgery, Harvard Medical School, Boston, MA, United States
| | - Lukas D. Landegger
- Eaton-Peabody Laboratories, Department of Otolaryngology – Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, United States
- Department of Otolaryngology – Head and Neck Surgery, Harvard Medical School, Boston, MA, United States
- Department of Otorhinolaryngology-Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| | - Sasa Vasilijic
- Eaton-Peabody Laboratories, Department of Otolaryngology – Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, United States
- Department of Otolaryngology – Head and Neck Surgery, Harvard Medical School, Boston, MA, United States
| | - Hiroshi Hyakusoku
- Eaton-Peabody Laboratories, Department of Otolaryngology – Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, United States
- Department of Otolaryngology – Head and Neck Surgery, Harvard Medical School, Boston, MA, United States
- Department of Otorhinolaryngology, Yokosuka Kyosai Hospital, Kanagawa, Japan
| | - Sachiyo Katsumi
- Eaton-Peabody Laboratories, Department of Otolaryngology – Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, United States
- Department of Otolaryngology – Head and Neck Surgery, Harvard Medical School, Boston, MA, United States
| | - Charles E. McKenna
- Department of Chemistry, University of Southern California, Los Angeles, CA, United States
| | - Albert S. B. Edge
- Eaton-Peabody Laboratories, Department of Otolaryngology – Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, United States
- Department of Otolaryngology – Head and Neck Surgery, Harvard Medical School, Boston, MA, United States
- Speech and Hearing Bioscience and Technology Program, Harvard Medical School, Boston, MA, United States
- Harvard Stem Cell Institute, Cambridge, MA, United States
| | - Konstantina M. Stankovic
- Eaton-Peabody Laboratories, Department of Otolaryngology – Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, United States
- Department of Otolaryngology – Head and Neck Surgery, Harvard Medical School, Boston, MA, United States
- Speech and Hearing Bioscience and Technology Program, Harvard Medical School, Boston, MA, United States
- Harvard Stem Cell Institute, Cambridge, MA, United States
- Program in Therapeutic Science, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
39
|
Peter MN, Paasche G, Reich U, Lenarz T, Warnecke A. Differential Effects of Low- and High-Dose Dexamethasone on Electrically Induced Damage of the Cultured Organ of Corti. Neurotox Res 2020; 38:487-497. [PMID: 32495312 PMCID: PMC7334252 DOI: 10.1007/s12640-020-00228-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 05/02/2020] [Accepted: 05/19/2020] [Indexed: 12/29/2022]
Abstract
An increased number of patients with residual hearing are undergoing cochlear implantation. A subset of these experience delayed hearing loss post-implantation, and the aetiology of this loss is not well understood. Our previous studies suggest that electrical stimulation can induce damage to hair cells in organ of Corti (OC) organotypic cultures. Dexamethasone has the potential to protect residual hearing due to its multiple effects on cells and tissue (e.g., anti-inflammatory, free radical scavenger). We therefore hypothesized that dexamethasone treatment could prevent electrical stimulation induced changes in the OC. Organ of Corti explants from neonatal rats (P2–4) were cultured for 24 h with two different concentrations of dexamethasone. Thereafter, OC were subjected to a charge-balanced biphasic pulsed electrical stimulation (0.44–2 mA) for a further 24 h. Unstimulated dexamethasone-treated OC served as controls. Outcome analysis included immunohistochemical labelling of ribbon synapses, histochemical analysis of free reactive oxygen species and morphological analysis of stereocilia bundles. Overall, the protective effects of dexamethasone on electrically induced damage in cochlear explants were moderate. High-dose dexamethasone protected bundle integrity at higher current levels. Low-dose dexamethasone tended to increase ribbon density in the apical region.
Collapse
Affiliation(s)
- Marvin N Peter
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Gerrit Paasche
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.,Cluster of Excellence "Hearing4all" of the German Research Foundation, Hannover, Germany
| | - Uta Reich
- Department of Otorhinolaryngology, Head and Neck Surgery, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Thomas Lenarz
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.,Cluster of Excellence "Hearing4all" of the German Research Foundation, Hannover, Germany
| | - Athanasia Warnecke
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany. .,Cluster of Excellence "Hearing4all" of the German Research Foundation, Hannover, Germany.
| |
Collapse
|
40
|
Tserga E, Damberg P, Canlon B, Cederroth CR. Auditory synaptopathy in mice lacking the glutamate transporter GLAST and its impact on brain activity. PROGRESS IN BRAIN RESEARCH 2020; 262:245-261. [PMID: 33931183 DOI: 10.1016/bs.pbr.2020.04.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Neurotransmission of acoustic signals from the hair cells to the auditory nerve relies on a tightly controlled communication between pre-synaptic ribbons and post-synaptic glutamatergic terminals. After noise overexposure, de-afferentation occurs as a consequence of excessive glutamate release. What maintains synaptic integrity in the cochlea is poorly understood. The objective of this study is to evaluate the role of GLAST in maintaining synaptic integrity in the cochlea in absence or presence of noise, and its impact on sound-evoked brain activity using manganese-enhanced MRI (MeMRI). The glutamate aspartate transporter GLAST is present in supporting cells near the afferent synapse and its genetic deletion leads to greater synaptic swelling after noise overexposure. At baseline, GLAST knockout (GLAST KO) mice displayed two-fold lower wave 1 amplitude of the auditory brainstem response (ABR) when compared to their wild-type littermates in spite of similar ABR and distortion product otoacoustic emissions (DPOAE) thresholds. While the abundance of ribbons was not affected by the loss of GLAST function, the number of paired synapses was halved in GLAST KO mice, suggestive of a pre-existing auditory synaptopathy. Immediately after the noise exposure ABR thresholds rose by 41-62dB to a similar degree in GLAST WT and KO mice and DPOAE remained unaffected. In the acute phase following noise exposure, GLAST KO mice showed near complete de-afferentation unlike WT mice which maintained four to seven paired synapses per IHC. Brain activity using MeMRI found noise exposure to cause greater activity in the inferior colliculus in GLAST KO but not in WT mice. No changes in brain activity was found in GLAST KO mice at baseline in spite of affected afferent synapses, suggesting that auditory synaptopathy may not be sufficient to alter brain activity in the absence of noise exposure.
Collapse
Affiliation(s)
- Evangelia Tserga
- Laboratory of Experimental Audiology, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Peter Damberg
- Karolinska Experimental Research and Imaging Center, Karolinska University Hospital, Stockholm, Sweden
| | - Barbara Canlon
- Laboratory of Experimental Audiology, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Christopher R Cederroth
- Laboratory of Experimental Audiology, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
41
|
Leake PA, Akil O, Lang H. Neurotrophin gene therapy to promote survival of spiral ganglion neurons after deafness. Hear Res 2020; 394:107955. [PMID: 32331858 DOI: 10.1016/j.heares.2020.107955] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 03/16/2020] [Accepted: 03/26/2020] [Indexed: 12/13/2022]
Abstract
Hearing impairment is a major health and economic concern worldwide. Currently, the cochlear implant (CI) is the standard of care for remediation of severe to profound hearing loss, and in general, contemporary CIs are highly successful. But there is great variability in outcomes among individuals, especially in children, with many CI users deriving much less or even marginal benefit. Much of this variability is related to differences in auditory nerve survival, and there has been substantial interest in recent years in exploring potential therapies to improve survival of the cochlear spiral ganglion neurons (SGN) after deafness. Preclinical studies using osmotic pumps and other approaches in deafened animal models to deliver neurotrophic factors (NTs) directly to the cochlea have shown promising results, especially with Brain-Derived Neurotrophic Factor (BDNF). More recent studies have focused on the use of NT gene therapy to force expression of NTs by target cells within the cochlea. This could provide the means for a one-time treatment to promote long-term NT expression and improve neural survival after deafness. This review summarizes the evidence for the efficacy of exogenous NTs in preventing SGN degeneration after hearing loss and reviews the animal research to date suggesting that NT gene therapy can elicit long-term NT expression in the cochlea, resulting in significantly improved SGN and radial nerve fiber survival after deafness. In addition, we discuss NT gene therapy in other non-auditory applications and consider some of the remaining issues with regard to selecting optimal vectors, timing of treatment, and place/method of delivery, etc. that must be resolved prior to considering clinical application.
Collapse
Affiliation(s)
- Patricia A Leake
- S & I Epstein Laboratory, Dept. of Otolaryngology Head and Neck Surgery, University of California San Francisco, 2340 Sutter Street, Room N331, San Francisco, CA, 94115-1330, USA.
| | - Omar Akil
- S & I Epstein Laboratory, Dept. of Otolaryngology Head and Neck Surgery, University of California San Francisco, 2340 Sutter Street, Room N331, San Francisco, CA, 94115-1330, USA
| | - Hainan Lang
- Dept. of Pathology and Laboratory Medicine, Medical University of South Carolina, 165 Ashley Avenue, Room RS613, Charleston, SC, 29414, USA
| |
Collapse
|
42
|
Smith DR, Dumont CM, Park J, Ciciriello AJ, Guo A, Tatineni R, Cummings BJ, Anderson AJ, Shea LD. Polycistronic Delivery of IL-10 and NT-3 Promotes Oligodendrocyte Myelination and Functional Recovery in a Mouse Spinal Cord Injury Model. Tissue Eng Part A 2020; 26:672-682. [PMID: 32000627 DOI: 10.1089/ten.tea.2019.0321] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
One million estimated cases of spinal cord injury (SCI) have been reported in the United States and repairing an injury has constituted a difficult clinical challenge. The complex, dynamic, inhibitory microenvironment postinjury, which is characterized by proinflammatory signaling from invading leukocytes and lack of sufficient factors that promote axonal survival and elongation, limits regeneration. Herein, we investigated the delivery of polycistronic vectors, which have the potential to coexpress factors that target distinct barriers to regeneration, from a multiple channel poly(lactide-co-glycolide) (PLG) bridge to enhance spinal cord regeneration. In this study, we investigated polycistronic delivery of IL-10 that targets proinflammatory signaling, and NT-3 that targets axonal survival and elongation. A significant increase was observed in the density of regenerative macrophages for IL-10+NT-3 condition relative to conditions without IL-10. Furthermore, combined delivery of IL-10+NT-3 produced a significant increase of axonal density and notably myelinated axons compared with all other conditions. A significant increase in functional recovery was observed for IL-10+NT-3 delivery at 12 weeks postinjury that was positively correlated to oligodendrocyte myelinated axon density, suggesting oligodendrocyte-mediated myelination as an important target to improve functional recovery. These results further support the use of multiple channel PLG bridges as a growth supportive substrate and platform to deliver bioactive agents to modulate the SCI microenvironment and promote regeneration and functional recovery. Impact statement Spinal cord injury (SCI) results in a complex microenvironment that contains multiple barriers to regeneration and functional recovery. Multiple factors are necessary to address these barriers to regeneration, and polycistronic lentiviral gene therapy represents a strategy to locally express multiple factors simultaneously. A bicistronic vector encoding IL-10 and NT-3 was delivered from a poly(lactide-co-glycolide) bridge, which provides structural support that guides regeneration, resulting in increased axonal growth, myelination, and subsequent functional recovery. These results demonstrate the opportunity of targeting multiple barriers to SCI regeneration for additive effects.
Collapse
Affiliation(s)
- Dominique R Smith
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Courtney M Dumont
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan.,Department of Biomedical Engineering, University of Miami, Coral Gables, Florida.,Biomedical Nanotechnology Institute at University of Miami (BioNIUM), University of Miami, Miami, Florida
| | - Jonghyuck Park
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky.,Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Andrew J Ciciriello
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan.,Department of Biomedical Engineering, University of Miami, Coral Gables, Florida
| | - Amina Guo
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Ravindra Tatineni
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Brian J Cummings
- Institute for Memory Impairments and Neurological Disorders (iMIND), University of California, Irvine, California.,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California.,Department of Anatomy and Neurobiology, University of California, Irvine, California.,Department of Physical Medicine and Rehabilitation, University of California, Irvine, California
| | - Aileen J Anderson
- Institute for Memory Impairments and Neurological Disorders (iMIND), University of California, Irvine, California.,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California.,Department of Anatomy and Neurobiology, University of California, Irvine, California.,Department of Physical Medicine and Rehabilitation, University of California, Irvine, California
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan.,Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
43
|
Protection of cochlear synapses from noise-induced excitotoxic trauma by blockade of Ca 2+-permeable AMPA receptors. Proc Natl Acad Sci U S A 2020; 117:3828-3838. [PMID: 32015128 DOI: 10.1073/pnas.1914247117] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Exposure to loud sound damages the postsynaptic terminals of spiral ganglion neurons (SGNs) on cochlear inner hair cells (IHCs), resulting in loss of synapses, a process termed synaptopathy. Glutamatergic neurotransmission via α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA)-type receptors is required for synaptopathy, and here we identify a possible involvement of GluA2-lacking Ca2+-permeable AMPA receptors (CP-AMPARs) using IEM-1460, which has been shown to block GluA2-lacking AMPARs. In CBA/CaJ mice, a 2-h exposure to 100-dB sound pressure level octave band (8 to 16 kHz) noise results in no permanent threshold shift but does cause significant synaptopathy and a reduction in auditory brainstem response (ABR) wave-I amplitude. Chronic intracochlear perfusion of IEM-1460 in artificial perilymph (AP) into adult CBA/CaJ mice prevented the decrease in ABR wave-I amplitude and the synaptopathy relative to intracochlear perfusion of AP alone. Interestingly, IEM-1460 itself did not affect the ABR threshold, presumably because GluA2-containing AMPARs can sustain sufficient synaptic transmission to evoke low-threshold responses during blockade of GluA2-lacking AMPARs. On individual postsynaptic densities, we observed GluA2-lacking nanodomains alongside regions with robust GluA2 expression, consistent with the idea that individual synapses have both CP-AMPARs and Ca2+-impermeable AMPARs. SGNs innervating the same IHC differ in their relative vulnerability to noise. We found local heterogeneity among synapses in the relative abundance of GluA2 subunits that may underlie such differences in vulnerability. We propose a role for GluA2-lacking CP-AMPARs in noise-induced cochlear synaptopathy whereby differences among synapses account for differences in excitotoxic susceptibility. These data suggest a means of maintaining normal hearing thresholds while protecting against noise-induced synaptopathy, via selective blockade of CP-AMPARs.
Collapse
|
44
|
Holt AG, Kühl A, Braun RD, Altschuler R. The rat as a model for studying noise injury and otoprotection. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2019; 146:3681. [PMID: 31795688 DOI: 10.1121/1.5131344] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
A major challenge for those studying noise-induced injury pre-clinically is the selection of an animal model. Noise injury models are particularly relevant in an age when people are constantly bombarded by loud noise due to occupation and/or recreation. The rat has been widely used for noise-related morphological, physiological, biochemical, and molecular assessment. Noise exposure resulting in a temporary (TTS) or permanent threshold shift (PTS) yields trauma in peripheral and central auditory related pathways. While the precise nature of noise-related injuries continues to be delineated, both PTS and TTS (with or without hidden hearing loss) result in homeostatic changes implicated in conditions such as tinnitus and hyperacusis. Compared to mice, rats generally tolerate exposure to loud sounds reasonably well, often without exhibiting other physical non-inner ear related symptoms such as death, loss of consciousness, or seizures [Skradski, Clark, Jiang, White, Fu, and Ptacek (2001). Neuron 31, 537-544; Faingold (2002). Hear. Res. 168, 223-237; Firstova, Abaimov, Surina, Poletaeva, Fedotova, and Kovalev (2012). Bull Exp. Biol. Med. 154, 196-198; De Sarro, Russo, Citraro, and Meldrum (2017). Epilepsy Behav. 71, 165-173]. This ability of the rat to thrive following noise exposure permits study of long-term effects. Like the mouse, the rat also offers a well-characterized genome allowing genetic manipulations (i.e., knock-out, viral-based gene expression modulation, and optogenetics). Rat models of noise-related injury also provide valuable information for understanding mechanistic changes to identify therapeutic targets for treatment. This article provides a framework for selection of the rat as a model for noise injury studies.
Collapse
Affiliation(s)
- Avril Genene Holt
- Department of Ophthalmology, Visual, and Anatomical Sciences (OVAS), School of Medicine, Wayne State University, 550 East Canfield Avenue, 454 Lande Building, Detroit, Michigan 48201, USA
| | - André Kühl
- Department of Ophthalmology, Visual, and Anatomical Sciences (OVAS), School of Medicine, Wayne State University, 550 East Canfield Avenue, 454 Lande Building, Detroit, Michigan 48201, USA
| | - Rod D Braun
- Department of Ophthalmology, Visual, and Anatomical Sciences (OVAS), School of Medicine, Wayne State University, 550 East Canfield Avenue, 454 Lande Building, Detroit, Michigan 48201, USA
| | - Richard Altschuler
- Department of Otolaryngology; Cell and Developmental Biology, Kresge Hearing Research Institute, University of Michigan, 1150 West Medical Center Drive, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
45
|
Szobota S, Mathur PD, Siegel S, Black K, Saragovi HU, Foster AC. BDNF, NT-3 and Trk receptor agonist monoclonal antibodies promote neuron survival, neurite extension, and synapse restoration in rat cochlea ex vivo models relevant for hidden hearing loss. PLoS One 2019; 14:e0224022. [PMID: 31671109 PMCID: PMC6822712 DOI: 10.1371/journal.pone.0224022] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 10/03/2019] [Indexed: 12/21/2022] Open
Abstract
Neurotrophins and their mimetics are potential treatments for hearing disorders because of their trophic effects on spiral ganglion neurons (SGNs) whose connections to hair cells may be compromised in many forms of hearing loss. Studies in noise or ototoxin-exposed animals have shown that local delivery of NT-3 or BDNF has beneficial effects on SGNs and hearing. We evaluated several TrkB or TrkC monoclonal antibody agonists and small molecules, along with BDNF and NT-3, in rat cochlea ex vivo models. The TrkB agonists BDNF and a monoclonal antibody, M3, had the greatest effects on SGN survival, neurite outgrowth and branching. In organotypic cochlear explants, BDNF and M3 enhanced synapse formation between SGNs and inner hair cells and restored these connections after excitotoxin-induced synaptopathy. Loss of these synapses has recently been implicated in hidden hearing loss, a condition characterized by difficulty hearing speech in the presence of background noise. The unique profile of M3 revealed here warrants further investigation, and the broad activity profile of BDNF observed underpins its continued development as a hearing loss therapeutic.
Collapse
Affiliation(s)
- Stephanie Szobota
- Otonomy, Inc., San Diego, California, United States of America
- * E-mail:
| | | | - Sairey Siegel
- Otonomy, Inc., San Diego, California, United States of America
| | | | - H. Uri Saragovi
- Lady Davis Institute-Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Alan C. Foster
- Otonomy, Inc., San Diego, California, United States of America
| |
Collapse
|
46
|
Liang Q, Shen N, Lai B, Xu C, Sun Z, Wang Z, Li S. Electrical Stimulation Degenerated Cochlear Synapses Through Oxidative Stress in Neonatal Cochlear Explants. Front Neurosci 2019; 13:1073. [PMID: 31680814 PMCID: PMC6803620 DOI: 10.3389/fnins.2019.01073] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 09/24/2019] [Indexed: 11/30/2022] Open
Abstract
Neurostimulation devices use electrical stimulation (ES) to substitute, supplement or modulate neural function. However, the impact of ES on their modulating structures is largely unknown. For example, recipients of cochlear implants using electroacoustic stimulation experienced delayed loss of residual hearing over time after ES, even though ES had no impact on the morphology of hair cells. In this study, using a novel model of cochlear explant culture with charge-balanced biphasic ES, we found that ES did not change the quantity and morphology of hair cells but decreased the number of inner hair cell (IHC) synapses and the density of spiral ganglion neuron (SGN) peripheral fibers. Inhibiting calcium influx with voltage-dependent calcium channel (VDCC) blockers attenuated the loss of SGN peripheral fibers and IHC synapses induced by ES. ES increased ROS/RNS in cochlear explants, but the inhibition of calcium influx abolished this effect. Glutathione peroxidase 1 (GPx1) and GPx2 in cochlear explants decreased under ES and ebselen abolished this effect and attenuated the loss of SGN peripheral fibers. This finding demonstrated that ES induced the degeneration of SGN peripheral fibers and IHC synapses in a current intensity- and duration-dependent manner in vitro. Calcium influx resulting in oxidative stress played an important role in this process. Additionally, ebselen might be a potential protector of ES-induced cochlear synaptic degeneration.
Collapse
Affiliation(s)
- Qiong Liang
- Department of Otolaryngology, Eye and ENT Hospital of Fudan University, Shanghai, China.,National Health Commission Key Laboratory of Hearing Medicine, Shanghai, China
| | - Na Shen
- Department of Otolaryngology, Eye and ENT Hospital of Fudan University, Shanghai, China.,National Health Commission Key Laboratory of Hearing Medicine, Shanghai, China.,Department of Otolaryngology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Bin Lai
- State Key Laboratory of Medical Neurobiology, Shanghai, China
| | - Changjian Xu
- Shanghai Cochlear Engineering Technology Research Center, Shanghai, China
| | - Zengjun Sun
- Shanghai Cochlear Engineering Technology Research Center, Shanghai, China
| | - Zhengmin Wang
- Department of Otolaryngology, Eye and ENT Hospital of Fudan University, Shanghai, China.,National Health Commission Key Laboratory of Hearing Medicine, Shanghai, China
| | - Shufeng Li
- Department of Otolaryngology, Eye and ENT Hospital of Fudan University, Shanghai, China.,National Health Commission Key Laboratory of Hearing Medicine, Shanghai, China
| |
Collapse
|
47
|
Han BR, Lin SC, Espinosa K, Thorne PR, Vlajkovic SM. Inhibition of the Adenosine A 2A Receptor Mitigates Excitotoxic Injury in Organotypic Tissue Cultures of the Rat Cochlea. Cells 2019; 8:cells8080877. [PMID: 31408967 PMCID: PMC6721830 DOI: 10.3390/cells8080877] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/09/2019] [Accepted: 08/09/2019] [Indexed: 12/20/2022] Open
Abstract
The primary loss of cochlear glutamatergic afferent nerve synapses due to noise or ageing (cochlear neuropathy) often presents as difficulties in speech discrimination in noisy conditions (hidden hearing loss (HHL)). Currently, there is no treatment for this condition. Our previous studies in mice with genetic deletion of the adenosine A2A receptor (A2AR) have demonstrated better preservation of cochlear afferent synapses and spiral ganglion neurons after noise exposure compared to wildtype mice. This has informed our current targeted approach to cochlear neuroprotection based on pharmacological inhibition of the A2AR. Here, we have used organotypic tissue culture of the Wistar rat cochlea at postnatal day 6 (P6) to model excitotoxic injury induced by N-methyl-d-aspartate (NMDA)/kainic acid (NK) treatment for 2 h. The excitotoxic injury was characterised by a reduction in the density of neural processes immediately after NK treatment and loss of afferent synapses in the presence of intact sensory hair cells. The administration of istradefylline (a clinically approved A2AR antagonist) reduced deafferentation of inner hair cells and improved the survival of afferent synapses after excitotoxic injury. This study thus provides evidence that A2AR inhibition promotes cochlear recovery from excitotoxic injury, and may have implications for the treatment of cochlear neuropathy and prevention of HHL.
Collapse
Affiliation(s)
- Belinda Rx Han
- Department of Physiology and The Eisdell Moore Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Shelly Cy Lin
- Department of Physiology and The Eisdell Moore Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Kristan Espinosa
- Department of Physiology and The Eisdell Moore Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Peter R Thorne
- Department of Physiology and The Eisdell Moore Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Srdjan M Vlajkovic
- Department of Physiology and The Eisdell Moore Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| |
Collapse
|
48
|
Sheth S, Sheehan K, Dhukhwa A, Al Aameri RFH, Mamillapalli C, Mukherjea D, Rybak LP, Ramkumar V. Oral Administration of Caffeine Exacerbates Cisplatin-Induced Hearing Loss. Sci Rep 2019; 9:9571. [PMID: 31267026 PMCID: PMC6606569 DOI: 10.1038/s41598-019-45964-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/14/2019] [Indexed: 12/31/2022] Open
Abstract
Adenosine A1 receptors (A1AR) are well characterized for their role in cytoprotection. Previous studies have demonstrated the presence of these receptors in the cochlea where their activation were shown to suppress cisplatin-induced inflammatory response and the resulting ototoxicity. Inhibition of A1AR by caffeine, a widely consumed psychoactive substance, could antagonize the endogenous protective role of these receptors in cochlea and potentiate cisplatin-induced hearing loss. This hypothesis was tested in a rat model of cisplatin ototoxicity following oral administration of caffeine. We report here that single-dose administration of caffeine exacerbates cisplatin-induced hearing loss without increasing the damage to outer hair cells (OHCs), but increased synaptopathy and inflammation in the cochlea. These effects of caffeine were mediated by its blockade of A1AR, as co-administration of R-PIA, an A1AR agonist, reversed the detrimental actions of caffeine and cisplatin on hearing loss. Multiple doses of caffeine exacerbated cisplatin ototoxicity which was associated with damage to OHCs and cochlear synaptopathy. These findings highlight a possible drug-drug interaction between caffeine and cisplatin for ototoxicity and suggest that caffeine consumption should be cautioned in cancer patients treated with a chemotherapeutic regimen containing cisplatin.
Collapse
Affiliation(s)
- Sandeep Sheth
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Kelly Sheehan
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Asmita Dhukhwa
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Raheem F H Al Aameri
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Chaitanya Mamillapalli
- Department of Internal Medicine (Division of Endocrinology), Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Debashree Mukherjea
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Leonard P Rybak
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Vickram Ramkumar
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States.
| |
Collapse
|
49
|
Kaur T, Clayman AC, Nash AJ, Schrader AD, Warchol ME, Ohlemiller KK. Lack of Fractalkine Receptor on Macrophages Impairs Spontaneous Recovery of Ribbon Synapses After Moderate Noise Trauma in C57BL/6 Mice. Front Neurosci 2019; 13:620. [PMID: 31263398 PMCID: PMC6585312 DOI: 10.3389/fnins.2019.00620] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 05/29/2019] [Indexed: 01/08/2023] Open
Abstract
Noise trauma causes loss of synaptic connections between cochlear inner hair cells (IHCs) and the spiral ganglion neurons (SGNs). Such synaptic loss can trigger slow and progressive degeneration of SGNs. Macrophage fractalkine signaling is critical for neuron survival in the injured cochlea, but its role in cochlear synaptopathy is unknown. Fractalkine, a chemokine, is constitutively expressed by SGNs and signals via its receptor CX3CR1 that is expressed on macrophages. The present study characterized the immune response and examined the function of fractalkine signaling in degeneration and repair of cochlear synapses following noise trauma. Adult mice wild type, heterozygous and knockout for CX3CR1 on a C57BL/6 background were exposed for 2 h to an octave band noise at 90 dB SPL. Noise exposure caused temporary shifts in hearing thresholds without any evident loss of hair cells in CX3CR1 heterozygous mice that have intact fractalkine signaling. Enhanced macrophage migration toward the IHC-synaptic region was observed immediately after exposure in all genotypes. Synaptic immunolabeling revealed a rapid loss of ribbon synapses throughout the basal turn of the cochlea of all genotypes. The damaged synapses spontaneously recovered in mice with intact CX3CR1. However, CX3CR1 knockout (KO) animals displayed enhanced synaptic degeneration that correlated with attenuated suprathreshold neural responses at higher frequencies. Exposed CX3CR1 KO mice also exhibited increased loss of IHCs and SGN cell bodies compared to exposed heterozygous mice. These results indicate that macrophages can promote repair of damaged synapses after moderate noise trauma and that repair requires fractalkine signaling.
Collapse
Affiliation(s)
- Tejbeer Kaur
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States
| | - Anna C Clayman
- Program in Audiology and Communication Sciences, Washington University School of Medicine, St. Louis, MO, United States
| | - Andrew J Nash
- Program in Audiology and Communication Sciences, Washington University School of Medicine, St. Louis, MO, United States
| | - Angela D Schrader
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States
| | - Mark E Warchol
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States
| | - Kevin K Ohlemiller
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States.,Program in Audiology and Communication Sciences, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
50
|
Coate TM, Scott MK, Gurjar MC. Current concepts in cochlear ribbon synapse formation. Synapse 2019; 73:e22087. [PMID: 30592086 PMCID: PMC6573016 DOI: 10.1002/syn.22087] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 12/11/2022]
Abstract
In mammals, hair cells and spiral ganglion neurons (SGNs) in the cochlea together are sophisticated "sensorineural" structures that transduce auditory information from the outside world into the brain. Hair cells and SGNs are joined by glutamatergic ribbon-type synapses composed of a molecular machinery rivaling in complexity the mechanoelectric transduction components found at the apical side of the hair cell. The cochlear hair cell ribbon synapse has received much attention lately because of recent and important findings related to its damage (sometimes termed "synaptopathy") as a result of noise overexposure. During development, ribbon synapses between type I SGNs and inner hair cells form in the time window between birth and hearing onset and is a process coordinated with type I SGN myelination, spontaneous activity, synaptic pruning, and innervation by efferents. In this review, we highlight new findings regarding the diversity of type I SGNs and inner hair cell synapses, and the molecular mechanisms of selective hair cell targeting. Also discussed are cell adhesion molecules and protein constituents of the ribbon synapse, and how these factors participate in ribbon synapse formation. We also note interesting new insights into the morphological development of type II SGNs, and the potential for cochlear macrophages as important players in protecting SGNs. We also address recent studies demonstrating that the structural and physiological profiles of the type I SGNs do not reach full maturity until weeks after hearing onset, suggesting a protracted development that is likely modulated by activity.
Collapse
Affiliation(s)
- Thomas M. Coate
- Georgetown University, Department of Biology, 37th and O St. NW. Washington, DC. 20007. USA
| | - M. Katie Scott
- Department of Biological Sciences and Purdue Institute of Integrative Neuroscience, Purdue University, West Lafayette, Indiana 47907. USA
| | - Mansa C. Gurjar
- Georgetown University, Department of Biology, 37th and O St. NW. Washington, DC. 20007. USA
| |
Collapse
|