1
|
Ege T, Tao L, North BJ. The Role of Molecular and Cellular Aging Pathways on Age-Related Hearing Loss. Int J Mol Sci 2024; 25:9705. [PMID: 39273652 PMCID: PMC11396656 DOI: 10.3390/ijms25179705] [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: 07/27/2024] [Revised: 08/27/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024] Open
Abstract
Aging, a complex process marked by molecular and cellular changes, inevitably influences tissue and organ homeostasis and leads to an increased onset or progression of many chronic diseases and conditions, one of which is age-related hearing loss (ARHL). ARHL, known as presbycusis, is characterized by the gradual and irreversible decline in auditory sensitivity, accompanied by the loss of auditory sensory cells and neurons, and the decline in auditory processing abilities associated with aging. The extended human lifespan achieved by modern medicine simultaneously exposes a rising prevalence of age-related conditions, with ARHL being one of the most significant. While our understanding of the molecular basis for aging has increased over the past three decades, a further understanding of the interrelationship between the key pathways controlling the aging process and the development of ARHL is needed to identify novel targets for the treatment of AHRL. The dysregulation of molecular pathways (AMPK, mTOR, insulin/IGF-1, and sirtuins) and cellular pathways (senescence, autophagy, and oxidative stress) have been shown to contribute to ARHL. However, the mechanistic basis for these pathways in the initiation and progression of ARHL needs to be clarified. Therefore, understanding how longevity pathways are associated with ARHL will directly influence the development of therapeutic strategies to treat or prevent ARHL. This review explores our current understanding of the molecular and cellular mechanisms of aging and hearing loss and their potential to provide new approaches for early diagnosis, prevention, and treatment of ARHL.
Collapse
Affiliation(s)
| | - Litao Tao
- Biomedical Sciences Department, School of Medicine, Creighton University, Omaha, NE 68178, USA;
| | - Brian J. North
- Biomedical Sciences Department, School of Medicine, Creighton University, Omaha, NE 68178, USA;
| |
Collapse
|
2
|
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
|
3
|
Self-complementarity in adeno-associated virus enhances transduction and gene expression in mouse cochlear tissues. PLoS One 2020; 15:e0242599. [PMID: 33227033 PMCID: PMC7682903 DOI: 10.1371/journal.pone.0242599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 11/06/2020] [Indexed: 12/03/2022] Open
Abstract
Sensorineural hearing loss is one of the most common disabilities worldwide. Such prevalence necessitates effective tools for studying the molecular workings of cochlear cells. One prominent and effective vector for expressing genes of interest in research models is adeno-associated virus (AAV). However, AAV efficacy in transducing cochlear cells can vary for a number of reasons including serotype, species, and methodology, and oftentimes requires high multiplicity of infection which can damage the sensory cells. Reports in other systems suggest multiple approaches can be used to enhance AAV transduction including self-complementary vector design and pharmacological inhibition of degradation. Here we produced AAV to drive green fluorescent protein (GFP) expression in explanted neonatal mouse cochleae. Treatment with eeyarestatin I, tyrphostin 23, or lipofectamine 2000 did not result in increased transduction, however, self-complementary vector design resulted in significantly more GFP positive cells when compared to single-stranded controls. Similarly, self-complementary AAV2 vectors demonstrated enhanced transduction efficiency compared to single stranded AAV2 when injected via the posterior semicircular canal, in vivo. Self-complementary vectors for AAV1, 8, and 9 serotypes also demonstrated robust GFP transduction in cochlear cells in vivo, though these were not directly compared to single stranded vectors. These findings suggest that second-strand synthesis may be a rate limiting step in AAV transduction of cochlear tissues and that self-complementary AAV can be used to effectively target large numbers of cochlear cells in vitro and in vivo.
Collapse
|
4
|
Valentini C, Szeto B, Kysar JW, Lalwani AK. Inner Ear Gene Delivery: Vectors and Routes. HEARING BALANCE AND COMMUNICATION 2020; 18:278-285. [PMID: 33604229 DOI: 10.1080/21695717.2020.1807261] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Objectives Current treatments for hearing loss offer some functional improvements in hearing, but do not restore normal hearing. The aim of this review is to highlight recent advances in viral and non-viral vectors for gene therapy and to discuss approaches for overcoming barriers inherent to inner ear delivery of gene products. Data Sources The databases used were Medline, EMBASE, Web of Science, and Google Scholar. Search terms were [("cochlea*" or "inner ear" or "transtympanic" or "intratympanic" or "intracochlear" or "hair cells" or "spiral ganglia" or "Organ of Corti") and ("gene therapy" or "gene delivery")]. The references section of resulting articles was also used to identify relevant studies. Results Both viral and non-viral vectors play important roles in advancing gene delivery to the inner ear. The round window membrane is one significant barrier to gene delivery that intratympanic delivery methods attempt to overcome through diffusion and intracochlear delivery methods bypass completely. Conclusions Gene therapy for hearing loss is a promising treatment for restoring hearing function by addressing innate defects. Recent technological advances in inner ear drug delivery techniques pose exciting opportunities for progress in gene therapy.
Collapse
Affiliation(s)
- Chris Valentini
- Department of Otolaryngology -- Head and Neck Surgery, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Betsy Szeto
- Department of Otolaryngology -- Head and Neck Surgery, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Jeffrey W Kysar
- Department of Otolaryngology -- Head and Neck Surgery, Columbia University Vagelos College of Physicians and Surgeons, New York, NY.,Department of Mechanical Engineering, School of Engineering, Columbia University, New York, New York
| | - Anil K Lalwani
- Department of Otolaryngology -- Head and Neck Surgery, Columbia University Vagelos College of Physicians and Surgeons, New York, NY.,Department of Mechanical Engineering, School of Engineering, Columbia University, New York, New York
| |
Collapse
|
5
|
Szeto B, Chiang H, Valentini C, Yu M, Kysar JW, Lalwani AK. Inner ear delivery: Challenges and opportunities. Laryngoscope Investig Otolaryngol 2020; 5:122-131. [PMID: 32128438 PMCID: PMC7042639 DOI: 10.1002/lio2.336] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/21/2019] [Accepted: 11/21/2019] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES The treatment of inner ear disorders remains challenging due to anatomic barriers intrinsic to the bony labyrinth. The purpose of this review is to highlight recent advances and strategies for overcoming these barriers and to discuss promising future avenues for investigation. DATA SOURCES The databases used were PubMed, EMBASE, and Web of Science. RESULTS Although some studies aimed to improve systemic delivery using nanoparticle systems, the majority enhanced local delivery using hydrogels, nanoparticles, and microneedles. Developments in direct intracochlear delivery include intracochlear injection and intracochlear implants. CONCLUSIONS In the absence of a systemic drug that targets only the inner ear, the best alternative is local delivery that harnesses a combination of new strategies to overcome anatomic barriers. The combination of microneedle technology with hydrogel and nanoparticle delivery is a promising area for future investigation. LEVEL OF EVIDENCE NA.
Collapse
Affiliation(s)
- Betsy Szeto
- Department of Otolaryngology–Head and Neck SurgeryColumbia University Vagelos College of Physicians and SurgeonsNew YorkNew York
| | - Harry Chiang
- Department of Otolaryngology–Head and Neck SurgeryColumbia University Vagelos College of Physicians and SurgeonsNew YorkNew York
| | - Chris Valentini
- Department of Otolaryngology–Head and Neck SurgeryColumbia University Vagelos College of Physicians and SurgeonsNew YorkNew York
| | - Michelle Yu
- Department of Otolaryngology–Head and Neck SurgeryColumbia University Vagelos College of Physicians and SurgeonsNew YorkNew York
| | - Jeffrey W. Kysar
- Department of Otolaryngology–Head and Neck SurgeryColumbia University Vagelos College of Physicians and SurgeonsNew YorkNew York
- Department of Mechanical Engineering, School of EngineeringColumbia UniversityNew YorkNew York
| | - Anil K. Lalwani
- Department of Otolaryngology–Head and Neck SurgeryColumbia University Vagelos College of Physicians and SurgeonsNew YorkNew York
- Department of Mechanical Engineering, School of EngineeringColumbia UniversityNew YorkNew York
| |
Collapse
|
6
|
Gu X, Chai R, Guo L, Dong B, Li W, Shu Y, Huang X, Li H. Transduction of Adeno-Associated Virus Vectors Targeting Hair Cells and Supporting Cells in the Neonatal Mouse Cochlea. Front Cell Neurosci 2019; 13:8. [PMID: 30733670 PMCID: PMC6353798 DOI: 10.3389/fncel.2019.00008] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 01/10/2019] [Indexed: 02/05/2023] Open
Abstract
Adeno-associated virus (AAV) is the preferred vector for gene therapy of hereditary deafness, and different viral serotypes, promoters and transduction pathways can influence the targeting of AAV to different types of cells and the expression levels of numerous exogenous genes. To determine the transduction and expression patterns of AAV with different serotypes or promoters in hair cells and supporting cells in the neonatal mouse cochlea, we examined the expression of enhanced green fluorescent protein (eGFP) for five different types of AAV vectors [serotypes 2, 9, and Anc80L65 with promoter cytomegalovirus (CMV)-beta-Globin and serotypes 2 and 9 with promoter chicken beta-actin (CBA)] in in vitro cochlear explant cultures and we tested the transduction of AAV2/2-CBA, AAV2/9-CBA, and AAV2/Anc80L65-CMV by in vivo microinjection into the scala media of the cochlea. We found that each AAV vector had its own transduction and expression characteristics in hair cells and supporting cells in different regions of the cochlea. There was a tonotopic gradient for the in vitro transduction of AAV2/2-CBA, AAV2/9-CBA, AAV2/2-CMV, and AAV2/9-CMV in outer hair cells (OHCs), with more OHCs expressing eGFP at the base of the cochlea than at the apex. AAV2/2-CBA in vitro and AAV2/Anc80L65-CMV in vivo induced more supporting cells expressing eGFP at the apex than in the base. We found that AAV vectors with different promoters had different expression efficacies in hair cells and supporting cells of the auditory epithelium. The CMV-beta-Globin promoter could drive the expression of the delivered construct more efficiently in hair cells, while the CBA promoter was more efficient in supporting cells. The in vitro and in vivo experiments both demonstrated that AAV2/Anc80L65-CMV was a very promising vector for gene therapy of deafness because of its high transduction rates in hair cells. These results might be useful for selecting the appropriate vectors for gene delivery into different types of inner ear cells and thus improving the effectiveness of gene therapy.
Collapse
Affiliation(s)
- Xi Gu
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China
| | - Luo Guo
- ENT Institute and Otorhinolaryngology Department, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Biao Dong
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Wenyan Li
- ENT Institute and Otorhinolaryngology Department, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Yilai Shu
- ENT Institute and Otorhinolaryngology Department, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| | - Xinsheng Huang
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huawei Li
- ENT Institute and Otorhinolaryngology Department, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Cochlear Implant, Shanghai, China.,The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Kanzaki S. Gene Delivery into the Inner Ear and Its Clinical Implications for Hearing and Balance. Molecules 2018; 23:molecules23102507. [PMID: 30274337 PMCID: PMC6222543 DOI: 10.3390/molecules23102507] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 09/04/2018] [Accepted: 09/14/2018] [Indexed: 11/16/2022] Open
Abstract
The inner ear contains many types of cell, including sensory hair cells and neurons. If these cells are damaged, they do not regenerate. Inner ear disorders have various etiologies. Some are related to aging or are idiopathic, as in sudden deafness. Others occur due to acoustic trauma, exposure to ototoxic drugs, viral infections, immune responses, or endolymphatic hydrops (Meniere's disease). For these disorders, inner ear regeneration therapy is expected to be a feasible alternative to cochlear implants for hearing recovery. Recently, the mechanisms underlying inner ear regeneration have been gradually clarified. Inner ear cell progenitors or stem cells have been identified. Factors necessary for regeneration have also been elucidated from the mechanism of hair cell generation. Inducing differentiation of endogenous stem cells or inner ear stem cell transplantation is expected. In this paper, we discuss recent approaches to hair cell proliferation and differentiation for inner ear regeneration. We discuss the future road map for clinical application. The therapies mentioned above require topical administration of transgenes or drug onto progenitors of sensory cells. Developing efficient and safe modes of administration is clinically important. In this regard, we also discuss our development of an inner ear endoscope to facilitate topical administration.
Collapse
Affiliation(s)
- Sho Kanzaki
- Department of Otolaryngology Head and Neck Surgery, School of Medicine, KEIO University, Tokyo 160-8582, Japan.
| |
Collapse
|
8
|
Peyvandi AA, Abbaszadeh HA, Roozbahany NA, Pourbakht A, Khoshsirat S, Niri HH, Peyvandi H, Niknazar S. Deferoxamine promotes mesenchymal stem cell homing in noise-induced injured cochlea through PI3K/AKT pathway. Cell Prolif 2018; 51:e12434. [PMID: 29341316 DOI: 10.1111/cpr.12434] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 12/06/2017] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Over 5% of the world's population suffers from disabling hearing loss. Stem cell homing in target tissue is an important aspect of cell-based therapy, which its augmentation increases cell therapy efficiency. Deferoxamine (DFO) can induce the Akt activation, and phosphorylation status of AKT (p-AKT) upregulates CXC chemokine receptor-4 (CXCR4) expression. We examined whether DFO can enhance mesenchymal stem cells (MSCs) homing in noise-induced damaged cochlea by PI3K/AKT dependent mechanism. MATERIALS AND METHODS Mesenchymal stem cells were treated with DFO. AKT, p-AKT protein and hypoxia inducible factor 1- α (HIF-1α) and CXCR4 gene and protein expression was evaluated by RT- PCR and Western blot analysis. For in vivo assay, rats were assigned to control, sham, noise exposure groups without any treatment or receiving normal, DFO-treated and DFO +LY294002 (The PI3K inhibitor)-treated MSCs. Following chronic exposure to 115 dB white noise, MSCs were injected into the rat cochlea through the round window. Number of Hoechst- labelled cells was determined in the endolymph after 24 hours. RESULTS Deferoxamine increased P-AKT, HIF-1α and CXCR4 expression in MSCs compared to non-treated cells. DFO pre-conditioning significantly increased the homing ability of MSCs into injured ear compared to normal MSCs. These effects of DFO were blocked by LY294002. CONCLUSIONS Pre-conditioning of MSCs by DFO before transplantation can improve stem cell homing in the damaged cochlea through PI3K/AKT pathway activation.
Collapse
Affiliation(s)
- A A Peyvandi
- Hearing Disorders Research Center, Loghman Hakim Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - H-A Abbaszadeh
- Hearing Disorders Research Center, Loghman Hakim Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - N Ahmady Roozbahany
- Hearing Disorders Research Center, Loghman Hakim Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,G. Raymond Chang School, Ryerson University, Toronto, Canada
| | - A Pourbakht
- Department of Audiology, School of Rehabilitation Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - S Khoshsirat
- Hearing Disorders Research Center, Loghman Hakim Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - H Haddadzade Niri
- Department of Audiology, School of Rehabilitation Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - H Peyvandi
- Hearing Disorders Research Center, Loghman Hakim Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Yale University, New Haven, CT, USA
| | - S Niknazar
- Hearing Disorders Research Center, Loghman Hakim Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Shibata SB, Yoshimura H, Ranum PT, Goodwin AT, Smith RJH. Intravenous rAAV2/9 injection for murine cochlear gene delivery. Sci Rep 2017; 7:9609. [PMID: 28852025 PMCID: PMC5575199 DOI: 10.1038/s41598-017-09805-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/28/2017] [Indexed: 01/08/2023] Open
Abstract
Gene therapy for genetic deafness is a promising approach by which to prevent hearing loss or to restore hearing after loss has occurred. Although a variety of direct approaches to introduce viral particles into the inner ear have been described, presumed physiological barriers have heretofore precluded investigation of systemic gene delivery to the cochlea. In this study, we sought to characterize systemic delivery of a rAAV2/9 vector as a non-invasive means of cochlear transduction. In wild-type neonatal mice (postnatal day 0-1), we show that intravenous injection of rAAV2/9 carrying an eGFP-reporter gene results in binaural transduction of inner hair cells, spiral ganglion neurons and vestibular hair cells. Transduction efficiency increases in a dose-dependent manner. Inner hair cells are transduced in an apex-to-base gradient, with transduction reaching 96% in the apical turn. Hearing acuity in treated animals is unaltered at postnatal day 30. Transduction is influenced by viral serotype and age at injection, with less efficient cochlear transduction observed with systemic delivery of rAAV2/1 and in juvenile mice with rAAV2/9. Collectively, these data validate intravenous delivery of rAAV2/9 as a novel and atraumatic technique for inner ear transgene delivery in early postnatal mice.
Collapse
Affiliation(s)
- Seiji B Shibata
- Department of Otolaryngology - Head and Neck Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.,Molecular Otolaryngology and Renal Research Laboratories, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Hidekane Yoshimura
- Molecular Otolaryngology and Renal Research Laboratories, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.,Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Nagano, 390-8621, Japan
| | - Paul T Ranum
- Molecular Otolaryngology and Renal Research Laboratories, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.,Interdisciplinary Graduate Program in Molecular & Cellular Biology, The University of Iowa Graduate College, University of Iowa, Iowa City, IA, 52242, USA
| | - Alexander T Goodwin
- Molecular Otolaryngology and Renal Research Laboratories, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Richard J H Smith
- Department of Otolaryngology - Head and Neck Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA. .,Molecular Otolaryngology and Renal Research Laboratories, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA. .,Interdisciplinary Graduate Program in Molecular & Cellular Biology, The University of Iowa Graduate College, University of Iowa, Iowa City, IA, 52242, USA. .,Iowa Institute of Human Genetics, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
10
|
Chien WW, McDougald DS, Roy S, Fitzgerald TS, Cunningham LL. Cochlear gene transfer mediated by adeno-associated virus: Comparison of two surgical approaches. Laryngoscope 2015; 125:2557-64. [PMID: 25891801 DOI: 10.1002/lary.25317] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2015] [Indexed: 11/10/2022]
Abstract
OBJECTIVES/HYPOTHESIS Gene therapy offers the possibility of delivering corrective genetic materials to the cochlea, potentially improving hearing. In animals, the most commonly used surgical methods for viral gene therapy delivery to the cochlea are the round window and the cochleostomy approaches. However, the patterns of viral infection and the effects on hearing have not been directly compared between these two approaches. In this study, we compare the patterns of cochlear infection and effects on hearing between these two surgical approaches using adeno-associated virus serotype 2/8 (AAV8) as the gene delivery vehicle. STUDY DESIGN Animal study and basic science research. METHODS One- to two-month-old CBA/J mice were used in this study. AAV8-green fluorescent protein (GFP) was delivered to the cochlea by either the round window or the cochleostomy approach (described below). Auditory brainstem response was used to examine hearing thresholds before and after surgery. Animals were examined at 1, 2, 3, and 4 weeks after surgery for the patterns of cochlear infection and hearing loss. RESULTS Cochlear gene transfer was successful through both surgical approaches. In both approaches, AAV8-GFP mostly infected the inner hair cells. There was occasional low-level infection of the outer hair cells and supporting cells. The two surgical approaches resulted in comparable viral infection efficiencies. The round window approach resulted in less surgical trauma, as indicated by hearing loss, than the cochleostomy approach. CONCLUSIONS Adeno-associated virus-mediated gene transfer to the cochlea can be accomplished using either the round window or the cochleostomy surgical approach. The round window approach resulted in less hearing loss compared to the cochleostomy approach. LEVEL OF EVIDENCE NA.
Collapse
Affiliation(s)
- Wade W Chien
- National Institute on Deafness and Other Communication Disorders (w.w.c., d.s.m., s.r., t.s.f., l.l.c), National Institutes of Health, Bethesda, Maryland, U.S.A.,Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland, U.S.A
| | - Devin S McDougald
- National Institute on Deafness and Other Communication Disorders (w.w.c., d.s.m., s.r., t.s.f., l.l.c), National Institutes of Health, Bethesda, Maryland, U.S.A
| | - Soumen Roy
- National Institute on Deafness and Other Communication Disorders (w.w.c., d.s.m., s.r., t.s.f., l.l.c), National Institutes of Health, Bethesda, Maryland, U.S.A
| | - Tracy S Fitzgerald
- National Institute on Deafness and Other Communication Disorders (w.w.c., d.s.m., s.r., t.s.f., l.l.c), National Institutes of Health, Bethesda, Maryland, U.S.A
| | - Lisa L Cunningham
- National Institute on Deafness and Other Communication Disorders (w.w.c., d.s.m., s.r., t.s.f., l.l.c), National Institutes of Health, Bethesda, Maryland, U.S.A
| |
Collapse
|
11
|
Akil O, Rouse SL, Chan DK, Lustig LR. Surgical method for virally mediated gene delivery to the mouse inner ear through the round window membrane. J Vis Exp 2015:52187. [PMID: 25867531 PMCID: PMC4401361 DOI: 10.3791/52187] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Gene therapy, used to achieve functional recovery from sensorineural deafness, promises to grant better understanding of the underlying molecular and genetic mechanisms that contribute to hearing loss. Introduction of vectors into the inner ear must be done in a way that widely distributes the agent throughout the cochlea while minimizing injury to the existing structures. This manuscript describes a post-auricular surgical approach that can be used for mouse cochlear therapy using molecular, pharmacologic, and viral delivery to mice postnatal day 10 and older via the round window membrane (RWM). This surgical approach enables rapid and direct delivery into the scala tympani while minimizing blood loss and avoiding animal mortality. This technique involves negligible or no damage to essential structures of the inner and middle ear as well as neck muscles while wholly preserving hearing. To demonstrate the efficacy of this surgical technique, the vesicular glutamate transporter 3 knockout (VGLUT3 KO) mice will be used as an example of a mouse model of congenital deafness that recovers hearing after delivery of VGLUT3 to the inner ear using an adeno-associated virus (AAV-1).
Collapse
Affiliation(s)
- Omar Akil
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco;
| | - Stephanie L Rouse
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco
| | - Dylan K Chan
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco
| | - Lawrence R Lustig
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco
| |
Collapse
|
12
|
Differential effects of AAV.BDNF and AAV.Ntf3 in the deafened adult guinea pig ear. Sci Rep 2015; 5:8619. [PMID: 25726967 DOI: 10.1038/srep08619] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 01/27/2015] [Indexed: 01/14/2023] Open
Abstract
Cochlear hair cell loss results in secondary regression of peripheral auditory fibers (PAFs) and loss of spiral ganglion neurons (SGNs). The performance of cochlear implants (CI) in rehabilitating hearing depends on survival of SGNs. Here we compare the effects of adeno-associated virus vectors with neurotrophin gene inserts, AAV.BDNF and AAV.Ntf3, on guinea pig ears deafened systemically (kanamycin and furosemide) or locally (neomycin). AAV.BDNF or AAV.Ntf3 was delivered to the guinea pig cochlea one week following deafening and ears were assessed morphologically 3 months later. At that time, neurotrophins levels were not significantly elevated in the cochlear fluids, even though in vitro and shorter term in vivo experiments demonstrate robust elevation of neurotrophins with these viral vectors. Nevertheless, animals receiving these vectors exhibited considerable re-growth of PAFs in the basilar membrane area. In systemically deafened animals there was a negative correlation between the presence of differentiated supporting cells and PAFs, suggesting that supporting cells influence the outcome of neurotrophin over-expression aimed at enhancing the cochlear neural substrate. Counts of SGN in Rosenthal's canal indicate that BDNF was more effective than NT-3 in preserving SGNs. The results demonstrate that a transient elevation in neurotrophin levels can sustain the cochlear neural substrate in the long term.
Collapse
|
13
|
Wang Y, Sun Y, Chang Q, Ahmad S, Zhou B, Kim Y, Li H, Lin X. Early postnatal virus inoculation into the scala media achieved extensive expression of exogenous green fluorescent protein in the inner ear and preserved auditory brainstem response thresholds. J Gene Med 2013; 15:123-33. [PMID: 23413036 DOI: 10.1002/jgm.2701] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2012] [Revised: 01/24/2013] [Accepted: 01/29/2013] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Gene transfer into the inner ear is a promising approach for treating sensorineural hearing loss. The special electrochemical environment of the scala media raises a formidable challenge for effective gene delivery at the same time as keeping normal cochlear function intact. The present study aimed to define a suitable strategy for preserving hearing after viral inoculation directly into the scala media performed at various postnatal developmental stages. METHODS We assessed transgene expression of green fluorescent protein (GFP) mediated by various types of adeno-associated virus (AAV) and lentivirus (LV) in the mouse cochlea. Auditory brainstem responses were measured 30 days after inoculation to assess effects on hearing. RESULTS Patterns of GFP expression confirmed extensive exogenous gene expression in various types of cells lining the endolymphatic space. The use of different viral vectors and promoters resulted in specific cellular GFP expression patterns. AAV2/1 with cytomegalovirus promoter apparently gave the best results for GFP expression in the supporting cells. Histological examination showed normal cochlear morphology and no hair cell loss after either AAV or LV injections. We found that hearing thresholds were not significantly changed when the injections were performed in mice younger than postnatal day 5, regardless of the type of virus tested. CONCLUSIONS Viral inoculation and expression in the inner ear for the restoration of hearing must not damage cochlear function. Using normal hearing mice as a model, we have achieved this necessary step, which is required for the treatment of many types of congenital deafness that require early intervention.
Collapse
Affiliation(s)
- Yunfeng Wang
- Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, PR China
| | | | | | | | | | | | | | | |
Collapse
|
14
|
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
|
15
|
XIA LI, YIN SHANKAI. Local gene transfection in the cochlea (Review). Mol Med Rep 2013; 8:3-10. [DOI: 10.3892/mmr.2013.1496] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 12/13/2012] [Indexed: 11/06/2022] Open
|
16
|
Budenz CL, Pfingst BE, Raphael Y. The use of neurotrophin therapy in the inner ear to augment cochlear implantation outcomes. Anat Rec (Hoboken) 2012; 295:1896-908. [PMID: 23044834 DOI: 10.1002/ar.22586] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 07/24/2012] [Indexed: 12/12/2022]
Abstract
Severe to profound deafness is most often secondary to a loss of or injury to cochlear mechanosensory cells, and there is often an associated loss of the peripheral auditory neural structures, specifically the spiral ganglion neurons and peripheral auditory fibers. Cochlear implantation is currently our best hearing rehabilitation strategy for severe to profound deafness. These implants work by directly electrically stimulating the remnant auditory neural structures within the deafened cochlea. When administered to the deafened cochlea in animal models, neurotrophins, specifically brain derived neurotrophic factor and neurotrophin-3, have been shown to dramatically improve spiral ganglion neuron survival and stimulate peripheral auditory fiber regrowth. In animal models, neurotrophins administered in combination with cochlear implantation has resulted in significant improvements in the electrophysiological and psychophysical measures of outcome. While further research must be done before these therapies can be applied clinically, neurotrophin therapies for the inner ear show great promise in enhancing CI outcomes and the treatment of hearing loss.
Collapse
Affiliation(s)
- Cameron L Budenz
- Kresge Hearing Research Institute, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | |
Collapse
|
17
|
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
|
18
|
Shibata SB, Budenz CL, Bowling SA, Pfingst BE, Raphael Y. Nerve maintenance and regeneration in the damaged cochlea. Hear Res 2011; 281:56-64. [PMID: 21596129 PMCID: PMC3196294 DOI: 10.1016/j.heares.2011.04.019] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Revised: 04/22/2011] [Accepted: 04/23/2011] [Indexed: 12/22/2022]
Abstract
Following the onset of sensorineural hearing loss, degeneration of mechanosensitive hair cells and spiral ganglion cells (SGCs) in humans and animals occurs to variable degrees, with a trend for greater neural degeneration with greater duration of deafness. Emergence of the cochlear implant prosthesis has provided much needed aid to many hearing impaired patients and has become a well-recognized therapy worldwide. However, ongoing peripheral nerve fiber regression and subsequent degeneration of SGC bodies can reduce the neural targets of cochlear implant stimulation and diminish its function. There is increasing interest in bio-engineering approaches that aim to enhance cochlear implant efficacy by preventing SGC body degeneration and/or regenerating peripheral nerve fibers into the deaf sensory epithelium. We review the advancements in maintaining and regenerating nerves in damaged animal cochleae, with an emphasis on the therapeutic capacity of neurotrophic factors delivered to the inner ear after an insult. Additionally, we summarize the histological process of neuronal degeneration in the inner ear and describe different animal models that have been employed to study this mechanism. Research on enhancing the biological infrastructure of the deafened cochlea in order to improve cochlear implant efficacy is of immediate clinical importance.
Collapse
Affiliation(s)
- Seiji B. Shibata
- Kresge Hearing Research Institute, Department of Otolaryngology, The University of Michigan, Ann Arbor, MI, 48109-5648, USA
| | - Cameron L. Budenz
- Kresge Hearing Research Institute, Department of Otolaryngology, The University of Michigan, Ann Arbor, MI, 48109-5648, USA
| | - Sara A. Bowling
- Kresge Hearing Research Institute, Department of Otolaryngology, The University of Michigan, Ann Arbor, MI, 48109-5648, USA
| | - Bryan E. Pfingst
- Kresge Hearing Research Institute, Department of Otolaryngology, The University of Michigan, Ann Arbor, MI, 48109-5648, USA
| | - Yehoash Raphael
- Kresge Hearing Research Institute, Department of Otolaryngology, The University of Michigan, Ann Arbor, MI, 48109-5648, USA
| |
Collapse
|
19
|
Wise AK, Tu T, Atkinson PJ, Flynn BO, Sgro BE, Hume C, O'Leary SJ, Shepherd RK, Richardson RT. The effect of deafness duration on neurotrophin gene therapy for spiral ganglion neuron protection. Hear Res 2011; 278:69-76. [PMID: 21557994 PMCID: PMC3152700 DOI: 10.1016/j.heares.2011.04.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Revised: 03/17/2011] [Accepted: 04/22/2011] [Indexed: 01/31/2023]
Abstract
A cochlear implant can restore hearing function by electrically exciting spiral ganglion neurons (SGNs) in the deaf cochlea. However, following deafness SGNs undergo progressive degeneration ultimately leading to their death. One significant cause of SGN degeneration is the loss of neurotrophic support that is normally provided by cells within the organ of Corti (OC). The administration of exogenous neurotrophins (NTs) can protect SGNs from degeneration but the effects are short-lived once the source of NTs has been exhausted. NT gene therapy, whereby cells within the cochlea are transfected with genes enabling them to produce NTs, is one strategy for providing a cellular source of NTs that may provide long-term support for SGNs. As the SGNs normally innervate sensory cells within the OC, targeting residual OC cells for gene therapy in the deaf cochlea may provide a source of NTs for SGN protection and targeted regrowth of their peripheral fibers. However, the continual degeneration of the OC over extended periods of deafness may deplete the cellular targets for NT gene therapy and hence limit the effectiveness of this method in preventing SGN loss. This study examined the effects of deafness duration on the efficacy of NT gene therapy in preventing SGN loss in guinea pigs that were systemically deafened with aminoglycosides. Adenoviral vectors containing green fluorescent protein (GFP) with or without genes for Brain Derived Neurotrophic Factor (BDNF) and Neurotrophin-3 (NT3) were injected into the scala media (SM) compartment of cochleae that had been deafened for one, four or eight weeks prior to the viral injection. The results showed that viral transfection of cells within the SM was still possible even after severe degeneration of the OC. Supporting cells (pillar and Deiters' cells), cells within the stria vascularis, the spiral ligament, endosteal cells lining the scala compartments and interdental cells in the spiral limbus were transfected. However, the level of transfection was remarkably lower following longer durations of deafness. There was a significant increase in SGN survival in the entire basal turn for cochleae that received NT gene therapy compared to the untreated contralateral control cochleae for the one week deaf group. In the four week deaf group significant SGN survival was observed in the lower basal turn only. There was no increase in SGN survival for the eight week deaf group in any cochlear region. These findings indicated that the efficacy of NT gene therapy diminished with increasing durations of deafness leading to reduced benefits in terms of SGN protection. Clinically, there remains a window of opportunity in which NT gene therapy can provide ongoing trophic support for SGNs.
Collapse
Affiliation(s)
- Andrew K Wise
- The Bionic Ear Institute, 384-388 Albert Street, East Melbourne, Victoria 3002, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Efficient cochlear gene transfection in guinea-pigs with adeno-associated viral vectors by partial digestion of round window membrane. Gene Ther 2011; 19:255-63. [PMID: 21697953 DOI: 10.1038/gt.2011.91] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The auditory portion of the inner ear, the cochlea, is an ideal organ for local gene transfection owing to its relative isolation. Various carriers have been tested for cochlear gene transfection. To date, viral vectors appear to have much higher transfection efficacy than non-viral mechanisms. Among these vectors, recombinant adeno-associated virus (rAAV) vectors have several advantages such as being non-pathogenic and the ability to produce prolonged gene expression in various cell types. However, rAAV vectors cannot pass through the intact round window membrane (RWM), otherwise a very attractive approach to access the human inner ear. In this study, performed in guinea-pigs, we describe a method to increase the permeability of RWM to rAAV vectors by partial digestion with collagenase solution. Elevated delivery of rAAV across the partially digested RWM increased transfection efficacy to a satisfactory level, even though it was still lower than that achieved by direct cochleostomy injection. Functional tests (auditory brainstem responses) showed that this enzymatic manipulation did not cause permanent hearing loss if applied appropriately. Morphological observations suggested that the damage to RWM caused by partial digestion healed within four weeks. Taken together, these findings suggest that partial digestion of the RWM is a safe and effective method for increasing the transfection of cochlear sensory cells with rAAV.
Collapse
|
21
|
Wang H, Murphy R, Taaffe D, Yin S, Xia L, Hauswirth WW, Bance M, Robertson GS, Wang J. Efficient cochlear gene transfection in guinea-pigs with adeno-associated viral vectors by partial digestion of round window membrane. Gene Ther 2011. [PMID: 21697953 DOI: 10.1038/gt.2011.91gt201191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The auditory portion of the inner ear, the cochlea, is an ideal organ for local gene transfection owing to its relative isolation. Various carriers have been tested for cochlear gene transfection. To date, viral vectors appear to have much higher transfection efficacy than non-viral mechanisms. Among these vectors, recombinant adeno-associated virus (rAAV) vectors have several advantages such as being non-pathogenic and the ability to produce prolonged gene expression in various cell types. However, rAAV vectors cannot pass through the intact round window membrane (RWM), otherwise a very attractive approach to access the human inner ear. In this study, performed in guinea-pigs, we describe a method to increase the permeability of RWM to rAAV vectors by partial digestion with collagenase solution. Elevated delivery of rAAV across the partially digested RWM increased transfection efficacy to a satisfactory level, even though it was still lower than that achieved by direct cochleostomy injection. Functional tests (auditory brainstem responses) showed that this enzymatic manipulation did not cause permanent hearing loss if applied appropriately. Morphological observations suggested that the damage to RWM caused by partial digestion healed within four weeks. Taken together, these findings suggest that partial digestion of the RWM is a safe and effective method for increasing the transfection of cochlear sensory cells with rAAV.
Collapse
Affiliation(s)
- H Wang
- Department of Otolaryngology, Affiliated Sixth People's Hospital of Shanghai Jiao Tong University, Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Adeno-associated virus-mediated gene delivery into the scala media of the normal and deafened adult mouse ear. Gene Ther 2011; 18:569-78. [PMID: 21209625 PMCID: PMC3085601 DOI: 10.1038/gt.2010.175] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Murine models are ideal for studying cochlear gene transfer, as many hearing loss-related mutations have been discovered and mapped within the mouse genome. However, because of the small size and delicate nature, the membranous labyrinth of the mouse is a challenging target for the delivery of viral vectors. To minimize injection trauma, we developed a procedure for the controlled release of adeno-associated viruses (AAVs) into the scala media of adult mice. This procedure poses minimal risk of injury to structures of the cochlea and middle ear, and allows for near-complete preservation of low and middle frequency hearing. In this study, transduction efficiency and cellular specificity of AAV vectors (serotypes 1, 2, 5, 6 and 8) were investigated in normal and drug-deafened ears. Using the cytomegalovirus promoter to drive gene expression, a variety of cell types were transduced successfully, including sensory hair cells and supporting cells, as well as cells in the auditory nerve and spiral ligament. Among all five serotypes, inner hair cells were the most effectively transduced cochlear cell type. All five serotypes of AAV vectors transduced cells of the auditory nerve, though serotype 8 was the most efficient vector for transduction. Our findings indicate that efficient AAV inoculation (via the scala media) can be performed in adult mouse ears, with hearing preservation a realistic goal. The procedure we describe may also have applications for intra-endolymphatic drug delivery in many mouse models of human deafness.
Collapse
|
23
|
Wu J, Liu B, Fan J, Zhu Q, Wu J. Study of protective effect on rat cochlear spiral ganglion after blast exposure by adenovirus-mediated human β-nerve growth factor gene. Am J Otolaryngol 2011; 32:8-12. [PMID: 20022668 DOI: 10.1016/j.amjoto.2009.08.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Accepted: 08/30/2009] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To study whether adenovirus-mediated human β-nerve growth factor (Ad-hNGFβ) gene has any protective effect on rat cochlear spiral ganglion after blast exposure. METHODS Deafness was induced by blast exposure (172.0 dB) in 20 healthy rats. Seven days after blast exposure, Ad-hNGFβ was infused into the perilymphatic space of 10 animals as the hNGFβ/blast group, and artificial perilymph fluid (APF) was infused into the perilymphatic space of 10 animals as the APF/blast control group. An additional control group consisted of 10 healthy rats which received Ad-hNGFβ target gene with no blast exposure (hNGFβ/control group). Auditory functions were monitored by thresholds of auditory brain stem responses (ABR). At weeks 1, 4, and 8 postoperatively, the animals were killed, and the cochleae were removed for immunohistochemical, hematoxylin and eosin staining study. RESULTS The ABR threshold shifts in the hNGFβ/blast group were significantly smaller than that of APF/blast control group. There were no significant differences of the ABR values between before and after operation in the hNGFβ/control group. Expression of Ad-hNGFβ protein was detected in each turn of the cochlea in the first week, with almost equal intensity in all turns. In the fourth week, the reactive intensity decreased. In the eighth week, no reaction was detectable. The results of hematoxylin and eosin stain showed that the number of spiral ganglions in the hNGFβ/blast group was significantly greater than that of the APF/blast control group in the 4th week (P < .01). CONCLUSION Adenovirus-mediated human β-nerve growth factor can be expressed at a high level and for a relatively long period in the blast impaired cochlea, suggesting that Ad-hNGFβ has a protective effect on rat cochlear spiral ganglion cells after blast exposure.
Collapse
Affiliation(s)
- Jian Wu
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China.
| | | | | | | | | |
Collapse
|
24
|
Lei L, Han D. Efficient transduction of spiral ganglion cells using adenovirus type 5 vector in the rat. Acta Otolaryngol 2010; 130:810-4. [PMID: 20205620 DOI: 10.3109/00016480903510742] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONCLUSIONS The adenovirus-5 vector specifically transduced spiral ganglion cells with high efficiency, suggesting that it is a potential gene therapeutic tool for the survival of spiral ganglion cells with secondary injury. OBJECTIVES This study aimed to find a suitable viral vector allowing efficient transduction to spiral ganglion cells. METHODS Lentivirus, adeno-associated virus-2 and adenovirus-5 constructs habouring green fluorescence protein (GFP) gene were injected into scala tympani via the round window membrane of rat. Distribution and fluorescence intensity of GFP within the cochlea were estimated using a fluorescence microscope. RESULTS The GFP expressions mediated by all three viral vectors were observed in multiple cell types of the cochlea. Compared with the other two viral vectors, the adenovirus-5 vector efficiently transduced cochlear spiral ganglion cells in vivo and was still present 2 weeks after viral vector injection.
Collapse
Affiliation(s)
- Li Lei
- Department of Otolaryngology Head and Neck surgery, Affiliated Beijng Tongren Hospital, Capital Medical University, PR China
| | | |
Collapse
|
25
|
Wise AK, Hume CR, Flynn BO, Jeelall YS, Suhr CL, Sgro BE, O'Leary SJ, Shepherd RK, Richardson RT. Effects of localized neurotrophin gene expression on spiral ganglion neuron resprouting in the deafened cochlea. Mol Ther 2010; 18:1111-22. [PMID: 20216530 DOI: 10.1038/mt.2010.28] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
A cochlear implant may be used to electrically stimulate spiral ganglion neurons (SGNs) in people with severe sensorineural hearing loss (SNHL). However, these neurons progressively degenerate after SNHL due to loss of neurotrophins normally supplied by sensory hair cells (HCs). Experimentally, exogenous neurotrophin administration prevents SGN degeneration but can also result in abnormal resprouting of their peripheral fibers. This study aimed to create a target-derived neurotrophin source to increase neuron survival and redirect fiber resprouting following SNHL. Adenoviral (Ad) vectors expressing green fluorescent protein (GFP) alone or in combination with brain-derived neurotrophic factor (BDNF) or neurotrophin-3 (NT3) were injected into the cochlear scala tympani or scala media of guinea-pigs (GPs) deafened via aminoglycosides for 1 week. After 3 weeks, cochleae were examined for gene expression, neuron survival, and the projection of peripheral fibers in response to gene expression. Injection of vectors into the scala media resulted in more localized gene expression than scala tympani injection with gene expression consistently observed within the partially degenerated organ of Corti. There was also greater neuron survival and evidence of localized fiber responses to neurotrophin-expressing cells within the organ of Corti from scala media injections (P < 0.05), a first step in promoting organized resprouting of auditory peripheral fibers via gene therapy.
Collapse
Affiliation(s)
- Andrew K Wise
- Bionic Ear Institute, East Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Shibata SB, Cortez SR, Beyer LA, Wiler JA, Di Polo A, Pfingst BE, Raphael Y. Transgenic BDNF induces nerve fiber regrowth into the auditory epithelium in deaf cochleae. Exp Neurol 2010; 223:464-72. [PMID: 20109446 DOI: 10.1016/j.expneurol.2010.01.011] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2009] [Revised: 01/20/2010] [Accepted: 01/21/2010] [Indexed: 12/26/2022]
Abstract
Sensory organs typically use receptor cells and afferent neurons to transduce environmental signals and transmit them to the CNS. When sensory cells are lost, nerves often regress from the sensory area. Therapeutic and regenerative approaches would benefit from the presence of nerve fibers in the tissue. In the hearing system, retraction of afferent innervation may accompany the degeneration of auditory hair cells that is associated with permanent hearing loss. The only therapy currently available for cases with severe or complete loss of hair cells is the cochlear implant auditory prosthesis. To enhance the therapeutic benefits of a cochlear implant, it is necessary to attract nerve fibers back into the cochlear epithelium. Here we show that forced expression of the neurotrophin gene BDNF in epithelial or mesothelial cells that remain in the deaf ear induces robust regrowth of nerve fibers towards the cells that secrete the neurotrophin, and results in re-innervation of the sensory area. The process of neurotrophin-induced neuronal regeneration is accompanied by significant preservation of the spiral ganglion cells. The ability to regrow nerve fibers into the basilar membrane area and protect the auditory nerve will enhance performance of cochlear implants and augment future cell replacement therapies such as stem cell implantation or induced transdifferentiation. This model also provides a general experimental stage for drawing nerve fibers into a tissue devoid of neurons, and studying the interaction between the nerve fibers and the tissue.
Collapse
Affiliation(s)
- Seiji B Shibata
- Kresge Hearing Research Institute, Department of Otolaryngology, The University of Michigan, 1150 W. Med. Cntr. Dr., Ann Arbor, MI 48109-5648, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
O'Leary SJ, Richardson RR, McDermott HJ. Principles of design and biological approaches for improving the selectivity of cochlear implant electrodes. J Neural Eng 2009; 6:055002. [PMID: 19721188 DOI: 10.1088/1741-2560/6/5/055002] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The perceptual performance of cochlear implant recipients seems to have reached a plateau in recent years. This may be attributable to inadequate neural selectivity of available intracochlear electrodes, caused by current spread and electrode interactions. Attempts to improve electrode selectivity have included manipulating the number and configuration of electrodes that are stimulated at any one time, displacing perilymph from the cochlea to restrict current flow along the cochlea, and reducing the distance between electrodes and neurons. One experimental approach by which the distance between neurons and electrodes may be reduced is to use neurotrophic factors to promote the regeneration of the peripheral dendrites of auditory neurons and guide them towards intracochlear electrodes. The likely requirements of a system for regenerating auditory neurons towards the cochlear electrode include either a stable release of neurotrophin, or transient neurotrophin followed by electrical stimulation; a close proximity of electrode to osseous spiral lamina or a polymer to bridge the gap between the two; guidance signals to attract neurons towards the electrode; patterning of the electrode surface to direct dendrites to electrode contacts and a 'stop' signal to arrest regeneration once the electrode has been reached.
Collapse
Affiliation(s)
- Stephen J O'Leary
- Department of Otolaryngology, University of Melbourne, Royal Victorian Eye and Ear Hospital, Australia.
| | | | | |
Collapse
|
28
|
VAN LAER LUT, VAN CAMP GUY. Autosomal Dominant Nonsyndromic Hearing Impairment: an Overview. ACTA ACUST UNITED AC 2009. [DOI: 10.1080/16513860310003111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
29
|
Abstract
Gene transfer into the cells of the cochlea is useful for both research and therapy. Bovine adeno-associated virus (BAAV) is a new viral vector with potential for long-term gene expression with little or no side effects. In this study, we assessed transgene expression using BAAV with beta-actin-GFP as a reporter gene, in the cochleae of normal and deafened guinea pigs. We used two different routes to inoculate the cochlea: scala media (SM) or scala tympani (ST). Auditory brainstem response assessments were carried out before inoculation, 7 days after inoculation and immediately before killing, to assess the functional consequences of the treatment. We observed threshold shifts because of the surgical invasion, but no apparent pathology associated with the virus. Fourteen days after the injection, animals were killed and cochleae assessed histologically. Epi-fluorescence showed that BAAV transduced the supporting cells of both normal and deafened animals through SM and ST inoculations. Transgene expression in cells of the membranous labyrinth after ST inoculation is an important outcome because of the greater feasibility of this route for future clinical application. BAAV facilitates efficient transduction of the membranous labyrinth epithelium with minimum pathogenicity and may become clinically applicable for inner ear gene therapy.
Collapse
|
30
|
Richardson RT, Wise AK, Andrew JK, O'Leary SJ. Novel drug delivery systems for inner ear protection and regeneration after hearing loss. Expert Opin Drug Deliv 2009; 5:1059-76. [PMID: 18817513 DOI: 10.1517/17425247.5.10.1059] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND A cochlear implant, the only current treatment for restoring auditory perception after severe or profound sensorineural hearing loss (SNHL), works by electrically stimulating spiral ganglion neurons (SGNs). However, gradual degeneration of SGNs associated with SNHL can compromise the efficacy of the device. OBJECTIVE To review novel drug delivery systems for preserving and/or regenerating sensory cells in the cochlea after SNHL. METHODS The effectiveness of traditional cochlear drug delivery systems is compared to newer techniques such as cell, polymer and gene transfer technologies. Special requirements for local drug delivery to the cochlea are discussed, such as protecting residual hearing and site-specific drug delivery for cell preservation and regeneration. RESULTS/CONCLUSIONS Drug delivery systems with the potential for immediate clinical translation, as well as those that will contribute to the future of hearing preservation or cochlear cellular regeneration, are identified.
Collapse
Affiliation(s)
- Rachael T Richardson
- Bionic Ear Institute, 384 Albert Street, East Melbourne, Victoria 3002, Australia.
| | | | | | | |
Collapse
|
31
|
Study on neural stem cell transplantation into natural rat cochlea via round window. Am J Otolaryngol 2009; 30:8-16. [PMID: 19027507 DOI: 10.1016/j.amjoto.2007.12.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2007] [Revised: 12/28/2007] [Accepted: 12/29/2007] [Indexed: 11/21/2022]
Abstract
OBJECTIVE The aim of the study is to investigate the survival of neural stem cells (NSCs) in normal rat cochlea and their potential effect on auditory function and cochlea structures via round window transplantation. METHODS In comparison with the normal rats without any transplantation (group III), normal rat cochleae were transplanted with NSCs infected with adenovirus carrying green fluorescence protein (GFP) gene (group I) or the artificial perilymph (group II) via round windows. Auditory functions were monitored by thresholds of auditory brain stem responses (ABRs); the cochlea structures were examined by hematoxylin and eosin staining; survivals of implanted NSCs were determined by the expression of GFP; survivals of hair cells were accessed by whole mount preparation; and ultrastructures of hair cells were examined by scanning electron microscopy. RESULT There were significant differences in the click-ABR thresholds in rats among all 3 groups neither at pretransplantation nor at posttransplantation; there were no significant differences in these values before and after transplantation in the same rats from each group. After transplantation, the cochlea structures were normal in both group I and group II. Grafted NSCs were visualized by the GFP expression in every turn of the cochlea in all animals of group I. There were no significant differences in the losses of outer hair cells (OHCs) among 3 groups. The inner hair cells and most OHCs were normal in every turns of cochleae of all groups. CONCLUSION Neural stem cells survived in normal rat cochlea after transplantation via round window and showed no obvious effects on auditory functions and inner ear pathologic examination of the rat cochlea.
Collapse
|
32
|
Konishi M, Kawamoto K, Izumikawa M, Kuriyama H, Yamashita T. Gene transfer into guinea pig cochlea using adeno-associated virus vectors. J Gene Med 2008; 10:610-8. [PMID: 18338819 DOI: 10.1002/jgm.1189] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Several genes are candidates for treating inner ear diseases. For clinical applications, minimally invasive approaches to the inner ear are desirable along with minimal side-effects. METHODS Adeno-associated virus (AAV) was used as a vector into the guinea pig inner ear. Six AAV-cytomegalovirus hybrids (AAV-2/1, -2/2, -2/5, -2/7, -2/8 and -2/9) were infused into perilymph of the cochlea basal turn, an approach that could be used in cochlear implant surgery. At 7 days after injection, distribution of gene expression, hearing and morphology were evaluated. Adenoviral vector was also used to compare distributions of gene expression. Moreover, distribution of cell surface receptors of AAV in the cochlea was examined using immunohistochemistry. RESULTS Using the perilymphatic approach, adenovirus could be transferred to mesothelial cells lining the perilymph, but not sensory cells. Conversely, all AAV serotypes displayed tissue tropism to inner hair cells, with AAV-2/2 showing particularly efficient transfer to sensory cells. This tissue tropism of AAV could not be explained by the distribution of AAV receptors. Hearing and morphology were largely unaffected. CONCLUSIONS Our results indicate that AAV vector can be safely applied to the inner ear and AAV-2/2 offers a good tool for transferring transgenes into sensory cells of the inner ear efficiently without toxicity.
Collapse
Affiliation(s)
- Masaya Konishi
- Departments of Otolaryngology and Head-Neck Surgery, Kansai Medical University, Moriguchi, Osaka, Japan.
| | | | | | | | | |
Collapse
|
33
|
Iizuka T, Kanzaki S, Mochizuki H, Inoshita A, Narui Y, Furukawa M, Kusunoki T, Saji M, Ogawa K, Ikeda K. Noninvasive in vivo delivery of transgene via adeno-associated virus into supporting cells of the neonatal mouse cochlea. Hum Gene Ther 2008; 19:384-90. [PMID: 18439125 DOI: 10.1089/hum.2007.167] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
There are a number of genetic diseases that affect the cochlea early in life, which require normal gene transfer in the early developmental stage to prevent deafness. The delivery of adenovirus (AdV) and adeno-associated virus (AAV) was investigated to elucidate the efficiency and cellular specificity of transgene expression in the neonatal mouse cochlea. The extent of AdV transfection is comparable to that obtained with adult mice. AAV-directed gene transfer after injection into the scala media through a cochleostomy showed transgene expression in the supporting cells, inner hair cells (IHCs), and lateral wall with resulting hearing loss. On the other hand, gene expression was observed in Deiters cells, IHCs, and lateral wall without hearing loss after the application of AAV into the scala tympani through the round window. These findings indicate that injection of AAV into the scala tympani of the neonatal mouse cochlea therefore has the potential to efficiently and noninvasively introduce transgenes to the cochlear supporting cells, and this modality is thus considered to be a promising strategy to prevent hereditary prelingual deafness.
Collapse
Affiliation(s)
- Takashi Iizuka
- Department of Otorhinolaryngology, Juntendo University School of Medicine, Tokyo 113-8421, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Transfer of exogenous genetic material into the mammalian inner ear using viral vectors has been characterized over the last decade. A number of different viral vectors have been shown to transfect the varying cell types of the nonprimate mammalian inner ear. Several routes of delivery have been identified for introduction of vectors into the inner ear while minimizing injury to existing structures and at the same time ensuring widespread distribution of the agent throughout the cochlea and the rest of the inner ear. These studies raise the possibility that gene transfer may be developed as a potential strategy for treating inner ear dysfunction in humans. Furthermore, a recent report showing successful transfection of excised human vestibular epithelia offers proof of principle that viral gene transfer is a viable strategy for introduction and expression of exogenous genetic material to restore function to the inner ear. Human vestibular epithelia were harvested from patients undergoing labyrinthectomy, either for intractable Ménière's disease or vestibular schwannoma resection, and cultured for as long as 5 days. In those experiments, recombinant, multiply-deleted, replication-deficient adenoviral vectors were used to transfect and express a reporter gene as well as the functionally relevant gene, wild-type KCNQ4, a potassium channel gene that when mutated causes the autosomal dominant HL DFNA2.Here, we review the current state of viral-mediated gene transfer in the inner ear and discuss different viral vectors, routes of delivery, and potential applications of gene therapy. Emphasis is placed on experiments demonstrating viral transfection of human inner ear tissue and implications of these findings and for the future of gene therapy in the human inner ear.
Collapse
|
35
|
Hendricks JL, Chikar JA, Crumling MA, Raphael Y, Martin DC. Localized cell and drug delivery for auditory prostheses. Hear Res 2008; 242:117-31. [PMID: 18573323 DOI: 10.1016/j.heares.2008.06.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2007] [Revised: 05/09/2008] [Accepted: 06/02/2008] [Indexed: 12/20/2022]
Abstract
Localized cell and drug delivery to the cochlea and central auditory pathway can improve the safety and performance of implanted auditory prostheses (APs). While generally successful, these devices have a number of limitations and adverse effects including limited tonal and dynamic ranges, channel interactions, unwanted stimulation of non-auditory nerves, immune rejection, and infections including meningitis. Many of these limitations are associated with the tissue reactions to implanted auditory prosthetic devices and the gradual degeneration of the auditory system following deafness. Strategies to reduce the insertion trauma, degeneration of target neurons, fibrous and bony tissue encapsulation, and immune activation can improve the viability of tissue required for AP function as well as improve the resolution of stimulation for reduced channel interaction and improved place-pitch and level discrimination. Many pharmaceutical compounds have been identified that promote the viability of auditory tissue and prevent inflammation and infection. Cell delivery and gene therapy have provided promising results for treating hearing loss and reversing degeneration. Currently, many clinical and experimental methods can produce extremely localized and sustained drug delivery to address AP limitations. These methods provide better control over drug concentrations while eliminating the adverse effects of systemic delivery. Many of these drug delivery techniques can be integrated into modern auditory prosthetic devices to optimize the tissue response to the implanted device and reduce the risk of infection or rejection. Together, these methods and pharmaceutical agents can be used to optimize the tissue-device interface for improved AP safety and effectiveness.
Collapse
Affiliation(s)
- Jeffrey L Hendricks
- Department of Biomedical Engineering, The University of Michigan, 1107 Gerstacker Building, 2200 Bonisteel Boulevard, Ann Arbor, MI 48109-2099, USA.
| | | | | | | | | |
Collapse
|
36
|
[Expression patterns of non-viral transfection with GFP in the organ of Corti in vitro and in vivo. Gene therapy of the inner ear with non-viral vectors]. HNO 2008; 56:524-9. [PMID: 18409074 DOI: 10.1007/s00106-008-1738-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Diseases of the inner ear such as presbycusis, tinnitus, sudden hearing loss, and vertigo affect many patients, but so far there are no specific therapy options. Gene therapy might become a potential modality of treatment. Viral vectors are standard in animal models to date. Future considerations, however, call for a further evaluation of non-viral transfection methods. MATERIAL AND METHODS The non-viral transfection agents Metafectene, Superfect, Effectene, and Mirus TransIT were incubated with a plasmid coding for GFP. In vivo, the plasmid-agent mix was injected via the membrane of the round window, and 48 h later the inner ear was perfused, harvested, decalcified, and histologically evaluated for GFP expression. RESULTS Cationic lipids (Metafectene) and dendrimers (Superfect) were able to transfect cells in the area of the organ of Corti and lead to GFP expression. The polyamine (Mirus TransIT) did show expression of GFP in the area of Rosenthal's canal and in the area of the inner hair cell. The combination of a non-liposomal lipid with a DNA condensing component (Effectene) did not show transfection of the organ of Corti. In the area of the spiral ganglia cells, GFP expression was seen with all the transfection agents. CONCLUSIONS Non-viral transfection agents are able to introduce a reporter gene in cells of the inner ear in vitro and in vivo. There are, however, differences in the efficiency of the transfection. They might be an alternative in gene therapy of the inner ear. Further investigations to elucidate their potential are needed.
Collapse
|
37
|
Pietola L, Aarnisalo AA, Joensuu J, Pellinen R, Wahlfors J, Jero J. HOX-GFP and WOX-GFP lentivirus vectors for inner ear gene transfer. Acta Otolaryngol 2008; 128:613-20. [PMID: 18568493 DOI: 10.1080/00016480701663409] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
CONCLUSION GFP transgene was expressed in the lining cells of the perilymphatic space. Lentivirus vectors are safe and cause only minimal inflammatory reaction. Transgene products can be delivered into the perilymph by utilizing lentivirus vectors. OBJECTIVES To analyze the efficiency and safety of lentiviral vectors HOX-GFP and WOX-GFP in intracochlear gene transfer. MATERIALS AND METHODS Lentivirus vectors were tested for their transduction efficiency in vivo in CD-1 mice. Half of the animals were pretreated with kanamycin. Lentivirus vector or saline (1 microl) was injected into the inner ear. All the animals were sacrificed 14 days after the surgery and the cochleae and selected organs were analyzed immunohistochemically. RESULTS HOX-GFP and WOX-GFP expression was restricted to the lining cells of the scala tympani and scala vestibuli. No GFP expression was seen in the organ of Corti or the spiral ganglion. Aminoglycoside treatment had no effect on the expression of these vectors. The distant spread of lentivirus vectors was minimal; only the liver of one animal showed some GFP expression. Inflammatory reaction caused by these vectors was mild. Few inflammatory cells were found in the perilymphatic space of the cochlea and in the vestibular organ.
Collapse
|
38
|
Xia A, Wooltorton JRA, Palmer DJ, Ng P, Pereira FA, Eatock RA, Oghalai JS. Functional prestin transduction of immature outer hair cells from normal and prestin-null mice. J Assoc Res Otolaryngol 2008; 9:307-20. [PMID: 18506528 DOI: 10.1007/s10162-008-0121-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2007] [Accepted: 04/04/2008] [Indexed: 10/22/2022] Open
Abstract
Prestin is a membrane protein in the outer hair cell (OHC) that has been shown to be essential for electromotility. OHCs from prestin-null mice do not express prestin, do not have a nonlinear capacitance (the electrical signature of electromotility), and are smaller in size than wild-type OHCs. We sought to determine whether prestin-null OHCs can be transduced to incorporate functional prestin protein in a normal fashion. A recombinant helper-dependent adenovirus expressing prestin and green fluorescent protein (HDAd-prestin-GFP) was created and tested in human embryonic kidney cells (HEK cells). Transduced HEK cells demonstrated membrane expression of prestin and nonlinear capacitance. HDAd-prestin-GFP was then applied to cochlear sensory epithelium explants harvested from wild-type and prestin-null mice at postnatal days 2-3, the age at which native prestin is just beginning to become functional in wild-type mice. At postnatal days 4-5, we investigated transduced OHCs for (1) their prestin expression pattern as revealed by immunofluorescence; (2) their cell surface area as measured by linear capacitance; and (3) their prestin function as indicated by nonlinear capacitance. HDAd-prestin-GFP efficiently transduced OHCs of both genotypes and prestin protein localized to the plasma membrane. Whole-cell voltage clamp studies revealed a nonlinear capacitance in transduced wild-type and prestin-null OHCs, but not in non-transduced cells of either genotype. Prestin transduction did not increase the linear capacitance (cell surface area) for either genotype. In peak nonlinear capacitance, voltage at peak nonlinear capacitance, charge density of the nonlinear capacitance, and shape of the voltage-capacitance curves, the transduced cells of the two genotypes resembled each other and previously reported data from adult wild-type mouse OHCs. Thus, prestin introduced into prestin-deficient OHCs segregates normally to the cell membrane and generates a normal nonlinear capacitance, indicative of normal prestin function.
Collapse
Affiliation(s)
- Anping Xia
- Bobby R. Alford Department of Otolaryngology, Head and Neck Surgery, Baylor College of Medicine, One Baylor Plaza, NA102, Houston, TX 77030, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Pettingill LN, Richardson RT, Wise AK, O'Leary SJ, Shepherd RK. Neurotrophic factors and neural prostheses: potential clinical applications based upon findings in the auditory system. IEEE Trans Biomed Eng 2007; 54:1138-48. [PMID: 17551571 PMCID: PMC1886005 DOI: 10.1109/tbme.2007.895375] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Spiral ganglion neurons (SGNs) are the target cells of the cochlear implant, a neural prosthesis designed to provide important auditory cues to severely or profoundly deaf patients. The ongoing degeneration of SGNs that occurs following a sensorineural hearing loss is, therefore, considered a limiting factor in cochlear implant efficacy. We review neurobiological techniques aimed at preventing SGN degeneration using exogenous delivery of neurotrophic factors. Application of these proteins prevents SGN degeneration and can enhance neurite outgrowth. Furthermore, chronic electrical stimulation of SGNs increases neurotrophic factor-induced survival and is correlated with functional benefits. The application of neurotrophic factors has the potential to enhance the benefits that patients can derive from cochlear implants; moreover, these techniques may be relevant for use with neural prostheses in other neurological conditions.
Collapse
MESH Headings
- Animals
- Cell Survival/drug effects
- Cell Survival/physiology
- Cochlea/drug effects
- Cochlea/physiopathology
- Cochlear Implants/trends
- Combined Modality Therapy
- Disease Models, Animal
- Electric Stimulation Therapy/instrumentation
- Electric Stimulation Therapy/methods
- Electrodes, Implanted
- Evoked Potentials, Auditory, Brain Stem/drug effects
- Evoked Potentials, Auditory, Brain Stem/physiology
- Hearing Loss, Sensorineural/pathology
- Hearing Loss, Sensorineural/physiopathology
- Hearing Loss, Sensorineural/therapy
- Membrane Potentials/physiology
- Nerve Degeneration/drug therapy
- Nerve Degeneration/physiopathology
- Nerve Degeneration/prevention & control
- Nerve Growth Factors/administration & dosage
- Neurons, Afferent/drug effects
- Neurons, Afferent/physiology
- Recovery of Function/drug effects
- Recovery of Function/physiology
- Spiral Ganglion/drug effects
- Spiral Ganglion/physiology
- Treatment Outcome
Collapse
|
40
|
Aarnisalo AA, Pietola L, Joensuu J, Isosomppi J, Aarnisalo P, Dinculescu A, Lewin AS, Flannery J, Hauswirth WW, Sankila EM, Jero J. Anti-clarin-1 AAV-delivered ribozyme induced apoptosis in the mouse cochlea. Hear Res 2007; 230:9-16. [PMID: 17493778 DOI: 10.1016/j.heares.2007.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2006] [Revised: 03/19/2007] [Accepted: 03/20/2007] [Indexed: 02/08/2023]
Abstract
Usher syndrome type 3 is caused by mutations in the USH3A gene, which encodes the protein clarin-1. Clarin-1 is a member of the tetraspanin superfamily (TM4SF) of transmembrane proteins, expressed in the organ of Corti and spiral ganglion cells of the mouse ear. We have examined whether the AAV-mediated anti-clarin ribozyme delivery causes apoptotic cell death in vivo in the organ of Corti. We used an AAV-2 vector delivered hammerhead ribozyme, AAV-CBA-Rz, which specifically recognizes and cleaves wild type mouse clarin-1 mRNA. Cochleae of CD-1 mice were injected either with 1mul of the AAV-CBA-Rz, or control AAV vectors containing the green fluorescent protein (GFP) marker gene (AAV-CBA-GFP). Additional controls were performed with saline only. At one-week and one-month post-injection, the animals were sacrificed and the cochleae were studied by histology and fluorescence imaging. Mice injected with AAV-CBA-GFP displayed GFP reporter expression of varying fluorescence intensity throughout the length of the cochlea in the outer and inner hair cells and stria vascularis, and to a lesser extent, in vestibular epithelial cells. GFP expression was not detectable in the spiral ganglion. The pro-apoptotic effect of AAV-CBA-delivered anti-clarin-1 ribozymes was evaluated by TUNEL-staining. We observed in the AAV-CBA-Rz, AAV-CBA-GFP and saline control groups apoptotic nuclei in the outer and inner hair cells and in the stria vascularis one week after the microinjection. The vestibular epithelium was also observed to contain apoptotic cells. No TUNEL-positive spiral ganglion neurons were detected. After one-month post-injection, the AAV-CBA-Rz-injected group had significantly more apoptotic outer and inner hair cells and cells of the stria vascularis than the AAV-CBA-GFP group. In this study, we demonstrate that AAV-CBA mediated clarin-1 ribozyme may induce apoptosis of the cochlear hair cells and cells of the stria vascularis. Surprisingly, we did not observe apoptosis in spiral ganglion cells, which should also be susceptible to clarin-1 mRNA cleavage. This result may be due to the injection technique, the promoter used, or tropism of the AAV serotype 2 viral vector. These results suggest the role of apoptosis in the progression of USH3A hearing loss warrants further evaluation.
Collapse
Affiliation(s)
- A A Aarnisalo
- Department Otorhinolaryngology, University of Helsinki, Helsinki, Finland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Stone IM, Lurie DI, Kelley MW, Poulsen DJ. Adeno-associated virus-mediated gene transfer to hair cells and support cells of the murine cochlea. Mol Ther 2005; 11:843-8. [PMID: 15922954 DOI: 10.1016/j.ymthe.2005.02.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2004] [Revised: 02/03/2005] [Accepted: 02/03/2005] [Indexed: 11/19/2022] Open
Abstract
More than 28 million Americans suffer from various forms of hearing loss. The lack of effective treatments for many forms of hearing disorders has prompted interest in the potential application of gene delivery techniques to treat both inherited and pathological hearing disorders. However, to develop a gene therapy strategy that will successfully treat hearing disorders, appropriate vectors that are capable of transducing cochlear hair cells and support cells must be identified. In the present study, we examined the efficiency with which AAV vectors (serotypes 1, 2, and 5) transduce hair cells and support cells in cochlear explants from P0 and E13 mice. We further examined the ability of the CBA and GFAP promoters to drive expression of a GFP marker gene in hair cells and support cells. Robust GFP expression was observed in hair cells and support cells following transduction of primary murine cochlear explants with AAV serotypes 1 and 2, but not serotype 5. The CBA promoter predominantly drove GFP expression in hair cells. In contrast, strong expression from the GFAP promoter was observed primarily in support cells. Thus, using AAV vectors and specific promoters, cell-type-specific expression of transgenes can be established within the cochlea.
Collapse
Affiliation(s)
- Ida M Stone
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, No. 1552, Missoula, MT 59812, USA
| | | | | | | |
Collapse
|
42
|
Liu Y, Okada T, Sheykholeslami K, Shimazaki K, Nomoto T, Muramatsu SI, Kanazawa T, Takeuchi K, Ajalli R, Mizukami H, Kume A, Ichimura K, Ozawa K. Specific and efficient transduction of cochlear inner hair cells with recombinant adeno-associated virus type 3 vector. Mol Ther 2005; 12:725-33. [PMID: 16169458 DOI: 10.1016/j.ymthe.2005.03.021] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2004] [Revised: 03/23/2005] [Accepted: 03/24/2005] [Indexed: 11/17/2022] Open
Abstract
Recombinant adeno-associated virus (AAV) vectors are of interest for cochlear gene therapy because of their ability to mediate the efficient transfer and long-term stable expression of therapeutic genes in a wide variety of postmitotic tissues with minimal vector-related cytotoxicity. In the present study, seven AAV serotypes (AAV1-5, 7, 8) were used to construct vectors. The expression of EGFP by the chicken beta-actin promoter associated with the cytomegalovirus immediate-early enhancer in cochlear cells showed that each of these serotypes successfully targets distinct cochlear cell types. In contrast to the other serotypes, the AAV3 vector specifically transduced cochlear inner hair cells with high efficiency in vivo, while the AAV1, 2, 5, 7, and 8 vectors also transduced these and other cell types, including spiral ganglion and spiral ligament cells. There was no loss of cochlear function with respect to evoked auditory brain-stem responses over the range of frequencies tested after the injection of AAV vectors. These findings are of value for further molecular studies of cochlear inner hair cells and for gene replacement strategies to correct recessive genetic hearing loss due to monogenic mutations in these cells.
Collapse
Affiliation(s)
- Yuhe Liu
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical School, Minami-kawachi, Kawachi, Tochigi, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
With the completion of the sequencing of the human genome, the field of medicine is undergoing a dramatic and fundamental change. The identification of our genes and the proteins they encode and the mechanisms of mutations that are pathogenic will allow us to devise revolutionary new ways to diagnose, treat and prevent the thousands of disorders that affect us. Certainly, disorders of the auditory system are no exception. Revealing the molecular mechanisms of hearing and understanding the role of each player in the intricate auditory network could enable us to employ gene- or cell-based therapy to cure or prevent hearing loss. To this end, much emphasis has been placed on the identification and characterization of genes involved in human deafness, as well as research on mouse models for deafness. Ultimately, the effect of genomics on medicine will be dramatic, providing us with the ability to cure sensory defects, a tangible goal that is now within our reach.
Collapse
Affiliation(s)
- Orna Atar
- Department of Human Genetics and Molecular Medicine, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | |
Collapse
|
44
|
Di Pasquale G, Rzadzinska A, Schneider ME, Bossis I, Chiorini JA, Kachar B. A Novel Bovine Virus Efficiently Transduces Inner Ear Neuroepithelial Cells. Mol Ther 2005; 11:849-55. [PMID: 15922955 DOI: 10.1016/j.ymthe.2005.02.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2004] [Revised: 02/03/2005] [Accepted: 02/03/2005] [Indexed: 11/22/2022] Open
Abstract
Disruption of the cellular composition or arrangement of the sensory epithelia due to hair cell or supporting cell damage leads to hearing loss and vestibular dysfunctions. These peripheral hearing disorders make good targets for gene therapy; however, development requires efficient gene transfer methods for the inner ear. Here we characterized the cellular tropism of a novel adeno-associated bovine virus vector (BAAV) in cultured rat inner ear epithelia. To help identify transduced cells, we used beta-actin-GFP as a reporter gene. We found that BAAV efficiently transduced auditory and vestibular hair cells as well as all types of supporting cells with no apparent pathological effects. The number of transduced hair cells significantly increased in both a dose- and a time-dependent manner. Transduction was independent of the cells' maturation state and was observed in both P2 and P10 cultures. Interestingly, even after several days of incubation with BAAV, hair cells demonstrated varying progression of beta-actin-GFP incorporation into the stereocilia. This suggests that the onset of viral transduction can occur throughout the course of the experiment. Of the other tested AAVs, AAV2 and AAV5 transduced only a small percentage of inner and vestibular hair cells, respectively, whereas no transduction was detected with AAV4.
Collapse
Affiliation(s)
- Giovanni Di Pasquale
- Gene Therapy and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
45
|
Duan M, Venail F, Spencer N, Mezzina M. Treatment of peripheral sensorineural hearing loss: gene therapy. Gene Ther 2004; 11 Suppl 1:S51-6. [PMID: 15454957 DOI: 10.1038/sj.gt.3302369] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Noise, chemicals and genetic defects are all common causes of irreversible hearing loss, which at present have no cure. Gene therapy may soon be utilized in both the protection and the treatment of these exogenous and endogenous sources of hearing loss. Gene therapy technology is rapidly developing and the inner ear is a particularly feasible model for gene therapy. This review outlines our current understanding of the mechanisms behind deafness and prospects for treatment, discusses the inner ear model in detail and reviews the efforts that have been made in inner ear gene therapy. Finally, the proposed next steps will be discussed. The viral mediated delivery of neurotrophins and antioxidants offers imminent promise in preventing and treating exogenous hearing loss and improving cochlear implant therapy.
Collapse
Affiliation(s)
- M Duan
- Department of Clinical Neuroscience and Center for Hearing and Communication Research, Building MI-ENT, Karolinska Hospital, Stockholm, Sweden
| | | | | | | |
Collapse
|
46
|
Abstract
Vectors derived from adeno-associated viruses (AAV) represent a promising tool for retinal gene transfer in pre-clinical and clinical settings. AAV vectors efficiently transduce dividing and non-dividing cells, escape cellular immunity and result in long-non-term transduction. In addition, they may be targeted to specific retinal cell types by taking advantage of surface proteins from various AAV serotypes thus limiting transfer of therapeutic genes to those cells requiring correction. This review will provide an overview of the properties of AAV vectors followed by a detailed report of their use in retinal gene transfer for mendelian and non-mendelian disorders.
Collapse
Affiliation(s)
- Enrico M Surace
- Telethon Institute of Genetics and Medicine, Via P. Castellino 111, Naples 80131, Italy
| | | |
Collapse
|
47
|
Affiliation(s)
- Anil K Lalwani
- Laboratory of Molecular Otology, Department of Otolaryngology, New York University School of Medicine, New York, USA.
| | | |
Collapse
|
48
|
Shi W, Teschendorf C, Muzyczka N, Siemann DW. Gene therapy delivery of endostatin enhances the treatment efficacy of radiation. Radiother Oncol 2003; 66:1-9. [PMID: 12559515 DOI: 10.1016/s0167-8140(02)00280-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND AND PURPOSE To evaluate whether sustained expression of mouse endostatin by adeno-associated virus (AAV)-mediated gene transfer can enhance the treatment efficacy of ionizing radiation. MATERIALS AND METHODS Mouse endostatin was cloned into recombinant AAV (rAAV) under the control of CMV beta-actin promoter. Recombinant mouse endostatin expressed via AAV gene transfer was tested for biological activity in endothelial cells. The impact of elevated serum levels of endostatin on tumor-induced angiogenesis was evaluated using an in vivo angiogenesis assay. The anti-tumor efficacy of combining rAAV-mediated endostatin delivery with radiation was evaluated in a human colorectal tumor model (HT29). RESULTS Recombinant mouse endostatin expressed through an AAV vector (rAAV-mEndo) inhibited endothelial cell proliferation (by 40-45%) and migration (by 22-33%). Intramuscular injection of rAAV-mEndo (1x10(9) i.u.) led to a sustained serum endostatin level of approximately 500 ng/ml. Compared to control animals this endostatin level was sufficient to inhibit tumor cell-induced vessel formation (37 vs. 28.5, P<0.05) and delay the growth of HT29 xenografts (time from 200 to 1,000 mm(3), 21 vs. 34.5 days, P<0.05). When combined with ionizing radiation, elevated serum endostatin levels significantly enhanced the time for tumors to grow from 200 to 1,000 mm(3) (radiation, 34 days; endostatin plus radiation, 50 days, P<0.05). CONCLUSION The delivery of endostatin via rAAV vectors may provide an effective means of enhancing the anti-tumor efficacy of radiation therapy.
Collapse
Affiliation(s)
- Wenyin Shi
- Department of Pharmacology and Experimental Therapeutics, University of Florida, Gainesville, FL 32610, USA
| | | | | | | |
Collapse
|
49
|
Abstract
Inoculation of adenovirus vectors in vivo has induced transgene expression in a variety of cochlear cells, but not hair cells or supporting cells in most previous studies. Specific hair cell inoculation by viral vectors has not been demonstrated in the mature guinea pig cochlea in vitro. We injected an Adex1CAlacZ into the mature guinea pig cochlear explants, which were incubated for 24-72 h. We found many lacZ-positive cells in a variety of tissues including the spiral ganglion and stria vascularis. Transgene expression was also found in the outer hair cells and supporting cells, such as the Deiters cells and pillar cells. These findings indicate that adenovirus vectors can be transfected into mature guinea pig hair cells and supporting cells in culture.
Collapse
Affiliation(s)
- Tatsuya Yamasoba
- Department of Otolaryngology, University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-8655, Japan.
| | | | | |
Collapse
|
50
|
Lalwani AK, Han JJ, Castelein CM, Carvalho GJ, Mhatre AN. In vitro and in vivo assessment of the ability of adeno-associated virus-brain-derived neurotrophic factor to enhance spiral ganglion cell survival following ototoxic insult. Laryngoscope 2002; 112:1325-34. [PMID: 12172239 DOI: 10.1097/00005537-200208000-00001] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES/HYPOTHESIS Auditory dysfunction following ototoxic insult results from loss of cochlear hair cells. Secondary degeneration of auditory neurons ensues from withdrawal of neurotrophic support from hair cells and can be prevented with administration of neurotrophins. Administration of adeno-associated virus containing the gene for brain-derived neurotrophic factor will promote spiral ganglion neuron survival after the destruction of hair cells. METHODS Prevention of aminoglycoside-induced spiral ganglion neuron loss through the expression of brain-derived neurotrophic factor mediated by means of the adeno-associated virus was tested in vitro in cochlear explants and in vivo in mammalian cochlea. RESULTS Neuronal survival was significantly enhanced in adeno-associated virus-brain-derived neurotrophic factor transfected rat cochlear explants compared with control samples (30% vs. 19%, P <.05) following exposure to aminoglycoside. Following deafening with aminoglycoside and loop diuretic and introduction of adeno-associated virus-brain-derived neurotrophic factor through osmotic minipump, the experimental group of animals infused with adeno-associated virus-brain-derived neurotrophic factor displayed enhanced spiral ganglion neuron survival in the basal turn of the cochlea when compared with the control group infused with adeno-associated virus containing green fluorescent protein reporter gene. CONCLUSIONS Administration of adeno-associated virus-brain-derived neurotrophic factor enhances spiral ganglion neuron survival following ototoxic exposure in vitro and in vivo. These studies lay the groundwork for further exploration of its application as an adjunct therapy for patients undergoing cochlear implantation because the success of implantation depends directly on the population of neurons available for electrical stimulation.
Collapse
Affiliation(s)
- Anil K Lalwani
- Laboratory of Molecular Otology, Epstein Laboratories, Department of Otolaryngology-Head and Neck Surgery, University of California at San Francisco, 94143-0342, USA.
| | | | | | | | | |
Collapse
|