1
|
Qiu Y, Xie L, Wang X, Xu K, Bai X, Chen S, Sun Y. Abnormal Innervation, Demyelination, and Degeneration of Spiral Ganglion Neurons as Well as Disruption of Heminodes are Involved in the Onset of Deafness in Cx26 Null Mice. Neurosci Bull 2024; 40:1093-1103. [PMID: 38311706 PMCID: PMC11306449 DOI: 10.1007/s12264-023-01167-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 09/14/2023] [Indexed: 02/06/2024] Open
Abstract
GJB2 gene mutations are the most common causes of autosomal recessive non-syndromic hereditary deafness. For individuals suffering from severe to profound GJB2-related deafness, cochlear implants have emerged as the sole remedy for auditory improvement. Some previous studies have highlighted the crucial role of preserving cochlear neural components in achieving favorable outcomes after cochlear implantation. Thus, we generated a conditional knockout mouse model (Cx26-CKO) in which Cx26 was completely deleted in the cochlear supporting cells driven by the Sox2 promoter. The Cx26-CKO mice showed severe hearing loss and massive loss of hair cells and Deiter's cells, which represented the extreme form of human deafness caused by GJB2 gene mutations. In addition, multiple pathological changes in the peripheral auditory nervous system were found, including abnormal innervation, demyelination, and degeneration of spiral ganglion neurons as well as disruption of heminodes in Cx26-CKO mice. These findings provide invaluable insights into the deafness mechanism and the treatment for severe deafness in Cx26-null mice.
Collapse
Affiliation(s)
- Yue Qiu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Le Xie
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaohui Wang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kai Xu
- Department of Otolaryngology, Nanchang University, Nanchang, 330006, China
| | - Xue Bai
- Department of Otolaryngology, Nanchang University, Nanchang, 330006, China
| | - Sen Chen
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Yu Sun
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
2
|
Huey EL, Turecek J, Delisle MM, Mazor O, Romero GE, Dua M, Sarafis ZK, Hobble A, Booth KT, Goodrich LV, Corey DP, Ginty DD. The auditory midbrain mediates tactile vibration sensing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.584077. [PMID: 38496510 PMCID: PMC10942453 DOI: 10.1101/2024.03.08.584077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Vibrations are ubiquitous in nature, shaping behavior across the animal kingdom. For mammals, mechanical vibrations acting on the body are detected by mechanoreceptors of the skin and deep tissues and processed by the somatosensory system, while sound waves traveling through air are captured by the cochlea and encoded in the auditory system. Here, we report that mechanical vibrations detected by the body's Pacinian corpuscle neurons, which are unique in their ability to entrain to high frequency (40-1000 Hz) environmental vibrations, are prominently encoded by neurons in the lateral cortex of the inferior colliculus (LCIC) of the midbrain. Remarkably, most LCIC neurons receive convergent Pacinian and auditory input and respond more strongly to coincident tactile-auditory stimulation than to either modality alone. Moreover, the LCIC is required for behavioral responses to high frequency mechanical vibrations. Thus, environmental vibrations captured by Pacinian corpuscles are encoded in the auditory midbrain to mediate behavior.
Collapse
|
3
|
Schvartz-Leyzac KC, Colesa DJ, Swiderski DL, Raphael Y, Pfingst BE. Cochlear Health and Cochlear-implant Function. J Assoc Res Otolaryngol 2023; 24:5-29. [PMID: 36600147 PMCID: PMC9971430 DOI: 10.1007/s10162-022-00882-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 11/24/2022] [Indexed: 01/06/2023] Open
Abstract
The cochlear implant (CI) is widely considered to be one of the most innovative and successful neuroprosthetic treatments developed to date. Although outcomes vary, CIs are able to effectively improve hearing in nearly all recipients and can substantially improve speech understanding and quality of life for patients with significant hearing loss. A wealth of research has focused on underlying factors that contribute to success with a CI, and recent evidence suggests that the overall health of the cochlea could potentially play a larger role than previously recognized. This article defines and reviews attributes of cochlear health and describes procedures to evaluate cochlear health in humans and animal models in order to examine the effects of cochlear health on performance with a CI. Lastly, we describe how future biologic approaches can be used to preserve and/or enhance cochlear health in order to maximize performance for individual CI recipients.
Collapse
Affiliation(s)
- Kara C Schvartz-Leyzac
- Department of Otolaryngology-Head and Neck Surgery, Michigan Medicine, Kresge Hearing Research Institute, University of Michigan, 1150 Medical Center Drive, Ann Arbor, MI, 48109-5616, USA
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, 135 Rutledge Ave, Charleston, SC, 29425, USA
| | - Deborah J Colesa
- Department of Otolaryngology-Head and Neck Surgery, Michigan Medicine, Kresge Hearing Research Institute, University of Michigan, 1150 Medical Center Drive, Ann Arbor, MI, 48109-5616, USA
| | - Donald L Swiderski
- Department of Otolaryngology-Head and Neck Surgery, Michigan Medicine, Kresge Hearing Research Institute, University of Michigan, 1150 Medical Center Drive, Ann Arbor, MI, 48109-5616, USA
| | - Yehoash Raphael
- Department of Otolaryngology-Head and Neck Surgery, Michigan Medicine, Kresge Hearing Research Institute, University of Michigan, 1150 Medical Center Drive, Ann Arbor, MI, 48109-5616, USA
| | - Bryan E Pfingst
- Department of Otolaryngology-Head and Neck Surgery, Michigan Medicine, Kresge Hearing Research Institute, University of Michigan, 1150 Medical Center Drive, Ann Arbor, MI, 48109-5616, USA.
| |
Collapse
|
4
|
Connexin Mutations and Hereditary Diseases. Int J Mol Sci 2022; 23:ijms23084255. [PMID: 35457072 PMCID: PMC9027513 DOI: 10.3390/ijms23084255] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/04/2022] [Accepted: 04/09/2022] [Indexed: 02/01/2023] Open
Abstract
Inherited diseases caused by connexin mutations are found in multiple organs and include hereditary deafness, congenital cataract, congenital heart diseases, hereditary skin diseases, and X-linked Charcot–Marie–Tooth disease (CMT1X). A large number of knockout and knock-in animal models have been used to study the pathology and pathogenesis of diseases of different organs. Because the structures of different connexins are highly homologous and the functions of gap junctions formed by these connexins are similar, connexin-related hereditary diseases may share the same pathogenic mechanism. Here, we analyze the similarities and differences of the pathology and pathogenesis in animal models and find that connexin mutations in gap junction genes expressed in the ear, eye, heart, skin, and peripheral nerves can affect cellular proliferation and differentiation of corresponding organs. Additionally, some dominant mutations (e.g., Cx43 p.Gly60Ser, Cx32 p.Arg75Trp, Cx32 p.Asn175Asp, and Cx32 p.Arg142Trp) are identified as gain-of-function variants in vivo, which may play a vital role in the onset of dominant inherited diseases. Specifically, patients with these dominant mutations receive no benefits from gene therapy. Finally, the complete loss of gap junctional function or altered channel function including permeability (ions, adenosine triphosphate (ATP), Inositol 1,4,5-trisphosphate (IP3), Ca2+, glucose, miRNA) and electric activity are also identified in vivo or in vitro.
Collapse
|
5
|
Zhang L, Chen S, Sun Y. Mechanism and Prevention of Spiral Ganglion Neuron Degeneration in the Cochlea. Front Cell Neurosci 2022; 15:814891. [PMID: 35069120 PMCID: PMC8766678 DOI: 10.3389/fncel.2021.814891] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 12/09/2021] [Indexed: 12/14/2022] Open
Abstract
Sensorineural hearing loss (SNHL) is one of the most prevalent sensory deficits in humans, and approximately 360 million people worldwide are affected. The current treatment option for severe to profound hearing loss is cochlear implantation (CI), but its treatment efficacy is related to the survival of spiral ganglion neurons (SGNs). SGNs are the primary sensory neurons, transmitting complex acoustic information from hair cells to second-order sensory neurons in the cochlear nucleus. In mammals, SGNs have very limited regeneration ability, and SGN loss causes irreversible hearing loss. In most cases of SNHL, SGN damage is the dominant pathogenesis, and it could be caused by noise exposure, ototoxic drugs, hereditary defects, presbycusis, etc. Tremendous efforts have been made to identify novel treatments to prevent or reverse the damage to SGNs, including gene therapy and stem cell therapy. This review summarizes the major causes and the corresponding mechanisms of SGN loss and the current protection strategies, especially gene therapy and stem cell therapy, to promote the development of new therapeutic methods.
Collapse
Affiliation(s)
- Li Zhang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sen Chen
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Sun
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Otorhinolaryngology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Tisi A, Rovers J, Vink HA, Ramekers D, Maccarone R, Versnel H. No Protective Effects of Hair Cells or Supporting Cells in Ototoxically Deafened Guinea Pigs upon Administration of BDNF. Brain Sci 2021; 12:2. [PMID: 35053747 PMCID: PMC8773526 DOI: 10.3390/brainsci12010002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 11/16/2022] Open
Abstract
We investigated whether treatment with brain-derived neurotrophic factor (BDNF), which is known to protect spiral ganglion cells (SGCs), could also protect hair cells (HCs) and supporting cells (SCs) in the organ of Corti of a guinea pig model of sensorineural hearing loss. Hearing loss was induced by administration of kanamycin/furosemide and two BDNF treatments were performed: (1) by gelatin sponge (BDNF-GS) with acute cochlear implantation (CI), and (2) through a mini-osmotic pump (BDNF-OP) with chronic CI. Outer HCs (OHCs), inner HCs (IHCs), Border, Phalangeal, Pillar, Deiters', and Hensen's cells were counted. The BDNF-GS cochleas had significantly fewer OHCs compared to the untreated ones, while the IHC and SC numbers did not differ between treated and untreated cochleas. The BDNF-OP group showed similar cell numbers to the untreated group. SGC packing density was not correlated with the total number of SCs for either BDNF group. Our data suggest that: (1) BDNF does not prevent cell death in the organ of Corti, and that the protection of SGCs could result from a direct targeting by BDNF; (2) BDNF might induce a different function/activity of the remaining cells in the organ of Corti (independently from cell number).
Collapse
Affiliation(s)
- Annamaria Tisi
- Department of Applied Clinical Sciences and Biotechnology, University of L′Aquila, 67100 L′Aquila, Italy; (A.T.); (R.M.)
| | - Jochebed Rovers
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Utrecht, Utrecht University, Room G.02.531, P.O. Box 85500, 3508 GA Utrecht, The Netherlands; (J.R.); (H.A.V.); (D.R.)
| | - Henk A. Vink
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Utrecht, Utrecht University, Room G.02.531, P.O. Box 85500, 3508 GA Utrecht, The Netherlands; (J.R.); (H.A.V.); (D.R.)
- UMC Utrecht Brain Center, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands
| | - Dyan Ramekers
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Utrecht, Utrecht University, Room G.02.531, P.O. Box 85500, 3508 GA Utrecht, The Netherlands; (J.R.); (H.A.V.); (D.R.)
- UMC Utrecht Brain Center, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands
| | - Rita Maccarone
- Department of Applied Clinical Sciences and Biotechnology, University of L′Aquila, 67100 L′Aquila, Italy; (A.T.); (R.M.)
| | - Huib Versnel
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Utrecht, Utrecht University, Room G.02.531, P.O. Box 85500, 3508 GA Utrecht, The Netherlands; (J.R.); (H.A.V.); (D.R.)
- UMC Utrecht Brain Center, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
7
|
Guo J, Ma X, Skidmore JM, Cimerman J, Prieskorn DM, Beyer LA, Swiderski DL, Dolan DF, Martin DM, Raphael Y. GJB2 gene therapy and conditional deletion reveal developmental stage-dependent effects on inner ear structure and function. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 23:319-333. [PMID: 34729379 PMCID: PMC8531464 DOI: 10.1016/j.omtm.2021.09.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/24/2021] [Indexed: 11/15/2022]
Abstract
Pathogenic variants in GJB2, the gene encoding connexin 26, are the most common cause of autosomal-recessive hereditary deafness. Despite this high prevalence, pathogenic mechanisms leading to GJB2-related deafness are not well understood, and cures are absent. Humans with GJB2-related deafness retain at least some auditory hair cells and neurons, and their deafness is usually stable. In contrast, mice with conditional loss of Gjb2 in supporting cells exhibit extensive loss of hair cells and neurons and rapidly progress to profound deafness, precluding the application of therapies that require intact cochlear cells. In an attempt to design a less severe Gjb2 animal model, we generated mice with inducible Sox10iCre ERT2 -mediated loss of Gjb2. Tamoxifen injection led to reduced connexin 26 expression and impaired function, but cochlear hair cells and neurons survived for 2 months, allowing phenotypic rescue attempts within this time. AAV-mediated gene transfer of GJB2 in mature mutant ears did not demonstrate threshold improvement and in some animals exacerbated hearing loss and resulted in hair cell loss. We conclude that Sox10iCre ERT2 ;Gjb2 flox/flox mice are valuable for studying the biology of connexin 26 in the cochlea. In particular, these mice may be useful for evaluating gene therapy vectors and development of therapies for GJB2-related deafness.
Collapse
Affiliation(s)
- Jingying Guo
- Kresge Hearing Research Institute, Otolaryngology, Head and Neck Surgery, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA.,Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaobo Ma
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jennifer M Skidmore
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Jelka Cimerman
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Diane M Prieskorn
- Kresge Hearing Research Institute, Otolaryngology, Head and Neck Surgery, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Lisa A Beyer
- Kresge Hearing Research Institute, Otolaryngology, Head and Neck Surgery, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Donald L Swiderski
- Kresge Hearing Research Institute, Otolaryngology, Head and Neck Surgery, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - David F Dolan
- Kresge Hearing Research Institute, Otolaryngology, Head and Neck Surgery, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Donna M Martin
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA.,Department of Human Genetics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Yehoash Raphael
- Kresge Hearing Research Institute, Otolaryngology, Head and Neck Surgery, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
8
|
Szeto B, Valentini C, Aksit A, Werth EG, Goeta S, Brown LM, Olson ES, Kysar JW, Lalwani AK. Impact of Systemic versus Intratympanic Dexamethasone Administration on the Perilymph Proteome. J Proteome Res 2021; 20:4001-4009. [PMID: 34291951 DOI: 10.1021/acs.jproteome.1c00322] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Glucocorticoids are the first-line treatment for sensorineural hearing loss, but little is known about the mechanism of their protective effect or the impact of route of administration. The recent development of hollow microneedles enables safe and reliable sampling of perilymph for proteomic analysis. Using these microneedles, we investigate the effect of intratympanic (IT) versus intraperitoneal (IP) dexamethasone administration on guinea pig perilymph proteome. Guinea pigs were treated with IT dexamethasone (n = 6), IP dexamethasone (n = 8), or untreated for control (n = 8) 6 h prior to aspiration. The round window membrane (RWM) was accessed via a postauricular approach, and hollow microneedles were used to perforate the RWM and aspirate 1 μL of perilymph. Perilymph samples were analyzed by liquid chromatography-mass spectrometry-based label-free quantitative proteomics. Mass spectrometry raw data files have been deposited in an international public repository (MassIVE proteomics repository at https://massive.ucsd.edu/) under data set # MSV000086887. In the 22 samples of perilymph analyzed, 632 proteins were detected, including the inner ear protein cochlin, a perilymph marker. Of these, 14 proteins were modulated by IP, and three proteins were modulated by IT dexamethasone. In both IP and IT dexamethasone groups, VGF nerve growth factor inducible was significantly upregulated compared to control. The remaining adjusted proteins modulate neurons, inflammation, or protein synthesis. Proteome analysis facilitated by the use of hollow microneedles shows that route of dexamethasone administration impacts changes seen in perilymph proteome. Compared to IT administration, the IP route was associated with greater changes in protein expression, including proteins involved in neuroprotection, inflammatory pathway, and protein synthesis. Our findings show that microneedles can mediate safe and effective intracochlear sampling and hold promise for inner ear diagnostics.
Collapse
Affiliation(s)
- Betsy Szeto
- Department of Otolaryngology-Head and Neck Surgery, Columbia University Vagelos College of Physicians and Surgeons, New York, New York 10032, United States
| | - Chris Valentini
- Department of Otolaryngology-Head and Neck Surgery, Columbia University Vagelos College of Physicians and Surgeons, New York, New York 10032, United States
| | - Aykut Aksit
- Department of Mechanical Engineering, Columbia University, New York, New York 10027, United States
| | - Emily G Werth
- Quantitative Proteomics and Metabolomics Center, Department of Biological Sciences, Columbia University, New York, New York 10027, United States
| | - Shahar Goeta
- Quantitative Proteomics and Metabolomics Center, Department of Biological Sciences, Columbia University, New York, New York 10027, United States
| | - Lewis M Brown
- Quantitative Proteomics and Metabolomics Center, Department of Biological Sciences, Columbia University, New York, New York 10027, United States
| | - Elizabeth S Olson
- Department of Otolaryngology-Head and Neck Surgery, Columbia University Vagelos College of Physicians and Surgeons, New York, New York 10032, United States.,Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| | - Jeffrey W Kysar
- Department of Otolaryngology-Head and Neck Surgery, Columbia University Vagelos College of Physicians and Surgeons, New York, New York 10032, United States.,Department of Mechanical Engineering, Columbia University, New York, New York 10027, United States
| | - Anil K Lalwani
- Department of Otolaryngology-Head and Neck Surgery, Columbia University Vagelos College of Physicians and Surgeons, New York, New York 10032, United States.,Department of Mechanical Engineering, Columbia University, New York, New York 10027, United States
| |
Collapse
|
9
|
Bankoti K, Generotti C, Hwa T, Wang L, O'Malley BW, Li D. Advances and challenges in adeno-associated viral inner-ear gene therapy for sensorineural hearing loss. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:209-236. [PMID: 33850952 PMCID: PMC8010215 DOI: 10.1016/j.omtm.2021.03.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
There is growing attention and effort focused on treating the root cause of sensorineural hearing loss rather than managing associated secondary characteristic features. With recent substantial advances in understanding sensorineural hearing-loss mechanisms, gene delivery has emerged as a promising strategy for the biological treatment of hearing loss associated with genetic dysfunction. There are several successful and promising proof-of-principle examples of transgene deliveries in animal models; however, there remains substantial further progress to be made in these avenues before realizing their clinical application in humans. Herein, we review different aspects of development, ongoing preclinical studies, and challenges to the clinical transition of transgene delivery of the inner ear toward the restoration of lost auditory and vestibular function.
Collapse
Affiliation(s)
- Kamakshi Bankoti
- Department of Otorhinolaryngology, Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Charles Generotti
- Department of Otorhinolaryngology, Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tiffany Hwa
- Department of Otorhinolaryngology, Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lili Wang
- Department of Medicine, Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bert W O'Malley
- Department of Otorhinolaryngology, Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daqing Li
- Department of Otorhinolaryngology, Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
10
|
Ding N, Lee S, Lieber-Kotz M, Yang J, Gao X. Advances in genome editing for genetic hearing loss. Adv Drug Deliv Rev 2021; 168:118-133. [PMID: 32387678 DOI: 10.1016/j.addr.2020.05.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 04/27/2020] [Accepted: 05/04/2020] [Indexed: 02/07/2023]
Abstract
According to the World Health Organization, hearing loss affects over 466 million people worldwide and is the most common human sensory impairment. It is estimated that genetic factors contribute to the causation of approximately 50% of congenital hearing loss. Yet, curative approaches to reversing or preventing genetic hearing impairment are still limited. The clustered regularly interspaced short palindromic repeats-associated protein 9 (CRISPR-Cas9) systems enable programmable and targeted gene editing in highly versatile manners and offer new gene therapy strategies for genetic hearing loss. Here, we summarize the most common deafness-associated genes, illustrate recent strategies undertaken by using CRISPR-Cas9 systems for targeted gene editing and further compare the CRISPR strategies to non-CRISPR gene therapies. We also examine the merits of different vehicles and delivery forms of genome editing agents. Lastly, we describe the development of animal models that could facilitate the eventual clinical applications of the CRISPR technology to the treatment of genetic hearing diseases.
Collapse
|
11
|
He L, Guo JY, Liu K, Wang GP, Gong SS. Research progress on flat epithelium of the inner ear. Physiol Res 2020; 69:775-785. [PMID: 32901490 DOI: 10.33549/physiolres.934447] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Sensorineural hearing loss and vertigo, resulting from lesions in the sensory epithelium of the inner ear, have a high incidence worldwide. The sensory epithelium of the inner ear may exhibit extreme degeneration and is transformed to flat epithelium (FE) in humans and mice with profound sensorineural hearing loss and/or vertigo. Various factors, including ototoxic drugs, noise exposure, aging, and genetic defects, can induce FE. Both hair cells and supporting cells are severely damaged in FE, and the normal cytoarchitecture of the sensory epithelium is replaced by a monolayer of very thin, flat cells of irregular contour. The pathophysiologic mechanism of FE is unclear but involves robust cell division. The cellular origin of flat cells in FE is heterogeneous; they may be transformed from supporting cells that have lost some features of supporting cells (dedifferentiation) or may have migrated from the flanking region. The epithelial-mesenchymal transition may play an important role in this process. The treatment of FE is challenging given the severe degeneration and loss of both hair cells and supporting cells. Cochlear implant or vestibular prosthesis implantation, gene therapy, and stem cell therapy show promise for the treatment of FE, although many challenges remain to be overcome.
Collapse
Affiliation(s)
- L He
- Department of Otolaryngology-Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China. ,
| | | | | | | | | |
Collapse
|
12
|
Murillo-Cuesta S, Artuch R, Asensio F, de la Villa P, Dierssen M, Enríquez JA, Fillat C, Fourcade S, Ibáñez B, Montoliu L, Oliver E, Pujol A, Salido E, Vallejo M, Varela-Nieto I. The Value of Mouse Models of Rare Diseases: A Spanish Experience. Front Genet 2020; 11:583932. [PMID: 33173540 PMCID: PMC7591746 DOI: 10.3389/fgene.2020.583932] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/14/2020] [Indexed: 11/13/2022] Open
Abstract
Animal models are invaluable for biomedical research, especially in the context of rare diseases, which have a very low prevalence and are often complex. Concretely mouse models provide key information on rare disease mechanisms and therapeutic strategies that cannot be obtained by using only alternative methods, and greatly contribute to accelerate the development of new therapeutic options for rare diseases. Despite this, the use of experimental animals remains controversial. The combination of respectful management, ethical laws and transparency regarding animal experimentation contributes to improve society’s opinion about biomedical research and positively impacts on research quality, which eventually also benefits patients. Here we present examples of current advances in preclinical research in rare diseases using mouse models, together with our perspective on future directions and challenges.
Collapse
Affiliation(s)
- Silvia Murillo-Cuesta
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.,Instituto de Investigaciones Biomédicas Alberto Sols (IIBM), Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid, Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Rafael Artuch
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.,Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain
| | - Fernando Asensio
- Gregorio Marañón Institute for Health Research (IISGM), Madrid, Spain
| | - Pedro de la Villa
- Faculty of Medicine, University of Alcalá (UAH), Alcalá de Henares, Spain
| | - Mara Dierssen
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.,Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Jose Antonio Enríquez
- Spanish National Center for Cardiovascular Research (CNIC), Institute of Health Carlos III, Madrid, Spain.,Biomedical Research Networking Center on Frailty and Healthy Ageing (CIBERFES), Institute of Health Carlos III, Madrid, Spain
| | - Cristina Fillat
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Stéphane Fourcade
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Borja Ibáñez
- Spanish National Center for Cardiovascular Research (CNIC), Institute of Health Carlos III, Madrid, Spain.,Biomedical Research Networking Center on Cardiovascular Diseases (CIBERCV), Institute of Health Carlos III, Madrid, Spain.,Cardiology Department, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS-FJD), Madrid, Spain
| | - Lluis Montoliu
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.,National Center for Biotechnology (CNB), Spanish National Research Council, Madrid, Spain
| | - Eduardo Oliver
- Spanish National Center for Cardiovascular Research (CNIC), Institute of Health Carlos III, Madrid, Spain.,Biomedical Research Networking Center on Cardiovascular Diseases (CIBERCV), Institute of Health Carlos III, Madrid, Spain
| | - Aurora Pujol
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Catalan Institution of Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Eduardo Salido
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.,Unidad de Investigación, Hospital Universitario de Canarias, Instituto de Tecnologías Biomédicas (ITB), La Laguna, Spain
| | - Mario Vallejo
- Instituto de Investigaciones Biomédicas Alberto Sols (IIBM), Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid, Madrid, Spain.,Biomedical Research Networking Center on Diabetes and Metabolic Diseases (CIBERDEM), Institute of Health Carlos III, Madrid, Spain
| | - Isabel Varela-Nieto
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.,Instituto de Investigaciones Biomédicas Alberto Sols (IIBM), Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid, Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| |
Collapse
|
13
|
Aaron KA, Kim GS, Cheng AG. Advances in Inner Ear Therapeutics for Hearing Loss in Children. CURRENT OTORHINOLARYNGOLOGY REPORTS 2020; 8:285-294. [PMID: 36090148 PMCID: PMC9455742 DOI: 10.1007/s40136-020-00300-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Purpose of review Hearing loss is a common congenital sensory disorder with various underlying causes. Here, we review and focus on genetic, infectious, and ototoxic causes and recent advances in inner ear therapeutics. Recent findings While hearing aids and cochlear implantation are the mainstay of treatment for pediatric hearing loss, novel biological therapeutics are being explored. Recent preclinical studies report positive results in viral-mediated gene transfer techniques and surgical approaches to the inner ear for genetic hearing loss. Novel pharmacologic agents, on the other hand, show promising results in reducing aminoglycoside and cisplatin ototoxicity. Clinical trials are underway to evaluate the efficacy of antivirals for cytomegalovirus-related hearing loss, and its pathogenesis and other potential therapeutics are currently under investigation. Summary Individualized therapies for genetic and infectious causes of sensorineural hearing loss in animal models as well as pediatric patients show promising results, with their potential efficacy being active areas of research.
Collapse
Affiliation(s)
- Ksenia A. Aaron
- Department of Otolaryngology – Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, 94305, USA
| | - Grace S. Kim
- Department of Otolaryngology – Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, 94305, USA
| | - Alan G. Cheng
- Department of Otolaryngology – Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, 94305, USA
| |
Collapse
|
14
|
Luo L, Ambrozkiewicz MC, Benseler F, Chen C, Dumontier E, Falkner S, Furlanis E, Gomez AM, Hoshina N, Huang WH, Hutchison MA, Itoh-Maruoka Y, Lavery LA, Li W, Maruo T, Motohashi J, Pai ELL, Pelkey KA, Pereira A, Philips T, Sinclair JL, Stogsdill JA, Traunmüller L, Wang J, Wortel J, You W, Abumaria N, Beier KT, Brose N, Burgess HA, Cepko CL, Cloutier JF, Eroglu C, Goebbels S, Kaeser PS, Kay JN, Lu W, Luo L, Mandai K, McBain CJ, Nave KA, Prado MA, Prado VF, Rothstein J, Rubenstein JL, Saher G, Sakimura K, Sanes JR, Scheiffele P, Takai Y, Umemori H, Verhage M, Yuzaki M, Zoghbi HY, Kawabe H, Craig AM. Optimizing Nervous System-Specific Gene Targeting with Cre Driver Lines: Prevalence of Germline Recombination and Influencing Factors. Neuron 2020; 106:37-65.e5. [PMID: 32027825 PMCID: PMC7377387 DOI: 10.1016/j.neuron.2020.01.008] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 11/12/2019] [Accepted: 01/10/2020] [Indexed: 12/17/2022]
Abstract
The Cre-loxP system is invaluable for spatial and temporal control of gene knockout, knockin, and reporter expression in the mouse nervous system. However, we report varying probabilities of unexpected germline recombination in distinct Cre driver lines designed for nervous system-specific recombination. Selective maternal or paternal germline recombination is showcased with sample Cre lines. Collated data reveal germline recombination in over half of 64 commonly used Cre driver lines, in most cases with a parental sex bias related to Cre expression in sperm or oocytes. Slight differences among Cre driver lines utilizing common transcriptional control elements affect germline recombination rates. Specific target loci demonstrated differential recombination; thus, reporters are not reliable proxies for another locus of interest. Similar principles apply to other recombinase systems and other genetically targeted organisms. We hereby draw attention to the prevalence of germline recombination and provide guidelines to inform future research for the neuroscience and broader molecular genetics communities.
Collapse
Affiliation(s)
- Lin Luo
- Djavad Mowafaghian Centre for Brain Health and Department of Psychiatry, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC V6T 2B5, Canada
| | - Mateusz C. Ambrozkiewicz
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Göttingen, Germany,Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany
| | - Fritz Benseler
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Göttingen, Germany
| | - Cui Chen
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Emilie Dumontier
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | | | | | | | - Naosuke Hoshina
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Wei-Hsiang Huang
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA,Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, The Research Institute of the McGill University Health Centre, Montreal, QC H3G 1A4, Canada
| | - Mary Anne Hutchison
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yu Itoh-Maruoka
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 1-5-6 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Laura A. Lavery
- Department of Molecular and Human Genetics, Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX 77003, USA,Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Wei Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Tomohiko Maruo
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 1-5-6 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan,Department of Biochemistry, Tokushima University Graduate School of Medical Sciences, 3-18-15, Kuramoto-cho, Tokushima 770-8503, Japan,Department of Biochemistry, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan
| | - Junko Motohashi
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Emily Ling-Lin Pai
- Nina Ireland Laboratory of Developmental Neurobiology, Department of Psychiatry, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA,Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kenneth A. Pelkey
- Section on Cellular and Synaptic Physiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ariane Pereira
- Department of Neurobiology and Department of Ophthalmology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Thomas Philips
- Department of Neurology and Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jennifer L. Sinclair
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Jeff A. Stogsdill
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02139, USA
| | | | - Jiexin Wang
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Joke Wortel
- Department of Functional Genomics and Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam and University Medical Center Amsterdam, de Boelelaan 1085, 1081 HV Amsterdam, the Netherlands
| | - Wenjia You
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA,Departments of Genetics, Harvard Medical School, Boston, MA 02115, USA,Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Nashat Abumaria
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China,Department of Laboratory Animal Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Kevin T. Beier
- Department of Physiology and Biophysics, Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA 92697, USA
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Göttingen, Germany
| | - Harold A. Burgess
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Constance L. Cepko
- Departments of Genetics, Harvard Medical School, Boston, MA 02115, USA,Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jean-François Cloutier
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Cagla Eroglu
- Department of Cell Biology, Department of Neurobiology, and Duke Institute for Brain Sciences, Regeneration Next Initiative, Duke University Medical Center, Durham, NC 27710, USA
| | - Sandra Goebbels
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Göttingen, Germany
| | - Pascal S. Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Jeremy N. Kay
- Department of Neurobiology and Department of Ophthalmology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Wei Lu
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Liqun Luo
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Kenji Mandai
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 1-5-6 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan,Department of Biochemistry, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan
| | - Chris J. McBain
- Section on Cellular and Synaptic Physiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Göttingen, Germany
| | - Marco A.M. Prado
- Robarts Research Institute, Department of Anatomy and Cell Biology, and Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5B7, Canada,Brain and Mind Institute, The University of Western Ontario, London, ON N6A 5B7, Canada
| | - Vania F. Prado
- Robarts Research Institute, Department of Anatomy and Cell Biology, and Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5B7, Canada,Brain and Mind Institute, The University of Western Ontario, London, ON N6A 5B7, Canada
| | - Jeffrey Rothstein
- Department of Neurology and Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - John L.R. Rubenstein
- Nina Ireland Laboratory of Developmental Neurobiology, Department of Psychiatry, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA,Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Gesine Saher
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Göttingen, Germany
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Joshua R. Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | | | - Yoshimi Takai
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 1-5-6 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Hisashi Umemori
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Matthijs Verhage
- Department of Functional Genomics and Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam and University Medical Center Amsterdam, de Boelelaan 1085, 1081 HV Amsterdam, the Netherlands
| | - Michisuke Yuzaki
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Huda Yahya Zoghbi
- Department of Molecular and Human Genetics, Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX 77003, USA,Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hiroshi Kawabe
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Göttingen, Germany; Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 1-5-6 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Department of Gerontology, Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, 2-2 Minatojima-minamimachi Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| | - Ann Marie Craig
- Djavad Mowafaghian Centre for Brain Health and Department of Psychiatry, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC V6T 2B5, Canada.
| |
Collapse
|
15
|
Van De Water TR. Historical Aspects of Gene Therapy and Stem Cell Therapy in the Treatment of Hearing and Balance Disorder. Anat Rec (Hoboken) 2020; 303:390-407. [DOI: 10.1002/ar.24332] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/18/2019] [Accepted: 11/22/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Thomas R. Van De Water
- Cochlear Implant Research Program, Department of Otolaryngology, University of Miami Ear InstituteUniversity of Miami Miller School of Medicine Miami Florida
| |
Collapse
|
16
|
Büning H, Schambach A, Morgan M, Rossi A, Wichova H, Staecker H, Warnecke A, Lenarz T. Challenges and advances in translating gene therapy for hearing disorders. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2020. [DOI: 10.1080/23808993.2020.1707077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Hildegard Büning
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research, Braunschweig, Germany
- REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael Morgan
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Axel Rossi
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Helena Wichova
- Department of Otolaryngology Head and Neck Surgery, University of Kansas School of Medicine, Kansas City, USA
| | - Hinrich Staecker
- Department of Otolaryngology Head and Neck Surgery, University of Kansas School of Medicine, Kansas City, USA
| | - Athanasia Warnecke
- Department of Otolaryngology, Hannover Medical School, 30625 Hannover, Germany
- Hearing4all Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Thomas Lenarz
- Department of Otolaryngology, Hannover Medical School, 30625 Hannover, Germany
- Hearing4all Cluster of Excellence, Hannover Medical School, Hannover, Germany
| |
Collapse
|
17
|
Szobota S, Mathur PD, Siegel S, Black K, Saragovi HU, Foster AC. BDNF, NT-3 and Trk receptor agonist monoclonal antibodies promote neuron survival, neurite extension, and synapse restoration in rat cochlea ex vivo models relevant for hidden hearing loss. PLoS One 2019; 14:e0224022. [PMID: 31671109 PMCID: PMC6822712 DOI: 10.1371/journal.pone.0224022] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 10/03/2019] [Indexed: 12/21/2022] Open
Abstract
Neurotrophins and their mimetics are potential treatments for hearing disorders because of their trophic effects on spiral ganglion neurons (SGNs) whose connections to hair cells may be compromised in many forms of hearing loss. Studies in noise or ototoxin-exposed animals have shown that local delivery of NT-3 or BDNF has beneficial effects on SGNs and hearing. We evaluated several TrkB or TrkC monoclonal antibody agonists and small molecules, along with BDNF and NT-3, in rat cochlea ex vivo models. The TrkB agonists BDNF and a monoclonal antibody, M3, had the greatest effects on SGN survival, neurite outgrowth and branching. In organotypic cochlear explants, BDNF and M3 enhanced synapse formation between SGNs and inner hair cells and restored these connections after excitotoxin-induced synaptopathy. Loss of these synapses has recently been implicated in hidden hearing loss, a condition characterized by difficulty hearing speech in the presence of background noise. The unique profile of M3 revealed here warrants further investigation, and the broad activity profile of BDNF observed underpins its continued development as a hearing loss therapeutic.
Collapse
Affiliation(s)
- Stephanie Szobota
- Otonomy, Inc., San Diego, California, United States of America
- * E-mail:
| | | | - Sairey Siegel
- Otonomy, Inc., San Diego, California, United States of America
| | | | - H. Uri Saragovi
- Lady Davis Institute-Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Alan C. Foster
- Otonomy, Inc., San Diego, California, United States of America
| |
Collapse
|
18
|
Bommakanti K, Iyer JS, Stankovic KM. Cochlear histopathology in human genetic hearing loss: State of the science and future prospects. Hear Res 2019; 382:107785. [PMID: 31493568 PMCID: PMC6778517 DOI: 10.1016/j.heares.2019.107785] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/30/2019] [Accepted: 08/15/2019] [Indexed: 12/22/2022]
Abstract
Sensorineural hearing loss (SNHL) is an extraordinarily common disability, affecting 466 million people across the globe. Half of these incidents are attributed to genetic mutations that disrupt the structure and function of the cochlea. The human cochlea's interior cannot be imaged or biopsied without damaging hearing; thus, everything known about the morphologic correlates of hereditary human deafness comes from histopathologic studies conducted in either cadaveric human temporal bone specimens or animal models of genetic deafness. The purpose of the present review is to a) summarize the findings from all published histopathologic studies conducted in human temporal bones with known SNHL-causing genetic mutations, and b) compare the reported phenotypes of human vs. mouse SNHL caused by the same genetic mutation. The fact that human temporal bone histopathologic analysis has been reported for only 22 of the nearly 200 identified deafness-causing genes suggests a great need for alternative and improved techniques for studying human hereditary deafness; in light of this, the present review concludes with a summary of promising future directions, specifically in the fields of high resolution cochlear imaging, intracochlear fluid biopsy, and gene therapy.
Collapse
Affiliation(s)
- Krishna Bommakanti
- Department of Otolaryngology, Harvard Medical School, Boston, MA, USA; Eaton Peabody Laboratories and Department of Otolaryngology, Massachusetts Eye and Ear, Boston, MA, USA; University of California San Diego School of Medicine, San Diego, CA, USA
| | - Janani S Iyer
- Department of Otolaryngology, Harvard Medical School, Boston, MA, USA; Eaton Peabody Laboratories and Department of Otolaryngology, Massachusetts Eye and Ear, Boston, MA, USA; Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, MA, USA; Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
| | - Konstantina M Stankovic
- Department of Otolaryngology, Harvard Medical School, Boston, MA, USA; Eaton Peabody Laboratories and Department of Otolaryngology, Massachusetts Eye and Ear, Boston, MA, USA; Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, MA, USA; Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
19
|
Naz S, Friedman TB. Growth factor and receptor malfunctions associated with human genetic deafness. Clin Genet 2019; 97:138-155. [PMID: 31506927 DOI: 10.1111/cge.13641] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/22/2019] [Accepted: 09/03/2019] [Indexed: 12/12/2022]
Abstract
A variety of different signaling pathways are necessary for development and maintenance of the human auditory system. Normal hearing allows for the detection of soft sounds within the frequency range of 20 to 20 000 Hz, but more importantly to perceive the human voice frequency band of 250 to 6000 Hz. Loss of hearing is common, and is a clinically heterogeneous disorder that can be caused by environmental factors such as exposure to loud noise, infections and ototoxic drugs. In addition, variants of hundreds of genes have been reported to disrupt processes required for hearing. Noncoding regulatory variants and variants of additional genes necessary for hearing remain to be discovered as many individuals with inherited deafness are without a genetic diagnosis, despite the advent of whole exome sequencing. Here, we discuss in detail some of these deafness-causing variants of genes encoding a ligand or its receptor. Spotlighted in this review are three growth factor-receptor-pairs EDN3/EDNRB, HGF/MET and JAG/NOTCH, which individually are necessary for normal hearing. We also offer our perspective on unanswered questions, future challenges and potential opportunities for treatments emerging from molecular genetic and mechanistic studies of deafness due to these causes.
Collapse
Affiliation(s)
- Sadaf Naz
- School of Biological Sciences, University of the Punjab, Lahore, Pakistan
| | - Thomas B Friedman
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
20
|
Advances in cochlear implantation for hereditary deafness caused by common mutations in deafness genes. JOURNAL OF BIO-X RESEARCH 2019. [DOI: 10.1097/jbr.0000000000000037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|
21
|
Celaya AM, Sánchez-Pérez I, Bermúdez-Muñoz JM, Rodríguez-de la Rosa L, Pintado-Berninches L, Perona R, Murillo-Cuesta S, Varela-Nieto I. Deficit of mitogen-activated protein kinase phosphatase 1 (DUSP1) accelerates progressive hearing loss. eLife 2019; 8:39159. [PMID: 30938680 PMCID: PMC6464786 DOI: 10.7554/elife.39159] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 04/01/2019] [Indexed: 12/12/2022] Open
Abstract
Mitogen-activated protein kinases (MAPK) such as p38 and the c-Jun N-terminal kinases (JNKs) are activated during the cellular response to stress signals. Their activity is regulated by the MAPK-phosphatase 1 (DUSP1), a key component of the anti-inflammatory response. Stress kinases are well-described elements of the response to otic injury and the otoprotective potential of JNK inhibitors is being tested in clinical trials. By contrast, there are no studies exploring the role of DUSP1 in hearing and hearing loss. Here we show that Dusp1 expression is age-regulated in the mouse cochlea. Dusp1 gene knock-out caused premature progressive hearing loss, as confirmed by auditory evoked responses in Dusp1-/- mice. Hearing loss correlated with cell death in hair cells, degeneration of spiral neurons and increased macrophage infiltration. Dusp1-/- mouse cochleae showed imbalanced redox status and dysregulated expression of cytokines. These data suggest that DUSP1 is essential for cochlear homeostasis in the response to stress during ageing.
Collapse
Affiliation(s)
- Adelaida M Celaya
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), CIBER, ISCIII, Madrid, Spain
| | - Isabel Sánchez-Pérez
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), CIBER, ISCIII, Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain.,Biochemistry Department, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain.,Biomedicine Unit UCLM-CSIC, Madrid, Spain
| | - Jose M Bermúdez-Muñoz
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), CIBER, ISCIII, Madrid, Spain
| | - Lourdes Rodríguez-de la Rosa
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), CIBER, ISCIII, Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Laura Pintado-Berninches
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Rosario Perona
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), CIBER, ISCIII, Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Silvia Murillo-Cuesta
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), CIBER, ISCIII, Madrid, Spain
| | - Isabel Varela-Nieto
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), CIBER, ISCIII, Madrid, Spain
| |
Collapse
|
22
|
Chang SY, Park YH, Carpena NT, Pham TT, Chung PS, Jung JY, Lee MY. Photobiomodulation promotes adenoviral gene transduction in auditory cells. Lasers Med Sci 2018; 34:367-375. [PMID: 30105484 DOI: 10.1007/s10103-018-2605-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 07/31/2018] [Indexed: 12/20/2022]
Abstract
Gene therapy is the delivery of a therapeutic gene into target cells to treat disorders by replacing disease-causing mutated genes with healthy ones. Gene therapy of the inner ear has been recently described, with applications for sensorineural hearing loss. However, gene delivery to the location of the inner ear, and thus efficacy of therapy, is challenging. Photobiomodulation (PBM) with a low-level laser has been suggested to have a therapeutic effect and has the potential to augment gene therapy. To investigate whether PBM improves the rate of adenovirus (Ad)-mediated viral delivery, we compared low-level laser therapy (LLLT) and non-LLLT HEI-OC1 cells treated with an Ad viral vector carrying green fluorescent protein (GFP). Cultured HEI-OC1 cells were divided into six groups: no treatment control, LLLT only, 1 μL Ad-GFP, 3 μL Ad-GFP, 1 μL Ad-GFP + LLLT, and 3 μL Ad-GFP + LLLT (LLLT: 808 nm at 15 mW for 15 min). Cells were irradiated twice: at 2 h and again at 24 h. A nonparametric Mann-Whitney U test was used to statistically analyze differences between the control and treatment groups. The viral inoculations used in this study did not change the amount of viable HEI-OC1 cells (N = 4-8). The 1 μL Ad-GFP + LLLT and 3 μL Ad-GFP + LLLT groups showed an increased density of GFP-positive cells compared to 1 μL and 3 μL Ad-GFP cells (N = 5-8, 1 μL: p = 0.0159; 3 μL: p = 0.0168,). The quantitative analysis of the epifluorescence of the 1 μL Ad-GFP + LLLT, and 3 μL Ad-GFP + LLLT groups revealed increased GFP expression/cell compared to 1 μL and 3 μL Ad-GFP cells (N = 6-15, 1 μL: p = 0.0082; 3 μL: p = 0.0012). The RT-qPCR results were consistent (N = 4-5, p = 0.0159). These findings suggest that PBM may enhance the gene delivery of Ad-mediated viral transduction, and the combination of the two may be a promising tool for gene therapy for sensorineural hearing loss.
Collapse
Affiliation(s)
- So-Young Chang
- Beckman Laser Institute Korea, College of Medicine, Dankook University, Cheonan, South Korea
| | - Yong-Ho Park
- Department of Otolaryngology-Head & Neck Surgery, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Nathaniel T Carpena
- Department of Otolaryngology-Head & Neck Surgery, College of Medicine, Dankook University, Cheonan, South Korea
| | - Tiffany T Pham
- Beckman Laser Institute, University of California Irvine, Irvine, CA, USA
| | - Phil-Sang Chung
- Beckman Laser Institute Korea, College of Medicine, Dankook University, Cheonan, South Korea.,Department of Otolaryngology-Head & Neck Surgery, College of Medicine, Dankook University, Cheonan, South Korea
| | - Jae Yun Jung
- Beckman Laser Institute Korea, College of Medicine, Dankook University, Cheonan, South Korea.,Department of Otolaryngology-Head & Neck Surgery, College of Medicine, Dankook University, Cheonan, South Korea
| | - Min Young Lee
- Beckman Laser Institute Korea, College of Medicine, Dankook University, Cheonan, South Korea. .,Department of Otolaryngology-Head & Neck Surgery, College of Medicine, Dankook University, Cheonan, South Korea.
| |
Collapse
|
23
|
Lee MY, Park YH. Potential of Gene and Cell Therapy for Inner Ear Hair Cells. BIOMED RESEARCH INTERNATIONAL 2018; 2018:8137614. [PMID: 30009175 PMCID: PMC6020521 DOI: 10.1155/2018/8137614] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 04/11/2018] [Accepted: 05/15/2018] [Indexed: 02/06/2023]
Abstract
Sensorineural hearing loss is caused by the loss of sensory hair cells (HCs) or a damaged afferent nerve pathway to the auditory cortex. The most common option for the treatment of sensorineural hearing loss is hearing rehabilitation using hearing devices. Various kinds of hearing devices are available but, despite recent advancements, their perceived sound quality does not mimic that of the "naïve" cochlea. Damage to crucial cochlear structures is mostly irreversible and results in permanent hearing loss. Cochlear HC regeneration has long been an important goal in the field of hearing research. However, it remains challenging because, thus far, no medical treatment has successfully regenerated cochlear HCs. Recent advances in genetic modulation and developmental techniques have led to novel approaches to generating HCs or protecting against HC loss, to preserve hearing. In this review, we present and review the current status of two different approaches to restoring or protecting hearing, gene therapy, including the newly introduced CRISPR/Cas9 genome editing, and stem cell therapy, and suggest the future direction.
Collapse
Affiliation(s)
- Min Yong Lee
- Department of Otorhinolaryngology and Head & Neck Surgery, Dankook University Hospital, Cheonan, Chungnam, Republic of Korea
| | - Yong-Ho Park
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Brain Research Institute, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
24
|
Kempfle JS, Nguyen K, Hamadani C, Koen N, Edge AS, Kashemirov BA, Jung DH, McKenna CE. Bisphosphonate-Linked TrkB Agonist: Cochlea-Targeted Delivery of a Neurotrophic Agent as a Strategy for the Treatment of Hearing Loss. Bioconjug Chem 2018; 29:1240-1250. [PMID: 29485861 DOI: 10.1021/acs.bioconjchem.8b00022] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Hearing loss affects more than two-thirds of the elderly population, and more than 17% of all adults in the U.S. Sensorineural hearing loss related to noise exposure or aging is associated with loss of inner ear sensory hair cells (HCs), cochlear spiral ganglion neurons (SGNs), and ribbon synapses between HCs and SGNs, stimulating intense interest in therapies to regenerate synaptic function. 7,8-Dihydroxyflavone (DHF) is a selective and potent agonist of tropomyosin receptor kinase B (TrkB) and protects the neuron from apoptosis. Despite evidence that TrkB agonists can promote survival of SGNs, local delivery of drugs such as DHF to the inner ear remains a challenge. We previously demonstrated in an animal model that a fluorescently labeled bisphosphonate, 6-FAM-Zol, administered to the round window membrane penetrated the membrane and diffused throughout the cochlea. Given their affinity for bone mineral, including cochlear bone, bisphosphonates offer an intriguing modality for targeted delivery of neurotrophic agents to the SGNs to promote survival, neurite outgrowth, and, potentially, regeneration of synapses between HCs and SGNs. The design and synthesis of a bisphosphonate conjugate of DHF (Ris-DHF) is presented, with a preliminary evaluation of its neurotrophic activity. Ris-DHF increases neurite outgrowth in vitro, maintains this ability after binding to hydroxyapatite, and regenerates synapses in kainic acid-damaged cochlear organ of Corti explants dissected in vitro with attached SGNs. The results suggest that bisphosphonate-TrkB agonist conjugates have promise as a novel approach to targeted delivery of drugs to treat sensorineural hearing loss.
Collapse
Affiliation(s)
- Judith S Kempfle
- Department of Otolaryngology and The Eaton-Peabody Laboratories , Massachusetts Eye and Ear Infirmary , Boston , Massachusetts 02114 , United States.,Department of Otology and Laryngology , Harvard Medical School , Boston , Massachusetts 02114 , United States.,Department of Otolaryngology , University of Tübingen Medical Center , Tübingen 72076 , Germany
| | - Kim Nguyen
- Department of Chemistry , University of Southern California , Los Angeles , California 90089-0744 , United States
| | - Christine Hamadani
- Department of Otolaryngology and The Eaton-Peabody Laboratories , Massachusetts Eye and Ear Infirmary , Boston , Massachusetts 02114 , United States.,Department of Otology and Laryngology , Harvard Medical School , Boston , Massachusetts 02114 , United States
| | - Nicholas Koen
- Department of Otolaryngology and The Eaton-Peabody Laboratories , Massachusetts Eye and Ear Infirmary , Boston , Massachusetts 02114 , United States.,Department of Otology and Laryngology , Harvard Medical School , Boston , Massachusetts 02114 , United States
| | - Albert S Edge
- Department of Otolaryngology and The Eaton-Peabody Laboratories , Massachusetts Eye and Ear Infirmary , Boston , Massachusetts 02114 , United States.,Department of Otology and Laryngology , Harvard Medical School , Boston , Massachusetts 02114 , United States
| | - Boris A Kashemirov
- Department of Chemistry , University of Southern California , Los Angeles , California 90089-0744 , United States
| | - David H Jung
- Department of Otolaryngology and The Eaton-Peabody Laboratories , Massachusetts Eye and Ear Infirmary , Boston , Massachusetts 02114 , United States.,Department of Otology and Laryngology , Harvard Medical School , Boston , Massachusetts 02114 , United States
| | - Charles E McKenna
- Department of Chemistry , University of Southern California , Los Angeles , California 90089-0744 , United States
| |
Collapse
|
25
|
Mao B, Moss CF, Wilkinson GS. Age-dependent gene expression in the inner ear of big brown bats (Eptesicus fuscus). PLoS One 2017; 12:e0186667. [PMID: 29073148 PMCID: PMC5658057 DOI: 10.1371/journal.pone.0186667] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Accepted: 10/05/2017] [Indexed: 12/25/2022] Open
Abstract
For echolocating bats, hearing is essential for survival. Specializations for detecting and processing high frequency sounds are apparent throughout their auditory systems. Recent studies on echolocating mammals have reported evidence of parallel evolution in some hearing-related genes in which distantly related groups of echolocating animals (bats and toothed whales), cluster together in gene trees due to apparent amino acid convergence. However, molecular adaptations can occur not only in coding sequences, but also in the regulation of gene expression. The aim of this study was to examine the expression of hearing-related genes in the inner ear of developing big brown bats, Eptesicus fuscus, during the period in which echolocation vocalizations increase dramatically in frequency. We found that seven genes were significantly upregulated in juveniles relative to adults, and that the expression of four genes through development correlated with estimated age. Compared to available data for mice, it appears that expression of some hearing genes is extended in juvenile bats. These results are consistent with a prolonged growth period required to develop larger cochlea relative to body size, a later maturation of high frequency hearing, and a greater dependence on high frequency hearing in echolocating bats.
Collapse
Affiliation(s)
- Beatrice Mao
- Department of Biology, College of Computer, Mathematical, and Natural Sciences, University of Maryland, College Park, Maryland, United States of America
| | - Cynthia F. Moss
- Department of Psychological and Brain Sciences, Zanvyl Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, Maryland, United States of America
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Gerald S. Wilkinson
- Department of Biology, College of Computer, Mathematical, and Natural Sciences, University of Maryland, College Park, Maryland, United States of America
| |
Collapse
|
26
|
Ahmed H, Shubina-Oleinik O, Holt JR. Emerging Gene Therapies for Genetic Hearing Loss. J Assoc Res Otolaryngol 2017; 18:649-670. [PMID: 28815315 PMCID: PMC5612923 DOI: 10.1007/s10162-017-0634-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 07/04/2017] [Indexed: 12/31/2022] Open
Abstract
Gene therapy, or the treatment of human disease using genetic material, for inner ear dysfunction is coming of age. Recent progress in developing gene therapy treatments for genetic hearing loss has demonstrated tantalizing proof-of-principle in animal models. While successful translation of this progress into treatments for humans awaits, there is growing interest from patients, scientists, clinicians, and industry. Nonetheless, it is clear that a number of hurdles remain, and expectations for total restoration of auditory function should remain tempered until these challenges have been overcome. Here, we review progress, prospects, and challenges for gene therapy in the inner ear. We focus on technical aspects, including routes of gene delivery to the inner ear, choice of vectors, promoters, inner ear targets, therapeutic strategies, preliminary success stories, and points to consider for translating of these successes to the clinic.
Collapse
Affiliation(s)
- Hena Ahmed
- Departments of Otolaryngology and Neurology, F.M. Kirby Neurobiology Center Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Olga Shubina-Oleinik
- Departments of Otolaryngology and Neurology, F.M. Kirby Neurobiology Center Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jeffrey R Holt
- Departments of Otolaryngology and Neurology, F.M. Kirby Neurobiology Center Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
27
|
Richardson RT, Thompson AC, Wise AK, Needham K. Challenges for the application of optical stimulation in the cochlea for the study and treatment of hearing loss. Expert Opin Biol Ther 2016; 17:213-223. [PMID: 27960585 DOI: 10.1080/14712598.2017.1271870] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Electrical stimulation has long been the most effective strategy for evoking neural activity from bionic devices and has been used with great success in the cochlear implant to allow deaf people to hear speech and sound. Despite its success, the spread of electrical current stimulates a broad region of neural tissue meaning that contemporary devices have limited precision. Optical stimulation as an alternative has attracted much recent interest for its capacity to provide highly focused stimuli, and therefore, potentially improved sensory perception. Given its specificity of activation, optical stimulation may also provide a useful tool in the study of fundamental neuroanatomy and neurophysiological processes. Areas covered: This review examines the advances in optical stimulation - infrared, nanoparticle-enhanced, and optogenetic-based - and its application in the inner ear for the restoration of auditory function following hearing loss. Expert opinion: Initial outcomes suggest that optogenetic-based approaches hold the greatest potential and viability amongst optical techniques for application in the cochlea. The future success of this approach will be governed by advances in the targeted delivery of opsins to auditory neurons, improvements in channel kinetics, development of optical arrays, and innovation of opsins that activate within the optimal near-infrared therapeutic window.
Collapse
Affiliation(s)
- Rachael T Richardson
- a Bionics Institute , East Melbourne , Australia.,b Department of Medical Bionics , University of Melbourne , East Melbourne , Australia
| | | | - Andrew K Wise
- a Bionics Institute , East Melbourne , Australia.,b Department of Medical Bionics , University of Melbourne , East Melbourne , Australia
| | - Karina Needham
- d Department of Surgery (Otolaryngology) , University of Melbourne, Royal Victorian Eye & Ear Hospital , East Melbourne , Australia
| |
Collapse
|
28
|
Application of the Co-culture Membrane System Pointed to a Protective Role of Catestatin on Hippocampal Plus Hypothalamic Neurons Exposed to Oxygen and Glucose Deprivation. Mol Neurobiol 2016; 54:7369-7381. [PMID: 27815840 DOI: 10.1007/s12035-016-0240-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 10/17/2016] [Indexed: 12/30/2022]
Abstract
Depletion of oxygen and glucose even for brief periods is sufficient to cause cerebral ischemia, which is a predominant worldwide cause of motor deficits with the reduction of life quality and subsequently death. Hence, more insights regarding protective measures against ischemic events are becoming a major research goal. Among the many neuronal factors, N-methyl-D-aspartate receptors (NMDAR), orexinergic neuroreceptors (ORXR), and sympatho-inhibitory neuropeptide catestatin (CST) are widely involved with ischemic episodes. In this study, it was possible to induce in vitro ischemic conditions of the hamster (Mesocricetus auratus) hippocampal and hypothalamic neuronal cultures, grown on a newly compartmentalized membrane system, via oxygen and glucose deprivation (OGD). These cultures displayed notably differentiated NMDARergic and ORXergic receptor expression activities along with evident brain-derived neurotrophic factor (BDNF) plus orexin A (ORX-A) secretion, especially under co-cultured conditions. Interestingly, addition of CST in OGD-insulted hippocampal cells accounted for upregulated GluN1 and ORX1R transcripts that in the case of the latter neuroreceptor was very strongly (p < 0.001) increased when co-cultured with hypothalamic cells. Similarly, hypothalamic neurons supplied very evident upregulations of GluN1, ORX1R, and above all of GluN2A transcripts along with increased BDNF and ORX-A secretion in the presence of hippocampal cells. Overall, the preferential CST effects on BDNF plus ORX-A production together with altered NMDAR and ORXR levels, especially in co-cultured hypothalamic cells pointed to ORX-containing neurons as major protective constituents against ischemic damages thus opening new scenarios on the cross-talking roles of CST during ischemic disorders.
Collapse
|
29
|
Wise AK, Tan J, Wang Y, Caruso F, Shepherd RK. Improved Auditory Nerve Survival with Nanoengineered Supraparticles for Neurotrophin Delivery into the Deafened Cochlea. PLoS One 2016; 11:e0164867. [PMID: 27788219 PMCID: PMC5082918 DOI: 10.1371/journal.pone.0164867] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 10/03/2016] [Indexed: 11/23/2022] Open
Abstract
Cochlear implants electrically stimulate spiral ganglion neurons (SGNs) in order to provide speech cues to severe-profoundly deaf patients. In normal hearing cochleae the SGNs depend on endogenous neurotrophins secreted by sensory cells in the organ of Corti for survival. SGNs gradually degenerate following deafness and consequently there is considerable interest in developing clinically relevant strategies to provide exogenous neurotrophins to preserve SGN survival. The present study investigated the safety and efficacy of a drug delivery system for the cochlea using nanoengineered silica supraparticles. In the present study we delivered Brain-derived neurotrophic factor (BDNF) over a period of four weeks and evaluated SGN survival as a measure of efficacy. Supraparticles were bilaterally implanted into the basal turn of cochleae in profoundly deafened guinea pigs. One ear received BDNF-loaded supraparticles and the other ear control (unloaded) supraparticles. After one month of treatment the cochleae were examined histologically. There was significantly greater survival of SGNs in cochleae that received BDNF supraparticles compared to the contralateral control cochleae (repeated measures ANOVA, p = 0.009). SGN survival was observed over a wide extent of the cochlea. The supraparticles were well tolerated within the cochlea with a tissue response that was localised to the site of implantation in the cochlear base. Although mild, the tissue response was significantly greater in cochleae treated with BDNF supraparticles compared to the controls (repeated measures ANOVA, p = 0.003). These data support the clinical potential of this technology particularly as the supraparticles can be loaded with a variety of therapeutic drugs.
Collapse
Affiliation(s)
- Andrew K. Wise
- The Bionics Institute, 384–388 Albert Street, East Melbourne, Melbourne, Australia
- The Department of Medical Bionics, University of Melbourne, Melbourne, Australia
- Department of Otolaryngology, University of Melbourne, Melbourne, Australia
- * E-mail:
| | - Justin Tan
- Department of Otolaryngology, University of Melbourne, Melbourne, Australia
| | - Yajun Wang
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering, the University of Melbourne, Melbourne, Australia
| | - Frank Caruso
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering, the University of Melbourne, Melbourne, Australia
| | - Robert K. Shepherd
- The Bionics Institute, 384–388 Albert Street, East Melbourne, Melbourne, Australia
- The Department of Medical Bionics, University of Melbourne, Melbourne, Australia
- Department of Otolaryngology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
30
|
Viral-mediated Ntf3 overexpression disrupts innervation and hearing in nondeafened guinea pig cochleae. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 3:16052. [PMID: 27525291 PMCID: PMC4972090 DOI: 10.1038/mtm.2016.52] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 05/13/2016] [Accepted: 06/11/2016] [Indexed: 01/21/2023]
Abstract
Synaptopathy in the cochlea occurs when the connection between inner hair cells and the auditory nerve is disrupted, leading to impaired hearing and nerve degeneration. Experiments using transgenic mice have shown that overexpression of NT3 by supporting cells repairs synaptopathy caused by overstimulation. To accomplish such therapy in the clinical setting, it would be necessary to activate the neurotrophin receptor on auditory neurons by other means. Here we test the outcome of NT3 overexpression using viral-mediated gene transfer into the perilymph versus the endolymph of the normal guinea pig cochlea. We inoculated two different Ntf3 viral vectors, adenovirus (Adv) or adeno-associated virus (AAV) into the perilymph, to facilitate transgene expression in the mesothelial cells and cochlear duct epithelium, respectively. We assessed outcomes by comparing Auditory brainstem response (ABR) thresholds prior to that at baseline to thresholds at 1 and 3 weeks after inoculation, and then performed histologic evaluation of hair cells, nerve endings, and synaptic ribbons. We observed hearing threshold shifts as well as disorganization of peripheral nerve endings and disruption of synaptic connections between inner hair cells and peripheral nerve endings with both vectors. The data suggest that elevation of NT3 levels in the cochlear fluids can disrupt innervation and degrade hearing.
Collapse
|
31
|
Kurioka T, Lee MY, Heeringa AN, Beyer LA, Swiderski DL, Kanicki AC, Kabara LL, Dolan DF, Shore SE, Raphael Y. Selective hair cell ablation and noise exposure lead to different patterns of changes in the cochlea and the cochlear nucleus. Neuroscience 2016; 332:242-57. [PMID: 27403879 DOI: 10.1016/j.neuroscience.2016.07.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 06/14/2016] [Accepted: 07/01/2016] [Indexed: 01/21/2023]
Abstract
In experimental animal models of auditory hair cell (HC) loss, insults such as noise or ototoxic drugs often lead to secondary changes or degeneration in non-sensory cells and neural components, including reduced density of spiral ganglion neurons, demyelination of auditory nerve fibers and altered cell numbers and innervation patterns in the cochlear nucleus (CN). However, it is not clear whether loss of HCs alone leads to secondary degeneration in these neural components of the auditory pathway. To elucidate this issue, we investigated changes of central components after cochlear insults specific to HCs using diphtheria toxin receptor (DTR) mice expressing DTR only in HCs and exhibiting complete HC loss when injected with diphtheria toxin (DT). We showed that DT-induced HC ablation has no significant impacts on the survival of auditory neurons, central synaptic terminals, and myelin, despite complete HC loss and profound deafness. In contrast, noise exposure induced significant changes in synapses, myelin and CN organization even without loss of inner HCs. We observed a decrease of neuronal size in the auditory pathway, including peripheral axons, spiral ganglion neurons, and CN neurons, likely due to loss of input from the cochlea. Taken together, selective HC ablation and noise exposure showed different patterns of pathology in the auditory pathway and the presence of HCs is not essential for the maintenance of central synaptic connectivity and myelination.
Collapse
Affiliation(s)
- Takaomi Kurioka
- Kresge Hearing Research Institute, Department of Otolaryngology-Head and Neck Surgery, University of Michigan, Ann Arbor, MI, USA; Department of Otorhinolaryngology-Head and Neck Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Min Young Lee
- Kresge Hearing Research Institute, Department of Otolaryngology-Head and Neck Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Amarins N Heeringa
- Kresge Hearing Research Institute, Department of Otolaryngology-Head and Neck Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Lisa A Beyer
- Kresge Hearing Research Institute, Department of Otolaryngology-Head and Neck Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Donald L Swiderski
- Kresge Hearing Research Institute, Department of Otolaryngology-Head and Neck Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Ariane C Kanicki
- Kresge Hearing Research Institute, Department of Otolaryngology-Head and Neck Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Lisa L Kabara
- Kresge Hearing Research Institute, Department of Otolaryngology-Head and Neck Surgery, University of Michigan, Ann Arbor, MI, USA
| | - David F Dolan
- Kresge Hearing Research Institute, Department of Otolaryngology-Head and Neck Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Susan E Shore
- Kresge Hearing Research Institute, Department of Otolaryngology-Head and Neck Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Yehoash Raphael
- Kresge Hearing Research Institute, Department of Otolaryngology-Head and Neck Surgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
32
|
ACEMg Diet Supplement Modifies Progression of Hereditary Deafness. Sci Rep 2016; 6:22690. [PMID: 26965868 PMCID: PMC4786814 DOI: 10.1038/srep22690] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/19/2016] [Indexed: 11/08/2022] Open
Abstract
Dietary supplements consisting of beta-carotene (precursor to vitamin A), vitamins C and E and the mineral magnesium (ACEMg) can be beneficial for reducing hearing loss due to aminoglycosides and overstimulation. This regimen also slowed progression of deafness for a boy with GJB2 (CONNEXIN 26) mutations. To assess the potential for treating GJB2 and other forms of hereditary hearing loss with ACEMg, we tested the influence of ACEMg on the cochlea and hearing of mouse models for two human mutations: GJB2, the leading cause of childhood deafness, and DIAPH3, a cause of auditory neuropathy. One group of mice modeling GJB2 (Gjb2-CKO) received ACEMg diet starting shortly after they were weaned (4 weeks) until 16 weeks of age. Another group of Gjb2-CKO mice received ACEMg in utero and after weaning. The ACEMg diet was given to mice modeling DIAPH3 (Diap3-Tg) after weaning (4 weeks) until 12 weeks of age. Control groups received food pellets without the ACEMg supplement. Hearing thresholds measured by auditory brainstem response were significantly better for Gjb2-CKO mice fed ACEMg than for the control diet group. In contrast, Diap3-Tg mice displayed worse thresholds than controls. These results indicate that ACEMg supplementation can influence the progression of genetic hearing loss.
Collapse
|
33
|
Lee MY, Takada T, Takada Y, Kappy MD, Beyer LA, Swiderski DL, Godin AL, Brewer S, King WM, Raphael Y. Mice with conditional deletion of Cx26 exhibit no vestibular phenotype despite secondary loss of Cx30 in the vestibular end organs. Hear Res 2015; 328:102-12. [PMID: 26232528 DOI: 10.1016/j.heares.2015.07.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 07/22/2015] [Accepted: 07/23/2015] [Indexed: 01/19/2023]
Abstract
Connexins are components of gap junctions which facilitate transfer of small molecules between cells. One member of the connexin family, Connexin 26 (Cx26), is prevalent in gap junctions in sensory epithelia of the inner ear. Mutations of GJB2, the gene encoding Cx26, cause significant hearing loss in humans. The vestibular system, however, does not usually show significant functional deficits in humans with this mutation. Mouse models for loss of Cx26 function demonstrate hearing loss and cochlear pathology but the extent of vestibular dysfunction and organ pathology are less well characterized. To understand the vestibular effects of Cx26 mutations, we evaluated vestibular function and histology of the vestibular sensory epithelia in a conditional knockout (CKO) mouse with Cx26 loss of function. Transgenic C57BL/6 mice, in which cre-Sox10 drives excision of the Cx26 gene from non-sensory cells flanking the sensory epithelium of the inner ear (Gjb2-CKO), were compared to age-matched wild types. Animals were sacrificed at ages between 4 and 40 weeks and their cochlear and vestibular sensory organs harvested for histological examination. Cx26 immunoreactivity was prominent in the peripheral vestibular system and the cochlea of wild type mice, but absent in the Gjb2-CKO specimens. The hair cell population in the cochleae of the Gjb2-CKO mice was severely depleted but in the vestibular organs it was intact, despite absence of Cx26 expression. The vestibular organs appeared normal at the latest time point examined, 40 weeks. To determine whether compensation by another connexin explains survival of the normal vestibular sensory epithelium, we evaluated the presence of Cx30 in the Gjb2-CKO mouse. We found that Cx30 labeling was normal in the cochlea, but it was decreased or absent in the vestibular system. The vestibular phenotype of the mutants was not different from wild-types as determined by time on the rotarod, head stability tests and physiological responses to vestibular stimulation. Thus presence of Cx30 in the cochlea does not compensate for Cx26 loss, and the absence of both connexins from vestibular sensory epithelia is no more injurious than the absence of one of them. Further studies to uncover the physiological foundation for this difference between the cochlea and the vestibular organs may help in designing treatments for GJB2 mutations.
Collapse
Affiliation(s)
- Min Young Lee
- Kresge Hearing Research Institute, Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109-5648, USA
| | - Tomoko Takada
- Kresge Hearing Research Institute, Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109-5648, USA
| | - Yohei Takada
- Kresge Hearing Research Institute, Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109-5648, USA
| | - Michelle D Kappy
- Kresge Hearing Research Institute, Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109-5648, USA
| | - Lisa A Beyer
- Kresge Hearing Research Institute, Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109-5648, USA
| | - Donald L Swiderski
- Kresge Hearing Research Institute, Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109-5648, USA
| | - Ashley L Godin
- Kresge Hearing Research Institute, Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109-5648, USA
| | - Shannon Brewer
- Kresge Hearing Research Institute, Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109-5648, USA
| | - W Michael King
- Kresge Hearing Research Institute, Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109-5648, USA
| | - Yehoash Raphael
- Kresge Hearing Research Institute, Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109-5648, USA.
| |
Collapse
|
34
|
Liu H, Zhao L. Recording potentials from scala media, saccule and utricle in mice. J Otol 2015; 10:87-91. [PMID: 29937788 PMCID: PMC6002562 DOI: 10.1016/j.joto.2015.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Objective To describe a protocol for recording electrical potentials from the scala media, saccule, and utricle in mice. Method CBA/J mice were used and potentials were recorded with glass electrodes inserted through the basilar membrane using a patch clamp system. Results Resting potentials were successfully recorded from the scala media, saccule and utricle using described protocols. Conclusions With the method described, one can measure resting potentials from the scala media, saccule and utricle, as well as cochlear microphonics (CM) and even auditory nerve compound action potentials (CAP), in a single mouse.
Collapse
Affiliation(s)
- Huizhan Liu
- Hair Cell Biophysics Laboratory, Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE 68178, USA.,Department of Otolaryngology, Head and Neck Surgery, Institute of Otolaryngology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Lidong Zhao
- Department of Otolaryngology, Head and Neck Surgery, Institute of Otolaryngology, Chinese PLA General Hospital, Beijing, 100853, China
| |
Collapse
|
35
|
Pfingst BE, Zhou N, Colesa DJ, Watts MM, Strahl SB, Garadat SN, Schvartz-Leyzac KC, Budenz CL, Raphael Y, Zwolan TA. Importance of cochlear health for implant function. Hear Res 2015; 322:77-88. [PMID: 25261772 PMCID: PMC4377117 DOI: 10.1016/j.heares.2014.09.009] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 08/14/2014] [Accepted: 09/16/2014] [Indexed: 01/17/2023]
Abstract
Amazing progress has been made in providing useful hearing to hearing-impaired individuals using cochlear implants, but challenges remain. One such challenge is understanding the effects of partial degeneration of the auditory nerve, the target of cochlear implant stimulation. Here we review studies from our human and animal laboratories aimed at characterizing the health of the implanted cochlea and the auditory nerve. We use the data on cochlear and neural health to guide rehabilitation strategies. The data also motivate the development of tissue-engineering procedures to preserve or build a healthy cochlea and improve performance obtained by cochlear implant recipients or eventually replace the need for a cochlear implant. This article is part of a Special Issue entitled .
Collapse
Affiliation(s)
- Bryan E Pfingst
- Department of Otolaryngology, University of Michigan, Ann Arbor, MI, USA.
| | - Ning Zhou
- Department of Otolaryngology, University of Michigan, Ann Arbor, MI, USA; East Carolina University, Greenville, NC, USA
| | - Deborah J Colesa
- Department of Otolaryngology, University of Michigan, Ann Arbor, MI, USA
| | - Melissa M Watts
- Department of Otolaryngology, University of Michigan, Ann Arbor, MI, USA
| | | | - Soha N Garadat
- Department of Otolaryngology, University of Michigan, Ann Arbor, MI, USA; The University of Jordan, Amman, Jordan
| | | | - Cameron L Budenz
- Department of Otolaryngology, University of Michigan, Ann Arbor, MI, USA
| | - Yehoash Raphael
- Department of Otolaryngology, University of Michigan, Ann Arbor, MI, USA
| | - Teresa A Zwolan
- Department of Otolaryngology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
36
|
Differential effects of AAV.BDNF and AAV.Ntf3 in the deafened adult guinea pig ear. Sci Rep 2015; 5:8619. [PMID: 25726967 DOI: 10.1038/srep08619] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 01/27/2015] [Indexed: 01/14/2023] Open
Abstract
Cochlear hair cell loss results in secondary regression of peripheral auditory fibers (PAFs) and loss of spiral ganglion neurons (SGNs). The performance of cochlear implants (CI) in rehabilitating hearing depends on survival of SGNs. Here we compare the effects of adeno-associated virus vectors with neurotrophin gene inserts, AAV.BDNF and AAV.Ntf3, on guinea pig ears deafened systemically (kanamycin and furosemide) or locally (neomycin). AAV.BDNF or AAV.Ntf3 was delivered to the guinea pig cochlea one week following deafening and ears were assessed morphologically 3 months later. At that time, neurotrophins levels were not significantly elevated in the cochlear fluids, even though in vitro and shorter term in vivo experiments demonstrate robust elevation of neurotrophins with these viral vectors. Nevertheless, animals receiving these vectors exhibited considerable re-growth of PAFs in the basilar membrane area. In systemically deafened animals there was a negative correlation between the presence of differentiated supporting cells and PAFs, suggesting that supporting cells influence the outcome of neurotrophin over-expression aimed at enhancing the cochlear neural substrate. Counts of SGN in Rosenthal's canal indicate that BDNF was more effective than NT-3 in preserving SGNs. The results demonstrate that a transient elevation in neurotrophin levels can sustain the cochlear neural substrate in the long term.
Collapse
|
37
|
Spontaneous regeneration of cochlear supporting cells after neonatal ablation ensures hearing in the adult mouse. Proc Natl Acad Sci U S A 2014; 111:16919-24. [PMID: 25385613 DOI: 10.1073/pnas.1408064111] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Supporting cells in the cochlea play critical roles in the development, maintenance, and function of sensory hair cells and auditory neurons. Although the loss of hair cells or auditory neurons results in sensorineural hearing loss, the consequence of supporting cell loss on auditory function is largely unknown. In this study, we specifically ablated inner border cells (IBCs) and inner phalangeal cells (IPhCs), the two types of supporting cells surrounding inner hair cells (IHCs) in mice in vivo. We demonstrate that the organ of Corti has the intrinsic capacity to replenish IBCs/IPhCs effectively during early postnatal development. Repopulation depends on the presence of hair cells and cells within the greater epithelial ridge and is independent of cell proliferation. This plastic response in the neonatal cochlea preserves neuronal survival, afferent innervation, and hearing sensitivity in adult mice. In contrast, the capacity for IBC/IPhC regeneration is lost in the mature organ of Corti, and consequently IHC survival and hearing sensitivity are impaired significantly, demonstrating that there is a critical period for the regeneration of cochlear supporting cells. Our findings indicate that the quiescent neonatal organ of Corti can replenish specific supporting cells completely after loss in vivo to guarantee mature hearing function.
Collapse
|
38
|
Kölliker's organ and the development of spontaneous activity in the auditory system: implications for hearing dysfunction. BIOMED RESEARCH INTERNATIONAL 2014; 2014:367939. [PMID: 25210710 PMCID: PMC4156998 DOI: 10.1155/2014/367939] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 08/07/2014] [Indexed: 11/25/2022]
Abstract
Prior to the “onset of hearing,” developing cochlear inner hair cells (IHCs) and primary auditory neurons undergo experience-independent activity, which is thought to be important in retaining and refining neural connections in the absence of sound. One of the major hypotheses regarding the origin of such activity involves a group of columnar epithelial supporting cells forming Kölliker's organ, which is only present during this critical period of auditory development. There is strong evidence for a purinergic signalling mechanism underlying such activity. ATP released through connexin hemichannels may activate P2 purinergic receptors in both Kölliker's organ and the adjacent IHCs, leading to generation of electrical activity throughout the auditory system. However, recent work has suggested an alternative origin, by demonstrating the ability of IHCs to generate this spontaneous activity without activation by ATP. Regardless, developmental abnormalities of Kölliker's organ may lead to congenital hearing loss, considering that mutations in ion channels (hemichannels, gap junctions, and calcium channels) involved in Kölliker's organ activity share strong links with such types of deafness.
Collapse
|