1
|
Vitry S, Mendia C, Maudoux A, El-Amraoui A. Advancing precision ear medicine: leveraging animal models for disease insights and therapeutic innovations. Mamm Genome 2025:10.1007/s00335-025-10126-y. [PMID: 40263131 DOI: 10.1007/s00335-025-10126-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/28/2025] [Indexed: 04/24/2025]
Abstract
Gene therapy offers significant promise for treating inner ear disorders, but its clinical translation requires robust preclinical validation, often reliant on animal models. This review examines the role of these models in advancing gene therapeutics for inherited inner ear disorders, focusing on successes, challenges, and treatment solutions. By providing a precise understanding of disease mechanisms, these models offer a versatile preclinical platform that is essential for assessing and validating therapies. Successful gene supplementation and editing have shown potential in restoring hearing and balance functions and preventing their decline. However, challenges such as limitations in gene delivery methods, surgical access, immune responses, and discrepancies in disease manifestation between animal models and humans hinder clinical translation. Current efforts are dedicated to developing innovative strategies aimed at enhancing the efficiency of gene delivery, overcoming physical barriers such as the blood-labyrinth barrier, improving target specificity, and maximizing therapeutic efficacy while minimizing adverse immune responses. Diverse gene supplementation and editing strategies, along with evolving technologies, hold promise for maximizing therapeutic outcomes using disease relevant models. The future of inner ear gene therapeutics will hinge on personalized therapies and team science fueling interdisciplinary collaborations among researchers, clinicians, companies, and regulatory agencies to expedite the translation from bench to bedside and unlock the immense potential of precision medicine in the inner ear.
Collapse
Affiliation(s)
- Sandrine Vitry
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Progressive Sensory Disorders, Pathophysiology and Therapy, F-75012, Paris, France.
- Unit Progressive Sensory Disorders, Pathophysiology and Therapy, Institut Pasteur, Institut de l'Audition, Paris, France.
| | - Clara Mendia
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Progressive Sensory Disorders, Pathophysiology and Therapy, F-75012, Paris, France
- Collège Doctoral, Sorbonne Université, 75005, Paris, France
| | - Audrey Maudoux
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Progressive Sensory Disorders, Pathophysiology and Therapy, F-75012, Paris, France
- Otolaryngology Department, Assistance Publique des Hôpitaux de Paris, Robert Debré University Hospital-APHP, Paris, France
| | - Aziz El-Amraoui
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, Progressive Sensory Disorders, Pathophysiology and Therapy, F-75012, Paris, France.
- Unit Progressive Sensory Disorders, Pathophysiology and Therapy, Institut Pasteur, Institut de l'Audition, Paris, France.
| |
Collapse
|
2
|
Maniaci A, La Via L, Lechien JR, Sangiorgio G, Iannella G, Magliulo G, Pace A, Mat Q, Lavalle S, Lentini M. Hearing Loss and Oxidative Stress: A Comprehensive Review. Antioxidants (Basel) 2024; 13:842. [PMID: 39061910 PMCID: PMC11274311 DOI: 10.3390/antiox13070842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Hearing loss is a prevalent condition affecting millions of people worldwide. Hearing loss has been linked to oxidative stress as a major factor in its onset and progression. The goal of this thorough analysis is to investigate the connection between oxidative stress and hearing loss, with an emphasis on the underlying mechanisms and possible treatments. The review addressed the many forms of hearing loss, the role of reactive oxygen species (ROS) in causing damage to the cochlea, and the auditory system's antioxidant defensive mechanisms. The review also goes over the available data that support the use of antioxidants and other methods to lessen hearing loss brought on by oxidative stress. We found that oxidative stress is implicated in multiple types of hearing loss, including age-related, noise-induced, and ototoxic hearing impairment. The cochlea's unique anatomical and physiological characteristics, such as high metabolic activity and limited blood supply, make it particularly susceptible to oxidative damage. Antioxidant therapies have shown promising results in both animal models and clinical studies for preventing and mitigating hearing loss. Emerging therapeutic approaches, including targeted drug delivery systems and gene therapy, offer new possibilities for addressing oxidative stress in the auditory system. The significance of this review lies in its comprehensive analysis of the intricate relationship between oxidative stress and hearing loss. By synthesizing current knowledge and identifying gaps in understanding, this review provides valuable insights for both researchers and clinicians. It highlights the potential of antioxidant-based interventions and emphasizes the need for further research into personalized treatment strategies. Our findings on oxidative stress mechanisms may also affect clinical practice and future research directions. This review serves as a foundation for developing novel therapeutic approaches and may inform evidence-based strategies for the prevention and treatment of hearing loss, ultimately contributing to improved quality of life for millions affected by this condition worldwide.
Collapse
Affiliation(s)
- A. Maniaci
- Faculty of Medicine and Surgery, University of Enna Kore, 94100 Enna, Italy;
- ASP Ragusa-Hospital Giovanni Paolo II, 97100 Ragusa, Italy;
- Otology Study Group, Young Otolaryngologists-International Federation of Otorhinolaryngological Societies, 13005 Paris, France; (J.R.L.); (G.I.); (A.P.); (Q.M.)
| | - L. La Via
- Department of Anaesthesia and Intensive Care, University Hospital Policlinico-San Marco, 95125 Catania, Italy
| | - J. R. Lechien
- Otology Study Group, Young Otolaryngologists-International Federation of Otorhinolaryngological Societies, 13005 Paris, France; (J.R.L.); (G.I.); (A.P.); (Q.M.)
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, UMONS Research Institute for Health Sciences and Technology, University of Mons (UMons), 7000 Mons, Belgium
- Department of Otolaryngology-Head & Neck Surgery, Foch Hospital, School of Medicine, UFR Simone Veil, Université Versailles Saint-Quentin-en-Yvelines (Paris Saclay University), 78180 Paris, France
- Department of Otolaryngology-Head & Neck Surgery, EpiCURA Hospital, 7301 Hornu, Belgium
| | - G. Sangiorgio
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy;
| | - G. Iannella
- Otology Study Group, Young Otolaryngologists-International Federation of Otorhinolaryngological Societies, 13005 Paris, France; (J.R.L.); (G.I.); (A.P.); (Q.M.)
- Department of ‘Organi di Senso’, University “Sapienza”, 00185 Rome, Italy;
| | - G. Magliulo
- Department of ‘Organi di Senso’, University “Sapienza”, 00185 Rome, Italy;
| | - A. Pace
- Otology Study Group, Young Otolaryngologists-International Federation of Otorhinolaryngological Societies, 13005 Paris, France; (J.R.L.); (G.I.); (A.P.); (Q.M.)
- Department of ‘Organi di Senso’, University “Sapienza”, 00185 Rome, Italy;
| | - Q. Mat
- Otology Study Group, Young Otolaryngologists-International Federation of Otorhinolaryngological Societies, 13005 Paris, France; (J.R.L.); (G.I.); (A.P.); (Q.M.)
- Department of Otorhinolaryngology, C.H.U. Charleroi, Chaussée de Bruxelles 140, 6042 Charleroi, Belgium
| | - S. Lavalle
- Faculty of Medicine and Surgery, University of Enna Kore, 94100 Enna, Italy;
| | - M. Lentini
- ASP Ragusa-Hospital Giovanni Paolo II, 97100 Ragusa, Italy;
| |
Collapse
|
3
|
Teraoka M, Hato N, Inufusa H, You F. Role of Oxidative Stress in Sensorineural Hearing Loss. Int J Mol Sci 2024; 25:4146. [PMID: 38673731 PMCID: PMC11050000 DOI: 10.3390/ijms25084146] [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: 02/20/2024] [Revised: 03/27/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Hearing is essential for communication, and its loss can cause a serious disruption to one's social life. Hearing loss is also recognized as a major risk factor for dementia; therefore, addressing hearing loss is a pressing global issue. Sensorineural hearing loss, the predominant type of hearing loss, is mainly due to damage to the inner ear along with a variety of pathologies including ischemia, noise, trauma, aging, and ototoxic drugs. In addition to genetic factors, oxidative stress has been identified as a common mechanism underlying several cochlear pathologies. The cochlea, which plays a major role in auditory function, requires high-energy metabolism and is, therefore, highly susceptible to oxidative stress, particularly in the mitochondria. Based on these pathological findings, the potential of antioxidants for the treatment of hearing loss has been demonstrated in several animal studies. However, results from human studies are insufficient, and future clinical trials are required. This review discusses the relationship between sensorineural hearing loss and reactive oxidative species (ROS), with particular emphasis on age-related hearing loss, noise-induced hearing loss, and ischemia-reperfusion injury. Based on these mechanisms, the current status and future perspectives of ROS-targeted therapy for sensorineural hearing loss are described.
Collapse
Affiliation(s)
- Masato Teraoka
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Ehime University, Toon 791-0295, Ehime, Japan;
| | - Naohito Hato
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Ehime University, Toon 791-0295, Ehime, Japan;
| | - Haruhiko Inufusa
- Division of Anti-Oxidant Research, Life Science Research Center, Gifu University, Yanagito 1-1, Gifu 501-1194, Japan; (H.I.); (F.Y.)
| | - Fukka You
- Division of Anti-Oxidant Research, Life Science Research Center, Gifu University, Yanagito 1-1, Gifu 501-1194, Japan; (H.I.); (F.Y.)
| |
Collapse
|
4
|
Nassauer L, Staecker H, Huang P, Renslo B, Goblet M, Harre J, Warnecke A, Schott JW, Morgan M, Galla M, Schambach A. Protection from cisplatin-induced hearing loss with lentiviral vector-mediated ectopic expression of the anti-apoptotic protein BCL-XL. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102157. [PMID: 38450280 PMCID: PMC10915631 DOI: 10.1016/j.omtn.2024.102157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 02/15/2024] [Indexed: 03/08/2024]
Abstract
Cisplatin is a highly effective chemotherapeutic agent, but it can cause sensorineural hearing loss (SNHL) in patients. Cisplatin-induced ototoxicity is closely related to the accumulation of reactive oxygen species (ROS) and subsequent death of hair cells (HCs) and spiral ganglion neurons (SGNs). Despite various strategies to combat ototoxicity, only one therapeutic agent has thus far been clinically approved. Therefore, we have developed a gene therapy concept to protect cochlear cells from cisplatin-induced toxicity. Self-inactivating lentiviral (LV) vectors were used to ectopically express various antioxidant enzymes or anti-apoptotic proteins to enhance the cellular ROS scavenging or prevent apoptosis in affected cell types. In direct comparison, anti-apoptotic proteins mediated a stronger reduction in cytotoxicity than antioxidant enzymes. Importantly, overexpression of the most promising candidate, Bcl-xl, achieved an up to 2.5-fold reduction in cisplatin-induced cytotoxicity in HEI-OC1 cells, phoenix auditory neurons, and primary SGN cultures. BCL-XL protected against cisplatin-mediated tissue destruction in cochlear explants. Strikingly, in vivo application of the LV BCL-XL vector improved hearing and increased HC survival in cisplatin-treated mice. In conclusion, we have established a preclinical gene therapy approach to protect mice from cisplatin-induced ototoxicity that has the potential to be translated to clinical use in cancer patients.
Collapse
Affiliation(s)
- Larissa Nassauer
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Hinrich Staecker
- Department of Otolaryngology-Head and Neck Surgery, University of Kansas School of Medicine, Kansas City, KS 66160, USA
| | - Peixin Huang
- Department of Otolaryngology-Head and Neck Surgery, University of Kansas School of Medicine, Kansas City, KS 66160, USA
| | - Bryan Renslo
- Department of Otolaryngology-Head and Neck Surgery, University of Kansas School of Medicine, Kansas City, KS 66160, USA
| | - Madeleine Goblet
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, 30625 Hannover, Germany
- Cluster of Excellence “Hearing4all”, Hannover Medical School, 30625 Hannover, Germany
| | - Jennifer Harre
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, 30625 Hannover, Germany
- Cluster of Excellence “Hearing4all”, Hannover Medical School, 30625 Hannover, Germany
| | - Athanasia Warnecke
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, 30625 Hannover, Germany
- Cluster of Excellence “Hearing4all”, Hannover Medical School, 30625 Hannover, Germany
| | - Juliane W. Schott
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Michael Morgan
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Melanie Galla
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
5
|
Han L, Wang Z, Wang D, Gao Z, Hu S, Shi D, Shu Y. Mechanisms and otoprotective strategies of programmed cell death on aminoglycoside-induced ototoxicity. Front Cell Dev Biol 2024; 11:1305433. [PMID: 38259515 PMCID: PMC10800616 DOI: 10.3389/fcell.2023.1305433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 12/14/2023] [Indexed: 01/24/2024] Open
Abstract
Aminoglycosides are commonly used for the treatment of life-threatening bacterial infections, however, aminoglycosides may cause irreversible hearing loss with a long-term clinical therapy. The mechanism and prevention of the ototoxicity of aminoglycosides are still limited although amounts of studies explored widely. Specifically, advancements in programmed cell death (PCD) provide more new perspectives. This review summarizes the general signal pathways in programmed cell death, including apoptosis, autophagy, and ferroptosis, as well as the mechanisms of aminoglycoside-induced ototoxicity. Additionally, novel interventions, especially gene therapy strategies, are also investigated for the prevention or treatment of aminoglycoside-induced hearing loss with prospective clinical applications.
Collapse
Affiliation(s)
- Lei Han
- Department of Otorhinolaryngology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Zijing Wang
- Department of Otorhinolaryngology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Daqi Wang
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Ziwen Gao
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Shaowei Hu
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Dazhi Shi
- Department of Otorhinolaryngology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yilai Shu
- Department of Otorhinolaryngology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Li H, Oh SH, Shin HC, Suh MW. Intratympanic Administration of Dieckol Prevents Ototoxic Hearing Loss. Mar Drugs 2022; 20:md20100622. [PMID: 36286446 PMCID: PMC9604621 DOI: 10.3390/md20100622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/22/2022] [Accepted: 09/24/2022] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE Systemic administration of dieckol reportedly ameliorates acute hearing loss. In this study, dieckol was delivered to the inner ear by the intratympanic route. The functional and anatomic effects and safety of dieckol were assessed using the rat ototoxicity model. MATERIALS AND METHODS Dieckol in a high-molecular-weight hyaluronic acid vehicle (dieckol+vehicle group) or vehicle without dieckol (vehicle-only group) were randomly delivered into 12 ears intratympanically. Ototoxic hearing loss was induced by intravenous administration of cisplatin, gentamicin, and furosemide. The hearing threshold and surviving outer hair cells (OHC) were enumerated. Biocompatibility was assessed by serial endoscopy of the tympanic membrane (TM), and the histology of the TM and the base of bulla (BB) mucosa was quantitatively assessed. RESULTS The hearing threshold was significantly better (difference of 20 dB SPL) in the dieckol+vehicle group than in the vehicle-only group. The number of surviving OHCs was significantly greater in the dieckol+vehicle group than in the vehicle-only group. There were no signs of inflammation or infection in the ear. The thickness of the TM and the BB mucosa did not differ between the two groups. CONCLUSION Intratympanic local delivery of dieckol may be a safe and effective method to prevent ototoxic hearing loss.
Collapse
Affiliation(s)
- Hui Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul 03080, Korea
| | - Seung Ha Oh
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul 03080, Korea
| | - Hyeon-Cheol Shin
- Center for Molecular Intelligence, The State University of New York, Incheon 21985, Korea
| | - Myung-Whan Suh
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul 03080, Korea
- Correspondence: ; Tel.: +82-2-2072-3649; Fax: +82-2-745-2387
| |
Collapse
|
7
|
Li M, Mu Y, Cai H, Wu H, Ding Y. Application of New Materials in Auditory Disease Treatment. Front Cell Neurosci 2022; 15:831591. [PMID: 35173583 PMCID: PMC8841849 DOI: 10.3389/fncel.2021.831591] [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: 12/08/2021] [Accepted: 12/22/2021] [Indexed: 11/16/2022] Open
Abstract
Auditory diseases are disabling public health problems that afflict a significant number of people worldwide, and they remain largely incurable until now. Driven by continuous innovation in the fields of chemistry, physics, and materials science, novel materials that can be applied to hearing diseases are constantly emerging. In contrast to conventional materials, new materials are easily accessible, inexpensive, non-invasive, with better acoustic therapy effects and weaker immune rejection after implantation. When new materials are used to treat auditory diseases, the wound healing, infection prevention, disease recurrence, hair cell regeneration, functional recovery, and other aspects have been significantly improved. Despite these advances, clinical success has been limited, largely due to issues regarding a lack of effectiveness and safety. With ever-developing scientific research, more novel materials will be facilitated into clinical use in the future.
Collapse
|
8
|
Nelson L, Johns JD, Gu S, Hoa M. Utilizing Single Cell RNA-Sequencing to Implicate Cell Types and Therapeutic Targets for SSNHL in the Adult Cochlea. Otol Neurotol 2021; 42:e1410-e1421. [PMID: 34510123 PMCID: PMC8595752 DOI: 10.1097/mao.0000000000003356] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To identify genes implicated in sudden sensorineural hearing loss (SSNHL) and localize their expression in the cochlea to further explore potential pathogenic mechanisms and therapeutic targets. STUDY DESIGN Systematic literature review and bioinformatics analysis. DATA SOURCES The following sources were searched from inception through July 2, 2020: PubMed-NCBI, MEDLINE, Embase, CINAHL, Cochrane Library, ClinicalTrials.gov, OpenGrey, GreyNet, GreyLiterature Report, and European Union Clinical Trials Registry. PubMed-NCBI and MEDLINE were additionally searched for human temporal bone histopathologic studies related to SSNHL. METHODS Literature review of candidate SSNHL genes was conducted according to PRISMA guidelines. Existing temporal bone studies from SSNHL patients were analyzed to identify the most commonly affected inner ear structures. Previously published single-cell and single-nucleus RNA-Seq datasets of the adult mouse stria vascularis, as well as postnatal day 7 and 15 mouse cochlear hair cells and supporting cells, were utilized for localization of the SSNHL-related genes curated through literature review. CONCLUSIONS We report 92 unique single nucleotide polymorphisms (SNPs) in 76 different genes that have been investigated in relation to SSNHL in the literature. We demonstrate that a subset of these genes are expressed by cell types in the adult mouse stria vascularis and organ of Corti, consistent with findings from temporal bone studies in human subjects with SSNHL. We highlight several potential genetic targets relevant to current and possible future SSNHL treatments.
Collapse
Affiliation(s)
- Lacey Nelson
- Georgetown University School of Medicine, Washington, D.C
| | - J. Dixon Johns
- Department of Otolaryngology–Head and Neck Surgery, Georgetown University Medical Center, Washington, DC
| | - Shoujun Gu
- Auditory Development and Restoration Program, National Institute on Deafness and Other Communication Disorders, Bethesda, MD
| | - Michael Hoa
- Department of Otolaryngology–Head and Neck Surgery, Georgetown University Medical Center, Washington, DC
- Auditory Development and Restoration Program, National Institute on Deafness and Other Communication Disorders, Bethesda, MD
| |
Collapse
|
9
|
Varela-Nieto I, Murillo-Cuesta S, Rodríguez-de la Rosa L, Oset-Gasque MJ, Marco-Contelles J. Use of Radical Oxygen Species Scavenger Nitrones to Treat Oxidative Stress-Mediated Hearing Loss: State of the Art and Challenges. Front Cell Neurosci 2021; 15:711269. [PMID: 34539349 PMCID: PMC8440819 DOI: 10.3389/fncel.2021.711269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/11/2021] [Indexed: 12/20/2022] Open
Abstract
Nitrones are potent antioxidant molecules able to reduce oxidative stress by trapping reactive oxygen and nitrogen species. The antioxidant potential of nitrones has been extensively tested in multiple models of human diseases. Sensorineural hearing loss has a heterogeneous etiology, genetic alterations, aging, toxins or exposure to noise can cause damage to hair cells at the organ of Corti, the hearing receptor. Noxious stimuli share a battery of common mechanisms by which they cause hair cell injury, including oxidative stress, the generation of free radicals and redox imbalance. Therefore, targeting oxidative stress-mediated hearing loss has been the subject of much attention. Here we review the chemistry of nitrones, the existing literature on their use as antioxidants and the general state of the art of antioxidant treatments for hearing loss.
Collapse
Affiliation(s)
- Isabel Varela-Nieto
- Institute for Biomedical Research “Alberto Sols,” Spanish National Research Council (CSIC)-Autonomous University of Madrid, Madrid, Spain
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain
- Hospital La Paz Institute for Health Research, Madrid, Spain
| | - Silvia Murillo-Cuesta
- Institute for Biomedical Research “Alberto Sols,” Spanish National Research Council (CSIC)-Autonomous University of Madrid, Madrid, Spain
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain
- Hospital La Paz Institute for Health Research, Madrid, Spain
| | - Lourdes Rodríguez-de la Rosa
- Institute for Biomedical Research “Alberto Sols,” Spanish National Research Council (CSIC)-Autonomous University of Madrid, Madrid, Spain
- Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain
- Hospital La Paz Institute for Health Research, Madrid, Spain
| | - María Jesús Oset-Gasque
- Department of Biochemistry and Molecular Biology, School of Pharmacy, Complutense University of Madrid, Madrid, Spain
- Institute of Neurochemistry Research, Complutense University of Madrid, Madrid, Spain
| | - José Marco-Contelles
- Laboratory of Medicinal Chemistry, Institute of General Organic Chemistry, CSIC, Madrid, Spain
| |
Collapse
|
10
|
SOD2 Alleviates Hearing Loss Induced by Noise and Kanamycin in Mitochondrial DNA4834-deficient Rats by Regulating PI3K/MAPK Signaling. Curr Med Sci 2021; 41:587-596. [PMID: 34169429 DOI: 10.1007/s11596-021-2376-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 02/16/2021] [Indexed: 10/21/2022]
Abstract
Superoxide dismutase 2 (SOD2)-mediated gene therapy has significant protective effects against kanamycin-induced hearing loss and hair cell loss in the inner ear, but the underlying mechanisms are still unclear. Herein, an in vivo aging model of mitochondrial DNA (mtDNA)4834 deletion mutation was established using D-galactose, and the effects of noise or kanamycin on inner ear injury was investigated. Rats subjected to mtDNA4834 mutation via D-galactose administration showed hearing loss characterized by the disruption of inner ear structure (abnormal cell morphology, hair cell lysis, and the absence of the organ of Corti), increased SOD2 promoter methylation, and an increase in the degree of apoptosis. Exposure to noise or kanamycin further contributed to the effects of D-galactose. SOD2 overexpression induced by viral injection accordingly counteracted the effects of noise and kanamycin and ameliorated the symptoms of hearing loss, suggesting the critical involvement of SOD2 in preventing deafness and hearing-related conditions. The PI3K and MAPK signaling pathways were also regulated by noise/kanamycin exposure and/or SOD2 overexpression, indicating that they may be involved in the therapeutic effect of SOD2 against age-related hearing loss.
Collapse
|
11
|
Crane R, Conley SM, Al-Ubaidi MR, Naash MI. Gene Therapy to the Retina and the Cochlea. Front Neurosci 2021; 15:652215. [PMID: 33815052 PMCID: PMC8010260 DOI: 10.3389/fnins.2021.652215] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/22/2021] [Indexed: 12/20/2022] Open
Abstract
Vision and hearing disorders comprise the most common sensory disorders found in people. Many forms of vision and hearing loss are inherited and current treatments only provide patients with temporary or partial relief. As a result, developing genetic therapies for any of the several hundred known causative genes underlying inherited retinal and cochlear disorders has been of great interest. Recent exciting advances in gene therapy have shown promise for the clinical treatment of inherited retinal diseases, and while clinical gene therapies for cochlear disease are not yet available, research in the last several years has resulted in significant advancement in preclinical development for gene delivery to the cochlea. Furthermore, the development of somatic targeted genome editing using CRISPR/Cas9 has brought new possibilities for the treatment of dominant or gain-of-function disease. Here we discuss the current state of gene therapy for inherited diseases of the retina and cochlea with an eye toward areas that still need additional development.
Collapse
Affiliation(s)
- Ryan Crane
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Shannon M. Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Oklahoma Center for Neurosciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Muayyad R. Al-Ubaidi
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
- College of Optometry, University of Houston, Houston, TX, United States
- Depatment of Biology and Biochemistry, University of Houston, Houston, TX, United States
| | - Muna I. Naash
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
- College of Optometry, University of Houston, Houston, TX, United States
- Depatment of Biology and Biochemistry, University of Houston, Houston, TX, United States
| |
Collapse
|
12
|
Bermúdez‐Muñoz JM, Celaya AM, Hijazo‐Pechero S, Wang J, Serrano M, Varela‐Nieto I. G6PD overexpression protects from oxidative stress and age-related hearing loss. Aging Cell 2020; 19:e13275. [PMID: 33222382 PMCID: PMC7744953 DOI: 10.1111/acel.13275] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 09/03/2020] [Accepted: 10/18/2020] [Indexed: 12/17/2022] Open
Abstract
Aging of the auditory system is associated with the incremental production of reactive oxygen species (ROS) and the accumulation of oxidative damage in macromolecules, which contributes to cellular malfunction, compromises cell viability, and, ultimately, leads to functional decline. Cellular detoxification relies in part on the production of NADPH, which is an important cofactor for major cellular antioxidant systems. NADPH is produced principally by the housekeeping enzyme glucose‐6‐phosphate dehydrogenase (G6PD), which catalyzes the rate‐limiting step in the pentose phosphate pathway. We show here that G6PD transgenic mice (G6PD‐Tg), which show enhanced constitutive G6PD activity and NADPH production along life, have lower auditory thresholds than wild‐type mice during aging, together with preserved inner hair cell (IHC) and outer hair cell (OHC), OHC innervation, and a conserved number of synapses per IHC. Gene expression of antioxidant enzymes was higher in 3‐month‐old G6PD‐Tg mice than in wild‐type counterparts, whereas the levels of pro‐apoptotic proteins were lower. Consequently, nitration of proteins, mitochondrial damage, and TUNEL+ apoptotic cells were all lower in 9‐month‐old G6PD‐Tg than in wild‐type counterparts. Unexpectedly, G6PD overexpression triggered low‐grade inflammation that was effectively resolved in young mice, as shown by the absence of cochlear cellular damage and macrophage infiltration. Our results lead us to propose that NADPH overproduction from an early stage is an efficient mechanism to maintain the balance between the production of ROS and cellular detoxification power along aging and thus prevents hearing loss progression.
Collapse
Affiliation(s)
- Jose M. Bermúdez‐Muñoz
- Institute for Biomedical Research “Alberto Sols” (IIBM) Spanish National Research Council‐Autonomous University of Madrid (CSIC‐UAM Madrid Spain
- Rare Diseases Networking Biomedical Research Centre (CIBERER) CIBER Carlos III Institute of Health Madrid Spain
| | - Adelaida M. Celaya
- Institute for Biomedical Research “Alberto Sols” (IIBM) Spanish National Research Council‐Autonomous University of Madrid (CSIC‐UAM Madrid Spain
- Rare Diseases Networking Biomedical Research Centre (CIBERER) CIBER Carlos III Institute of Health Madrid Spain
| | - Sara Hijazo‐Pechero
- Institute for Biomedical Research “Alberto Sols” (IIBM) Spanish National Research Council‐Autonomous University of Madrid (CSIC‐UAM Madrid Spain
| | - Jing Wang
- INSERM ‐ UMR 1051 Institut des Neurosciences de Montpellier Montpellier France
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB) Barcelona Spain
| | - Isabel Varela‐Nieto
- Institute for Biomedical Research “Alberto Sols” (IIBM) Spanish National Research Council‐Autonomous University of Madrid (CSIC‐UAM Madrid Spain
- Rare Diseases Networking Biomedical Research Centre (CIBERER) CIBER Carlos III Institute of Health Madrid Spain
- Hospital La Paz Institute for Health Research (IdiPAZ) Madrid Spain
| |
Collapse
|
13
|
Zhong Z, Fu X, Li H, Chen J, Wang M, Gao S, Zhang L, Cheng C, Zhang Y, Li P, Zhang S, Qian X, Shu Y, Chai R, Gao X. Citicoline Protects Auditory Hair Cells Against Neomycin-Induced Damage. Front Cell Dev Biol 2020; 8:712. [PMID: 32984303 PMCID: PMC7487320 DOI: 10.3389/fcell.2020.00712] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/13/2020] [Indexed: 01/07/2023] Open
Abstract
Aminoglycoside-induced hair cell (HC) loss is one of the most important causes of hearing loss. After entering the inner ear, aminoglycosides induce the production of high levels of reactive oxygen species (ROS) that subsequently activate apoptosis in HCs. Citicoline, a nucleoside derivative, plays a therapeutic role in central nervous system injury and in neurodegenerative disease models, including addictive disorders, stroke, head trauma, and cognitive impairment in the elderly, and has been widely used in the clinic as an FDA approved drug. However, its effect on auditory HCs remains unknown. Here, we used HC-like HEI-OC-1 cells and whole organ explant cultured mouse cochleae to explore the effect of citicoline on aminoglycoside-induced HC damage. Consistent with previous reports, both ROS levels and apoptosis were significantly increased in neomycin-induced cochlear HCs and HEI-OC-1 cells compared to undamaged controls. Interestingly, we found that co-treatment with citicoline significantly protected against neomycin-induced HC loss in both HEI-OC-1 cells and whole organ explant cultured cochleae. Furthermore, we demonstrated that citicoline could significantly reduce neomycin-induced mitochondrial dysfunction and inhibit neomycin-induced ROS accumulation and subsequent apoptosis. Thus, we conclude that citicoline can protect against neomycin-induced HC loss by inhibiting ROS aggregation and thus preventing apoptosis in HCs, and this suggests that citicoline might serve as a potential therapeutic drug in the clinic to protect HCs.
Collapse
Affiliation(s)
- Zhenhua Zhong
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China.,Department of Otolaryngology, Head and Neck Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Xiaolong Fu
- MOE Key Laboratory for Developmental Genes and Human Disease, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Institute of Life Sciences, Southeast University, Nanjing, China
| | - He Li
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jie Chen
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Maohua Wang
- Department of Otolaryngology, Head and Neck Surgery, Xiangya School of Medicine, Central South University, Changsha, China
| | - Song Gao
- Department of Otolaryngology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Liyan Zhang
- MOE Key Laboratory for Developmental Genes and Human Disease, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Cheng Cheng
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Yuan Zhang
- MOE Key Laboratory for Developmental Genes and Human Disease, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Peipei Li
- School of Life Sciences, Shandong University, Jinan, China
| | - Shasha Zhang
- MOE Key Laboratory for Developmental Genes and Human Disease, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Xiaoyun Qian
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Yilai Shu
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Institute of Biomedical Sciences, NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Renjie Chai
- MOE Key Laboratory for Developmental Genes and Human Disease, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Institute of Life Sciences, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Xia Gao
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
Wang H, Zhao H, Sun K, Huang X, Jin L, Feng J. Evolutionary Basis of High-Frequency Hearing in the Cochleae of Echolocators Revealed by Comparative Genomics. Genome Biol Evol 2020; 12:3740-3753. [PMID: 31730196 PMCID: PMC7145703 DOI: 10.1093/gbe/evz250] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2019] [Indexed: 12/25/2022] Open
Abstract
High-frequency hearing is important for the survival of both echolocating bats and whales, but our understanding of its genetic basis is scattered and segmented. In this study, we combined RNA-Seq and comparative genomic analyses to obtain insights into the comprehensive gene expression profile of the cochlea and the adaptive evolution of hearing-related genes. A total of 144 genes were found to have been under positive selection in various species of echolocating bats and toothed whales, 34 of which were identified to be related to hearing behavior or auditory processes. Subsequently, multiple physiological processes associated with those genes were found to have adaptively evolved in echolocating bats and toothed whales, including cochlear bony development, antioxidant activity, ion balance, and homeostatic processes, along with signal transduction. In addition, abundant convergent/parallel genes and sites were detected between different pairs of echolocator species; however, no specific hearing-related physiological pathways were enriched by them and almost all of the convergent/parallel signals were selectively neutral, as previously reported. Notably, two adaptive parallel evolved sites in TECPR2 were shown to have been under positive selection, indicating their functional importance for the evolution of echolocation and high-frequency hearing in laryngeal echolocating bats. This study deepens our understanding of the genetic bases underlying high-frequency hearing in the cochlea of echolocating bats and toothed whales.
Collapse
Affiliation(s)
- Hui Wang
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China.,College of Life Science, Jilin Agricultural University, Changchun, China
| | - Hanbo Zhao
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China
| | - Keping Sun
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China
| | - Xiaobin Huang
- Vector Laboratory for Zoonosis Control and Prevention, Dali University, China
| | - Longru Jin
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China
| | - Jiang Feng
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China.,College of Life Science, Jilin Agricultural University, Changchun, China
| |
Collapse
|
17
|
Romito M, Pu Y, Stankovic KM, Psaltis D. Imaging hair cells through laser-ablated cochlear bone. BIOMEDICAL OPTICS EXPRESS 2019; 10:5974-5988. [PMID: 31799058 PMCID: PMC6865115 DOI: 10.1364/boe.10.005974] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/29/2019] [Accepted: 10/24/2019] [Indexed: 05/04/2023]
Abstract
We report an innovative technique for the visualization of cells through an overlying scattering medium by combining femtosecond laser bone ablation and two-photon excitation fluorescence (TPEF) microscopy. We demonstrate the technique by imaging hair cells in an intact mouse cochlea ex vivo. Intracochlear imaging is important for the assessment of hearing disorders. However, the small size of the cochlea and its encasement in the densest bone in the body present challenging obstacles, preventing the visualization of the intracochlear microanatomy using standard clinical imaging modalities. The controlled laser ablation reduces the optical scattering of the cochlear bone while the TPEF allows visualization of individual cells behind the bone. We implemented optical coherence tomography (OCT) simultaneously with the laser ablation to enhance the precision of the ablation and prevent inadvertent damage to the cells behind the bone.
Collapse
Affiliation(s)
- Marilisa Romito
- Optics Laboratory, School of Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Ye Pu
- Optics Laboratory, School of Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Konstantina M. Stankovic
- Department of Otolaryngology – Head and Neck Surgery, Harvard Medical School and Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Demetri Psaltis
- Optics Laboratory, School of Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|
18
|
Zhang S, Gao Y, He Q, Zhang Y, Han L, Jin M, Liu T, Liu K, Sun C. A new active peptide from Neptunea arthritica cumingii exerts protective effects against gentamicin-induced sensory-hair cell injury in zebrafish. Drug Chem Toxicol 2019; 45:161-169. [PMID: 31533491 DOI: 10.1080/01480545.2019.1665680] [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] [Indexed: 12/13/2022]
Abstract
Gentamicin is commonly used for effective treatment of severe Gram-negative bacterial infections. However, its use is being increasingly restricted owing to the ototoxic effects attributed to it. Gentamicin-induced ototoxicity is thought to be related with apoptosis induced by reactive oxygen species (ROS). In this study, we found a novel active peptide from Neptunea arthritica cumingii with otoprotective effects and no significant embryotoxic effects. The combined application of gentamicin and this novel active peptide helped sensory-hair cells to protect themselves from lethal ROS accumulation. This, in turn, reduced the expression of three genes (caspase-3, caspase-9, Bax), and thereby, the sensory-hair cell apoptosis promoted by ROS accumulation upon gentamicin administration. Our findings provided new insights into the prevention of gentamicin-induced hearing loss.
Collapse
Affiliation(s)
- Shanshan Zhang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Key Laboratory for Biosensors of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences) , Jinan , Shandong Province , PR China
| | - Yan Gao
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Key Laboratory for Biosensors of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences) , Jinan , Shandong Province , PR China
| | - Qiuxia He
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Key Laboratory for Biosensors of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences) , Jinan , Shandong Province , PR China
| | - Yun Zhang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Key Laboratory for Biosensors of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences) , Jinan , Shandong Province , PR China
| | - Liwen Han
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Key Laboratory for Biosensors of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences) , Jinan , Shandong Province , PR China
| | - Meng Jin
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Key Laboratory for Biosensors of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences) , Jinan , Shandong Province , PR China
| | - Tong Liu
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Key Laboratory for Biosensors of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences) , Jinan , Shandong Province , PR China
| | - Kechun Liu
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Key Laboratory for Biosensors of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences) , Jinan , Shandong Province , PR China
| | - Chen Sun
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Key Laboratory for Biosensors of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences) , Jinan , Shandong Province , PR China
| |
Collapse
|
19
|
Pickett SB, Raible DW. Water Waves to Sound Waves: Using Zebrafish to Explore Hair Cell Biology. J Assoc Res Otolaryngol 2019; 20:1-19. [PMID: 30635804 DOI: 10.1007/s10162-018-00711-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 12/19/2018] [Indexed: 01/09/2023] Open
Abstract
Although perhaps best known for their use in developmental studies, over the last couple of decades, zebrafish have become increasingly popular model organisms for investigating auditory system function and disease. Like mammals, zebrafish possess inner ear mechanosensory hair cells required for hearing, as well as superficial hair cells of the lateral line sensory system, which mediate detection of directional water flow. Complementing mammalian studies, zebrafish have been used to gain significant insights into many facets of hair cell biology, including mechanotransduction and synaptic physiology as well as mechanisms of both hereditary and acquired hair cell dysfunction. Here, we provide an overview of this literature, highlighting some of the particular advantages of using zebrafish to investigate hearing and hearing loss.
Collapse
Affiliation(s)
- Sarah B Pickett
- Department of Biological Structure, University of Washington, Health Sciences Building H-501, 1959 NE Pacific Street, Box 357420, Seattle, WA, 98195-7420, USA
- Graduate Program in Neuroscience, University of Washington, 1959 NE Pacific Street, Box 357270, Seattle, WA, 98195-7270, USA
| | - David W Raible
- Department of Biological Structure, University of Washington, Health Sciences Building H-501, 1959 NE Pacific Street, Box 357420, Seattle, WA, 98195-7420, USA.
- Graduate Program in Neuroscience, University of Washington, 1959 NE Pacific Street, Box 357270, Seattle, WA, 98195-7270, USA.
- Virginia Merrill Bloedel Hearing Research Center, University of Washington, 1701 NE Columbia Rd, Box 357923, Seattle, WA, 98195-7923, USA.
| |
Collapse
|
20
|
Vitamin C alleviates ototoxic effect caused by coadministration of amikacin and furosemide. Pharmacol Rep 2019; 71:351-356. [PMID: 30831441 DOI: 10.1016/j.pharep.2019.01.002] [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: 05/26/2018] [Revised: 12/05/2018] [Accepted: 01/04/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Drug-induced ototoxicity is still a main clinical problem in otolaryngology. It is widely known that aminoglycoside antibiotics combined with loop diuretics significantly contribute to permanent ototoxicity. The aim of this study was to find out whether ascorbic acid (vitamin C) is able to reverse or alleviate ototoxicity evoked by systemic (ip) administration of combination of amikacin and furosemide in experimental male albino Swiss mice. METHODS Ototoxic combination of amikacin and furosemide was isobolographically evaluated based on the hearing threshold decreasing doses by 20% and 50% (TDD20 and TDD50), respectively. Linear regression analysis was used to determine the TDD20 and TDD50 values for amikacin, furosemide, vitamin C administered alone and in combination (at the fixed-ratio of 1:1). RESULTS Vitamin C (in a dose of 500 mg/kg, ip) alleviated the impairment in hearing threshold evoked by combined ip administration of amikacin and furosemide (at the fixed-ratio of 1:1) in mice by reducing TDD50 values from 49.82 to 21.56 (p < 0.01). In contrast, vitamin C (500 mg/kg, ip) had no significant effect on TDD20 values for the combination of amikacin and furosemide at the fixed-ratio of 1:1. CONCLUSIONS Vitamin C administered together with ototoxic drug combination of amikacin and furosemide reduced ototoxicity evoked by this two-drug combination in the experimental mice.
Collapse
|
21
|
Puia-Dumitrescu M, Bretzius OM, Brown N, Fitz-Henley JA, Ssengonzi R, Wechsler CS, Gray KD, Benjamin DK, Smith PB, Clark RH, Gonzalez D, Hornik CP. Evaluation of Gentamicin Exposure in the Neonatal Intensive Care Unit and Hearing Function at Discharge. J Pediatr 2018; 203:131-136. [PMID: 30244991 PMCID: PMC6361629 DOI: 10.1016/j.jpeds.2018.07.101] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/25/2018] [Accepted: 07/30/2018] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To characterize the association between gentamicin dosing, duration of treatment, and ototoxicity in hospitalized infants. STUDY DESIGN This retrospective cohort study conducted at 330 neonatal intensive care units (2002-2014) included inborn infants exposed to gentamicin with available hearing screen results, and excluded infants with incomplete dosing data and major congenital anomalies. Our primary outcome was the final hearing screen result performed during hospitalization: abnormal (failed or referred for further testing in one or both ears) or normal (bilateral passed). The 4 measures of gentamicin exposure were highest daily dose, average daily dose, cumulative dose, and cumulative duration of exposure. We fitted separate multivariable logistic regression models adjusted for demographics, comorbidities, and other clinical events. RESULTS A total of 84 808 infants met inclusion/exclusion criteria; median (25th, 75th percentile) gestational age and birth weight were 35 weeks (33, 38) and 2480 g (1890, 3184), respectively. Failed hearing screens occurred in 3238 (3.8%) infants; failed screens were more likely in infants of lower gestational age and birth weight, who had longer hospital lengths of stay, higher rates of morbidities, and were small for gestational age. Median highest daily dose, average daily dose, and cumulative dose were 4.0 mg/kg/day (3.0, 4.0), 3.8 mg/kg/day (3.0, 4.0), and 12.1 mg/kg (9.1, 20.5), respectively. Median cumulative duration of exposure was 3 days (3, 6). In adjusted analysis, gentamicin dose and duration of therapy were not associated with hearing screen failure. CONCLUSIONS Gentamicin dosing and duration of treatment were not associated with increased odds of failed hearing screen at the time of discharge from initial neonatal intensive care unit stay.
Collapse
Affiliation(s)
- Mihai Puia-Dumitrescu
- Department of Pediatrics, Duke University Medical Center, Durham, NC; Duke Clinical Research Institute, Durham, NC
| | | | - Nia Brown
- Duke Clinical Research Institute, Durham, NC
| | | | | | | | - Keyaria D Gray
- Department of Pediatrics, Duke University Medical Center, Durham, NC
| | | | - P Brian Smith
- Department of Pediatrics, Duke University Medical Center, Durham, NC; Duke Clinical Research Institute, Durham, NC
| | - Reese H Clark
- Pediatrix-Obstetrix Center for Research and Education, Sunrise, FL
| | - Daniel Gonzalez
- Department of Pediatrics, University of North Carolina, Chapel Hill, NC
| | - Christoph P Hornik
- Department of Pediatrics, Duke University Medical Center, Durham, NC; Duke Clinical Research Institute, Durham, NC.
| |
Collapse
|
22
|
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: 2.0] [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
|
23
|
Wu X, Li X, Song Y, Li H, Bai X, Liu W, Han Y, Xu L, Li J, Zhang D, Wang H, Fan Z. Allicin protects auditory hair cells and spiral ganglion neurons from cisplatin - Induced apoptosis. Neuropharmacology 2017; 116:429-440. [DOI: 10.1016/j.neuropharm.2017.01.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 12/02/2016] [Accepted: 01/02/2017] [Indexed: 12/18/2022]
|
24
|
György B, Sage C, Indzhykulian AA, Scheffer DI, Brisson AR, Tan S, Wu X, Volak A, Mu D, Tamvakologos PI, Li Y, Fitzpatrick Z, Ericsson M, Breakefield XO, Corey DP, Maguire CA. Rescue of Hearing by Gene Delivery to Inner-Ear Hair Cells Using Exosome-Associated AAV. Mol Ther 2017; 25:379-391. [PMID: 28082074 DOI: 10.1016/j.ymthe.2016.12.010] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 11/29/2016] [Accepted: 12/05/2016] [Indexed: 12/12/2022] Open
Abstract
Adeno-associated virus (AAV) is a safe and effective vector for gene therapy for retinal disorders. Gene therapy for hearing disorders is not as advanced, in part because gene delivery to sensory hair cells of the inner ear is inefficient. Although AAV transduces the inner hair cells of the mouse cochlea, outer hair cells remain refractory to transduction. Here, we demonstrate that a vector, exosome-associated AAV (exo-AAV), is a potent carrier of transgenes to all inner ear hair cells. Exo-AAV1-GFP is more efficient than conventional AAV1-GFP, both in mouse cochlear explants in vitro and with direct cochlear injection in vivo. Exo-AAV shows no toxicity in vivo, as assayed by tests of auditory and vestibular function. Finally, exo-AAV1 gene therapy partially rescues hearing in a mouse model of hereditary deafness (lipoma HMGIC fusion partner-like 5/tetraspan membrane protein of hair cell stereocilia [Lhfpl5/Tmhs-/-]). Exo-AAV is a powerful gene delivery system for hair cell research and may be useful for gene therapy for deafness.
Collapse
Affiliation(s)
- Bence György
- Department of Neurobiology and Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Building 149, Charlestown, Boston, MA 02129, USA
| | - Cyrille Sage
- Department of Neurobiology and Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Artur A Indzhykulian
- Department of Neurobiology and Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Deborah I Scheffer
- Department of Neurobiology and Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Alain R Brisson
- Molecular Imaging and NanoBioTechnology, UMR-5248-CBMN CNRS-University of Bordeaux-IPB, Allée Geoffroy Saint-Hilaire, F-33600 Pessac, France
| | - Sisareuth Tan
- Molecular Imaging and NanoBioTechnology, UMR-5248-CBMN CNRS-University of Bordeaux-IPB, Allée Geoffroy Saint-Hilaire, F-33600 Pessac, France
| | - Xudong Wu
- Department of Neurobiology and Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Adrienn Volak
- Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Building 149, Charlestown, Boston, MA 02129, USA
| | - Dakai Mu
- Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Building 149, Charlestown, Boston, MA 02129, USA
| | - Panos I Tamvakologos
- Department of Neurobiology and Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Yaqiao Li
- Department of Neurobiology and Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Zachary Fitzpatrick
- Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Building 149, Charlestown, Boston, MA 02129, USA
| | - Maria Ericsson
- Department of Cell Biology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Xandra O Breakefield
- Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Building 149, Charlestown, Boston, MA 02129, USA; Program in Neuroscience, Harvard Medical School, Building 149, Charlestown, Boston, MA 02129, USA
| | - David P Corey
- Department of Neurobiology and Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA.
| | - Casey A Maguire
- Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Building 149, Charlestown, Boston, MA 02129, USA.
| |
Collapse
|
25
|
Richardson R, Smart M, Tracey-White D, Webster AR, Moosajee M. Mechanism and evidence of nonsense suppression therapy for genetic eye disorders. Exp Eye Res 2017; 155:24-37. [PMID: 28065590 DOI: 10.1016/j.exer.2017.01.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 11/24/2016] [Accepted: 01/04/2017] [Indexed: 01/09/2023]
Abstract
Between 5 and 70% of genetic disease is caused by in-frame nonsense mutations, which introduce a premature termination codon (PTC) within the disease-causing gene. Consequently, during translation, non-functional or gain-of-function truncated proteins of pathological significance, are formed. Approximately 50% of all inherited retinal disorders have been associated with PTCs, highlighting the importance of novel pharmacological or gene correction therapies in ocular disease. Pharmacological nonsense suppression of PTCs could delineate a therapeutic strategy that treats the mutation in a gene- and disease-independent manner. This approach aims to suppress the fidelity of the ribosome during protein synthesis so that a near-cognate aminoacyl-tRNA, which shares two of the three nucleotides of the PTC, can be inserted into the peptide chain, allowing translation to continue, and a full-length functional protein to be produced. Here we discuss the mechanisms and evidence of nonsense suppression agents, including the small molecule drug ataluren (or PTC124) and next generation 'designer' aminoglycosides, for the treatment of genetic eye disease.
Collapse
Affiliation(s)
- Rose Richardson
- Department of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology, London, UK
| | - Matthew Smart
- Department of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology, London, UK
| | - Dhani Tracey-White
- Department of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology, London, UK
| | - Andrew R Webster
- Department of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology, London, UK; Moorfields Eye Hospital NHS Foundation Trust, London, UK
| | - Mariya Moosajee
- Department of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology, London, UK; Moorfields Eye Hospital NHS Foundation Trust, London, UK.
| |
Collapse
|
26
|
Tavanai E, Mohammadkhani G. Role of antioxidants in prevention of age-related hearing loss: a review of literature. Eur Arch Otorhinolaryngol 2016; 274:1821-1834. [PMID: 27858145 DOI: 10.1007/s00405-016-4378-6] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 11/08/2016] [Indexed: 01/21/2023]
Abstract
Age-related hearing loss (ARHL), also known as presbycusis, is one of the most prevalent chronic degenerative conditions. It is characterized by a decline in auditory function. ARHL is caused by the interaction of multiple factors, including cochlear aging, environment, genetic predisposition, and health comorbidities. The primary pathology of ARHL includes the hair cells loss, stria vascularis atrophy, and loss of spiral ganglion neurons as well as the changes in central auditory pathways. The research to date suggests that oxidative stress and mitochondrial DNA deletion (mtDNA) play a major role in pathophysiology of ARHL. Therefore, similar to other otological conditions, several studies have also showed that antioxidants can slow ARHL, but some also indicate that antioxidant therapy is not a magic elixir that will prevent or treat hearing loss associated with aging completely, but why? All available clinical trials, including animal and human studies, in English language that examined the protective effects of antioxidants against ARHL were reviewed. Materials were obtained by searching ELSEVIER, PubMed, Scopus, Web of knowledge, Google Scholar databases, Clinical trials, and Cochrane database of systematic reviews. Although ARHL has been shown to be slowed by supplementation with antioxidants, particularly in laboratory animals, a few studies have investigated the effect of interventions against ARHL in humans. High-quality clinical trials are needed to investigate if ARHL can be delayed or prevented in humans. However, it seems that targeting several cell-death pathways is better than targeting the only oxidative stress pathway.
Collapse
Affiliation(s)
- Elham Tavanai
- Department of Audiology, School of Rehabilitation, Tehran University of Medical Sciences, Piche-Shemiran, Enghelab Ave, Tehran, Iran
| | - Ghassem Mohammadkhani
- Department of Audiology, School of Rehabilitation, Tehran University of Medical Sciences, Piche-Shemiran, Enghelab Ave, Tehran, Iran.
| |
Collapse
|
27
|
Kim YJ, Kim J, Kim YS, Shin B, Choo OS, Lee JJ, Choung YH. Connexin 43 Acts as a Proapoptotic Modulator in Cisplatin-Induced Auditory Cell Death. Antioxid Redox Signal 2016; 25:623-636. [PMID: 27122099 DOI: 10.1089/ars.2015.6412] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
AIMS Gap junction coupling is known to play a role in intercellular communication by the Good Samaritan effect or bystander effect. Nonjunctional connexins (Cxs) may also play certain gap junction-independent roles in cell death or survival. The purpose of the present study was to investigate the role of junctional and nonjunctional Cxs in ototoxic drug-induced auditory cell death by focusing on Cx43 in the cochlea. RESULTS Nonjunctional Cx43 conditions were prepared by low confluence culture (5 × 103/cm2) or a trafficking inhibitor, brefeldin A (BFA), in auditory cells, and short lengthened Cx43s with amino-terminal (NT; amino acids 1-256) or carboxy-terminal (CT; amino acids 257-382) were transfected into Cx-deficient HeLa cells to avoid gap junction formation. Knockdown of nonchannel Cx43 (small interfering RNA [siRNA]) inhibited Cis-diamminedichloroplatinum (cisplatin)-induced cell death regardless of gap junction formation; however, a gap junction blocker, 18 alpha-glycyrrhetinic acid (18α-GA), showed inhibitory effect only under the junctional condition. BFA did not show any additive influence on the inhibitory effect of siRNA Cx43. Shortened Cx43-transfected HeLa cells also resulted in a significant increase in cell death under cisplatin. In the animal studies with cisplatin-treated rats, hearing thresholds of auditory brainstem response were significantly preserved by a gap junction blocker, carbenoxolone, showing much more preserved stereocilia of hair cells in scanning electron microscopic findings. Innovation and Conclusion: Cx43 plays a proapoptotic role in cisplatin-induced auditory cell death in both junctional and nonjunctional conditions. Targeting the Cx-mediated signaling control may be helpful in designing new therapeutic strategies for drug-induced ototoxicity. Antioxid. Redox Signal. 25, 623-636.
Collapse
Affiliation(s)
- Yeon Ju Kim
- 1 Department of Otolaryngology, Ajou University School of Medicine , Suwon, Republic of Korea
| | - Jangho Kim
- 2 Department of Rural and Biosystems Engineering, Chonnam National University , Gwangju, Republic of Korea
| | - Young Sun Kim
- 1 Department of Otolaryngology, Ajou University School of Medicine , Suwon, Republic of Korea
| | - Beomyong Shin
- 3 Department of Medical Sciences, The Graduate School, Ajou University , Suwon, Republic of Korea
| | - Oak-Sung Choo
- 1 Department of Otolaryngology, Ajou University School of Medicine , Suwon, Republic of Korea
| | - Jong Joo Lee
- 1 Department of Otolaryngology, Ajou University School of Medicine , Suwon, Republic of Korea
| | - Yun-Hoon Choung
- 1 Department of Otolaryngology, Ajou University School of Medicine , Suwon, Republic of Korea.,3 Department of Medical Sciences, The Graduate School, Ajou University , Suwon, Republic of Korea
| |
Collapse
|
28
|
Esterberg R, Linbo T, Pickett SB, Wu P, Ou HC, Rubel EW, Raible DW. Mitochondrial calcium uptake underlies ROS generation during aminoglycoside-induced hair cell death. J Clin Invest 2016; 126:3556-66. [PMID: 27500493 DOI: 10.1172/jci84939] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 06/09/2016] [Indexed: 12/11/2022] Open
Abstract
Exposure to aminoglycoside antibiotics can lead to the generation of toxic levels of reactive oxygen species (ROS) within mechanosensory hair cells of the inner ear that have been implicated in hearing and balance disorders. Better understanding of the origin of aminoglycoside-induced ROS could focus the development of therapies aimed at preventing this event. In this work, we used the zebrafish lateral line system to monitor the dynamic behavior of mitochondrial and cytoplasmic oxidation occurring within the same dying hair cell following exposure to aminoglycosides. The increased oxidation observed in both mitochondria and cytoplasm of dying hair cells was highly correlated with mitochondrial calcium uptake. Application of the mitochondrial uniporter inhibitor Ru360 reduced mitochondrial and cytoplasmic oxidation, suggesting that mitochondrial calcium drives ROS generation during aminoglycoside-induced hair cell death. Furthermore, targeting mitochondria with free radical scavengers conferred superior protection against aminoglycoside exposure compared with identical, untargeted scavengers. Our findings suggest that targeted therapies aimed at preventing mitochondrial oxidation have therapeutic potential to ameliorate the toxic effects of aminoglycoside exposure.
Collapse
|
29
|
Taura A, Taura K, Koyama Y, Yamamoto N, Nakagawa T, Ito J, Ryan AF. Hair cell stereociliary bundle regeneration by espin gene transduction after aminoglycoside damage and hair cell induction by Notch inhibition. Gene Ther 2016; 23:415-23. [PMID: 26886463 PMCID: PMC4860107 DOI: 10.1038/gt.2016.12] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/27/2015] [Accepted: 10/23/2015] [Indexed: 12/20/2022]
Abstract
Once inner ear hair cells (HCs) are damaged by drugs, noise or aging, their apical structures including the stereociliary arrays are frequently the first cellular feature to be lost. Although this can be followed by progressive loss of HC somata, a significant number of HC bodies often remain even after stereociliary loss. However, in the absence of stereocilia they are nonfunctional. HCs can sometimes be regenerated by Atoh1 transduction or Notch inhibition, but they also may lack stereociliary bundles. It is therefore important to develop methods for the regeneration of stereocilia, in order to achieve HC functional recovery. Espin is an actin-bundling protein known to participate in sterociliary elongation during development. We evaluated stereociliary array regeneration in damaged vestibular sensory epithelia in tissue culture, using viral vector transduction of two espin isoforms. Utricular HCs were damaged with aminoglycosides. The utricles were then treated with a γ-secretase inhibitor, followed by espin or control transduction and histochemistry. Although γ-secretase inhibition increased the number of HCs, few had stereociliary arrays. In contrast, 46 h after espin1 transduction, a significant increase in hair-bundle-like structures was observed. These were confirmed to be immature stereociliary arrays by scanning electron microscopy. Increased uptake of FM1-43 uptake provided evidence of stereociliary function. Espin4 transduction had no effect. The results demonstrate that espin1 gene therapy can restore stereocilia on damaged or regenerated HCs.
Collapse
Affiliation(s)
- A Taura
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - K Taura
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Y Koyama
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - N Yamamoto
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - T Nakagawa
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - J Ito
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - A F Ryan
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Divison of Otolaryngology, Department of Surgery, UCSD School of Medicine, La Jolla, CA, USA.,VA Medical Center, San Diego, CA, USA.,Department of Neurosciences, UCSD School of Medicine, La Jolla, CA, USA
| |
Collapse
|
30
|
Kelly KM, Lalwani AK. On the Distant Horizon--Medical Therapy for Sensorineural Hearing Loss. Otolaryngol Clin North Am 2015; 48:1149-65. [PMID: 26409822 DOI: 10.1016/j.otc.2015.07.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Hearing loss is the most common sensory deficit in developed societies. Hearing impairment in children, particularly of prelingual onset, has been shown to negatively affect educational achievement, future employment and earnings, and even life expectancy. Sensorineural hearing loss (SNHL), which refers to defects within the cochlea or auditory nerve itself, far outweighs conductive causes for permanent hearing loss in both children and adults. The causes of SNHL in children are heterogeneous, including both congenital and acquired causes. This article identifies potential mechanisms of intervention both at the level of the hair cell and the spiral ganglion neurons.
Collapse
Affiliation(s)
- Kathleen M Kelly
- Department of Otolaryngology - Head and Neck Surgery, University of Texas Southwestern Medical Center, 5323 Harry Hinds Blvd, Dallas, TX 75390, USA
| | - Anil K Lalwani
- Department of Otolaryngology - Head and Neck Surgery, Columbia University Medical Center, Harkness Pavilion, 180 Fort Washington Avenue, Floor 7, New York, NY 10032, USA.
| |
Collapse
|
31
|
Kurioka T, Mizutari K, Niwa K, Fukumori T, Inoue M, Hasegawa M, Shiotani A. Hyaluronic acid pretreatment for Sendai virus-mediated cochlear gene transfer. Gene Ther 2015; 23:187-95. [PMID: 26361273 DOI: 10.1038/gt.2015.94] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 08/19/2015] [Accepted: 09/04/2015] [Indexed: 12/14/2022]
Abstract
Gene therapy with viral vectors is one of the most promising strategies for sensorineural hearing loss. However, safe and effective administration of the viral vector into cochlear tissue is difficult because of the anatomical isolation of the cochlea. We investigated the efficiency and safety of round window membrane (RWM) application of Sendai virus, one of the most promising non-genotoxic vectors, after pretreatment with hyaluronic acid (HA) on the RWM to promote efficient viral translocation into the cochlea. Sendai virus expressing the green fluorescent protein reporter gene was detected throughout cochlear tissues following application combined with HA pretreatment. Quantitative analysis revealed that maximum expression was reached 3 days after treatment. The efficiency of transgene expression was several 100-fold greater with HA pretreatment than that without. Furthermore, unlike the conventional intracochlear delivery methods, this approach did not cause hearing loss. These findings reveal the potential utility of gene therapy with Sendai virus and HA for treatment of sensorineural hearing loss.
Collapse
Affiliation(s)
- T Kurioka
- Department of Otolaryngology, Head and Neck Surgery, National Defense Medical College, Saitama, Japan
| | - K Mizutari
- Department of Otolaryngology, Head and Neck Surgery, National Defense Medical College, Saitama, Japan
| | - K Niwa
- Department of Otolaryngology, Head and Neck Surgery, National Defense Medical College, Saitama, Japan
| | - T Fukumori
- Department of Otolaryngology, Head and Neck Surgery, National Defense Medical College, Saitama, Japan
| | - M Inoue
- DNAVEC Corporation, Ibaraki, Japan
| | | | - A Shiotani
- Department of Otolaryngology, Head and Neck Surgery, National Defense Medical College, Saitama, Japan
| |
Collapse
|
32
|
El-Barbary MN, Ismail RIH, Ibrahim AAA. Gentamicin extended interval regimen and ototoxicity in neonates. Int J Pediatr Otorhinolaryngol 2015; 79:1294-8. [PMID: 26071016 DOI: 10.1016/j.ijporl.2015.05.036] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 04/27/2015] [Accepted: 05/22/2015] [Indexed: 11/28/2022]
Abstract
OBJECTIVES To assess the extended interval regimen gentamicin associated ototoxicity in neonatal intensive care unit using hearing tests. METHODS Two hundred and twenty neonates admitted to neonatal intensive care were assessed; 110 neonates who had received gentamicin and 110 neonates who had not received gentamicin served as control group. Gentamicin group were further subdivided according to the duration of treatment into 50 neonates who had received gentamicin for 5 days or less and 60 neonates who had received gentamicin for more than 5 days. TEOAEs were used for hearing screening. Auditory brain response was performed 3 months later for failed cases to confirm the hearing impairment. RESULTS Three neonates failed TEOAEs screening in each group but hearing impairment was confirmed in one neonate only (0.9%) in each group (gentamicin and control groups). Neonates who received gentamicin for more than 5 days showed comparable results as regard TEOAEs or ABR results with those who received gentamicin for 5 days or less, and control group. CONCLUSIONS Extended interval dosing of gentamicin therapy in neonates does not increase the incidence of hearing loss. This suggests that hearing loss in neonatal intensive care unit may be attributed to factors other than gentamicin treatment.
Collapse
Affiliation(s)
- Mohamed N El-Barbary
- Department of Pediatrics, Faculty of Medicine, Ain Shams University, Ramses Street, Cairo, 11566, Egypt
| | - Rania I H Ismail
- Department of Pediatrics, Faculty of Medicine, Ain Shams University, Ramses Street, Cairo, 11566, Egypt.
| | | |
Collapse
|
33
|
Impact of sparfloxacin on melanogenesis and antioxidant defense system in normal human melanocytes HEMa-LP – An in vitro study. Pharmacol Rep 2015; 67:38-43. [DOI: 10.1016/j.pharep.2014.07.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 07/25/2014] [Accepted: 07/28/2014] [Indexed: 12/17/2022]
|
34
|
Chen Y, Li L, Ni W, Zhang Y, Sun S, Miao D, Chai R, Li H. Bmi1 regulates auditory hair cell survival by maintaining redox balance. Cell Death Dis 2015; 6:e1605. [PMID: 25611380 PMCID: PMC4669747 DOI: 10.1038/cddis.2014.549] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 11/13/2014] [Accepted: 11/17/2014] [Indexed: 01/06/2023]
Abstract
Reactive oxygen species (ROS) accumulation are involved in noise- and ototoxic drug-induced hair cell loss, which is the major cause of hearing loss. Bmi1 is a member of the Polycomb protein family and has been reported to regulate mitochondrial function and ROS level in thymocytes and neurons. In this study, we reported the expression of Bmi1 in mouse cochlea and investigated the role of Bmi1 in hair cell survival. Bmi1 expressed in hair cells and supporting cells in mouse cochlea. Bmi1−/− mice displayed severe hearing loss and patched outer hair cell loss from postnatal day 22. Ototoxic drug-induced hair cells loss dramatically increased in Bmi1−/− mice compared with that in wild-type controls both in vivo and in vitro, indicating Bmi1−/− hair cells were significantly more sensitive to ototoxic drug-induced damage. Cleaved caspase-3 and TUNEL staining demonstrated that apoptosis was involved in the increased hair cell loss of Bmi1−/− mice. Aminophenyl fluorescein and MitoSOX Red staining showed the level of free radicals and mitochondrial ROS increased in Bmi1−/− hair cells due to the aggravated disequilibrium of antioxidant–prooxidant balance. Furthermore, the antioxidant N-acetylcysteine rescued Bmi1−/− hair cells from neomycin injury both in vitro and in vivo, suggesting that ROS accumulation was mainly responsible for the increased aminoglycosides sensitivity in Bmi1−/− hair cells. Our findings demonstrate that Bmi1 has an important role in hair cell survival by controlling redox balance and ROS level, thus suggesting that Bmi1 may work as a new therapeutic target for the prevention of hair cell death.
Collapse
Affiliation(s)
- Y Chen
- 1] Department of Otorhinolaryngology, Hearing Research Institute, Affiliated Eye and ENT Hospital of Fudan University, Shanghai 200031, China [2] Central Laboratory, Affiliated Eye and ENT Hospital of Fudan University, Shanghai 200031, China
| | - L Li
- Department of Otorhinolaryngology, Hearing Research Institute, Affiliated Eye and ENT Hospital of Fudan University, Shanghai 200031, China
| | - W Ni
- Department of Otorhinolaryngology, Hearing Research Institute, Affiliated Eye and ENT Hospital of Fudan University, Shanghai 200031, China
| | - Y Zhang
- 1] Department of Otorhinolaryngology, Hearing Research Institute, Affiliated Eye and ENT Hospital of Fudan University, Shanghai 200031, China [2] Central Laboratory, Affiliated Eye and ENT Hospital of Fudan University, Shanghai 200031, China [3] Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - S Sun
- 1] Department of Otorhinolaryngology, Hearing Research Institute, Affiliated Eye and ENT Hospital of Fudan University, Shanghai 200031, China [2] Central Laboratory, Affiliated Eye and ENT Hospital of Fudan University, Shanghai 200031, China
| | - D Miao
- State Key Laboratory of Reproductive Medicine, Research Center for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing 210096, China
| | - R Chai
- Co-innovation Center of Neuroregeneration, Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - H Li
- 1] Department of Otorhinolaryngology, Hearing Research Institute, Affiliated Eye and ENT Hospital of Fudan University, Shanghai 200031, China [2] Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China [3] State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| |
Collapse
|
35
|
|
36
|
Electroacoustic stimulation: now and into the future. BIOMED RESEARCH INTERNATIONAL 2014; 2014:350504. [PMID: 25276779 PMCID: PMC4168031 DOI: 10.1155/2014/350504] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 08/04/2014] [Indexed: 12/22/2022]
Abstract
Cochlear implants have provided hearing to hundreds of thousands of profoundly deaf people around the world. Recently, the eligibility criteria for cochlear implantation have been relaxed to include individuals who have some useful residual hearing. These recipients receive inputs from both electric and acoustic stimulation (EAS). Implant recipients who can combine these hearing modalities demonstrate pronounced benefit in speech perception, listening in background noise, and music appreciation over implant recipients that rely on electrical stimulation alone. The mechanisms bestowing this benefit are unknown, but it is likely that interaction of the electric and acoustic signals in the auditory pathway plays a role. Protection of residual hearing both during and following cochlear implantation is critical for EAS. A number of surgical refinements have been implemented to protect residual hearing, and the development of hearing-protective drug and gene therapies is promising for EAS recipients. This review outlines the current field of EAS, with a focus on interactions that are observed between these modalities in animal models. It also outlines current trends in EAS surgery and gives an overview of the drug and gene therapies that are clinically translatable and may one day provide protection of residual hearing for cochlear implant recipients.
Collapse
|
37
|
Wrześniok D, Otręba M, Beberok A, Buszman E. Impact of kanamycin on melanogenesis and antioxidant enzymes activity in melanocytes--an in vitro study. J Cell Biochem 2014; 114:2746-52. [PMID: 23804282 DOI: 10.1002/jcb.24623] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 06/20/2013] [Indexed: 01/16/2023]
Abstract
Aminoglycosides, broad spectrum aminoglycoside antibiotics, are used in various infections therapy due to their good antimicrobial characteristics. However, their adverse effects such as nephrotoxicity and auditory ototoxicity, as well as some toxic effects directed to pigmented tissues, complicate the use of these agents. This study was undertaken to investigate the effect of aminoglycoside antibiotic-kanamycin on viability, melanogenesis and antioxidant enzymes activity in cultured human normal melanocytes (HEMa-LP). It has been demonstrated that kanamycin induces concentration-dependent loss in melanocytes viability. The value of EC50 was found to be ~6.0 mM. Kanamycin suppressed melanin biosynthesis: antibiotic was shown to inhibit cellular tyrosinase activity and to reduce melanin content in normal human melanocytes. Significant changes in the cellular antioxidant enzymes: SOD, CAT and GPx were stated in melanocytes exposed to kanamycin. Moreover, it was observed that kanamycin caused depletion of antioxidant defense sytem. It is concluded that the inhibitory effect of kanamycin on melanogenesis and not sufficient antioxidant defense mechanism in melanocytes in vitro may explain the potential mechanisms of undesirable side effects of this drug directed to pigmented tissues in vivo.
Collapse
Affiliation(s)
- Dorota Wrześniok
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Medical University of Silesia, Jagiellońska 4, PL 41-200, Sosnowiec, Poland
| | | | | | | |
Collapse
|
38
|
Inoue A, Iwasaki S, Fujimoto C, Nakajima T, Yamasoba T. Developmental changes in the protective effect of exogenous brain-derived neurotrophic factor and neurotrophin-3 against ototoxic drugs in cultured rat vestibular ganglion neurons. Cell Tissue Res 2014; 356:299-308. [DOI: 10.1007/s00441-014-1813-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Accepted: 01/14/2014] [Indexed: 12/13/2022]
|
39
|
Tadros SF, D'Souza M, Zhu X, Frisina RD. Gene expression changes for antioxidants pathways in the mouse cochlea: relations to age-related hearing deficits. PLoS One 2014; 9:e90279. [PMID: 24587312 PMCID: PMC3938674 DOI: 10.1371/journal.pone.0090279] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 01/28/2014] [Indexed: 12/17/2022] Open
Abstract
Age-related hearing loss - presbycusis - is the number one neurodegenerative disorder and top communication deficit of our aged population. Like many aging disorders of the nervous system, damage from free radicals linked to production of reactive oxygen and/or nitrogen species (ROS and RNS, respectively) may play key roles in disease progression. The efficacy of the antioxidant systems, e.g., glutathione and thioredoxin, is an important factor in pathophysiology of the aging nervous system. In this investigation, relations between the expression of antioxidant-related genes in the auditory portion of the inner ear - cochlea, and age-related hearing loss was explored for CBA/CaJ mice. Forty mice were classified into four groups according to age and degree of hearing loss. Cochlear mRNA samples were collected and cDNA generated. Using Affymetrix® GeneChip, the expressions of 56 antioxidant-related gene probes were analyzed to estimate the differences in gene expression between the four subject groups. The expression of Glutathione peroxidase 6, Gpx6; Thioredoxin reductase 1, Txnrd1; Isocitrate dehydrogenase 1, Idh1; and Heat shock protein 1, Hspb1; were significantly different, or showed large fold-change differences between subject groups. The Gpx6, Txnrd1 and Hspb1 gene expression changes were validated using qPCR. The Gpx6 gene was upregulated while the Txnrd1 gene was downregulated with age/hearing loss. The Hspb1 gene was found to be downregulated in middle-aged animals as well as those with mild presbycusis, whereas it was upregulated in those with severe presbycusis. These results facilitate development of future interventions to predict, prevent or slow down the progression of presbycusis.
Collapse
Affiliation(s)
- Sherif F. Tadros
- International Center for Hearing & Speech Research, National Technical Institute for the Deaf, Rochester Institute of Technology, Rochester, New York, United States of America
- Otolaryngology Dept., University of Rochester Medical School, Rochester, New York, United States of America
| | - Mary D'Souza
- International Center for Hearing & Speech Research, National Technical Institute for the Deaf, Rochester Institute of Technology, Rochester, New York, United States of America
- Otolaryngology Dept., University of Rochester Medical School, Rochester, New York, United States of America
| | - Xiaoxia Zhu
- International Center for Hearing & Speech Research, National Technical Institute for the Deaf, Rochester Institute of Technology, Rochester, New York, United States of America
- Otolaryngology Dept., University of Rochester Medical School, Rochester, New York, United States of America
- Depts. Chemical & Biomedical Engineering, Communication Sciences & Disorders, and Global Center for Hearing & Speech Research, University of South Florida, Tampa, Florida, United States of America
| | - Robert D. Frisina
- International Center for Hearing & Speech Research, National Technical Institute for the Deaf, Rochester Institute of Technology, Rochester, New York, United States of America
- Otolaryngology Dept., University of Rochester Medical School, Rochester, New York, United States of America
- Depts. Chemical & Biomedical Engineering, Communication Sciences & Disorders, and Global Center for Hearing & Speech Research, University of South Florida, Tampa, Florida, United States of America
| |
Collapse
|
40
|
Kohrman DC, Raphael Y. Gene therapy for deafness. Gene Ther 2013; 20:1119-23. [PMID: 23864018 PMCID: PMC4113964 DOI: 10.1038/gt.2013.39] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 06/06/2013] [Accepted: 06/10/2013] [Indexed: 12/18/2022]
Abstract
Hearing loss is the most common sensory deficit in humans and can result from genetic, environmental or combined etiologies that prevent normal function of the cochlea, the peripheral sensory organ. Recent advances in understanding the genetic pathways that are critical for the development and maintenance of cochlear function, as well as the molecular mechanisms that underlie cell trauma and death, have provided exciting opportunities for modulating these pathways to correct genetic mutations, to enhance the endogenous protective pathways for hearing preservation and to regenerate lost sensory cells with the possibility of ameliorating hearing loss. A number of recent animal studies have used gene-based therapies in innovative ways toward realizing these goals. With further refinement, some of the protective and regenerative approaches reviewed here may become clinically applicable.
Collapse
Affiliation(s)
- D C Kohrman
- Department of Otolaryngology-Head and Neck Surgery, Kresge Hearing Research Institute, The University of Michigan, Ann Arbor, MI, USA
| | | |
Collapse
|
41
|
Wong ACY, Froud KE, Hsieh YSY. Noise-induced hearing loss in the 21 st century: A research and translational update. World J Otorhinolaryngol 2013; 3:58-70. [DOI: 10.5319/wjo.v3.i3.58] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 07/19/2013] [Indexed: 02/06/2023] Open
Abstract
Millions of people worldwide are exposed to harmful levels of noise daily in their work and leisure environment. This makes noise-induced hearing loss (NIHL) a major occupational health risk globally. NIHL is the second most common form of acquired hearing loss after age-related hearing loss and is itself a major contributing factor to presbycusis. Temporary threshold shifts, once thought to be relatively harmless and recoverable, are now known to cause permanent cochlear injury leading to permanent loss of hearing sensitivity. This article reviews the current understanding of the cellular and molecular pathophysiology of NIHL with latest findings from animal models. Therapeutic approaches to protect against or to mitigate NIHL are discussed based on their proposed action against these known mechanisms of cochlear injury. Successes in identifying genes that predispose individuals to NIHL by candidate gene association studies are discussed with matched gene knockout animal models. This links to exciting developments in experimental gene therapy to replace and regenerate lost hair cells and post-noise otoprotective therapies currently being investigated in clinical trials. The aim is to provide new insights into current and projected future strategies to manage NIHL; bench to bedside treatment is foreseeable in the next 5 to 10 years.
Collapse
|
42
|
Tian CJ, Kim SW, Kim YJ, Lim HJ, Park R, So HS, Choung YH. Red ginseng protects against gentamicin-induced balance dysfunction and hearing loss in rats through antiapoptotic functions of ginsenoside Rb1. Food Chem Toxicol 2013; 60:369-76. [PMID: 23933362 DOI: 10.1016/j.fct.2013.07.069] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Revised: 07/13/2013] [Accepted: 07/26/2013] [Indexed: 11/16/2022]
Abstract
The authors evaluated the protective effects of Korean red ginseng (KRG) against gentamicin (GM)-induced unilateral vestibular and hearing dysfunction and investigated its effective mechanism using in vitro cell cultures. Vestibular function was comprehensively evaluated by a scoring system that ranged from 0 (normal) to 3 (worst) points, using head tilt, tail hanging, and swimming tests. The GM group showed significantly more deteriorated vestibular function (0 point--5 rats, 1 point--1 rat, 2 points--3 rats, and 3 points--3 rats) than the KRG+GM group (0 point--9 rats and 1 point--1 rat) (p<0.01). The hearing thresholds were better in the KRG+GM group than in the GM group (p<0.05). Quantitative analysis of hair cell damage in the scanning electron microscopy was closely related with vestibular and hearing functional results. In vitro study showed that ginsenoside Rb1 (gRb1) attenuated reactive oxygen species production, suppressed JNK activation, up-regulated Bcl-xL and down-regulated Bax, cytochrome c, caspase 3, and cleaved poly (ADP-ribose) polymerase in GM-treated VOT-E36 cells. These findings suggest that KRG including gRb1 component protects against vestibular/hearing dysfunction by inhibiting apoptotic pathways when ototoxicity is induced by unilateral intratympanic injection with GM in rats.
Collapse
Affiliation(s)
- Chun Jie Tian
- Department of Otolaryngology, Ajou University School of Medicine, Suwon, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
43
|
Wei Y, Fu Y, Liu S, Xia G, Pan S. Effect of lentiviruses carrying enhanced green fluorescent protein injected into the scala media through a cochleostomy in rats. Am J Otolaryngol 2013; 34:301-7. [PMID: 23465349 DOI: 10.1016/j.amjoto.2012.12.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 12/24/2012] [Indexed: 10/27/2022]
Abstract
PURPOSE The purposes of the current study were to assess the feasibility of post-auricular microinjection of lentiviruses carrying enhanced green fluorescent protein (EGFP) into the scala media through cochleostomies in rats, determine the expression of viral gene in the cochlea, and record the post-operative changes in the number and auditory function of cochlear hair cells (HCs). METHODS Healthy rats were randomly divided into two groups. The left ears of the animals in group I were injected with lentivirus carrying EGFP (n=10) via scala media lateral wall cochleostomies, and the left ears of the animals in group II were similarly injected with artificial endolymph (n=10). Prior to and 30 days post-injection, auditory function was assessed with click-auditory brainstem response (ABR) testing, EGFP expression was determined with cochlear frozen sections under fluorescence microscopy, and survival of HCs was estimated based on whole mount preparations. RESULTS Thirty days after surgery, click-ABR testing revealed that there were significant differences in the auditory function, EGFP expression, and survival of HCs in the left ears before and after surgery in the same rats from each group. In group I, EGFP was noted in the strial marginal cells of the scala media, the organ of Corti, spiral nerves, and spiral ganglion cells. CONCLUSION Lentiviruses were successfully introduced into the scala media through cochleostomies in rats, and the EGFP reporter gene was efficiently expressed in the organ of Corti, spiral nerves, and spiral ganglion cells.
Collapse
|
44
|
Kanzaki S. [Feasibility of drug and gene therapy for sensorineural hearing loss]. Nihon Yakurigaku Zasshi 2013; 141:188-90. [PMID: 23575422 DOI: 10.1254/fpj.141.188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
45
|
A combination of cilostazol and Ginkgo biloba extract protects against cisplatin-induced Cochleo-vestibular dysfunction by inhibiting the mitochondrial apoptotic and ERK pathways. Cell Death Dis 2013; 4:e509. [PMID: 23429295 PMCID: PMC3734837 DOI: 10.1038/cddis.2013.33] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cisplatin (cis-diammine-dichloroplatinum; CDDP) is an anticancer drug that induces significant hearing loss and balance dysfunction as side effects. Cilostazol (CS, 6-[4-(1-cyclohexyl-1H-tetrazol-5-yl) butoxy]-3, 4-dihydro-2-(1H)-quinolinone) has neuroprotective and antioxidant effects, whereas Ginkgo biloba extract (GbE) has preventive effects on CDDP-induced hearing loss in rats, and GbE enhances the antiatherogenic effect of CS by inhibiting the generation of reactive oxygen species (ROS). The purpose of this study was to investigate the effects of renexin (RXN), which contains GbE and CS, against CDDP-induced cochleo-vestibular dysfunction in rats and to elucidate the mechanism underlying the protective effects of RXN on auditory cells. Rats intraperitoneally injected with CDDP exhibited an increase in hearing threshold and vestibular dysfunction, which agreed with hair cell damage in the Organ of Corti and otoliths. However, these impairments were significantly prevented in a dose-dependent manner by pre- and co-treatment with RXN, and these preventive effects in RXN-treated rats were more prominent than those in GbE-treated rats. In a CDDP pharmacokinetic study, platinum concentration was very similar between CDDP-only treated and RXN+CDDP cotreated rats. RXN markedly attenuated CDDP-induced intracellular ROS and significantly reduced CDDP-activated expression of p-extracellular regulated kinase (ERK), BAX, cytochrome c, cleaved caspase-3 and cleaved poly (ADP-ribose) polymerase, but increased BCL-XL expression. These results show that RXN may have a synergistic effect by strongly protecting hearing and vestibular dysfunction induced by CDDP by inhibiting ROS production, mitochondrial pathways and the ERK pathway, without interfering with CDDP pharmacokinetics. Therefore, RXN could potentially be used to reduce CDDP-related hearing loss and dizziness.
Collapse
|
46
|
Modulation of melanogenesis and antioxidant defense system in melanocytes by amikacin. Toxicol In Vitro 2013; 27:1102-8. [PMID: 23416261 DOI: 10.1016/j.tiv.2013.02.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 01/25/2013] [Accepted: 02/06/2013] [Indexed: 11/21/2022]
Abstract
Amikacin is principally used to treat infections caused by microorganisms resistant to other aminoglycosides. Ototoxicity is one of the side effects of amikacin, but the causative mechanism of damage to the ear has not been fully established. Thus, the aim of this work was to examine the impact of amikacin on the melanogenesis and antioxidant defense system in cultured human normal melanocytes (HEMa-LP). Amikacin induced the concentration - dependent loss in melanocytes viability. The value of EC50 was determined to be ~7.5 mM. The analyzed antibiotic inhibited melanin biosynthesis in concentration-dependent manner. Increasing the amikacin concentration also resulted in a decrease in cellular tyrosinase activity. To study the antioxidant defense system in melanocytes, the activities of superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx) in cells exposed to amikacin were determined. Significant changes in cellular antioxidant enzymes activities were observed. Modulation of melanogenesis and the antioxidant status of melanocytes resulting from the use of amikacin in vitro may explain a potential role of melanin and melanocytes in the mechanisms of aminoglycosides ototoxic effects in vivo.
Collapse
|
47
|
Pérez B, Rodríguez-Pombo P, Ugarte M, Desviat L. Readthrough strategies for therapeutic suppression of nonsense mutations in inherited metabolic disease. Mol Syndromol 2012; 3:230-6. [PMID: 23293581 PMCID: PMC3531923 DOI: 10.1159/000343086] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Inherited metabolic diseases (IMDs) belong to the group of rare diseases due to their low individual prevalence. Most of them are inherited in autosomal recessive fashion and represent good candidates for novel therapeutical strategies aimed at recovering partial enzyme function as they lack an effective treatment, and small levels of enzymatic activity have been shown to be associated with improved outcome and milder phenotypes. Recently, a novel therapeutic approach for genetic diseases has emerged, based on the ability of aminoglycosides and other compounds in allowing translation to proceed through a premature termination codon introduced by a nonsense mutation, which frequently constitute a significant fraction of the mutant alleles in a population. In this review we summarize the essentials of what is known as suppression therapy, the different compounds that have been identified by high-throughput screens or developed using a medicinal chemistry approach and the preclinical and clinical trials that are being conducted in general and in the field of IMDs in particular. Several IMDs have shown to be good models for evaluating readthrough compounds using patients' cells carrying nonsense mutations, monitoring for an increase in functional recovery and/or enzyme activity. Overall, the positive results obtained indicate the feasibility of the approach for different diseases and although the levels of protein function reached are low, they may be enough to alleviate the consequences of the pathology. Nonsense suppression thus represents a potential therapy or supplementary treatment for a number of IMD patients encouraging further clinical trials with readthrough drugs with improved functionality and low toxicity.
Collapse
Affiliation(s)
| | | | | | - L.R. Desviat
- Centro de Diagnóstico de Enfermedades Moleculares, y Centro de Biología Molecular Severo Ochoa, UAM-CSIC, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), y Instituto de Investigación Hospital Universitario La Paz (IDIPaz), Madrid, España
| |
Collapse
|
48
|
Keeling KM, Wang D, Conard SE, Bedwell DM. Suppression of premature termination codons as a therapeutic approach. Crit Rev Biochem Mol Biol 2012; 47:444-63. [PMID: 22672057 PMCID: PMC3432268 DOI: 10.3109/10409238.2012.694846] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In this review, we describe our current understanding of translation termination and pharmacological agents that influence the accuracy of this process. A number of drugs have been identified that induce suppression of translation termination at in-frame premature termination codons (PTCs; also known as nonsense mutations) in mammalian cells. We discuss efforts to utilize these drugs to suppress disease-causing PTCs that result in the loss of protein expression and function. In-frame PTCs represent a genotypic subset of mutations that make up ~11% of all known mutations that cause genetic diseases, and millions of patients have diseases attributable to PTCs. Current approaches aimed at reducing the efficiency of translation termination at PTCs (referred to as PTC suppression therapy) have the goal of alleviating the phenotypic consequences of a wide range of genetic diseases. Suppression therapy is currently in clinical trials for treatment of several genetic diseases caused by PTCs, and preliminary results suggest that some patients have shown clinical improvements. While current progress is promising, we discuss various approaches that may further enhance the efficiency of this novel therapeutic approach.
Collapse
Affiliation(s)
- Kim M. Keeling
- Dept. of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dan Wang
- Dept. of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sara E. Conard
- Dept. of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David M. Bedwell
- Dept. of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
49
|
Gene therapy for cisplatin-induced ototoxicity: a systematic review of in vitro and experimental animal studies. Otol Neurotol 2012; 33:302-10. [PMID: 22388732 DOI: 10.1097/mao.0b013e318248ee66] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Ototoxicity is a frequent adverse event of cisplatin treatment. No therapy is currently available for cisplatin-induced ototoxicity. A systematic review of experimental animal studies and in vitro experiments was conducted to evaluate gene therapy as a potential future therapeutic option. DATA SOURCES Eligible studies were identified through searches of electronic databases Ovid MEDLINE, Ovid MEDLINE In-Process, Embase, PubMed, Biosis Previews, Scopus, ISI Web of Science, and The Cochrane Library. STUDY SELECTION Articles obtained from the search were independently reviewed by 2 authors using specific criteria to identify experimental animal studies and in vitro experiments conducted to evaluate gene therapy for cisplatin-induced ototoxicity. No restriction was applied to publication dates or languages. DATA EXTRACTION Data extracted included experiment type, cell type, species, targeted gene, gene expression, method, administration, inner ear site evaluated, outcome measures for cytotoxicity, and significant results. RESULTS Fourteen articles were included in this review. In vitro and in vivo experiments have been performed to evaluate the potential of gene expression manipulation for cisplatin-induced ototoxicity. Twelve different genes were targeted including NTF3, GDNF, HO-1, XIAP, Trpv1, BCL2, Otos, Nfe2l2, Nox1, Nox3, Nox4, and Ctr1. All of the included articles demonstrated a benefit of gene therapy on cytotoxicity caused by cisplatin. CONCLUSION Experimental animal studies and in vitro experiments have demonstrated the efficacy of gene therapy for cisplatin-induced ototoxicity. However, further investigation regarding safety, immunogenicity, and consequences of genetic manipulation in the inner ear tissues must be completed to develop future therapeutic options.
Collapse
|
50
|
Sacheli R, Delacroix L, Vandenackerveken P, Nguyen L, Malgrange B. Gene transfer in inner ear cells: a challenging race. Gene Ther 2012; 20:237-47. [PMID: 22739386 DOI: 10.1038/gt.2012.51] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Recent advances in human genomics led to the identification of numerous defective genes causing deafness, which represent novel putative therapeutic targets. Future gene-based treatment of deafness resulting from genetic or acquired sensorineural hearing loss may include strategies ranging from gene therapy to antisense delivery. For successful development of gene therapies, a minimal requirement involves the engineering of appropriate gene carrier systems. Transfer of exogenous genetic material into the mammalian inner ear using viral or non-viral vectors has been characterized over the last decade. The nature of inner ear cells targeted, as well as the transgene expression level and duration, are highly dependent on the vector type, the route of administration and the strength of the promoter driving expression. This review summarizes and discusses recent advances in inner ear gene-transfer technologies aimed at examining gene function or identifying new treatment for inner ear disorders.
Collapse
Affiliation(s)
- R Sacheli
- GIGA-Neurosciences, Developmental Neurobiology Unit, University of Liège, Liège, Belgium
| | | | | | | | | |
Collapse
|