1
|
Li W, Xu B, Huang Y, Wang X, Yu D. Rodent models in sensorineural hearing loss research: A comprehensive review. Life Sci 2024; 358:123156. [PMID: 39442868 DOI: 10.1016/j.lfs.2024.123156] [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: 07/31/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
Sensorineural hearing loss (SNHL) constitutes a major global health challenge, affecting millions of individuals and substantially impairing social integration and quality of life. The complexity of the auditory system and the multifaceted nature of SNHL necessitate advanced methodologies to understand its etiology, progression, and potential therapeutic interventions. This review provides a comprehensive overview of the current animal models used in SNHL research, focusing on their selection based on specific characteristics and their contributions to elucidating pathophysiological mechanisms and evaluating novel treatment strategies. It discusses the most commonly used rodent models in hearing research, including mice, rats, guinea pigs, Mongolian gerbils, and chinchillas. Through a comparative analysis, this review underscores the importance of selecting models that align with specific research objectives in SNHL studies, discussing the advantages and limitations of each model. By advocating for a multidisciplinary approach that leverages the strengths of various animal models with technological advancements, this review aims to facilitate significant advancements in the prevention, diagnosis, and treatment of sensorineural hearing loss.
Collapse
Affiliation(s)
- Wenjing Li
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200100, PR China
| | - Baoying Xu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, PR China
| | - Yuqi Huang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, PR China
| | - Xueling Wang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200100, PR China
| | - Dehong Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, PR China.
| |
Collapse
|
2
|
Yilmaz NK, Ozen D, da Costa Monsanto R, Ocak E, Schuster AK, Shimura T, Cureoglu S. Efficiency of gene therapy for sensorineural hearing loss in mouse model: A meta-analysis. Laryngoscope Investig Otolaryngol 2024; 9:e70048. [PMID: 39655096 PMCID: PMC11626480 DOI: 10.1002/lio2.70048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024] Open
Abstract
Objectives Sensorineural hearing loss (SNHL) is a disorder characterized by the loss or impairment of cochlear hair cells or the auditory nerve. In recent years, gene therapy has emerged as a promising approach for SNHL treatment. The objective of this study is to evaluate the impact of gene therapy on the restoration or improvement of auditory function in mouse model with loss or impairment of hearing. Methods Studies with clear experimental designs, and auditory brainstem response (ABR) analysis as relevant outcome measures were included by searching PubMed, Scopus, and Web of Science databases. The PRISMA guideline was used for abstracting data and assessing data quality and validity. A quantitative synthesis was performed using a random effects model to examine the effect of gene therapy on auditory function in SNHL. Results Nine articles including 71 studies meeting the inclusion criteria were identified. These studies explored therapies targeting the TMC1, VGLUT3, USH1C, CLRN1, WHRN, and PJVK genes, with genetic material ranging from 1.8 × 1011 and 1.4 × 1014 gc/mL being delivered to the inner ear through round window membrane, cochleostomy, or posterior semicircular canal injection methods. The hearing test results showed a significant mean difference of 26.91 dB (95% CI: 22.01-31.85) in favor of the experimental group. Conclusions Although promising results have been obtained regarding the potential success of gene therapy in SNHL, further investigation is needed to explore the long-term effects of gene therapy, treatment response rates, and the relationships between different genetic mutation types.
Collapse
Affiliation(s)
- Nevra Keskin Yilmaz
- Department of Internal MedicineFaculty of Veterinary Medicine, Ankara UniversityAnkaraTurkey
- Department of Otolaryngology Head & Neck SurgeryUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Dogukan Ozen
- Department of BiostatisticsFaculty of Veterinary Medicine, Ankara UniversityAnkaraTurkey
| | | | - Emre Ocak
- Department of Otolaryngology Head & Neck SurgeryFaculty of Medicine, Ankara UniversityAnkaraTurkey
| | - Artur Koerig Schuster
- Postgraduate Program in Medicine: Surgical Sciences, Universidade Federal do Rio Grande do Sul (UFRGS)Porto AlegreRSBrazil
| | - Tomotaka Shimura
- Department of Otolaryngology Head & Neck SurgeryUniversity of MinnesotaMinneapolisMinnesotaUSA
- Department of OtorhinolaryngologyShowa University Fujigaoka HospitalYokohamaJapan
| | - Sebahattin Cureoglu
- Department of Otolaryngology Head & Neck SurgeryUniversity of MinnesotaMinneapolisMinnesotaUSA
| |
Collapse
|
3
|
Gil Y, Ryu J, Yang H, Ma Y, Nam KH, Jang SW, Shim S. Molecular Characterization of Subdomain Specification of Cochlear Duct Based on Foxg1 and Gata3. Int J Mol Sci 2024; 25:12700. [PMID: 39684410 DOI: 10.3390/ijms252312700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
The inner ear is one of the sensory organs of vertebrates and is largely composed of the vestibule, which controls balance, and the cochlea, which is responsible for hearing. In particular, a problem in cochlear development can lead to hearing loss. Although numerous studies have been conducted on genes involved in the development of the cochlea, many areas still need to be discovered regarding factors that control the patterning of the early cochlear duct. Herein, based on the dynamic expression pattern of FOXG1 in the apical and basal regions of the E13.5 cochlear duct, we identified detailed expression regions through an open-source analysis of single-cell RNA analysis data and demonstrated a clinical correlation with hearing loss. The distinct expression patterns of FOXG1 and GATA3 during the patterning process of the cochlear duct provide important clues to understanding how the fates of the apical and basal regions are divided. These results are expected to be extremely important not only for understanding the molecular mechanisms involved in the early development of the cochlear duct, but also for identifying potential genes that cause hearing loss.
Collapse
Affiliation(s)
- Yongjin Gil
- Department of Biochemistry, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Jiho Ryu
- Department of Biochemistry, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Hayoung Yang
- Department of Biochemistry, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Yechan Ma
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 138-736, Republic of Korea
| | - Ki-Hoan Nam
- Laboratory Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Sung-Wuk Jang
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 138-736, Republic of Korea
| | - Sungbo Shim
- Department of Biochemistry, Chungbuk National University, Cheongju 28644, Republic of Korea
| |
Collapse
|
4
|
Kong C, Yin G, Wang X, Sun Y. In Utero Gene Therapy and its Application in Genetic Hearing Loss. Adv Biol (Weinh) 2024; 8:e2400193. [PMID: 39007241 DOI: 10.1002/adbi.202400193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/03/2024] [Indexed: 07/16/2024]
Abstract
For monogenic genetic diseases, in utero gene therapy (IUGT) shows the potential for early prevention against irreversible and lethal pathological changes. Moreover, animal models have also demonstrated the effectiveness of IUGT in the treatment of coagulation disorders, hemoglobinopathies, neurogenetic disorders, and metabolic and pulmonary diseases. For major alpha thalassemia and severe osteogenesis imperfecta, in utero stem cell transplantation has entered the phase I clinical trial stage. Within the realm of the inner ear, genetic hearing loss significantly hampers speech, cognitive, and intellectual development in children. Nowadays, gene therapies offer substantial promise for deafness, with the success of clinical trials in autosomal recessive deafness 9 using AAV-OTOF gene therapy. However, the majority of genetic mutations that cause deafness affect the development of cochlear structures before the birth of fetuses. Thus, gene therapy before alterations in cochlear structure leading to hearing loss has promising applications. In this review, addressing advances in various fields of IUGT, the progress, and application of IUGT in the treatment of genetic hearing loss are focused, in particular its implementation methods and unique advantages.
Collapse
Affiliation(s)
- Chenyang Kong
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ge Yin
- 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
| | - Yu Sun
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Otorhinolaryngology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
5
|
Kim J, Eygeris Y, Ryals RC, Jozić A, Sahay G. Strategies for non-viral vectors targeting organs beyond the liver. NATURE NANOTECHNOLOGY 2024; 19:428-447. [PMID: 38151642 DOI: 10.1038/s41565-023-01563-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 11/01/2023] [Indexed: 12/29/2023]
Abstract
In recent years, nanoparticles have evolved to a clinical modality to deliver diverse nucleic acids. Rising interest in nanomedicines comes from proven safety and efficacy profiles established by continuous efforts to optimize physicochemical properties and endosomal escape. However, despite their transformative impact on the pharmaceutical industry, the clinical use of non-viral nucleic acid delivery is limited to hepatic diseases and vaccines due to liver accumulation. Overcoming liver tropism of nanoparticles is vital to meet clinical needs in other organs. Understanding the anatomical structure and physiological features of various organs would help to identify potential strategies for fine-tuning nanoparticle characteristics. In this Review, we discuss the source of liver tropism of non-viral vectors, present a brief overview of biological structure, processes and barriers in select organs, highlight approaches available to reach non-liver targets, and discuss techniques to accelerate the discovery of non-hepatic therapies.
Collapse
Affiliation(s)
- Jeonghwan Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
- College of Pharmacy, Yeungnam University, Gyeongsan, South Korea
| | - Yulia Eygeris
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
| | - Renee C Ryals
- Department of Ophthalmology, Casey Eye Institute, Oregon Health and Science University, Portland, OR, USA
| | - Antony Jozić
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA.
- Department of Ophthalmology, Casey Eye Institute, Oregon Health and Science University, Portland, OR, USA.
- Department of Biomedical Engineering, Robertson Life Sciences Building, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
6
|
Hahn R, Avraham KB. Gene Therapy for Inherited Hearing Loss: Updates and Remaining Challenges. Audiol Res 2023; 13:952-966. [PMID: 38131808 PMCID: PMC10740825 DOI: 10.3390/audiolres13060083] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/13/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
Hearing loss stands as the most prevalent sensory deficit among humans, posing a significant global health challenge. Projections indicate that by 2050, approximately 10% of the world's population will grapple with disabling hearing impairment. While approximately half of congenital hearing loss cases have a genetic etiology, traditional interventions such as hearing aids and cochlear implants do not completely restore normal hearing. The absence of biological treatment has prompted significant efforts in recent years, with a strong focus on gene therapy to address hereditary hearing loss. Although several studies have exhibited promising recovery from common forms of genetic deafness in mouse models, existing challenges must be overcome to make gene therapy applicable in the near future. Herein, we summarize the primary gene therapy strategies employed over past years, provide an overview of the recent achievements in preclinical studies for genetic hearing loss, and outline the current key obstacles to cochlear gene therapy.
Collapse
Affiliation(s)
| | - Karen B. Avraham
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel;
| |
Collapse
|
7
|
Abstract
The histories of targeted treatment trials in fragile X syndrome (FXS) are reviewed in animal studies and human trials. Advances in understanding the neurobiology of FXS have identified a number of pathways that are dysregulated in the absence of FMRP and are therefore pathways that can be targeted with new medication. The utilization of quantitative outcome measures to assess efficacy in multiple studies has improved the quality of more recent trials. Current treatment trials including the use of cannabidiol (CBD) topically and metformin orally have positive preliminary data, and both of these medications are available clinically. The use of the phosphodiesterase inhibitor (PDE4D), BPN1440, which raised the level of cAMP that is low in FXS has very promising results for improving cognition in adult males who underwent a controlled trial. There are many more targeted treatments that will undergo trials in FXS, so the future looks bright for new treatments.
Collapse
Affiliation(s)
- Devon Johnson
- MIND Institute, University of California Davis Health, Sacramento, CA, USA
| | - Courtney Clark
- MIND Institute, University of California Davis Health, Sacramento, CA, USA
| | - Randi Hagerman
- MIND Institute, University of California Davis Health, Sacramento, CA, USA
- Department of Pediatrics, University of California Davis Health, Sacramento, CA, USA
| |
Collapse
|
8
|
Ryu J, Statz JP, Chan W, Burch FC, Brigande JV, Kempton B, Porsov EV, Renner L, McGill T, Burwitz BJ, Hanna CB, Neuringer M, Hennebold JD. CRISPR/Cas9 editing of the MYO7A gene in rhesus macaque embryos to generate a primate model of Usher syndrome type 1B. Sci Rep 2022; 12:10036. [PMID: 35710827 PMCID: PMC9203743 DOI: 10.1038/s41598-022-13689-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/26/2022] [Indexed: 12/02/2022] Open
Abstract
Mutations in the MYO7A gene lead to Usher syndrome type 1B (USH1B), a disease characterized by congenital deafness, vision loss, and balance impairment. To create a nonhuman primate (NHP) USH1B model, CRISPR/Cas9 was used to disrupt MYO7A in rhesus macaque zygotes. The targeting efficiency of Cas9 mRNA and hybridized crRNA-tracrRNA (hyb-gRNA) was compared to Cas9 nuclease (Nuc) protein and synthetic single guide (sg)RNAs. Nuc/sgRNA injection led to higher editing efficiencies relative to mRNA/hyb-gRNAs. Mutations were assessed by preimplantation genetic testing (PGT) and those with the desired mutations were transferred into surrogates. A pregnancy was established from an embryo where 92.1% of the PGT sequencing reads possessed a single G insertion that leads to a premature stop codon. Analysis of single peripheral blood leukocytes from the infant revealed that half the cells possessed the homozygous single base insertion and the remaining cells had the wild-type MYO7A sequence. The infant showed sensitive auditory thresholds beginning at 3 months. Although further optimization is needed, our studies demonstrate that it is feasible to use CRISPR technologies for creating NHP models of human diseases.
Collapse
Affiliation(s)
- Junghyun Ryu
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, 97006, USA
| | - John P Statz
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, 97006, USA
- Division of Biological Sciences, University of Montana, Missoula, MT, 59812, USA
| | - William Chan
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, 97006, USA
- University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
| | - Fernanda C Burch
- Assisted Reproductive Technologies Core, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, 97006, USA
| | - John V Brigande
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Beth Kempton
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Edward V Porsov
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Lauren Renner
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, 97006, USA
| | - Trevor McGill
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, 97006, USA
- Department of Ophthalmology, Casey Eye Institute, Oregon Health and Science University, Beaverton, OR, 97006, USA
| | - Benjamin J Burwitz
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, 97006, USA
| | - Carol B Hanna
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, 97006, USA
- Assisted Reproductive Technologies Core, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, 97006, USA
| | - Martha Neuringer
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, 97006, USA
- Department of Ophthalmology, Casey Eye Institute, Oregon Health and Science University, Beaverton, OR, 97006, USA
| | - Jon D Hennebold
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, 97006, USA.
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR, 97239, USA.
| |
Collapse
|
9
|
Umugire A, Lee S, Lee CJ, Choi Y, Kim T, Cho HH. Hyaluronan synthase 1: A novel candidate gene associated with late-onset non-syndromic hereditary hearing loss. Clin Exp Otorhinolaryngol 2022; 15:220-229. [PMID: 35413171 PMCID: PMC9441500 DOI: 10.21053/ceo.2022.00038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/29/2022] [Indexed: 11/22/2022] Open
Abstract
Objectives Hyaluronan synthase 1 (HAS1) is a membrane-bound protein that is abundant in the epidermis and dermis, and it is important for skin function. However, its association with hearing loss has not yet been studied. Herein, we sought to evaluate the potential contribution of HAS1: c.1082G>A to genetic hearing loss. Methods We used whole-exome sequencing to analyze blood DNA samples of six patients of a family with autosomal dominant familial late-onset progressive hearing loss, which was revealed to be related to a variant of the HAS1 gene. Confirmatory Sanger sequencing was performed with samples from 10 members. A missense variant was detected in HAS1 (c.1082 G>A, p.Cys361Tyr). In silico analyses predicted this variant to result in the functional loss of HAS1. Immunostaining was conducted using wild-type mouse samples to verify HAS1 expression. Results Has1 was detected in an otocyst at E10.5. In the pup, Has1 expression was localized in the stria vascularis (SV), hair cells, supporting cells of the organ of Corti, and some spiral ganglion neurons. SV marginal cells markedly expressed Has1 in the adult stage. The hearing threshold in the Has1-depleted condition was investigated by accessing the International Mouse Phenotyping Consortium’s Auditory Brainstem Response (ABR) data. ABR of Has1 knock-out mice showed threshold elevations at 6, 12, and 18 kHz in young male adults. Conclusion HAS1 may have a close relationship with auditory function and genetic hearing loss. Further investigation is needed to reveal the precise role of HAS1 in the auditory system. HAS1 is a candidate gene for future hereditary hearing loss genetic testing.
Collapse
|
10
|
Elliott KL, Fritzsch B, Yamoah EN, Zine A. Age-Related Hearing Loss: Sensory and Neural Etiology and Their Interdependence. Front Aging Neurosci 2022; 14:814528. [PMID: 35250542 PMCID: PMC8891613 DOI: 10.3389/fnagi.2022.814528] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 01/03/2022] [Indexed: 12/19/2022] Open
Abstract
Age-related hearing loss (ARHL) is a common, increasing problem for older adults, affecting about 1 billion people by 2050. We aim to correlate the different reductions of hearing from cochlear hair cells (HCs), spiral ganglion neurons (SGNs), cochlear nuclei (CN), and superior olivary complex (SOC) with the analysis of various reasons for each one on the sensory deficit profiles. Outer HCs show a progressive loss in a basal-to-apical gradient, and inner HCs show a loss in a apex-to-base progression that results in ARHL at high frequencies after 70 years of age. In early neonates, SGNs innervation of cochlear HCs is maintained. Loss of SGNs results in a considerable decrease (~50% or more) of cochlear nuclei in neonates, though the loss is milder in older mice and humans. The dorsal cochlear nuclei (fusiform neurons) project directly to the inferior colliculi while most anterior cochlear nuclei reach the SOC. Reducing the number of neurons in the medial nucleus of the trapezoid body (MNTB) affects the interactions with the lateral superior olive to fine-tune ipsi- and contralateral projections that may remain normal in mice, possibly humans. The inferior colliculi receive direct cochlear fibers and second-order fibers from the superior olivary complex. Loss of the second-order fibers leads to hearing loss in mice and humans. Although ARHL may arise from many complex causes, HC degeneration remains the more significant problem of hearing restoration that would replace the cochlear implant. The review presents recent findings of older humans and mice with hearing loss.
Collapse
Affiliation(s)
- Karen L. Elliott
- Department of Biology, University of Iowa, Iowa City, IA, United States
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, IA, United States
- *Correspondence: Bernd Fritzsch
| | - Ebenezer N. Yamoah
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, United States
| | - Azel Zine
- LBN, Laboratory of Bioengineering and Nanoscience, University of Montpellier, Montpellier, France
| |
Collapse
|
11
|
Abstract
Congenital hearing loss is the most common birth defect, estimated to affect 2-3 in every 1000 births. Currently there is no cure for hearing loss. Treatment options are limited to hearing aids for mild and moderate cases, and cochlear implants for severe and profound hearing loss. Here we provide a literature overview of the environmental and genetic causes of congenital hearing loss, common animal models and methods used for hearing research, as well as recent advances towards developing therapies to treat congenital deafness. © 2021 The Authors.
Collapse
Affiliation(s)
- Justine M Renauld
- Department of Otolaryngology, Head & Neck Surgery, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Martin L Basch
- Department of Otolaryngology, Head & Neck Surgery, Case Western Reserve University School of Medicine, Cleveland, Ohio.,Department of Genetics and Genome Sciences, Case Western Reserve School of Medicine, Cleveland, Ohio.,Department of Biology, Case Western Reserve University, Cleveland, Ohio.,Department of Otolaryngology, Head & Neck Surgery, University Hospitals, Cleveland, Ohio
| |
Collapse
|
12
|
Roccio M. Directed differentiation and direct reprogramming: Applying stem cell technologies to hearing research. Stem Cells 2020; 39:375-388. [PMID: 33378797 DOI: 10.1002/stem.3315] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/24/2020] [Accepted: 12/01/2020] [Indexed: 12/26/2022]
Abstract
Hearing loss is the most widely spread sensory disorder in our society. In the majority of cases, it is caused by the loss or malfunctioning of cells in the cochlea: the mechanosensory hair cells, which act as primary sound receptors, and the connecting auditory neurons of the spiral ganglion, which relay the signal to upper brain centers. In contrast to other vertebrates, where damage to the hearing organ can be repaired through the activity of resident cells, acting as tissue progenitors, in mammals, sensory cell damage or loss is irreversible. The understanding of gene and cellular functions, through analysis of different animal models, has helped to identify causes of disease and possible targets for hearing restoration. Translation of these findings to novel therapeutics is, however, hindered by the lack of cellular assays, based on human sensory cells, to evaluate the conservation of molecular pathways across species and the efficacy of novel therapeutic strategies. In the last decade, stem cell technologies enabled to generate human sensory cell types in vitro, providing novel tools to study human inner ear biology, model disease, and validate therapeutics. This review focuses specifically on two technologies: directed differentiation of pluripotent stem cells and direct reprogramming of somatic cell types to sensory hair cells and neurons. Recent development in the field are discussed as well as how these tools could be implemented to become routinely adopted experimental models for hearing research.
Collapse
Affiliation(s)
- Marta Roccio
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich (USZ), and University of Zurich (UZH), Zurich, Switzerland
| |
Collapse
|
13
|
Breaking the sound barrier: Towards next-generation AAV vectors for gene therapy of hearing disorders. Hear Res 2020; 413:108092. [PMID: 33268240 DOI: 10.1016/j.heares.2020.108092] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 09/14/2020] [Accepted: 10/08/2020] [Indexed: 12/20/2022]
Abstract
Owing to the advances in transgenic animal technology and the advent of the next-generation sequencing era, over 120 genes causing hereditary hearing loss have been identified by now. In parallel, the field of human gene therapy continues to make exciting and rapid progress, culminating in the recent approval of several ex vivo and in vivo applications. Despite these encouraging developments and the growing interest in causative treatments for hearing disorders, gene therapeutic interventions in the inner ear remain in their infancy and await clinical translation. This review focuses on the adeno-associated virus (AAV), which nowadays represents one of the safest and most promising vectors in gene therapy. We first provide an overview of AAV biology and outline the principles of therapeutic gene transfer with recombinant AAV vectors, before pointing out major challenges and solutions for clinical translation including vector manufacturing and species translatability. Finally, we highlight seminal technologies for engineering and selection of next-generation "designer" AAV capsids, and illustrate their power and potential with recent examples of their application for inner ear gene transfer in animals.
Collapse
|
14
|
Thorpe RK, Smith RJH. Future directions for screening and treatment in congenital hearing loss. PRECISION CLINICAL MEDICINE 2020; 3:175-186. [PMID: 33209510 PMCID: PMC7653508 DOI: 10.1093/pcmedi/pbaa025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/06/2020] [Accepted: 07/12/2020] [Indexed: 02/06/2023] Open
Abstract
Hearing loss is the most common neurosensory deficit. It results from a variety of heritable and acquired causes and is linked to multiple deleterious effects on a child's development that can be ameliorated by prompt identification and individualized therapies. Diagnosing hearing loss in newborns is challenging, especially in mild or progressive cases, and its management requires a multidisciplinary team of healthcare providers comprising audiologists, pediatricians, otolaryngologists, and genetic counselors. While physiologic newborn hearing screening has resulted in earlier diagnosis of hearing loss than ever before, a growing body of knowledge supports the concurrent implementation of genetic and cytomegalovirus testing to offset the limitations inherent to a singular screening modality. In this review, we discuss the contemporary role of screening for hearing loss in newborns as well as future directions in its diagnosis and treatment.
Collapse
Affiliation(s)
- Ryan K Thorpe
- Molecular Otolaryngology and Renal Research Laboratories, Carver College of Medicine, University of Iowa, 375 Newton Rd, Iowa City, IA 52242, USA
- Department of Otolaryngology – Head and Neck Surgery, University of Iowa, 200 Hawkins Dr, Iowa City, IA 52242, USA
| | - Richard J H Smith
- Molecular Otolaryngology and Renal Research Laboratories, Carver College of Medicine, University of Iowa, 375 Newton Rd, Iowa City, IA 52242, USA
- Department of Otolaryngology – Head and Neck Surgery, University of Iowa, 200 Hawkins Dr, Iowa City, IA 52242, USA
- The Interdisciplinary Graduate Program in Genetics, University of Iowa, 375 Newton Rd, Iowa City, IA 52242, USA
- Iowa Institute of Human Genetics, University of Iowa, 375 Newton Rd, Iowa City, IA 52242, USA
| |
Collapse
|
15
|
Wang L, Kempton JB, Jiang H, Jodelka FM, Brigande AM, Dumont RA, Rigo F, Lentz JJ, Hastings ML, Brigande JV. Fetal antisense oligonucleotide therapy for congenital deafness and vestibular dysfunction. Nucleic Acids Res 2020; 48:5065-5080. [PMID: 32249312 PMCID: PMC7229850 DOI: 10.1093/nar/gkaa194] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/13/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
Disabling hearing loss impacts ∼466 million individuals worldwide with 34 million children affected. Gene and pharmacotherapeutic strategies to rescue auditory function in mouse models of human deafness are most effective when administered before hearing onset, after which therapeutic efficacy is significantly diminished or lost. We hypothesize that preemptive correction of a mutation in the fetal inner ear prior to maturation of the sensory epithelium will optimally restore sensory function. We previously demonstrated that transuterine microinjection of a splice-switching antisense oligonucleotide (ASO) into the amniotic cavity immediately surrounding the embryo on embryonic day 13-13.5 (E13-13.5) corrected pre-mRNA splicing in the juvenile Usher syndrome type 1c (Ush1c) mouse mutant. Here, we show that this strategy only marginally rescues hearing and partially rescues vestibular function. To improve therapeutic outcomes, we microinjected ASO directly into the E12.5 inner ear. A single intra-otic dose of ASO corrects harmonin RNA splicing, restores harmonin protein expression in sensory hair cell bundles, prevents hair cell loss, improves hearing sensitivity, and ameliorates vestibular dysfunction. Improvements in auditory and vestibular function were sustained well into adulthood. Our results demonstrate that an ASO pharmacotherapeutic administered to a developing organ system in utero preemptively corrects pre-mRNA splicing to abrogate the disease phenotype.
Collapse
Affiliation(s)
- Lingyan Wang
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - J Beth Kempton
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - Han Jiang
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - Francine M Jodelka
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Alev M Brigande
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - Rachel A Dumont
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, CA 92010 USA
| | - Jennifer J Lentz
- Department of Otorhinolaryngology, Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Michelle L Hastings
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - John V Brigande
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
16
|
Yamoah EN, Li M, Shah A, Elliott KL, Cheah K, Xu PX, Phillips S, Young SM, Eberl DF, Fritzsch B. Using Sox2 to alleviate the hallmarks of age-related hearing loss. Ageing Res Rev 2020; 59:101042. [PMID: 32173536 PMCID: PMC7261488 DOI: 10.1016/j.arr.2020.101042] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 02/07/2023]
Abstract
Age-related hearing loss (ARHL) is the most prevalent sensory deficit. ARHL reduces the quality of life of the growing population, setting seniors up for the enhanced mental decline. The size of the needy population, the structural deficit, and a likely research strategy for effective treatment of chronic neurosensory hearing in the elderly are needed. Although there has been profound advancement in auditory regenerative research, there remain multiple challenges to restore hearing loss. Thus, additional investigations are required, using novel tools. We propose how the (1) flat epithelium, remaining after the organ of Corti has deteriorated, can be converted to the repaired-sensory epithelium, using Sox2. This will include (2) developing an artificial gene regulatory network transmitted by (3) large viral vectors to the flat epithelium to stimulate remnants of the organ of Corti to restore hair cells. We hope to unite with our proposal toward the common goal, eventually restoring a functional human hearing organ by transforming the flat epithelial cells left after the organ of Corti loss.
Collapse
Affiliation(s)
- Ebenezer N Yamoah
- Department of Physiology and Cell Biology, University of Nevada, Reno, USA
| | - Mark Li
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, USA
| | - Anit Shah
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, USA
| | - Karen L Elliott
- Department of Biology, CLAS, University of Iowa, Iowa City, USA
| | - Kathy Cheah
- Department of Biochemistry, Hong Kong University, Hong Kong, China
| | - Pin-Xian Xu
- Department of Biochemistry, Hong Kong University, Hong Kong, China
| | - Stacia Phillips
- Department of Biochemistry, Hong Kong University, Hong Kong, China
| | - Samuel M Young
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, USA; Department of Otolaryngology, Iowa Neuroscience Institute, University of Iowa, Iowa City, USA
| | - Daniel F Eberl
- Department of Biology, CLAS, University of Iowa, Iowa City, USA
| | - Bernd Fritzsch
- Department of Biology, CLAS, University of Iowa, Iowa City, USA.
| |
Collapse
|
17
|
Lee S, Dondzillo A, Gubbels SP, Raphael Y. Practical aspects of inner ear gene delivery for research and clinical applications. Hear Res 2020; 394:107934. [PMID: 32204962 DOI: 10.1016/j.heares.2020.107934] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 02/26/2020] [Accepted: 02/28/2020] [Indexed: 12/24/2022]
Abstract
The application of gene therapy is widely expanding in research and continuously improving in preparation for clinical applications. The inner ear is an attractive target for gene therapy for treating environmental and genetic diseases in both the auditory and vestibular systems. With the lack of spontaneous cochlear hair cell replacement, hair cell regeneration in adult mammals is among the most important goals of gene therapy. In addition, correcting gene defects can open up a new era for treating inner ear diseases. The relative isolation and small size of the inner ear dictate local administration routes and carefully calculated small volumes of reagents. In the current review, we will cover effective timing, injection routes and types of vectors for successful gene delivery to specific target cells within the inner ear. Differences between research purposes and clinical applications are also discussed.
Collapse
Affiliation(s)
- Sungsu Lee
- Kresge Hearing Research Institute, Department of Otolaryngology, Head and Neck Surgery, Michigan Medicine, Ann Arbor, MI, USA
| | - Anna Dondzillo
- Department of Otolaryngology, Head and Neck Surgery, University of Colorado School of Medicine, Aurora, CO, USA
| | - Samuel P Gubbels
- Department of Otolaryngology, Head and Neck Surgery, University of Colorado School of Medicine, Aurora, CO, USA
| | - Yehoash Raphael
- Kresge Hearing Research Institute, Department of Otolaryngology, Head and Neck Surgery, Michigan Medicine, Ann Arbor, MI, USA.
| |
Collapse
|
18
|
Fetal gene therapy and pharmacotherapy to treat congenital hearing loss and vestibular dysfunction. Hear Res 2020; 394:107931. [PMID: 32173115 DOI: 10.1016/j.heares.2020.107931] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/24/2020] [Accepted: 02/26/2020] [Indexed: 12/23/2022]
Abstract
Disabling hearing loss is expected to affect over 900 million people worldwide by 2050. The World Health Organization estimates that the annual economic impact of hearing loss globally is US$ 750 billion. The inability to hear may complicate effective interpersonal communication and negatively impact personal and professional relationships. Recent advances in the genetic diagnosis of inner ear disease have keenly focused attention on strategies to restore hearing and balance in individuals with defined gene mutations. Mouse models of human hearing loss serve as the primary approach to test gene therapies and pharmacotherapies. The goal of this review is to articulate the rationale for fetal gene therapy and pharmacotherapy to treat congenital hearing loss and vestibular dysfunction. The differential onset of hearing in mice and humans suggests that a prenatal window of therapeutic efficacy in humans may be optimal to restore sensory function. Mouse studies demonstrating the utility of early fetal intervention in the inner ear show promise. We focus on the modulation of gene expression through two strategies that have successfully treated deafness in animal models and have had clinical success for other conditions in humans: gene replacement and antisense oligonucleotide-mediated modulation of gene expression. The recent establishment of effective therapies targeting the juvenile and adult mouse provide informative counterexamples where intervention in the maturing and fully functional mouse inner ear may be effective. Distillation of the current literature leads to the conclusion that novel therapeutic strategies to treat genetic deafness and imbalance will soon translate to clinical trials.
Collapse
|
19
|
Johnson Chacko L, Sergi C, Eberharter T, Dudas J, Rask-Andersen H, Hoermann R, Fritsch H, Fischer N, Glueckert R, Schrott-Fischer A. Early appearance of key transcription factors influence the spatiotemporal development of the human inner ear. Cell Tissue Res 2020; 379:459-471. [PMID: 31788757 DOI: 10.1007/s00441-019-03115-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 09/22/2019] [Indexed: 12/11/2022]
Abstract
Expression patterns of transcription factors leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5), transforming growth factor-β-activated kinase-1 (TAK1), SRY (sex-determining region Y)-box 2 (SOX2), and GATA binding protein 3 (GATA3) in the developing human fetal inner ear were studied between the gestation weeks 9 and 12. Further development of cochlear apex between gestational weeks 11 and 16 (GW11 and GW16) was examined using transmission electron microscopy. LGR5 was evident in the apical poles of the sensory epithelium of the cochlear duct and the vestibular end organs at GW11. Immunostaining was limited to hair cells of the organ of Corti by GW12. TAK1 was immune positive in inner hair cells of the organ of Corti by GW12 and colocalized with p75 neurotrophic receptor expression. Expression for SOX2 was confined primarily to the supporting cells of utricle at the earliest stage examined at GW9. Intense expression for GATA3 was presented in the cochlear sensory epithelium and spiral ganglia at GW9. Expression of GATA3 was present along the midline of both the utricle and saccule in the zone corresponding to the striolar reversal zone where the hair cell phenotype switches from type I to type II. The spatiotemporal gradient of the development of the organ of Corti was also evident with the apex of the cochlea forming by GW16. It seems that highly specific staining patterns of several transcriptions factors are critical in guiding the genesis of the inner ear over development. Our findings suggest that the spatiotemporal gradient in cochlear development extends at least until gestational week 16.
Collapse
Affiliation(s)
- Lejo Johnson Chacko
- Department of Otorhinolaryngology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Consolato Sergi
- Department of Laboratory Medicine and Pathology and Department of Pediatrics, University of Alberta, 8440 112 St, NW, Edmonton, AB, T6G 2B7, Canada
| | - Theresa Eberharter
- Department of Otorhinolaryngology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Jozsef Dudas
- Department of Otorhinolaryngology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Helge Rask-Andersen
- Department of Surgical Sciences, Head and Neck Surgery, Section of Otolaryngology, Uppsala University Hospital, SE-751 85, Uppsala, Sweden
| | - Romed Hoermann
- Department of Anatomy, Histology & Embryology, Division of Clinical & Functional Anatomy, Medical University of Innsbruck, Muellerstrasse 59, 6020, Innsbruck, Austria
| | - Helga Fritsch
- Department of Anatomy, Histology & Embryology, Division of Clinical & Functional Anatomy, Medical University of Innsbruck, Muellerstrasse 59, 6020, Innsbruck, Austria
| | - Natalie Fischer
- University Clinics Innsbruck, Tirol Kliniken, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Rudolf Glueckert
- Department of Otorhinolaryngology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
- University Clinics Innsbruck, Tirol Kliniken, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Anneliese Schrott-Fischer
- Department of Otorhinolaryngology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| |
Collapse
|
20
|
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: 0.2] [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
|
21
|
Burton JA, Valero MD, Hackett TA, Ramachandran R. The use of nonhuman primates in studies of noise injury and treatment. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2019; 146:3770. [PMID: 31795680 PMCID: PMC6881191 DOI: 10.1121/1.5132709] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 07/25/2019] [Accepted: 07/30/2019] [Indexed: 05/10/2023]
Abstract
Exposure to prolonged or high intensity noise increases the risk for permanent hearing impairment. Over several decades, researchers characterized the nature of harmful noise exposures and worked to establish guidelines for effective protection. Recent laboratory studies, primarily conducted in rodent models, indicate that the auditory system may be more vulnerable to noise-induced hearing loss (NIHL) than previously thought, driving renewed inquiries into the harmful effects of noise in humans. To bridge the translational gaps between rodents and humans, nonhuman primates (NHPs) may serve as key animal models. The phylogenetic proximity of NHPs to humans underlies tremendous similarity in many features of the auditory system (genomic, anatomical, physiological, behavioral), all of which are important considerations in the assessment and treatment of NIHL. This review summarizes the literature pertaining to NHPs as models of hearing and noise-induced hearing loss, discusses factors relevant to the translation of diagnostics and therapeutics from animals to humans, and concludes with some of the practical considerations involved in conducting NHP research.
Collapse
Affiliation(s)
- Jane A Burton
- Neuroscience Graduate Program, Vanderbilt University, Nashville, Tennessee 37212, USA
| | - Michelle D Valero
- Eaton Peabody Laboratories at Massachusetts Eye and Ear, Boston, Massachusetts 02114, USA
| | - Troy A Hackett
- Department of Hearing and Speech Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - Ramnarayan Ramachandran
- Department of Hearing and Speech Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| |
Collapse
|
22
|
Roccio M, Edge ASB. Inner ear organoids: new tools to understand neurosensory cell development, degeneration and regeneration. Development 2019; 146:146/17/dev177188. [PMID: 31477580 DOI: 10.1242/dev.177188] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The development of therapeutic interventions for hearing loss requires fundamental knowledge about the signaling pathways controlling tissue development as well as the establishment of human cell-based assays to validate therapeutic strategies ex vivo Recent advances in the field of stem cell biology and organoid culture systems allow the expansion and differentiation of tissue-specific progenitors and pluripotent stem cells in vitro into functional hair cells and otic-like neurons. We discuss how inner ear organoids have been developed and how they offer for the first time the opportunity to validate drug-based therapies, gene-targeting approaches and cell replacement strategies.
Collapse
Affiliation(s)
- Marta Roccio
- Inner Ear Research Laboratory, Department of Biomedical Research (DBMR), University of Bern, Bern 3008, Switzerland .,Department of Otorhinolaryngology, Head & Neck Surgery, Inselspital, Bern University Hospital, University of Bern, Bern 3010, Switzerland
| | - Albert S B Edge
- Department of Otolaryngology, Harvard Medical School, Boston, MA 02115, USA.,Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA.,Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
23
|
Claussen AD, Vielman Quevedo R, Mostaert B, Kirk JR, Dueck WF, Hansen MR. A mouse model of cochlear implantation with chronic electric stimulation. PLoS One 2019; 14:e0215407. [PMID: 30998726 PMCID: PMC6472764 DOI: 10.1371/journal.pone.0215407] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/01/2019] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVES Cochlear implants provide an effective treatment option for those with severe hearing loss, including those with preserved low frequency hearing. However, certain issues can reduce implant efficacy including intracochlear tissue response and delayed loss of residual acoustic hearing. We describe a mouse model of cochlear implantation with chronic electric stimulation that can be used to study cochlear implant biology and related pathologies. METHODS Twelve normal hearing adult CBA/J mice underwent unilateral cochlear implantation and were evenly divided into one group receiving electric stimulation and one not. Serial impedance and neural response telemetry (NRT) measurements were made to assess implant functionality. Functionality was defined as having at least one electrode with an impedance ≤ 35 kOhms. Mouse cochleae were harvested for histology and 3D x-ray microscopy 21 days post-operatively, or, in case the implant was still functional, at a later time point when the implant failed. A separate experiment measured the hearing preservation rate in 7 adult CBA/J mice undergoing unilateral cochlear implantation with serial auditory brainstem response (ABR) and distortion product otoacoustic emissions (DPOAE). RESULTS Implants maintained functionality for a mean of 35 days in the non-stimulated group and 19.8 days in the stimulated group. Reliable NRT and behavioral responses to electric stimulation were recorded. A robust intracochlear peri-implant tissue response with neo-ossification was seen in all cochleae. Six of seven mice maintained intact low frequency hearing up to 6 weeks following cochlear implantation. CONCLUSIONS We demonstrate the feasibility of cochlear implantation and behaviorally significant electric stimulation in the mouse, with the potential for hearing preservation. This model may be combined with established mouse models of hearing loss and the large genetic and molecular research toolkit unique to the mouse for mechanistic and therapeutic investigations of cochlear implant biology.
Collapse
Affiliation(s)
- Alexander D. Claussen
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, IA, United States of America
| | - René Vielman Quevedo
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, IA, United States of America
| | - Brian Mostaert
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, IA, United States of America
| | | | | | - Marlan R. Hansen
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, IA, United States of America
- Department of Neurosurgery, University of Iowa, Iowa City, IA, United States of America
| |
Collapse
|
24
|
Engraftment of Human Stem Cell-Derived Otic Progenitors in the Damaged Cochlea. Mol Ther 2019; 27:1101-1113. [PMID: 31005598 DOI: 10.1016/j.ymthe.2019.03.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 12/20/2022] Open
Abstract
Most cases of sensorineural deafness are caused by degeneration of hair cells. Although stem/progenitor cell therapy is becoming a promising treatment strategy in a variety of organ systems, cell engraftment in the adult mammalian cochlea has not yet been demonstrated. In this study, we generated human otic progenitor cells (hOPCs) from induced pluripotent stem cells (iPSCs) in vitro and identified these cells by the expression of known otic markers. We showed successful cell transplantation of iPSC-derived-hOPCs in an in vivo adult guinea pig model of ototoxicity. The delivered hOPCs migrated throughout the cochlea, engrafted in non-sensory regions, and survived up to 4 weeks post-transplantation. Some of the engrafted hOPCs responded to environmental cues within the cochlear sensory epithelium and displayed molecular features of early sensory differentiation. We confirmed these results with hair cell progenitors derived from Atoh1-GFP mice as donor cells. These mouse otic progenitors transplanted using the same in vivo delivery system migrated into damaged cochlear sensory epithelium and adopted a partial sensory cell fate. This is the first report of the survival and differentiation of hOPCs in ototoxic-injured mature cochlear epithelium, and it should stimulate further research into cell-based therapies for treatment of deafness.
Collapse
|
25
|
Abstract
Antisense oligonucleotides (ASOs) have shown potential as therapeutic molecules for the treatment of inner ear dysfunction. The peripheral sensory organs responsible for both hearing and equilibrium are housed within the inner ear. Hearing loss and vestibular balance problems affect a large portion of the population and limited treatment options exist. Targeting ASOs to the inner ear as a therapeutic strategy has unique pharmacokinetic and drug delivery opportunities and challenges. Here, we review ASO technology, delivery, disease targets, and other key considerations for development of this therapeutic approach.
Collapse
Affiliation(s)
- Michelle L Hastings
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Science and Medicine, North Chicago, IL, 60064, USA.
| | - Timothy A Jones
- Department of Special Education and Communication Disorders, University of Nebraska-Lincoln, 304 Barkley Memorial Center, Lincoln, NE, 68583, USA
| |
Collapse
|