1
|
Singh J, Randle MR, Walters BJ, Cox BC. The transcription factor Pou4f3 is essential for the survival of postnatal and adult mouse cochlear hair cells and normal hearing. Front Cell Neurosci 2024; 18:1369282. [PMID: 38566840 PMCID: PMC10985149 DOI: 10.3389/fncel.2024.1369282] [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: 01/11/2024] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction Hair cells (HCs) of the cochlea are responsible for sound transduction and hearing perception in mammals. Genetic mutations in the transcription factor Pou4f3 cause non-syndromic autosomal dominant hearing loss in humans (DFNA15) which varies in the age of onset depending on the individual mutation. Mouse models with germline deletion or mutations in Pou4f3 have previously demonstrated its critical role in the maturation and survival of cochlear HCs during embryonic development. However, the role of Pou4f3 in auditory function and in the survival or maintenance of cochlear HCs after birth and during adulthood has not been studied. Methods Therefore, using the inducible CreER-loxP system, we deleted Pou4f3 from mouse cochlear HCs at different postnatal ages, relevant to specific stages of HC maturation and hearing function. Results and discussion Elevated auditory brainstem response thresholds and significant HC loss were detected in mice with Pou4f3 deletion compared to their control littermates, regardless of the age when Pou4f3 was deleted. However, HC loss occurred more rapidly when Pou4f3 was deleted from immature HCs. Additionally, HC loss caused by Pou4f3 deletion did not affect the number of cochlear supporting cells, but caused a delayed loss of spiral ganglion neurons at 4 months after the deletion. In conclusion, Pou4f3 is necessary for the survival of cochlear HCs and normal hearing at all postnatal ages regardless of their maturation state. Our data also suggest that Pou4f3 indirectly regulates the survival of spiral ganglion neurons.
Collapse
Affiliation(s)
- Jarnail Singh
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Michelle R. Randle
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Bradley J. Walters
- Department of Otolaryngology-Head and Neck Surgery, University of Mississippi Medical Center, Jackson, MS, United States
| | - Brandon C. Cox
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, United States
| |
Collapse
|
2
|
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
|
3
|
Lee JH, Park S, Perez-Flores MC, Chen Y, Kang M, Choi J, Levine L, Gratton MA, Zhao J, Notterpek L, Yamoah EN. Demyelination and Na + Channel Redistribution Underlie Auditory and Vestibular Dysfunction in PMP22-Null Mice. eNeuro 2024; 11:ENEURO.0462-23.2023. [PMID: 38378628 PMCID: PMC11059428 DOI: 10.1523/eneuro.0462-23.2023] [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: 11/05/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 02/22/2024] Open
Abstract
Altered expression of peripheral myelin protein 22 (PMP22) results in demyelinating peripheral neuropathy. PMP22 exhibits a highly restricted tissue distribution with marked expression in the myelinating Schwann cells of peripheral nerves. Auditory and vestibular Schwann cells and the afferent neurons also express PMP22, suggesting a unique role in hearing and balancing. Indeed, neuropathic patients diagnosed with PMP22-linked hereditary neuropathies often present with auditory and balance deficits, an understudied clinical complication. To investigate the mechanism by which abnormal expression of PMP22 may cause auditory and vestibular deficits, we studied gene-targeted PMP22-null mice. PMP22-null mice exhibit an unsteady gait, have difficulty maintaining balance, and live for only ∼3-5 weeks relative to unaffected littermates. Histological analysis of the inner ear revealed reduced auditory and vestibular afferent nerve myelination and profound Na+ channel redistribution without PMP22. Yet, Na+ current density was unaltered, in stark contrast to increased K+ current density. Atypical postsynaptic densities and a range of neuronal abnormalities in the organ of Corti were also identified. Analyses of auditory brainstem responses (ABRs) and vestibular sensory-evoked potential (VsEP) revealed that PMP22-null mice had auditory and vestibular hypofunction. These results demonstrate that PMP22 is required for hearing and balance, and the protein is indispensable for the formation and maintenance of myelin in the peripheral arm of the eighth nerve. Our findings indicate that myelin abnormalities and altered signal propagation in the peripheral arm of the auditory nerve are likely causes of auditory deficits in patients with PMP22-linked neuropathies.
Collapse
Affiliation(s)
- Jeong Han Lee
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, Reno 89557, Nevada
| | - Seojin Park
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, Reno 89557, Nevada
- Prestige BioPharma, Busan 67264, South Korea
| | - Maria C Perez-Flores
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, Reno 89557, Nevada
| | - Yingying Chen
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, Reno 89557, Nevada
| | - Mincheol Kang
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, Reno 89557, Nevada
- Prestige BioPharma, Busan 67264, South Korea
| | - Jinsil Choi
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, Reno 89557, Nevada
| | - Lauren Levine
- Program in Audiology and Communication Sciences, Washington University, St. Louis 63110, Missouri
| | | | - Jie Zhao
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, Reno 89557, Nevada
| | - Lucia Notterpek
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, Reno 89557, Nevada
| | - Ebenezer N Yamoah
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, Reno 89557, Nevada
| |
Collapse
|
4
|
Filova I, Pysanenko K, Tavakoli M, Vochyanova S, Dvorakova M, Bohuslavova R, Smolik O, Fabriciova V, Hrabalova P, Benesova S, Valihrach L, Cerny J, Yamoah EN, Syka J, Fritzsch B, Pavlinkova G. ISL1 is necessary for auditory neuron development and contributes toward tonotopic organization. Proc Natl Acad Sci U S A 2022; 119:e2207433119. [PMID: 36074819 PMCID: PMC9478650 DOI: 10.1073/pnas.2207433119] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 08/04/2022] [Indexed: 11/18/2022] Open
Abstract
A cardinal feature of the auditory pathway is frequency selectivity, represented in a tonotopic map from the cochlea to the cortex. The molecular determinants of the auditory frequency map are unknown. Here, we discovered that the transcription factor ISL1 regulates the molecular and cellular features of auditory neurons, including the formation of the spiral ganglion and peripheral and central processes that shape the tonotopic representation of the auditory map. We selectively knocked out Isl1 in auditory neurons using Neurod1Cre strategies. In the absence of Isl1, spiral ganglion neurons migrate into the central cochlea and beyond, and the cochlear wiring is profoundly reduced and disrupted. The central axons of Isl1 mutants lose their topographic projections and segregation at the cochlear nucleus. Transcriptome analysis of spiral ganglion neurons shows that Isl1 regulates neurogenesis, axonogenesis, migration, neurotransmission-related machinery, and synaptic communication patterns. We show that peripheral disorganization in the cochlea affects the physiological properties of hearing in the midbrain and auditory behavior. Surprisingly, auditory processing features are preserved despite the significant hearing impairment, revealing central auditory pathway resilience and plasticity in Isl1 mutant mice. Mutant mice have a reduced acoustic startle reflex, altered prepulse inhibition, and characteristics of compensatory neural hyperactivity centrally. Our findings show that ISL1 is one of the obligatory factors required to sculpt auditory structural and functional tonotopic maps. Still, upon Isl1 deletion, the ensuing central plasticity of the auditory pathway does not suffice to overcome developmentally induced peripheral dysfunction of the cochlea.
Collapse
Affiliation(s)
- Iva Filova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Kateryna Pysanenko
- Department of Auditory Neuroscience, Institute of Experimental Medicine Czech Academy of Sciences, 14220 Prague, Czechia
| | - Mitra Tavakoli
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Simona Vochyanova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Martina Dvorakova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Romana Bohuslavova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Ondrej Smolik
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Valeria Fabriciova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Petra Hrabalova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Sarka Benesova
- Laboratory of Gene Expression, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Lukas Valihrach
- Laboratory of Gene Expression, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Jiri Cerny
- Laboratory of Light Microscopy, Institute of Molecular Genetics Czech Academy of Sciences, 14220 Prague, Czechia
| | - Ebenezer N. Yamoah
- Department of Physiology, School of Medicine, University of Nevada, Reno, NV 89557
| | - Josef Syka
- Department of Auditory Neuroscience, Institute of Experimental Medicine Czech Academy of Sciences, 14220 Prague, Czechia
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, IA 52242-1324
- Department of Otolaryngology, University of Iowa, Iowa City, IA 52242-1324
| | - Gabriela Pavlinkova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| |
Collapse
|
5
|
Chen D, Jia G, Zhang Y, Mao H, Zhao L, Li W, Chen Y, Ni Y. Sox2 overexpression alleviates noise-induced hearing loss by inhibiting inflammation-related hair cell apoptosis. J Neuroinflammation 2022; 19:59. [PMID: 35227273 PMCID: PMC8883703 DOI: 10.1186/s12974-022-02414-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 02/09/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The transcription factor Sox2 plays important roles in the developmental processes of multiple organs and tissues. However, whether Sox2 can protect mature or terminally differentiated cells against injury is still unknown.
Methods
We investigated the roles of Sox2 in cochlear hair cells, which are terminally differentiated cells, using conditional transgenic mice and several hearing loss models.
Results
Sox2 overexpression dramatically mitigated the degree of cochlear hair cell loss when exposed to ototoxic drugs. Noise-induced apoptosis of cochlear hair cells and hearing loss were also significantly alleviated by Sox2 overexpression. Notably, noise-induced upregulation of pro-inflammatory factors such as TNF-α and IL6 was inhibited by Sox2 overexpression. Then we used lipopolysaccharide to clarify the effect of Sox2 on cochlear inflammation, and Sox2 overexpression significantly inhibited lipopolysaccharide-induced upregulation of pro-inflammatory factors and alleviated inflammation-related cochlear hair cell death.
Conclusions
These results demonstrate a novel protective role of Sox2 in mature and terminally differentiated cochlear hair cells by inhibiting inflammation.
Collapse
|
6
|
Li W, Quan Y, Huang M, Wei W, Shu Y, Li H, Chen ZY. A Novel in vitro Model Delineating Hair Cell Regeneration and Neural Reinnervation in Adult Mouse Cochlea. Front Mol Neurosci 2022; 14:757831. [PMID: 35082601 PMCID: PMC8785685 DOI: 10.3389/fnmol.2021.757831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 12/07/2021] [Indexed: 11/13/2022] Open
Abstract
The study of an adult mammalian auditory system, such as regeneration, has been hampered by the lack of an in vitro system in which hypotheses can be tested efficiently. This is primarily due to the fact that the adult inner ear is encased in the toughest bone of the body, whereas its removal leads to the death of the sensory epithelium in culture. We hypothesized that we could take advantage of the integral cochlear structure to maintain the overall inner ear architecture and improve sensory epithelium survival in culture. We showed that by culturing adult mouse cochlea with the (surrounding) bone intact, the supporting cells (SCs) survived and almost all hair cells (HCs) degenerated. To evaluate the utility of the explant culture system, we demonstrated that the overexpression of Atoh1, an HC fate-determining factor, is sufficient to induce transdifferentiation of adult SCs to HC-like cells (HCLCs). Transdifferentiation-derived HCLCs resemble developmentally young HCs and are able to attract adult ganglion neurites. Furthermore, using a damage model, we showed that degenerated adult ganglions respond to regenerated HCLCs by directional neurite outgrowth that leads to HCLC-neuron contacts, strongly supporting the intrinsic properties of the HCLCs in establishing HCLC-neuron connections. The adult whole cochlear explant culture is suitable for diverse studies of the adult inner ear including regeneration, HC-neuron pathways, and inner ear drug screening.
Collapse
Affiliation(s)
- Wenyan Li
- Department of Otolaryngology-Head and Neck Surgery, Graduate Program in Speech and Hearing Bioscience and Technology and Program in Neuroscience, Harvard Medical School, Boston, MA, United States
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, MA, United States
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yizhou Quan
- Department of Otolaryngology-Head and Neck Surgery, Graduate Program in Speech and Hearing Bioscience and Technology and Program in Neuroscience, Harvard Medical School, Boston, MA, United States
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, MA, United States
| | - Mingqian Huang
- Department of Otolaryngology-Head and Neck Surgery, Graduate Program in Speech and Hearing Bioscience and Technology and Program in Neuroscience, Harvard Medical School, Boston, MA, United States
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, MA, United States
| | - Wei Wei
- Department of Otolaryngology-Head and Neck Surgery, Graduate Program in Speech and Hearing Bioscience and Technology and Program in Neuroscience, Harvard Medical School, Boston, MA, United States
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, MA, United States
| | - Yilai Shu
- Department of Otolaryngology-Head and Neck Surgery, Graduate Program in Speech and Hearing Bioscience and Technology and Program in Neuroscience, Harvard Medical School, Boston, MA, United States
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, MA, United States
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Huawei Li
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- *Correspondence: Huawei Li,
| | - Zheng-Yi Chen
- Department of Otolaryngology-Head and Neck Surgery, Graduate Program in Speech and Hearing Bioscience and Technology and Program in Neuroscience, Harvard Medical School, Boston, MA, United States
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, MA, United States
- Zheng-Yi Chen,
| |
Collapse
|
7
|
Xu Z, Rai V, Zuo J. TUB and ZNF532 Promote the Atoh1-Mediated Hair Cell Regeneration in Mouse Cochleae. Front Cell Neurosci 2021; 15:759223. [PMID: 34819838 PMCID: PMC8606527 DOI: 10.3389/fncel.2021.759223] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/15/2021] [Indexed: 12/31/2022] Open
Abstract
Hair cell (HC) regeneration is a promising therapy for permanent sensorineural hearing loss caused by HC loss in mammals. Atoh1 has been shown to convert supporting cells (SCs) to HCs in neonatal cochleae; its combinations with other factors can improve the efficiency of HC regeneration. To identify additional transcription factors for efficient Atoh1-mediated HC regeneration, here we optimized the electroporation procedure for explant culture of neonatal mouse organs of Corti and tested multiple transcription factors, Six2, Ikzf2, Lbh, Arid3b, Hmg20 a, Tub, Sall1, and Znf532, for their potential to promote Atoh1-mediated conversion of SCs to HCs. These transcription factors are expressed highly in HCs but differentially compared to the converted HCs based on previous studies, and are also potential co-reprograming factors for Atoh1-mediated SC-to-HC conversion by literature review. P0.5 cochlear explants were electroporated with these transcription factors alone or jointly with Atoh1. We found that Sox2+ progenitors concentrated within the lateral greater epithelial ridge (GER) can be electroporated efficiently with minimal HC damage. Atoh1 ectopic expression promoted HC regeneration in Sox2+ lateral GER cells. Transcription factors Tub and Znf532, but not the other six tested, promoted the HC regeneration mediated by Atoh1, consistent with previous studies that Isl1 promotes Atoh1-mediated HC conversionex vivo and in vivo and that both Tub and Znf532 are downstream targets of Isl1. Thus, our studies revealed an optimized electroporation method that can transfect the Sox2+ lateral GER cells efficiently with minimal damage to the endogenous HCs. Our results also demonstrate the importance of the Isl1/Tub/Znf532 pathway in promoting Atoh1-mediated HC regeneration.
Collapse
Affiliation(s)
- Zhenhang Xu
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States.,Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, China
| | - Vikrant Rai
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| | - Jian Zuo
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| |
Collapse
|
8
|
Cochlear hair cells of echolocating bats are immune to intense noise. J Genet Genomics 2021; 48:984-993. [PMID: 34393089 DOI: 10.1016/j.jgg.2021.06.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/15/2021] [Accepted: 06/06/2021] [Indexed: 11/23/2022]
Abstract
Exposure to intense noise can damage cochlear hair cells, leading to hearing loss in mammals. To avoid this constraint, most mammals have evolved in relatively quiet environments. Echolocating bats, however, are naturally exposed to continuous intense sounds from their own and neighboring sonar emissions for maintaining sonar directionality and range. Here, we propose the presence of intense noise resistance in cochlear hair cells of echolocating bats against noise-induced hearing loss (NIHL). To test this hypothesis, we performed noise exposure experiments for laboratory mice, one nonecholocating bat species, and five echolocating bat species. Contrary to nonecholocating fruit bats and mice, the hearing and the cochlear hair cells of echolocating bats remained unimpaired after continuous intense noise exposure. The comparative analyses of cochleae transcriptomic data showed that several genes protecting cochlear hair cells from intense sounds were overexpressed in echolocating bats. Particularly, the experimental examinations revealed that ISL1 overexpression significantly improved the survival of cochlear hair cells. Our findings support the existence of protective effects in cochlear hair cells of echolocating bats against intense noises, which provides new insight into understanding the relationship between cochlear hair cells and intense noises, and preventing or ameliorating NIHL in mammals.
Collapse
|
9
|
Mao H, Chen Y. Noise-Induced Hearing Loss: Updates on Molecular Targets and Potential Interventions. Neural Plast 2021; 2021:4784385. [PMID: 34306060 PMCID: PMC8279877 DOI: 10.1155/2021/4784385] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/12/2021] [Indexed: 12/18/2022] Open
Abstract
Noise overexposure leads to hair cell loss, synaptic ribbon reduction, and auditory nerve deterioration, resulting in transient or permanent hearing loss depending on the exposure severity. Oxidative stress, inflammation, calcium overload, glutamate excitotoxicity, and energy metabolism disturbance are the main contributors to noise-induced hearing loss (NIHL) up to now. Gene variations are also identified as NIHL related. Glucocorticoid is the only approved medication for NIHL treatment. New pharmaceuticals targeting oxidative stress, inflammation, or noise-induced neuropathy are emerging, highlighted by the nanoparticle-based drug delivery system. Given the complexity of the pathogenesis behind NIHL, deeper and more comprehensive studies still need to be fulfilled.
Collapse
Affiliation(s)
- Huanyu Mao
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai 200031, China
| | - Yan Chen
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai 200031, China
| |
Collapse
|
10
|
Ma Q, Yang F, Mackintosh C, Jayani RS, Oh S, Jin C, Nair SJ, Merkurjev D, Ma W, Allen S, Wang D, Almenar-Queralt A, Garcia-Bassets I. Super-Enhancer Redistribution as a Mechanism of Broad Gene Dysregulation in Repeatedly Drug-Treated Cancer Cells. Cell Rep 2021; 31:107532. [PMID: 32320655 DOI: 10.1016/j.celrep.2020.107532] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 01/07/2020] [Accepted: 03/27/2020] [Indexed: 12/14/2022] Open
Abstract
Cisplatin is an antineoplastic drug administered at suboptimal and intermittent doses to avoid life-threatening effects. Although this regimen shortly improves symptoms in the short term, it also leads to more malignant disease in the long term. We describe a multilayered analysis ranging from chromatin to translation-integrating chromatin immunoprecipitation sequencing (ChIP-seq), global run-on sequencing (GRO-seq), RNA sequencing (RNA-seq), and ribosome profiling-to understand how cisplatin confers (pre)malignant features by using a well-established ovarian cancer model of cisplatin exposure. This approach allows us to segregate the human transcriptome into gene modules representing distinct regulatory principles and to characterize that the most cisplatin-disrupted modules are associated with underlying events of super-enhancer plasticity. These events arise when cancer cells initiate without ultimately ending the program of drug-stimulated death. Using a PageRank-based algorithm, we predict super-enhancer regulator ISL1 as a driver of this plasticity and validate this prediction by using CRISPR/dCas9-KRAB inhibition (CRISPRi) and CRISPR/dCas9-VP64 activation (CRISPRa) tools. Together, we propose that cisplatin reprograms cancer cells when inducing them to undergo near-to-death experiences.
Collapse
Affiliation(s)
- Qi Ma
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Feng Yang
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Carlos Mackintosh
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ranveer Singh Jayani
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Soohwan Oh
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Chunyu Jin
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sreejith Janardhanan Nair
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Daria Merkurjev
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Wubin Ma
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Stephanie Allen
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Dong Wang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Angels Almenar-Queralt
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ivan Garcia-Bassets
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
11
|
Iyer AA, Groves AK. Transcription Factor Reprogramming in the Inner Ear: Turning on Cell Fate Switches to Regenerate Sensory Hair Cells. Front Cell Neurosci 2021; 15:660748. [PMID: 33854418 PMCID: PMC8039129 DOI: 10.3389/fncel.2021.660748] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/08/2021] [Indexed: 12/15/2022] Open
Abstract
Non-mammalian vertebrates can restore their auditory and vestibular hair cells naturally by triggering the regeneration of adjacent supporting cells. The transcription factor ATOH1 is a key regulator of hair cell development and regeneration in the inner ear. Following the death of hair cells, supporting cells upregulate ATOH1 and give rise to new hair cells. However, in the mature mammalian cochlea, such natural regeneration of hair cells is largely absent. Transcription factor reprogramming has been used in many tissues to convert one cell type into another, with the long-term hope of achieving tissue regeneration. Reprogramming transcription factors work by altering the transcriptomic and epigenetic landscapes in a target cell, resulting in a fate change to the desired cell type. Several studies have shown that ATOH1 is capable of reprogramming cochlear non-sensory tissue into cells resembling hair cells in young animals. However, the reprogramming ability of ATOH1 is lost with age, implying that the potency of individual hair cell-specific transcription factors may be reduced or lost over time by mechanisms that are still not clear. To circumvent this, combinations of key hair cell transcription factors have been used to promote hair cell regeneration in older animals. In this review, we summarize recent findings that have identified and studied these reprogramming factor combinations for hair cell regeneration. Finally, we discuss the important questions that emerge from these findings, particularly the feasibility of therapeutic strategies using reprogramming factors to restore human hearing in the future.
Collapse
Affiliation(s)
- Amrita A. Iyer
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Program in Genetics & Genomics, Houston, TX, United States
| | - Andrew K. Groves
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Program in Genetics & Genomics, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
12
|
Pavlinkova G. Molecular Aspects of the Development and Function of Auditory Neurons. Int J Mol Sci 2020; 22:ijms22010131. [PMID: 33374462 PMCID: PMC7796308 DOI: 10.3390/ijms22010131] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 01/08/2023] Open
Abstract
This review provides an up-to-date source of information on the primary auditory neurons or spiral ganglion neurons in the cochlea. These neurons transmit auditory information in the form of electric signals from sensory hair cells to the first auditory nuclei of the brain stem, the cochlear nuclei. Congenital and acquired neurosensory hearing loss affects millions of people worldwide. An increasing body of evidence suggest that the primary auditory neurons degenerate due to noise exposure and aging more readily than sensory cells, and thus, auditory neurons are a primary target for regenerative therapy. A better understanding of the development and function of these neurons is the ultimate goal for long-term maintenance, regeneration, and stem cell replacement therapy. In this review, we provide an overview of the key molecular factors responsible for the function and neurogenesis of the primary auditory neurons, as well as a brief introduction to stem cell research focused on the replacement and generation of auditory neurons.
Collapse
Affiliation(s)
- Gabriela Pavlinkova
- BIOCEV, Institute of Biotechnology of the Czech Academy of Sciences, 25250 Vestec, Czech Republic
| |
Collapse
|
13
|
Niu Y, Xie C, Du Z, Zeng J, Chen H, Jin L, Zhang Q, Yu H, Wang Y, Ping J, Yang C, Liu X, Li Y, Zhou G. Genome-wide association study identifies 7q11.22 and 7q36.3 associated with noise-induced hearing loss among Chinese population. J Cell Mol Med 2020; 25:411-420. [PMID: 33242228 PMCID: PMC7810922 DOI: 10.1111/jcmm.16094] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/02/2020] [Accepted: 10/28/2020] [Indexed: 12/25/2022] Open
Abstract
Noise-induced hearing loss (NIHL) seriously affects the life quality of humans and causes huge economic losses to society. To identify novel genetic loci involved in NIHL, we conducted a genome-wide association study (GWAS) for this symptom in Chinese populations. GWAS scan was performed in 89 NIHL subjects (cases) and 209 subjects with normal hearing who have been exposed to a similar noise environment (controls), followed by a replication study consisting of 53 cases and 360 controls. We identified that four candidate pathways were nominally significantly associated with NIHL, including the Erbb, Wnt, hedgehog and intraflagellar transport pathways. In addition, two novel index single-nucleotide polymorphisms, rs35075890 in the intron of AUTS2 gene at 7q11.22 (combined P = 1.3 × 10-6 ) and rs10081191 in the intron of PTPRN2 gene at 7q36.3 (combined P = 2.1 × 10-6 ), were significantly associated with NIHL. Furthermore, the expression quantitative trait loci analyses revealed that in brain tissues, the genotypes of rs35075890 are significantly associated with the expression levels of AUTS2, and the genotypes of rs10081191 are significantly associated with the expressions of PTPRN2 and WDR60. In conclusion, our findings highlight two novel loci at 7q11.22 and 7q36.3 conferring susceptibility to NIHL.
Collapse
Affiliation(s)
- Yuguang Niu
- Department of Otolaryngology, the First Medical Center of PLA General Hospital, Beijing, China
| | - Chengyong Xie
- Medical College of Guizhou University, Guiyang city, China
| | - Zhenhua Du
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Jifeng Zeng
- Department of Otolaryngology, the No. 954 Hospital of PLA, Shannan City, China
| | - Hongxia Chen
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Liang Jin
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Qing Zhang
- Collaborative Innovation Center for Personalized Cancer Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing City, China
| | - Huiying Yu
- Outpatient Department, the Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Yahui Wang
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Jie Ping
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Chenning Yang
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xinyi Liu
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yuanfeng Li
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Gangqiao Zhou
- Medical College of Guizhou University, Guiyang city, China.,State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Institute of Radiation Medicine, Beijing, China.,Collaborative Innovation Center for Personalized Cancer Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing City, China
| |
Collapse
|
14
|
Taroc EZM, Katreddi RR, Forni PE. Identifying Isl1 Genetic Lineage in the Developing Olfactory System and in GnRH-1 Neurons. Front Physiol 2020; 11:601923. [PMID: 33192618 PMCID: PMC7609815 DOI: 10.3389/fphys.2020.601923] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 09/30/2020] [Indexed: 01/04/2023] Open
Abstract
During embryonic development, symmetric ectodermal thickenings [olfactory placodes (OP)] give rise to several cell types that comprise the olfactory system, such as those that form the terminal nerve ganglion (TN), gonadotropin releasing hormone-1 neurons (GnRH-1ns), and other migratory neurons in rodents. Even though the genetic heterogeneity among these cell types is documented, unidentified cell populations arising from the OP remain. One candidate to identify placodal derived neurons in the developing nasal area is the transcription factor Isl1, which was recently identified in GnRH-3 neurons of the terminal nerve in fish, as well as expression in neurons of the nasal migratory mass (MM). Here, we analyzed the Isl1 genetic lineage in chemosensory neuronal populations in the nasal area and migratory GnRH-1ns in mice using in situ hybridization, immunolabeling a Tamoxifen inducible Isl1CreERT and a constitutive Isl1Cre knock-in mouse lines. In addition, we also performed conditional Isl1 ablation in developing GnRH neurons. We found Isl1 lineage across non-sensory cells of the respiratory epithelium and sustentacular cells of OE and VNO. We identified a population of transient embryonic Isl1 + neurons in the olfactory epithelium and sparse Isl1 + neurons in postnatal VNO. Isl1 is expressed in almost all GnRH neurons and in approximately half of the other neuron populations in the MM. However, Isl1 conditional ablation alone does not significantly compromise GnRH-1 neuronal migration or GnRH-1 expression, suggesting compensatory mechanisms. Further studies will elucidate the functional and mechanistic role of Isl1 in development of migratory endocrine neurons.
Collapse
Affiliation(s)
- Ed Zandro M Taroc
- Department of Biological Sciences, The RNA Institute, and the Center for Neuroscience Research, University at Albany, State University of New York, Albany, NY, United States
| | - Raghu Ram Katreddi
- Department of Biological Sciences, The RNA Institute, and the Center for Neuroscience Research, University at Albany, State University of New York, Albany, NY, United States
| | - Paolo E Forni
- Department of Biological Sciences, The RNA Institute, and the Center for Neuroscience Research, University at Albany, State University of New York, Albany, NY, United States
| |
Collapse
|
15
|
Zhao X, Jin C, Dong T, Sun Z, Zheng X, Feng B, Cheng Z, Li X, Tao Y, Wu H. Characterization of promoters for adeno-associated virus mediated efficient Cas9 activation in adult Cas9 knock-in murine cochleae. Hear Res 2020; 394:107999. [PMID: 32611519 DOI: 10.1016/j.heares.2020.107999] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 04/30/2020] [Accepted: 05/20/2020] [Indexed: 11/27/2022]
Abstract
CRISPR/Cas9 gene editing enables the treatment of hearing loss in congenitally deaf neonatal mice via both viral and non-viral delivery. While adeno-associated virus (AAV)-mediated gene delivery systems have been shown to be effective tools for gene replacement in the inner ear, application of the AAV-mediated CRISPR/Cas9 gene-editing approach for this purpose is yet to be documented. Based on our previous findings, we focused on the effects of several AAVs delivered via canalostomy injection in adult mice. Among the AAVs examined, AAV8 showed the greatest efficiency and specificity in transducing inner hair cells (IHC). The ability of Cre-expressing AAV8 to activate Cas9 in floxed-Cas9 knock-in (Cas9 KI) mice was further evaluated. We compared the effects of six different promoters (CMV, CAG, hSyn, CaMKIIa, GFAP, and ALB) of AAV8 delivered to the inner ear of adult Cas9 KI mice. Our findings showed that three AAV groups (CMV, CAG and hSyn promoters) infected the inner ear efficiently with different tropisms. Notably, AAVs with CMV, CAG, and hSyn promoters infected diverse cell types in mature murine cochleae, including IHCs. In particular, AAV8-hSyn showed high affinity to IHCs and spiral ganglion neurons (SGN). Neither the AAV8 virus itself (except AAV8-CAG) nor the surgical procedures used caused damage to HCs or impaired normal hearing. Our findings indicated that injection of AAV-Cre into mature inner ear efficiently induces Cas9 activation to achieve safe and efficient gene editing and different constituent promoters confer diverse infection patterns in cochlea, expanding the repertoire of gene-editing tools for regulating gene expression in target cells of the inner ear as part of the collective effort to rescue genetic hearing loss and develop effective gene therapy techniques.
Collapse
Affiliation(s)
- Xingle Zhao
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, No.639, Zhizaoju Road, Shanghai, 200011, PR China; Ear Institute, Shanghai Jiaotong University School of Medicine, No.115, Jinzun Road, Shanghai, 200011, PR China; Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200011, PR China
| | - Chenxi Jin
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, No.639, Zhizaoju Road, Shanghai, 200011, PR China; Ear Institute, Shanghai Jiaotong University School of Medicine, No.115, Jinzun Road, Shanghai, 200011, PR China; Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200011, PR China
| | - Tingting Dong
- Ear Institute, Shanghai Jiaotong University School of Medicine, No.115, Jinzun Road, Shanghai, 200011, PR China; Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200011, PR China; Biobank of Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.115, Jinzun Road, Shanghai, 200011, PR China
| | - Zhuoer Sun
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, No.639, Zhizaoju Road, Shanghai, 200011, PR China; Ear Institute, Shanghai Jiaotong University School of Medicine, No.115, Jinzun Road, Shanghai, 200011, PR China; Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200011, PR China
| | - Xiaofei Zheng
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, No.639, Zhizaoju Road, Shanghai, 200011, PR China; Ear Institute, Shanghai Jiaotong University School of Medicine, No.115, Jinzun Road, Shanghai, 200011, PR China; Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200011, PR China
| | - Baoyi Feng
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, No.639, Zhizaoju Road, Shanghai, 200011, PR China; Ear Institute, Shanghai Jiaotong University School of Medicine, No.115, Jinzun Road, Shanghai, 200011, PR China; Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200011, PR China
| | - Zhenzhe Cheng
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, No.639, Zhizaoju Road, Shanghai, 200011, PR China; Ear Institute, Shanghai Jiaotong University School of Medicine, No.115, Jinzun Road, Shanghai, 200011, PR China; Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200011, PR China
| | - Xiang Li
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, No.639, Zhizaoju Road, Shanghai, 200011, PR China; Ear Institute, Shanghai Jiaotong University School of Medicine, No.115, Jinzun Road, Shanghai, 200011, PR China; Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200011, PR China
| | - Yong Tao
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, No.639, Zhizaoju Road, Shanghai, 200011, PR China; Ear Institute, Shanghai Jiaotong University School of Medicine, No.115, Jinzun Road, Shanghai, 200011, PR China; Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200011, PR China.
| | - Hao Wu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, No.639, Zhizaoju Road, Shanghai, 200011, PR China; Ear Institute, Shanghai Jiaotong University School of Medicine, No.115, Jinzun Road, Shanghai, 200011, PR China; Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200011, PR China.
| |
Collapse
|
16
|
Hosoya M, Fujioka M, Murayama AY, Okano H, Ogawa K. The common marmoset as suitable nonhuman alternative for the analysis of primate cochlear development. FEBS J 2020; 288:325-353. [PMID: 32323465 PMCID: PMC7818239 DOI: 10.1111/febs.15341] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/30/2020] [Accepted: 04/20/2020] [Indexed: 12/13/2022]
Abstract
Cochlear development is a complex process with precise spatiotemporal patterns. A detailed understanding of this process is important for studies of congenital hearing loss and regenerative medicine. However, much of our understanding of cochlear development is based on rodent models. Animal models that bridge the gap between humans and rodents are needed. In this study, we investigated the development of hearing organs in a small New World monkey species, the common marmoset (Callithrix jacchus). We describe the general stages of cochlear development in comparison with those of humans and mice. Moreover, we examined more than 25 proteins involved in cochlear development and found that expression patterns were generally conserved between rodents and primates. However, several proteins involved in supporting cell processes and neuronal development exhibited interspecific expression differences. Human fetal samples for studies of primate‐specific cochlear development are extremely rare, especially for late developmental stages. Our results support the use of the common marmoset as an effective alternative for analyses of primate cochlear development.
Collapse
Affiliation(s)
- Makoto Hosoya
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masato Fujioka
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Ayako Y Murayama
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan.,Laboratory for Marmoset Neural Architecture, Center for Brain Science, RIKEN, Wako, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan.,Laboratory for Marmoset Neural Architecture, Center for Brain Science, RIKEN, Wako, Japan
| | - Kaoru Ogawa
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
17
|
Adeno-associated virus vector enables safe and efficient Cas9 activation in neonatal and adult Cas9 knockin murine cochleae. Gene Ther 2020; 27:392-405. [PMID: 32005950 DOI: 10.1038/s41434-020-0124-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 11/30/2019] [Accepted: 01/22/2020] [Indexed: 12/26/2022]
Abstract
Adeno-associated virus (AAV)-mediated gene delivery systems have been shown to be effective tools for gene manipulation in the inner ear. For example, hair cells (HCs) and multiple other cell types can be transduced by the local injection of AAVs into the inner ear. However, application of the AAV-mediated CRISPR/Cas9 gene-editing approach to the inner ear in adult mice has not yet been studied. Based on our previous work, we investigated several AAV serotypes in neonatal and adult mice in parallel, and found that AAV8 had the top efficiency to transduce inner HCs. We then tested the ability of Cre-expressing AAV8 to activate Cas9 in floxed-Cas9 knockin mice, and observed significant Cas9 activation in the inner ear of both neonatal and adult animals. Neither the AAV8 virus itself nor the surgical procedures used to deliver it-cochleostomy for neonatal mice and canalostomy for adult mice-caused any damage to HCs or impaired normal hearing. Our studies indicate that the local injection of AAV8-Cre can induce Cas9 activation to perform safe and efficient gene editing in the inner ear, expanding the repertoire of gene-editing tools for regulating gene expression in the inner ear as a part of efforts to rescue genetic hearing loss, initiate regeneration of HCs, or develop gene therapy techniques.
Collapse
|
18
|
Levy S, Bargmann CI. An Adaptive-Threshold Mechanism for Odor Sensation and Animal Navigation. Neuron 2019; 105:534-548.e13. [PMID: 31761709 DOI: 10.1016/j.neuron.2019.10.034] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 05/31/2019] [Accepted: 10/27/2019] [Indexed: 01/01/2023]
Abstract
Identifying the environmental information and computations that drive sensory detection is key for understanding animal behavior. Using experimental and theoretical analysis of AWCON, a well-described olfactory neuron in C. elegans, here we derive a general and broadly useful model that matches stimulus history to odor sensation and behavioral responses. We show that AWCON sensory activity is regulated by an absolute signal threshold that continuously adapts to odor history, allowing animals to compare present and past odor concentrations. The model predicts sensory activity and probabilistic behavior during animal navigation in different odor gradients and across a broad stimulus regime. Genetic studies demonstrate that the cGMP-dependent protein kinase EGL-4 determines the timescale of threshold adaptation, defining a molecular basis for a critical model feature. The adaptive threshold model efficiently filters stimulus noise, allowing reliable sensation in fluctuating environments, and represents a feedforward sensory mechanism with implications for other sensory systems.
Collapse
Affiliation(s)
- Sagi Levy
- Lulu and Anthony Wang Laboratory of Neural Circuits and Behavior, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| | - Cornelia I Bargmann
- Lulu and Anthony Wang Laboratory of Neural Circuits and Behavior, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA; Chan Zuckerberg Initiative, Palo Alto, CA 94301, USA
| |
Collapse
|
19
|
Early functional alterations in membrane properties and neuronal degeneration are hallmarks of progressive hearing loss in NOD mice. Sci Rep 2019; 9:12128. [PMID: 31431657 PMCID: PMC6702171 DOI: 10.1038/s41598-019-48376-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 08/05/2019] [Indexed: 11/24/2022] Open
Abstract
Presbycusis or age-related hearing loss (ARHL) is the most common sensory deficit in the human population. A substantial component of the etiology stems from pathological changes in sensory and non-sensory cells in the cochlea. Using a non-obese diabetic (NOD) mouse model, we have characterized changes in both hair cells and spiral ganglion neurons that may be relevant for early signs of age-related hearing loss (ARHL). We demonstrate that hair cell loss is preceded by, or in parallel with altered primary auditory neuron functions, and latent neurite retraction at the hair cell-auditory neuron synapse. The results were observed first in afferent inner hair cell synapse of type I neurites, followed by type II neuronal cell-body degeneration. Reduced membrane excitability and loss of postsynaptic densities were some of the inaugural events before any outward manifestation of hair bundle disarray and hair cell loss. We have identified profound alterations in type I neuronal membrane properties, including a reduction in membrane input resistance, prolonged action potential latency, and a decrease in membrane excitability. The resting membrane potential of aging type I neurons in the NOD, ARHL model, was significantly hyperpolarized, and analyses of the underlying membrane conductance showed a significant increase in K+ currents. We propose that attempts to alleviate some forms of ARHL should include early targeted primary latent neural degeneration for effective positive outcomes.
Collapse
|
20
|
Gu X, Chai R, Guo L, Dong B, Li W, Shu Y, Huang X, Li H. Transduction of Adeno-Associated Virus Vectors Targeting Hair Cells and Supporting Cells in the Neonatal Mouse Cochlea. Front Cell Neurosci 2019; 13:8. [PMID: 30733670 PMCID: PMC6353798 DOI: 10.3389/fncel.2019.00008] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 01/10/2019] [Indexed: 02/05/2023] Open
Abstract
Adeno-associated virus (AAV) is the preferred vector for gene therapy of hereditary deafness, and different viral serotypes, promoters and transduction pathways can influence the targeting of AAV to different types of cells and the expression levels of numerous exogenous genes. To determine the transduction and expression patterns of AAV with different serotypes or promoters in hair cells and supporting cells in the neonatal mouse cochlea, we examined the expression of enhanced green fluorescent protein (eGFP) for five different types of AAV vectors [serotypes 2, 9, and Anc80L65 with promoter cytomegalovirus (CMV)-beta-Globin and serotypes 2 and 9 with promoter chicken beta-actin (CBA)] in in vitro cochlear explant cultures and we tested the transduction of AAV2/2-CBA, AAV2/9-CBA, and AAV2/Anc80L65-CMV by in vivo microinjection into the scala media of the cochlea. We found that each AAV vector had its own transduction and expression characteristics in hair cells and supporting cells in different regions of the cochlea. There was a tonotopic gradient for the in vitro transduction of AAV2/2-CBA, AAV2/9-CBA, AAV2/2-CMV, and AAV2/9-CMV in outer hair cells (OHCs), with more OHCs expressing eGFP at the base of the cochlea than at the apex. AAV2/2-CBA in vitro and AAV2/Anc80L65-CMV in vivo induced more supporting cells expressing eGFP at the apex than in the base. We found that AAV vectors with different promoters had different expression efficacies in hair cells and supporting cells of the auditory epithelium. The CMV-beta-Globin promoter could drive the expression of the delivered construct more efficiently in hair cells, while the CBA promoter was more efficient in supporting cells. The in vitro and in vivo experiments both demonstrated that AAV2/Anc80L65-CMV was a very promising vector for gene therapy of deafness because of its high transduction rates in hair cells. These results might be useful for selecting the appropriate vectors for gene delivery into different types of inner ear cells and thus improving the effectiveness of gene therapy.
Collapse
Affiliation(s)
- Xi Gu
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China
| | - Luo Guo
- ENT Institute and Otorhinolaryngology Department, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Biao Dong
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Wenyan Li
- ENT Institute and Otorhinolaryngology Department, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Yilai Shu
- ENT Institute and Otorhinolaryngology Department, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Xinsheng Huang
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huawei Li
- ENT Institute and Otorhinolaryngology Department, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Cochlear Implant, Shanghai, China.,The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Liu W, Wang C, Yu H, Liu S, Yang J. Expression of acetylated tubulin in the postnatal developing mouse cochlea. Eur J Histochem 2018; 62. [PMID: 30088716 PMCID: PMC6119817 DOI: 10.4081/ejh.2018.2942] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 08/01/2018] [Indexed: 11/24/2022] Open
Abstract
Microtubules are an essential component of the cytoskeleton of a eukaryotic cell. The post-translational tubulin modifications play an important role in regulating microtubule properties, acetylation tubulin is one of the major post-translational modifications of microtubules. Acetylation tubulin has also been shown to be expressed in the cochlea. However, the detailed expression profiles of acetylation tubulin protein during development have not yet been investigated in the postnatal mammalian cochlea. Here, we first examined the spatio-temporal expression of acetylated tubulin in the mouse cochlea during postnatal development. At postnatal day 1 (P1), acetylated tubulin was localized primarily to the auditory nerve inside the cochlea and their synaptic contacts with the inner and outer hair cells (IHCs and OHCs). In the organ of Corti, acetylated tubulin occurred first at the apex of pillar cells. At P5, acetylated tubulin first appeared in the phalangeal processes of Deiters’ cells. At P8, staining was maintained in the phalangeal processes of Deiters’ cells and neural elements. At P10, labeling in Deiters’ cells extended from the apices of OHCs to the basilar membrane, acetylated tubulin was expressed throughout the cytoplasm of inner and outer pillar cells. At P12, acetylated tubulin displayed prominent and homogeneous labeling along the full length of the pillar cells. Linear labeling was present mainly in the Deiters’ cell bodies underlying OHCs. Between P14 and P17, acetylated tubulin was strongly expressed in inner and outer pillar cells and Deiters’ cells in a similar pattern as observed in the adult, and labeling in these cells were arranged in bundles. In addition, acetylated tubulin was expressed in stria vascularis, root cell bodies, and a small number of fibrocytes of the spiral ligament until the adult. In the adult mouse cochlea, immunostaining continued to predominate in Deiters’ cells and pillar cells. In Deiters’ cells, immunolabeling formed cups securing OHCs basal portions, and continued presence of acetylated tubulin-labeled nerve terminals below IHCs was shown. Our results presented here underscored the essential role played by acetylated tubulin in postnatal cochlear development, auditory neurotransmission and cochlear mechanics.
Collapse
Affiliation(s)
- Wenjing Liu
- Shanghai Jiaotong University Ear Institute, Department of Otorhinolaryngology-Head and Neck Surgery.
| | | | | | | | | |
Collapse
|
22
|
Zhang W, Kim SM, Wang W, Cai C, Feng Y, Kong W, Lin X. Cochlear Gene Therapy for Sensorineural Hearing Loss: Current Status and Major Remaining Hurdles for Translational Success. Front Mol Neurosci 2018; 11:221. [PMID: 29997477 PMCID: PMC6028713 DOI: 10.3389/fnmol.2018.00221] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 06/06/2018] [Indexed: 12/19/2022] Open
Abstract
Sensorineural hearing loss (SNHL) affects millions of people. Genetic mutations play a large and direct role in both congenital and late-onset cases of SNHL (e.g., age-dependent hearing loss, ADHL). Although hearing aids can help moderate to severe hearing loss the only effective treatment for deaf patients is the cochlear implant (CI). Gene- and cell-based therapies potentially may preserve or restore hearing with more natural sound perception, since their theoretical frequency resolution power is much higher than that of cochlear implants. These biologically-based interventions also carry the potential to re-establish hearing without the need for implanting any prosthetic device; the convenience and lower financial burden afforded by such biologically-based interventions could potentially benefit far more SNHL patients. Recently major progress has been achieved in preclinical studies of cochlear gene therapy. This review critically evaluates recent advances in the preclinical trials of gene therapies for SNHL and the major remaining challenges for the development and eventual clinical translation of this novel therapy. The cochlea bears many similarities to the eye for translational studies of gene therapies. Experience gained in ocular gene therapy trials, many of which have advanced to clinical phase III, may provide valuable guidance in improving the chance of success for cochlear gene therapy in human trials. A discussion on potential implications of translational knowledge gleaned from large numbers of advanced clinical trials of ocular gene therapy is therefore included.
Collapse
Affiliation(s)
- Wenjuan Zhang
- Department of Otolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sun Myoung Kim
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, GA, United States
| | - Wenwen Wang
- Department of Otolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | - Yong Feng
- Xiangya School of Medicine, Changsha, China
| | - Weijia Kong
- Department of Otolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Lin
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
23
|
Tao Y, Huang M, Shu Y, Ruprecht A, Wang H, Tang Y, Vandenberghe LH, Wang Q, Gao G, Kong WJ, Chen ZY. Delivery of Adeno-Associated Virus Vectors in Adult Mammalian Inner-Ear Cell Subtypes Without Auditory Dysfunction. Hum Gene Ther 2018; 29:492-506. [PMID: 29130354 PMCID: PMC5909114 DOI: 10.1089/hum.2017.120] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/27/2017] [Indexed: 02/05/2023] Open
Abstract
Hearing loss, including genetic hearing loss, is one of the most common forms of sensory deficits in humans with limited options of treatment. Adeno-associated virus (AAV)-mediated gene transfer has been shown to recover auditory functions effectively in mouse models of genetic deafness when delivered at neonatal stages. However, the mouse cochlea is still developing at those time points, whereas in humans, the newborn inner ears are already fully mature. For effective gene therapy to treat genetic deafness, it is necessary to determine whether AAV-mediated therapy can be equally effective in the fully mature mouse inner ear without causing damage to the inner ear. This study tested several AAV serotypes by canalostomy in adult mice. It is shown that most AAVs transduce the sensory inner hair cells efficiently, but are less efficient at transducing outer hair cells. A subset of AAVs also transduces non-sensory cochlear cell types. Neither the surgical procedure of canalostomy nor the AAV serotypes damage hair cells or impair normal hearing. The studies indicate that canalostomy can be a viable route for safe and efficient gene delivery, and they expand the repertoire of AAVs to target diverse cell types in the adult inner ear.
Collapse
Affiliation(s)
- Yong Tao
- Department of Otolaryngology and Program in Neuroscience, Harvard Medical School and Eaton Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingqian Huang
- Department of Otolaryngology and Program in Neuroscience, Harvard Medical School and Eaton Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
| | - Yilai Shu
- Department of Otolaryngology and Program in Neuroscience, Harvard Medical School and Eaton Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
- Department of Otolaryngology—Head and Neck Surgery, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Adam Ruprecht
- Department of Otolaryngology and Program in Neuroscience, Harvard Medical School and Eaton Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
| | - Hongyang Wang
- Department of Otolaryngology and Program in Neuroscience, Harvard Medical School and Eaton Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
- Chinese PLA Institute of Otolaryngology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Yong Tang
- Department of Otolaryngology and Program in Neuroscience, Harvard Medical School and Eaton Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
- Department of Ear, Nose and Throat, People's Hospital of Jilin Province, Changchun, China
| | - Luk H. Vandenberghe
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute and Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
- Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Qiuju Wang
- Chinese PLA Institute of Otolaryngology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Guangping Gao
- Horae Gene Therapy Center and Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wei-Jia Kong
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng-Yi Chen
- Department of Otolaryngology and Program in Neuroscience, Harvard Medical School and Eaton Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
| |
Collapse
|
24
|
Gao X, Tao Y, Lamas V, Huang M, Yeh WH, Pan B, Hu YJ, Hu JH, Thompson DB, Shu Y, Li Y, Wang H, Yang S, Xu Q, Polley DB, Liberman MC, Kong WJ, Holt JR, Chen ZY, Liu DR. Treatment of autosomal dominant hearing loss by in vivo delivery of genome editing agents. Nature 2017; 553:217-221. [PMID: 29258297 PMCID: PMC5784267 DOI: 10.1038/nature25164] [Citation(s) in RCA: 385] [Impact Index Per Article: 48.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 11/24/2017] [Indexed: 02/07/2023]
Abstract
Although genetic factors contribute to almost half of all deafness cases,
treatment options for genetic deafness are limited1–5. We developed a genome editing approach to target a
dominantly inherited form of genetic deafness. Here we show that cationic
lipid-mediated in vivo delivery of Cas9:guide RNA complexes can
ameliorate hearing loss in a mouse model of human genetic deafness. We designed
and validated in vitro and in primary fibroblasts genome
editing agents that preferentially disrupt the dominant deafness-associated
allele in the Tmc1 (transmembrane channel-like 1) Beethoven
(Bth) mouse model, even though the mutant
Bth allele differs from the wild-type allele at only a
single base pair. Injection of Cas9:guide RNA:lipid complexes targeting the
Bth allele into the cochlea of neonatal
Bth/+ mice substantially reduced progressive
hearing loss. We observed higher hair cell survival rates and lower auditory
brainstem response (ABR) thresholds in injected ears compared with uninjected
ears or ears injected with complexes that target an unrelated gene. Enhanced
acoustic reflex responses were observed among injected compared to uninjected
Bth/+ animals. These findings suggest protein:RNA
complex delivery of target gene-disrupting agents in vivo as a
potential strategy for the treatment of some autosomal dominant hearing loss
diseases.
Collapse
Affiliation(s)
- Xue Gao
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, USA.,Howard Hughes Medical Institute, Harvard University, Cambridge, Massachusetts 02138, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02141, USA
| | - Yong Tao
- Department of Otolaryngology and Program in Neuroscience, Harvard Medical School and Eaton Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts 02114, USA.,Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Veronica Lamas
- Department of Otolaryngology and Program in Neuroscience, Harvard Medical School and Eaton Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts 02114, USA
| | - Mingqian Huang
- Department of Otolaryngology and Program in Neuroscience, Harvard Medical School and Eaton Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts 02114, USA
| | - Wei-Hsi Yeh
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, USA.,Howard Hughes Medical Institute, Harvard University, Cambridge, Massachusetts 02138, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02141, USA.,Program in Speech and Hearing Bioscience and Technology, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Bifeng Pan
- Departments of Otolaryngology and Neurology, F.M. Kirby Neurobiology Center Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Yu-Juan Hu
- Department of Otolaryngology and Program in Neuroscience, Harvard Medical School and Eaton Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts 02114, USA.,Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Johnny H Hu
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, USA.,Howard Hughes Medical Institute, Harvard University, Cambridge, Massachusetts 02138, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02141, USA
| | - David B Thompson
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, USA.,Howard Hughes Medical Institute, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Yilai Shu
- Department of Otolaryngology and Program in Neuroscience, Harvard Medical School and Eaton Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts 02114, USA.,Department of Otolaryngology-Head and Neck Surgery, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yamin Li
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, USA
| | - Hongyang Wang
- Department of Otolaryngology and Program in Neuroscience, Harvard Medical School and Eaton Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts 02114, USA.,Department of Otolaryngology & Head Neck Surgery, Key Lab of Hearing Impairment Science of Ministry of Education, Key Lab of Hearing Impairment Prevention and Treatment of Beijing City, Chinese PLA Medical School, Beijing, China
| | - Shiming Yang
- Department of Otolaryngology & Head Neck Surgery, Key Lab of Hearing Impairment Science of Ministry of Education, Key Lab of Hearing Impairment Prevention and Treatment of Beijing City, Chinese PLA Medical School, Beijing, China
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, USA
| | - Daniel B Polley
- Department of Otolaryngology and Program in Neuroscience, Harvard Medical School and Eaton Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts 02114, USA
| | - M Charles Liberman
- Department of Otolaryngology and Program in Neuroscience, Harvard Medical School and Eaton Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts 02114, USA
| | - Wei-Jia Kong
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jeffrey R Holt
- Departments of Otolaryngology and Neurology, F.M. Kirby Neurobiology Center Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Zheng-Yi Chen
- Department of Otolaryngology and Program in Neuroscience, Harvard Medical School and Eaton Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts 02114, USA
| | - David R Liu
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, USA.,Howard Hughes Medical Institute, Harvard University, Cambridge, Massachusetts 02138, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02141, USA
| |
Collapse
|
25
|
Hearing sensitivity differs between zebrafish lines used in auditory research. Hear Res 2016; 341:220-231. [PMID: 27646864 DOI: 10.1016/j.heares.2016.09.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/23/2016] [Accepted: 09/15/2016] [Indexed: 11/20/2022]
Abstract
Zebrafish are increasingly used in auditory studies, in part due to the development of several transgenic lines that express hair cell-specific fluorescent proteins. However, it is largely unknown how transgene expression influences auditory phenotype. We previously observed reduced auditory sensitivity in adult Brn3c:mGFP transgenic zebrafish, which express membrane-bound green fluorescent protein (GFP) in sensory hair cells. Here, we examine the auditory sensitivity of zebrafish from multiple transgenic and background strains. We recorded auditory evoked potentials in adult animals and observed significantly higher auditory thresholds in three lines that express hair cell-specific GFP. There was no obvious correlation between hair cell density and auditory thresholds, suggesting that reduced sensitivity was not due to a reduction in hair cell density. FM1-43 uptake was reduced in Brn3c:mGFP fish but not in other lines, suggesting that a mechanotransduction defect may be responsible for the auditory phenotype in Brn3c animals, but that alternate mechanisms underlie the increased AEP thresholds in other lines. We found reduced prepulse inhibition (a measure of auditory-evoked behavior) in larval Brn3c animals, suggesting that auditory defects develop early in this line. We also found significant differences in auditory sensitivity between adults of different background strains, akin to strain differences observed in mouse models of auditory function. Our results suggest that researchers should exercise caution when selecting an appropriate zebrafish transgenic or background strain for auditory studies.
Collapse
|
26
|
Shu Y, Tao Y, Wang Z, Tang Y, Li H, Dai P, Gao G, Chen ZY. Identification of Adeno-Associated Viral Vectors That Target Neonatal and Adult Mammalian Inner Ear Cell Subtypes. Hum Gene Ther 2016; 27:687-99. [PMID: 27342665 DOI: 10.1089/hum.2016.053] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The mammalian inner ear consists of diverse cell types with important functions. Gene mutations in these diverse cell types have been found to underlie different forms of genetic hearing loss. Targeting these mutations for gene therapy development represents a future therapeutic strategy to treat hearing loss. Adeno-associated viral (AAV) vectors have become the vector of choice for gene delivery in animal models in vivo. To identify AAV vectors that target inner ear cell subtypes, we systemically screened 12 AAV vectors with different serotypes (AAV1, 2, 5, 6, 6.2, 7, 8, 9, rh.8, rh.10, rh.39, and rh.43) that carry a reporter gene GFP in neonatal and adult mice by microinjection in vivo. We found that most AAVs infect both neonatal and adult inner ear, with different specificities and expression levels. The inner ear cochlear sensory epithelial region, which includes auditory hair cells and supporting cells, is most frequently targeted for gene delivery. Expression of the transgene is sustained, and neonatal inner ear delivery does not adversely affect hearing. Adult inner ear injection of AAV has a similar infection pattern as the younger inner ear, with the exception that outer hair cell death caused by the injection procedure can lead to hearing loss. In the adult, more so than in the neonatal mice, cell types infected and efficiency of infection are correlated with the site of injection. Most infected cells survive in neonatal and adult inner ears. The study adds to the list of AAV vectors that transduce the mammalian inner ear efficiently, providing the tools that are important to study inner ear gene function and for the development of gene therapy to treat hearing loss.
Collapse
Affiliation(s)
- Yilai Shu
- 1 Department of Otolaryngology, Harvard Medical School and Eaton-Peabody Laboratories, Massachusetts Eye & Ear Infirmary, Boston, Massachusetts.,2 Department of Otolaryngology-Head and Neck Surgery, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China .,3 Key Laboratory of Hearing Medicine, National Health and Family Planning Commission, Shanghai, China
| | - Yong Tao
- 1 Department of Otolaryngology, Harvard Medical School and Eaton-Peabody Laboratories, Massachusetts Eye & Ear Infirmary, Boston, Massachusetts
| | - Zhengmin Wang
- 2 Department of Otolaryngology-Head and Neck Surgery, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China .,3 Key Laboratory of Hearing Medicine, National Health and Family Planning Commission, Shanghai, China
| | - Yong Tang
- 1 Department of Otolaryngology, Harvard Medical School and Eaton-Peabody Laboratories, Massachusetts Eye & Ear Infirmary, Boston, Massachusetts.,4 Department of Ear, Nose and Throat, People's Hospital of Jilin Province, Changchun, Jilin Province, China
| | - Huawei Li
- 2 Department of Otolaryngology-Head and Neck Surgery, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China .,3 Key Laboratory of Hearing Medicine, National Health and Family Planning Commission, Shanghai, China
| | - Pu Dai
- 5 Department of Otolaryngology, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Guangping Gao
- 6 Horae Gene Therapy Center and Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts.,7 State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Chengdu, Sichuan, China
| | - Zheng-Yi Chen
- 1 Department of Otolaryngology, Harvard Medical School and Eaton-Peabody Laboratories, Massachusetts Eye & Ear Infirmary, Boston, Massachusetts
| |
Collapse
|
27
|
Fergus DJ, Feng NY, Bass AH. Gene expression underlying enhanced, steroid-dependent auditory sensitivity of hair cell epithelium in a vocal fish. BMC Genomics 2015; 16:782. [PMID: 26466782 PMCID: PMC4607102 DOI: 10.1186/s12864-015-1940-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 09/19/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Successful animal communication depends on a receiver's ability to detect a sender's signal. Exemplars of adaptive sender-receiver coupling include acoustic communication, often important in the context of seasonal reproduction. During the reproductive summer season, both male and female midshipman fish (Porichthys notatus) exhibit similar increases in the steroid-dependent frequency sensitivity of the saccule, the main auditory division of the inner ear. This form of auditory plasticity enhances detection of the higher frequency components of the multi-harmonic, long-duration advertisement calls produced repetitively by males during summer nights of peak vocal and spawning activity. The molecular basis of this seasonal auditory plasticity has not been fully resolved. Here, we utilize an unbiased transcriptomic RNA sequencing approach to identify differentially expressed transcripts within the saccule's hair cell epithelium of reproductive summer and non-reproductive winter fish. RESULTS We assembled 74,027 unique transcripts from our saccular epithelial sequence reads. Of these, 6.4 % and 3.0 % were upregulated in the reproductive and non-reproductive saccular epithelium, respectively. Gene ontology (GO) term enrichment analyses of the differentially expressed transcripts showed that the reproductive saccular epithelium was transcriptionally, translationally, and metabolically more active than the non-reproductive epithelium. Furthermore, the expression of a specific suite of candidate genes, including ion channels and components of steroid-signaling pathways, was upregulated in the reproductive compared to the non-reproductive saccular epithelium. We found reported auditory functions for 14 candidate genes upregulated in the reproductive midshipman saccular epithelium, 8 of which are enriched in mouse hair cells, validating their hair cell-specific functions across vertebrates. CONCLUSIONS We identified a suite of differentially expressed genes belonging to neurotransmission and steroid-signaling pathways, consistent with previous work showing the importance of these characters in regulating hair cell auditory sensitivity in midshipman fish and, more broadly, vertebrates. The results were also consistent with auditory hair cells being generally more physiologically active when animals are in a reproductive state, a time of enhanced sensory-motor coupling between the auditory periphery and the upper harmonics of vocalizations. Together with several new candidate genes, our results identify discrete patterns of gene expression linked to frequency- and steroid-dependent plasticity of hair cell auditory sensitivity.
Collapse
Affiliation(s)
- Daniel J Fergus
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY, 14853, USA. .,Current Address: North Carolina Museum of Natural Sciences, Genomics and Microbiology, Raleigh, NC, 27601, USA.
| | - Ni Y Feng
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY, 14853, USA.
| | - Andrew H Bass
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
28
|
Abstract
Hair cells of the inner ear are essential for hearing and balance. As a consequence, pathogenic variants in genes specifically expressed in hair cells often cause hereditary deafness. Hair cells are few in number and not easily isolated from the adjacent supporting cells, so the biochemistry and molecular biology of hair cells can be difficult to study. To study gene expression in hair cells, we developed a protocol for hair cell isolation by FACS. With nearly pure hair cells and surrounding cells, from cochlea and utricle and from E16 to P7, we performed a comprehensive cell type-specific RNA-Seq study of gene expression during mouse inner ear development. Expression profiling revealed new hair cell genes with distinct expression patterns: some are specific for vestibular hair cells, others for cochlear hair cells, and some are expressed just before or after maturation of mechanosensitivity. We found that many of the known hereditary deafness genes are much more highly expressed in hair cells than surrounding cells, suggesting that genes preferentially expressed in hair cells are good candidates for unknown deafness genes.
Collapse
|
29
|
Abstract
The inability of mammals to regenerate auditory hair cells creates a pressing need to understand the means of enhancing hair cell survival following insult or injury. Hair cells are easily damaged by noise exposure, by ototoxic medications and as a consequence of aging processes, all of which lead to progressive and permanent hearing impairment as hair cells are lost. Significant efforts have been invested in designing strategies to prevent this damage from occurring since permanent hearing loss has a profound impact on communication and quality of life for patients. In this mini-review, we discuss recent progress in the use of antioxidants, anti-inflammatories and apoptosis inhibitors to enhance hair cell survival. We conclude by clarifying the distinction between protection and rescue strategies and by highlighting important areas of future research.
Collapse
|
30
|
Chiu KW, Lu LS, Wu CK. High pressure air jet in the endoscopic preparation room: risk of noise exposure on occupational health. BIOMED RESEARCH INTERNATIONAL 2015; 2015:610582. [PMID: 25710009 PMCID: PMC4331315 DOI: 10.1155/2015/610582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Accepted: 10/09/2014] [Indexed: 01/10/2023]
Abstract
After high-level disinfection of gastrointestinal endoscopes, they are hung to dry in order to prevent residual water droplets impact on patient health. To allow for quick drying and clinical reuse, some endoscopic units use a high pressure air jet (HPAJ) to remove the water droplets on the endoscopes. The purpose of this study was to evaluate the excessive noise exposure with the use of HPAJ in endoscopic preparation room and to investigate the risk to occupational health. Noise assessment was taken during 7 automatic endoscopic reprocessors (AERs) and combined with/without HPAJ use over an 8-hour time-weighted average (TWA). Analytical procedures of the NIOSH and the ISO for noise-induced hearing loss were estimated to develop analytic models. The peak of the noise spectrum of combined HPAJ and 7 AERs was significantly higher than that of the 7 AERs alone (108.3 ± 1.36 versus 69.3 ± 3.93 dBA, P < 0.0001). The risk of hearing loss (HL > 2.5 dB) was 2.15% at 90 dBA, 11.6% at 95 dBA, and 51.3% at 100 dBA. The odds ratio was 49.1 (95% CI: 11.9 to 203.6). The noise generated by the HPAJ to work over TWA seriously affected the occupational health and safety of those working in an endoscopic preparation room.
Collapse
Affiliation(s)
- King-Wah Chiu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, 123 Ta-Pei Road, Niao-Sung, Kaohsiung 833, Taiwan
| | - Lung-Sheng Lu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, 123 Ta-Pei Road, Niao-Sung, Kaohsiung 833, Taiwan
| | - Cheng-Kun Wu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, 123 Ta-Pei Road, Niao-Sung, Kaohsiung 833, Taiwan
| |
Collapse
|