1
|
Maniaci A, Briglia M, Allia F, Montalbano G, Romano GL, Zaouali MA, H’mida D, Gagliano C, Malaguarnera R, Lentini M, Graziano ACE, Giurdanella G. The Role of Pericytes in Inner Ear Disorders: A Comprehensive Review. BIOLOGY 2024; 13:802. [PMID: 39452111 PMCID: PMC11504721 DOI: 10.3390/biology13100802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/02/2024] [Accepted: 10/06/2024] [Indexed: 10/26/2024]
Abstract
Inner ear disorders, including sensorineural hearing loss, Meniere's disease, and vestibular neuritis, are prevalent conditions that significantly impact the quality of life. Despite their high incidence, the underlying pathophysiology of these disorders remains elusive, and current treatment options are often inadequate. Emerging evidence suggests that pericytes, a type of vascular mural cell specialized to maintain the integrity and function of the microvasculature, may play a crucial role in the development and progression of inner ear disorders. The pericytes are present in the microvasculature of both the cochlea and the vestibular system, where they regulate blood flow, maintain the blood-labyrinth barrier, facilitate angiogenesis, and provide trophic support to neurons. Understanding their role in inner ear disorders may provide valuable insights into the pathophysiology of these conditions and lead to the development of novel diagnostic and therapeutic strategies, improving the standard of living. This comprehensive review aims to provide a detailed overview of the role of pericytes in inner ear disorders, highlighting the anatomy and physiology in the microvasculature, and analyzing the mechanisms that contribute to the development of the disorders. Furthermore, we explore the potential pericyte-targeted therapies, including antioxidant, anti-inflammatory, and angiogenic approaches, as well as gene therapy strategies.
Collapse
Affiliation(s)
- Antonino Maniaci
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
- Department of Surgery, ENT Unit, Asp 7 Ragusa, 97100 Ragusa, Italy
| | - Marilena Briglia
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Fabio Allia
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Giuseppe Montalbano
- Zebrafish Neuromorphology Laboratory, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy;
| | - Giovanni Luca Romano
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Mohamed Amine Zaouali
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy, University of Monastir, Avicenne Street, 5019 Monastir, Tunisia;
| | - Dorra H’mida
- Department of Cytogenetics and Reproductive Biology, Farhat Hached Hospital, 4021 Sousse, Tunisia;
| | - Caterina Gagliano
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Roberta Malaguarnera
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Mario Lentini
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
- Department of Surgery, ENT Unit, Asp 7 Ragusa, 97100 Ragusa, Italy
| | - Adriana Carol Eleonora Graziano
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| | - Giovanni Giurdanella
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (A.M.); (M.B.); (F.A.); (G.L.R.); (C.G.); (R.M.); (G.G.)
| |
Collapse
|
2
|
Amiri M, Kaviari MA, Rostaminasab G, Barimani A, Rezakhani L. A novel cell-free therapy using exosomes in the inner ear regeneration. Tissue Cell 2024; 88:102373. [PMID: 38640600 DOI: 10.1016/j.tice.2024.102373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/01/2024] [Accepted: 04/03/2024] [Indexed: 04/21/2024]
Abstract
Cellular and molecular alterations associated with hearing loss are now better understood with advances in molecular biology. These changes indicate the participation of distinct damage and stress pathways that are unlikely to be fully addressed by conventional pharmaceutical treatment. Sensorineural hearing loss is a common and debilitating condition for which comprehensive pharmacologic intervention is not available. The complex and diverse molecular pathology that underlies hearing loss currently limits our ability to intervene with small molecules. The present review focuses on the potential for the use of extracellular vesicles in otology. It examines a variety of inner ear diseases and hearing loss that may be treatable using exosomes (EXOs). The role of EXOs as carriers for the treatment of diseases related to the inner ear as well as EXOs as biomarkers for the recognition of diseases related to the ear is discussed.
Collapse
Affiliation(s)
- Masoumeh Amiri
- Faculty of Medicine, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Mohammad Amin Kaviari
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran; Universal Scientific Education and Research Network (USERN) Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Gelavizh Rostaminasab
- Clinical Research Development Center, Imam Khomeini and Mohammad Kermanshahi and Farabi Hospitals, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amir Barimani
- Clinical Research Development Center, Imam Khomeini and Mohammad Kermanshahi and Farabi Hospitals, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Leila Rezakhani
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
3
|
Genetic insights, disease mechanisms, and biological therapeutics for Waardenburg syndrome. Gene Ther 2022; 29:479-497. [PMID: 33633356 DOI: 10.1038/s41434-021-00240-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/18/2021] [Accepted: 02/03/2021] [Indexed: 02/06/2023]
Abstract
Waardenburg syndrome (WS), also known as auditory-pigmentary syndrome, is the most common cause of syndromic hearing loss (HL), which accounts for approximately 2-5% of all patients with congenital hearing loss. WS is classified into four subtypes depending on the clinical phenotypes. Currently, pathogenic mutations of PAX3, MITF, SOX10, EDN3, EDNRB or SNAI2 are associated with different subtypes of WS. Although supportive techniques like hearing aids, cochlear implants, or other assistive listening devices can alleviate the HL symptom, there is no cure for WS to date. Recently major progress has been achieved in preclinical studies of genetic HL in animal models, including gene delivery and stem cell replacement therapies. This review focuses on the current understandings of pathogenic mechanisms and potential biological therapeutic approaches for HL in WS, providing strategies and directions for implementing WS biological therapies, as well as possible problems to be faced, in the future.
Collapse
|
4
|
Febles NK, Bauer MA, Ding B, Zhu X, Gallant ND, Frisina RD. A combinatorial approach to protect sensory tissue against cisplatin-induced ototoxicity. Hear Res 2022; 415:108430. [PMID: 35051751 PMCID: PMC8810742 DOI: 10.1016/j.heares.2022.108430] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 12/22/2021] [Accepted: 01/03/2022] [Indexed: 12/23/2022]
Abstract
Sensorineural Hearing Loss (SNHL) is a highly prevalent disorder involving permanent damage or loss to the inner ear's mechano-sensory hair cells and nerve fibers. Major contributing causes are ototoxic drugs, loud noises, and aging. Drug-induced hearing loss (DIHL), affects over 25% of patients treated with common therapeutics such as aminoglycoside antibiotics, loop diuretics or chemotherapeutics. A commonly used chemotherapeutic agent, cisplatin, is very effective for treating malignant tumors, but results in a majority of patients experiencing irreversible hearing loss and/or tinnitus. Additionally, since there is currently no FDA-approved treatments for SNHL, attenuation of ototoxicity is a major area of investigation in oncology, otolaryngology and hearing research. Several potential otoprotective agents have been investigated at the clinical trial stage, but none have progressed to a full FDA-approval. In this study, we investigated a combinatorial approach comprised of an antioxidant, a p53 inhibitor and a neurotrophin, as a multifactorial otoprotective treatment for cisplatin exposure. In vitro, HEI-OC1 cells, an immortalized organ of Corti epithelial cell line, pre-treated with this biotherapeutic cocktail had significantly reduced cisplatin-induced cell death, DNA fragmentation, and apoptotic activation. In an ex vivo study, rat pup D2-D3 organ of Corti explants, significant protection against cisplatin-based hair cell and neuronal loss was achieved by delivery of the same combinatorial pretreatment. Interestingly, the hair cell protection was localized to the basal and middle regions of the organ of Corti. Together, these findings highlight a novel approach to attenuate cisplatin ototoxicity and potentially prevent DIHL by addressing biological mechanisms of cisplatin ototoxicity.
Collapse
Affiliation(s)
- Nicole K Febles
- Department of Medical Engineering, University of South Florida, Tampa, FL 33620, USA; Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33602, USA
| | - Mark A Bauer
- Department of Medical Engineering, University of South Florida, Tampa, FL 33620, USA; Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33602, USA
| | - Bo Ding
- Department of Communicative Sciences and Disorders, University of South Florida, Tampa, FL 33602, USA; Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33602, USA
| | - Xiaoxia Zhu
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33602, USA
| | - Nathan D Gallant
- Department of Mechanical Engineering, University of South Florida, Tampa, FL 33602, USA.
| | - Robert D Frisina
- Department of Medical Engineering, University of South Florida, Tampa, FL 33620, USA; Department of Communicative Sciences and Disorders, University of South Florida, Tampa, FL 33602, USA; Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33602, USA.
| |
Collapse
|
5
|
Bergman JE, Davies C, Denton AJ, Ashman PE, Mittal R, Eshraghi AA. Advancements in Stem Cell Technology and Organoids for the Restoration of Sensorineural Hearing Loss. J Am Acad Audiol 2021; 32:636-645. [PMID: 34034344 DOI: 10.1055/s-0041-1728677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Sensorineural hearing loss (SNHL) is a significant cause of morbidity worldwide and currently has no curative treatment. Technological advancements in stem cell therapy have led to numerous studies that examine the generation of otic sensory cells from progenitors to restore inner ear function. Recently, organoids have emerged as a promising technique to further advance the process of creating functional replacement cells after irreversible hearing loss. Organoids are the three-dimensional generation of stem cells in culture to model the tissue organization and cellular components of the inner ear. Organoids have emerged as a promising technique to create functioning cochlear structures in vitro and may provide crucial information for the utilization of stem cells to restore SNHL. PURPOSE The purpose of this review is to discuss the recent advancements in stem cell-based regenerative therapy for SNHL. RESULTS Recent studies have improved our understanding about the developmental pathways involved in the generation of hair cells and spiral ganglion neurons. However, significant challenges remain in elucidating the molecular interactions and interplay required for stem cells to differentiate and function as otic sensory cells. A few of the challenges encountered with traditional stem cell therapy may be addressed with organoids. CONCLUSION Stem cell-based regenerative therapy holds a great potential for developing novel treatment modalities for SNHL. Further advancements are needed in addressing the challenges associated with stem cell-based regenerative therapy and promote their translation from bench to bedside.
Collapse
Affiliation(s)
- Jenna E Bergman
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida
| | - Camron Davies
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida
| | - Alexa J Denton
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida
| | - Peter E Ashman
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida
| | - Rahul Mittal
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida
| | - Adrien A Eshraghi
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida.,Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida.,Department of Biomedical Engineering, University of Miami, Coral Gables, Miami, Florida.,Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
6
|
Zhang S, Qiao Y, Wang Z, Zhuang J, Sun Y, Shang X, Li G. Identification of novel compound heterozygous variants in SLC19A2 and the genotype-phenotype associations in thiamine-responsive megaloblastic anemia. Clin Chim Acta 2021; 516:157-168. [PMID: 33571483 DOI: 10.1016/j.cca.2021.01.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 01/24/2021] [Accepted: 01/28/2021] [Indexed: 01/19/2023]
Abstract
BACKGROUND AND AIMS Thiamine-responsive megaloblastic anemia (TRMA), caused by SLC19A2 loss-of-function variants, is characterized by the triad of megaloblastic anemia, progressive sensorineural deafness, and non-type 1 diabetes mellitus. Here, we present the case of a Chinese infant with two novel variants segregating in compound heterozygous form in SLC19A2 and reviewed genotype-phenotype associations (GPAs) in patients with TRMA. MATERIALS AND METHODS Whole-exome sequencing was performed to establish a genetic diagnosis. The clinical manifestations and genetic variants were collected by performing a literature review. The bioinformatics software SIFT, PolyPhen2, and Mutation Taster was applied to predict variant effects and analyze GPAs. RESULTS Two novel variants segregating in compound heterozygous form in SLC19A2 (NM_006996.2: exon2:c.336_363del:p.W112fs; exon2:c.358G>T:p.G120X) was identified. Thiamine supplementation corrected anemia and diabetes mellitus but did not improve the hearing defect. In the literature, 183 patients with TRMA with 74 variants in SLC19A2 have been reported, with high incidence in the Middle East, South Asia, and the northern Mediterranean. Patients with biallelic premature termination codon variants presented with more severe phenotypes, and truncating sites on extracellular domains was a protective factor for the hemoglobin level at diagnosis. CONCLUSION Two novel compound heterozygous variants (NM_006996.2: exon2:c.336_363del:p.W112fs; exon2:c.358G>T:p.G120X) were identified, and GPAs in TRMA indicated the predictability of clinical manifestations.
Collapse
Affiliation(s)
- Shule Zhang
- Department of Pediatric Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 9677 Jingshi Road, Lixia Area, Jinan, Shandong 250021, China.
| | - Yu Qiao
- Department of Pediatric Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 9677 Jingshi Road, Lixia Area, Jinan, Shandong 250021, China; Department of Pediatric Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 9677 Jingshi Road, Lixia Area, Jinan, Shandong 250021, China.
| | - Zengmin Wang
- Department of Pediatric Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 9677 Jingshi Road, Lixia Area, Jinan, Shandong 250021, China; Department of Pediatric Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 9677 Jingshi Road, Lixia Area, Jinan, Shandong 250021, China.
| | - Jianxin Zhuang
- Department of Pediatric Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 9677 Jingshi Road, Lixia Area, Jinan, Shandong 250021, China; Department of Pediatric Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 9677 Jingshi Road, Lixia Area, Jinan, Shandong 250021, China.
| | - Yan Sun
- Department of Pediatric Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 9677 Jingshi Road, Lixia Area, Jinan, Shandong 250021, China; Department of Pediatric Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 9677 Jingshi Road, Lixia Area, Jinan, Shandong 250021, China.
| | - Xiaohong Shang
- Department of Pediatric Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 9677 Jingshi Road, Lixia Area, Jinan, Shandong 250021, China; Department of Pediatric Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 9677 Jingshi Road, Lixia Area, Jinan, Shandong 250021, China.
| | - Guimei Li
- Department of Pediatric Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 9677 Jingshi Road, Lixia Area, Jinan, Shandong 250021, China; Department of Pediatric Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 9677 Jingshi Road, Lixia Area, Jinan, Shandong 250021, China.
| |
Collapse
|
7
|
Whatley M, Francis A, Ng ZY, Khoh XE, Atlas MD, Dilley RJ, Wong EYM. Usher Syndrome: Genetics and Molecular Links of Hearing Loss and Directions for Therapy. Front Genet 2020; 11:565216. [PMID: 33193648 PMCID: PMC7642844 DOI: 10.3389/fgene.2020.565216] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/21/2020] [Indexed: 12/19/2022] Open
Abstract
Usher syndrome (USH) is an autosomal recessive (AR) disorder that permanently and severely affects the senses of hearing, vision, and balance. Three clinically distinct types of USH have been identified, decreasing in severity from Type 1 to 3, with symptoms of sensorineural hearing loss (SNHL), retinitis pigmentosa (RP), and vestibular dysfunction. There are currently nine confirmed and two suspected USH-causative genes, and a further three candidate loci have been mapped. The proteins encoded by these genes form complexes that play critical roles in the development and maintenance of cellular structures within the inner ear and retina, which have minimal capacity for repair or regeneration. In the cochlea, stereocilia are located on the apical surface of inner ear hair cells (HC) and are responsible for transducing mechanical stimuli from sound pressure waves into chemical signals. These signals are then detected by the auditory nerve fibers, transmitted to the brain and interpreted as sound. Disease-causing mutations in USH genes can destabilize the tip links that bind the stereocilia to each other, and cause defects in protein trafficking and stereocilia bundle morphology, thereby inhibiting mechanosensory transduction. This review summarizes the current knowledge on Usher syndrome with a particular emphasis on mutations in USH genes, USH protein structures, and functional analyses in animal models. Currently, there is no cure for USH. However, the genetic therapies that are rapidly developing will benefit from this compilation of detailed genetic information to identify the most effective strategies for restoring functional USH proteins.
Collapse
Affiliation(s)
- Meg Whatley
- Ear Science Institute Australia, Nedlands, WA, Australia
| | - Abbie Francis
- Ear Science Institute Australia, Nedlands, WA, Australia
- Emergency Medicine, The University of Western Australia, Nedlands, WA, Australia
| | - Zi Ying Ng
- Ear Science Institute Australia, Nedlands, WA, Australia
| | - Xin Ee Khoh
- Ear Science Institute Australia, Nedlands, WA, Australia
- School of Human Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Marcus D. Atlas
- Ear Science Institute Australia, Nedlands, WA, Australia
- Ear Sciences Centre, The University of Western Australia, Nedlands, WA, Australia
| | - Rodney J. Dilley
- Ear Science Institute Australia, Nedlands, WA, Australia
- Ear Sciences Centre, The University of Western Australia, Nedlands, WA, Australia
- Centre for Cell Therapy and Regenerative Medicine, The University of Western Australia, Perth, WA, Australia
| | - Elaine Y. M. Wong
- Ear Science Institute Australia, Nedlands, WA, Australia
- Ear Sciences Centre, The University of Western Australia, Nedlands, WA, Australia
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin University, Bentley, WA, Australia
| |
Collapse
|
8
|
Romito M, Pu Y, Stankovic KM, Psaltis D. Imaging hair cells through laser-ablated cochlear bone. BIOMEDICAL OPTICS EXPRESS 2019; 10:5974-5988. [PMID: 31799058 PMCID: PMC6865115 DOI: 10.1364/boe.10.005974] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/29/2019] [Accepted: 10/24/2019] [Indexed: 05/04/2023]
Abstract
We report an innovative technique for the visualization of cells through an overlying scattering medium by combining femtosecond laser bone ablation and two-photon excitation fluorescence (TPEF) microscopy. We demonstrate the technique by imaging hair cells in an intact mouse cochlea ex vivo. Intracochlear imaging is important for the assessment of hearing disorders. However, the small size of the cochlea and its encasement in the densest bone in the body present challenging obstacles, preventing the visualization of the intracochlear microanatomy using standard clinical imaging modalities. The controlled laser ablation reduces the optical scattering of the cochlear bone while the TPEF allows visualization of individual cells behind the bone. We implemented optical coherence tomography (OCT) simultaneously with the laser ablation to enhance the precision of the ablation and prevent inadvertent damage to the cells behind the bone.
Collapse
Affiliation(s)
- Marilisa Romito
- Optics Laboratory, School of Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Ye Pu
- Optics Laboratory, School of Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Konstantina M. Stankovic
- Department of Otolaryngology – Head and Neck Surgery, Harvard Medical School and Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Demetri Psaltis
- Optics Laboratory, School of Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|
9
|
Omichi R, Shibata SB, Morton CC, Smith RJH. Gene therapy for hearing loss. Hum Mol Genet 2019; 28:R65-R79. [PMID: 31227837 PMCID: PMC6796998 DOI: 10.1093/hmg/ddz129] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 04/15/2019] [Accepted: 06/07/2019] [Indexed: 12/26/2022] Open
Abstract
Sensorineural hearing loss (SNHL) is the most common sensory disorder. Its underlying etiologies include a broad spectrum of genetic and environmental factors that can lead to hearing loss that is congenital or late onset, stable or progressive, drug related, noise induced, age related, traumatic or post-infectious. Habilitation options typically focus on amplification using wearable or implantable devices; however exciting new gene-therapy-based strategies to restore and prevent SNHL are actively under investigation. Recent proof-of-principle studies demonstrate the potential therapeutic potential of molecular agents delivered to the inner ear to ameliorate different types of SNHL. Correcting or preventing underlying genetic forms of hearing loss is poised to become a reality. Herein, we review molecular therapies for hearing loss such as gene replacement, antisense oligonucleotides, RNA interference and CRISPR-based gene editing. We discuss delivery methods, techniques and viral vectors employed for inner ear gene therapy and the advancements in this field that are paving the way for basic science research discoveries to transition to clinical trials.
Collapse
Affiliation(s)
- Ryotaro Omichi
- Molecular Otolaryngology and Renal Research Laboratories, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Department of Otolaryngology—Head and Neck Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Seiji B Shibata
- Molecular Otolaryngology and Renal Research Laboratories, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Department of Otolaryngology—Head and Neck Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Cynthia C Morton
- Departments of Obstetrics and Gynecology and of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Manchester Centre for Audiology and Deafness, University of Manchester, Manchester Academic Health Science Centre, Manchester M139NT, UK
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Richard J H Smith
- Molecular Otolaryngology and Renal Research Laboratories, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Department of Otolaryngology—Head and Neck Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
10
|
Nacher-Soler G, Garrido JM, Rodríguez-Serrano F. Hearing regeneration and regenerative medicine: present and future approaches. Arch Med Sci 2019; 15:957-967. [PMID: 31360190 PMCID: PMC6657260 DOI: 10.5114/aoms.2019.86062] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Accepted: 05/28/2017] [Indexed: 01/04/2023] Open
Abstract
More than 5% of the world population lives with a hearing impairment. The main factors responsible for hearing degeneration are ototoxic drugs, aging, continued exposure to excessive noise and infections. The pool of adult stem cells in the inner ear drops dramatically after birth, and therefore an endogenous cellular source for regeneration is absent. Hearing loss can emerge after the degeneration of different cochlear components, so there are multiple targets to be reached, such as hair cells (HCs), spiral ganglion neurons (SGNs), supporting cells (SCs) and ribbon synapses. Important discoveries in the hearing regeneration field have been reported regarding stem cell transplantation, migration and survival; genetic systems for cell fate monitoring; and stem cell differentiation to HCs, SGNs and SCs using adult stem cells, embryonic stem cells and induced pluripotent stem cells. Moreover, some molecular mediators that affect the establishment of functional synapses have been identified. In this review, we will focus on reporting the state of the art in the regenerative medicine field for hearing recovery. Stem cell research has enabled remarkable advances in regeneration, particularly in neuronal cells and synapses. Despite the progress achieved, there are certain issues that need a deeper development to improve the results already obtained, or to develop new approaches aiming for the clinical application.
Collapse
Affiliation(s)
- German Nacher-Soler
- Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, Granada, Spain
| | - José Manuel Garrido
- Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, Granada, Spain
- Department of Cardiovascular Surgery, Virgen de las Nieves University Hospital, Granada, Spain
- Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, Spain
| | - Fernando Rodríguez-Serrano
- Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, Granada, Spain
- Biosanitary Research Institute of Granada (ibs.GRANADA), Granada, Spain
- Department of Human Anatomy and Embryology, University of Granada, Granada, Spain
| |
Collapse
|
11
|
Stem-cell therapy for hearing loss: are we there yet? Braz J Otorhinolaryngol 2019; 85:520-529. [PMID: 31186186 PMCID: PMC9443044 DOI: 10.1016/j.bjorl.2019.04.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 04/28/2019] [Indexed: 12/04/2022] Open
Abstract
Introduction Mammalian hair cells and auditory neurons do not show regenerative capacity. Hence, damage to these cell types is permanent and leads to hearing loss. However, there is no treatment that re-establishes auditory function. Regenerative therapies using stem cells represent a promising alternative. Objective This article aims to review the current literature about the main types of stem cells with potential for application in cell therapy for sensorineural hearing loss, the most relevant experiments already performed in animals, as well as the advances that have been recently made in the field. Methods Research included the databases PubMed/MEDLINE, Web of Science, Science Direct and SciELO, as well as gray literature. Search strategy included the following main terms: “stem cells”, “hair cells” and “auditory neurons”. Additionally, the main terms were combined with the following secondary terms: “mesenchymal”, “iPS”, “inner ear”, “auditory”. The research was conducted independently by three researchers. Results Differentiation of stem cells into hair cells and auditory neurons has a high success rate, reaching up to 82% for the first and 100% for the latter. Remarkably, these differentiated cells are able to interact with hair cells and auditory neurons of cochlear explants through formation of new synapses. When transplanted into the cochlea of animals with hearing loss, auditory restoration has been documented to date only in deafferented animals. Conclusion Advances have been more prominent in cases of auditory neuropathy, since partial improvement of auditory nerve conditions through cell-based therapy may increase the number of patients who can successfully receive cochlear implants.
Collapse
|
12
|
Czajkowski A, Mounier A, Delacroix L, Malgrange B. Pluripotent stem cell-derived cochlear cells: a challenge in constant progress. Cell Mol Life Sci 2019; 76:627-635. [PMID: 30341460 PMCID: PMC11105202 DOI: 10.1007/s00018-018-2950-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/27/2018] [Accepted: 10/15/2018] [Indexed: 12/31/2022]
Abstract
Hearing loss is a common affection mainly resulting from irreversible loss of the sensory hair cells of the cochlea; therefore, developing therapies to replace missing hair cells is essential. Understanding the mechanisms that drive their formation will not only help to unravel the molecular basis of deafness, but also give a roadmap for recapitulating hair cells development from cultured pluripotent stem cells. In this review, we provide an overview of the molecular mechanisms involved in hair cell production from both human and mouse embryonic stem cells. We then provide insights how this knowledge has been applied to differentiate induced pluripotent stem cells into otic progenitors and hair cells. Finally, we discuss the current limitations for properly obtaining functional hair cell in a Petri dish, as well as the difficulties that have to be overcome prior to consider stem cell therapy as a potential treatment for hearing loss.
Collapse
Affiliation(s)
- Amandine Czajkowski
- Developmental Neurobiology Unit, GIGA-Neurosciences, University of Liège, Quartier Hôpital (CHU), Avenue Hippocrate 15, Tour 4, 1er étage, Bât. B36, 4000, Liège, Belgium
| | - Anaïs Mounier
- Developmental Neurobiology Unit, GIGA-Neurosciences, University of Liège, Quartier Hôpital (CHU), Avenue Hippocrate 15, Tour 4, 1er étage, Bât. B36, 4000, Liège, Belgium
| | - Laurence Delacroix
- Developmental Neurobiology Unit, GIGA-Neurosciences, University of Liège, Quartier Hôpital (CHU), Avenue Hippocrate 15, Tour 4, 1er étage, Bât. B36, 4000, Liège, Belgium
| | - Brigitte Malgrange
- Developmental Neurobiology Unit, GIGA-Neurosciences, University of Liège, Quartier Hôpital (CHU), Avenue Hippocrate 15, Tour 4, 1er étage, Bât. B36, 4000, Liège, Belgium.
| |
Collapse
|
13
|
Musazzi UM, Franzé S, Cilurzo F. Innovative pharmaceutical approaches for the management of inner ear disorders. Drug Deliv Transl Res 2018; 8:436-449. [PMID: 28462501 DOI: 10.1007/s13346-017-0384-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The sense of hearing is essential for permitting human beings to interact with the environment, and its dysfunctions can strongly impact on the quality of life. In this context, the cochlea plays a fundamental role in the transformation of the airborne sound waves into electrical signals, which can be processed by the brain. However, several diseases and external stimuli (e.g., noise, drugs) can damage the sensorineural structures of cochlea, inducing progressive hearing dysfunctions until deafness. In clinical practice, the current pharmacological approaches to treat cochlear diseases are based on the almost exclusive use of systemic steroids. In the last decades, the efficacy of novel therapeutic molecules has been proven, taking advantage from a better comprehension of the pathological mechanisms underlying many cochlear diseases. In addition, the feasibility of intratympanic administration of drugs also permitted to overcome the pharmacokinetic limitations of the systemic drug administration, opening new frontiers in drug delivery to cochlea. Several innovative drug delivery systems, such as in situ gelling systems or nanocarriers, were designed, and their efficacy has been proven in vitro and in vivo in cochlear models. The current review aims to describe the art of state in the cochlear drug delivery, highlighting lights and shadows and discussing the most critical aspects still pending in the field.
Collapse
Affiliation(s)
- Umberto M Musazzi
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via G. Colombo, 71, 20133, Milan, Italy.
| | - Silvia Franzé
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via G. Colombo, 71, 20133, Milan, Italy
| | - Francesco Cilurzo
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Via G. Colombo, 71, 20133, Milan, Italy
| |
Collapse
|
14
|
Matsuoka AJ, Sayed ZA, Stephanopoulos N, Berns EJ, Wadhwani AR, Morrissey ZD, Chadly DM, Kobayashi S, Edelbrock AN, Mashimo T, Miller CA, McGuire TL, Stupp SI, Kessler JA. Creating a stem cell niche in the inner ear using self-assembling peptide amphiphiles. PLoS One 2017; 12:e0190150. [PMID: 29284013 PMCID: PMC5746215 DOI: 10.1371/journal.pone.0190150] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 12/09/2017] [Indexed: 11/23/2022] Open
Abstract
The use of human embryonic stem cells (hESCs) for regeneration of the spiral ganglion will require techniques for promoting otic neuronal progenitor (ONP) differentiation, anchoring of cells to anatomically appropriate and specific niches, and long-term cell survival after transplantation. In this study, we used self-assembling peptide amphiphile (PA) molecules that display an IKVAV epitope (IKVAV-PA) to create a niche for hESC-derived ONPs that supported neuronal differentiation and survival both in vitro and in vivo after transplantation into rodent inner ears. A feature of the IKVAV-PA gel is its ability to form organized nanofibers that promote directed neurite growth. Culture of hESC-derived ONPs in IKVAV-PA gels did not alter cell proliferation or viability. However, the presence of IKVAV-PA gels increased the number of cells expressing the neuronal marker beta-III tubulin and improved neurite extension. The self-assembly properties of the IKVAV-PA gel allowed it to be injected as a liquid into the inner ear to create a biophysical niche for transplanted cells after gelation in vivo. Injection of ONPs combined with IKVAV-PA into the modiolus of X-SCID rats increased survival and localization of the cells around the injection site compared to controls. Human cadaveric temporal bone studies demonstrated the technical feasibility of a transmastoid surgical approach for clinical intracochlear injection of the IKVAV-PA/ONP combination. Combining stem cell transplantation with injection of self-assembling PA gels to create a supportive niche may improve clinical approaches to spiral ganglion regeneration.
Collapse
Affiliation(s)
- Akihiro J. Matsuoka
- Department of Otolaryngology and Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
- Department of Communication Sciences and Disorders, Northwestern University, Evanston, Illinois, United States of America
- Hugh Knowles Center for Hearing Research, Northwestern University, Evanston, Illinois, United States of America
- * E-mail:
| | - Zafar A. Sayed
- Department of Otolaryngology and Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Nicholas Stephanopoulos
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois, United States of America
| | - Eric J. Berns
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, United States of America
| | - Anil R. Wadhwani
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Zachery D. Morrissey
- Department of Otolaryngology and Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Duncan M. Chadly
- Department of Otolaryngology and Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Shun Kobayashi
- Department of Otolaryngology and Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Alexandra N. Edelbrock
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, United States of America
| | - Tomoji Mashimo
- The Institute of Experimental Animal Sciences, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Charles A. Miller
- Department of Otolaryngology and Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Tammy L. McGuire
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Samuel I. Stupp
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois, United States of America
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, United States of America
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
- Department of Chemistry, Northwestern University, Evanston, Illinois, United States of America
- Department of Materials Science and Engineering, Northwestern University, Evanston, Illinois, United States of America
| | - John A. Kessler
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| |
Collapse
|
15
|
A Simple Model for Inducing Optimal Increase of SDF-1 with Aminoglycoside Ototoxicity. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4630241. [PMID: 29430461 PMCID: PMC5752978 DOI: 10.1155/2017/4630241] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/29/2017] [Accepted: 09/17/2017] [Indexed: 11/17/2022]
Abstract
Objectives As a homing factor of stem cell, stromal derived factor-1 (SDF-1) is important for the regenerative research in ototoxicity. Mice models with aminoglycoside ototoxicity have been widely used to study the regeneration capacity of MSCs in repair of cochlear injury. We developed a mouse model with maximal increase in SDF-1 levels in the inner ear, according to the “one-shot” doses of kanamycin and furosemide. Methods C57BL/6 mice had kanamycin (420, 550, and 600 mg/kg) dissolved in PBS, followed by an intraperitoneal injection of furosemide (130 mg/kg). The injuries of inner ear were measured with hearing thresholds, histology, and outer hair cell counts at 0, 3, 5, 7, 10, and 14 days before the sacrifice. The levels of SDF-1 in the inner ear were tested by real-time RT-PCR and immunohistochemistry. Results There were a significant reduction in hearing thresholds and a maximal increase of SDF-1 levels in the furosemide 130 mg/kg + kanamycin 550 mg/kg group, but severe hearing deterioration over time was observed in the furosemide 130 mg/kg + kanamycin 600 mg/kg group and four mice were dead. SDF-1 was detected mostly in the stria vascularis and organ of Corti showing the highest increase in expression. Conclusion We observed optimal induction of the stem cell homing factor in the newly generated aminoglycoside-induced ototoxicity mouse model using a “one-shot” protocol. This study regarding high SDF-1 levels in our mouse model of ototoxicity would play a major role in the development of therapeutic agents using MSC homing.
Collapse
|
16
|
Mittal R, Jung HD, Mittal J, Eshraghi AA. A perspective on stem cell therapy for ear disorders. J Cell Physiol 2017; 233:1823-1824. [PMID: 28608552 DOI: 10.1002/jcp.26048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 06/12/2017] [Indexed: 12/29/2022]
Abstract
The use of stem cells in cell-based therapy is an emerging concept for the treatment of ear disorders. Tympanic membrane perforation (TMP) and inner ear disorders are some of the most commonly presented otologic disorders that can benefit from advances in cell-based therapy. Studies have already demonstrated that stem cell-based therapy can potentially be an effective treatment modality for acute and chronic TMP. Recent studies have also shown promise in application of cell-based approach to treat inner ear dysfunction. In this perspective, we will discuss the recent advancements regarding the use of cell-based therapy for ear disorders.
Collapse
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Hyunseo D Jung
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Jeenu Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| | - Adrien A Eshraghi
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
17
|
Regenerative medicine in hearing recovery. Cytotherapy 2017; 19:909-915. [DOI: 10.1016/j.jcyt.2017.04.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/24/2017] [Accepted: 04/21/2017] [Indexed: 12/20/2022]
|
18
|
Revuelta M, Santaolalla F, Arteaga O, Alvarez A, Sánchez-del-Rey A, Hilario E. Recent advances in cochlear hair cell regeneration-A promising opportunity for the treatment of age-related hearing loss. Ageing Res Rev 2017; 36:149-155. [PMID: 28414155 DOI: 10.1016/j.arr.2017.04.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 04/05/2017] [Accepted: 04/10/2017] [Indexed: 01/22/2023]
Abstract
The objective of this paper is to review current information regarding the treatment of age-related hearing loss by using cochlear hair cell regeneration. Recent advances in the regeneration of the inner ear, including the usefulness of stem cells, are also presented. Based on the current literature, cochlear cell regeneration may well be possible in the short term and cochlear gene therapy may also be useful for the treatment of hearing loss associated with ageing. The present review provide further insight into the pathogenesis of Inner Ear senescence and aged-related hearing loss and facilitate the development of therapeutic strategies to repair hair cells damaged by ageing. More research will be needed in order to translate them into an effective treatment for deafness linked to cochlear senescence in humans.
Collapse
|
19
|
Ouji Y, Sakagami M, Omori H, Higashiyama S, Kawai N, Kitahara T, Wanaka A, Yoshikawa M. Efficient induction of inner ear hair cell-like cells from mouse ES cells using combination of Math1 transfection and conditioned medium from ST2 stromal cells. Stem Cell Res 2017; 23:50-56. [PMID: 28689068 DOI: 10.1016/j.scr.2017.06.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 06/28/2017] [Accepted: 06/29/2017] [Indexed: 12/22/2022] Open
Abstract
We sought to establish a more efficient technique for induction of inner ear hair cell-like cells (HC-like cells) from embryonic stem cells (ES cells) by using a combination of two previously reported methods; ST2 stromal cell-conditioned medium, known to be favorable for HC-like cell induction (HIST2 method), and ES cells with transfer of the Math1 gene (Math1-ES cells). Math1-ES cells carrying Tet-inducible Math1 were cultured for 14days with doxycycline in conditioned medium from cultures of ST2 stromal cells following formation of 4-day embryoid bodies (EBs). Although each of the previously introduced methods have been reported to induce approximately 20% HC-like cells and 10% HC-like cells in their respective populations in EB outgrowths at the end of the culture periods, the present combined method was able to generate approximately 30% HC-like cells expressing HC-related markers (myosin6, myosin7a, calretinin, α9AchR, Brn3c), which showed remarkable formation of stereocilia-like structures. Analysis of expressions of marker genes specific for cochlear (Lmod3, Emcn) and vestibular (Dnah5, Ptgds) cells indicated that our HIST2 method may lead to induction of cochlear- and vestibular-type cells. In addition, continuous Math1 induction by doxycycline without use of the HIST2 method preferentially induced cochlear markers with negligible effects on vestibular marker induction.
Collapse
Affiliation(s)
- Yukiteru Ouji
- Department of Pathogen, Infection and Immunity, Nara Medical University, Kashihara, Nara, Japan.
| | - Masaharu Sakagami
- Department of Pathogen, Infection and Immunity, Nara Medical University, Kashihara, Nara, Japan; Department of Otolaryngology - Head and Neck Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Hiroko Omori
- Core Instrumentation Facility, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Shinji Higashiyama
- Core Instrumentation Facility, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan; Central Instrumentation Laboratory, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Norikazu Kawai
- Department of Pathogen, Infection and Immunity, Nara Medical University, Kashihara, Nara, Japan
| | - Tadashi Kitahara
- Department of Otolaryngology - Head and Neck Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Akio Wanaka
- Department of Second Anatomy, Nara Medical University, Kashihara, Nara, Japan
| | - Masahide Yoshikawa
- Department of Pathogen, Infection and Immunity, Nara Medical University, Kashihara, Nara, Japan.
| |
Collapse
|
20
|
Warnecke A, Mellott AJ, Römer A, Lenarz T, Staecker H. Advances in translational inner ear stem cell research. Hear Res 2017; 353:76-86. [PMID: 28571616 DOI: 10.1016/j.heares.2017.05.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 05/01/2017] [Accepted: 05/23/2017] [Indexed: 12/16/2022]
Abstract
Stem cell research is expanding our understanding of developmental biology as well as promising the development of new therapies for a range of different diseases. Within hearing research, the use of stem cells has focused mainly on cell replacement. Stem cells however have a broad range of other potential applications that are just beginning to be explored in the ear. Mesenchymal stem cells are an adult derived stem cell population that have been shown to produce growth factors, modulate the immune system and can differentiate into a wide variety of tissue types. Potential advantages of mesenchymal/adult stem cells are that they have no ethical constraints on their use. However, appropriate regulatory oversight seems necessary in order to protect patients from side effects. Disadvantages may be the lack of efficacy in many preclinical studies. But if proven safe and efficacious, they are easily translatable to clinical trials. The current review will focus on the potential application on mesenchymal stem cells for the treatment of inner ear disorders.
Collapse
Affiliation(s)
- Athanasia Warnecke
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, Carl Neuberg-Str. 1, 30625, Hannover, Germany; Cluster of Excellence "Hearing4all" of the German Research Foundation, Germany
| | - Adam J Mellott
- Department of Plastic Surgery, University of Kansas School of Medicine, Kansas City, KS, USA
| | - Ariane Römer
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, Carl Neuberg-Str. 1, 30625, Hannover, Germany; Cluster of Excellence "Hearing4all" of the German Research Foundation, Germany
| | - Thomas Lenarz
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, Carl Neuberg-Str. 1, 30625, Hannover, Germany; Cluster of Excellence "Hearing4all" of the German Research Foundation, Germany
| | - Hinrich Staecker
- Department of Otolaryngology Head and Neck Surgery, University of Kansas School of Medicine, Kansas City, KS, USA.
| |
Collapse
|
21
|
Lee MY, Hackelberg S, Green KL, Lunghamer KG, Kurioka T, Loomis BR, Swiderski DL, Duncan RK, Raphael Y. Survival of human embryonic stem cells implanted in the guinea pig auditory epithelium. Sci Rep 2017; 7:46058. [PMID: 28387239 PMCID: PMC5384248 DOI: 10.1038/srep46058] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 03/10/2017] [Indexed: 01/05/2023] Open
Abstract
Hair cells in the mature cochlea cannot spontaneously regenerate. One potential approach for restoring hair cells is stem cell therapy. However, when cells are transplanted into scala media (SM) of the cochlea, they promptly die due to the high potassium concentration. We previously described a method for conditioning the SM to make it more hospitable to implanted cells and showed that HeLa cells could survive for up to a week using this method. Here, we evaluated the survival of human embryonic stem cells (hESC) constitutively expressing GFP (H9 Cre-LoxP) in deaf guinea pig cochleae that were pre-conditioned to reduce potassium levels. GFP-positive cells could be detected in the cochlea for at least 7 days after the injection. The cells appeared spherical or irregularly shaped, and some were aggregated. Flushing SM with sodium caprate prior to transplantation resulted in a lower proportion of stem cells expressing the pluripotency marker Oct3/4 and increased cell survival. The data demonstrate that conditioning procedures aimed at transiently reducing the concentration of potassium in the SM facilitate survival of hESCs for at least one week. During this time window, additional procedures can be applied to initiate the differentiation of the implanted hESCs into new hair cells.
Collapse
Affiliation(s)
- Min Young Lee
- Kresge Hearing Research Institute, Otolaryngology - Head and Neck Surgery, The University of Michigan Medical School, MSRB-3, Rm. 9301 1150 W. Medical Center Dr. Ann Arbor, MI 48109-5648, USA.,Department of Otorhinolaryngology and Head &Neck Surgery, Dankook University Hospital, 119, Dandae-ro, Dongnam-gu, Cheonan-si, Chungnam, 31116, Korea
| | - Sandra Hackelberg
- Kresge Hearing Research Institute, Otolaryngology - Head and Neck Surgery, The University of Michigan Medical School, MSRB-3, Rm. 9301 1150 W. Medical Center Dr. Ann Arbor, MI 48109-5648, USA
| | - Kari L Green
- Kresge Hearing Research Institute, Otolaryngology - Head and Neck Surgery, The University of Michigan Medical School, MSRB-3, Rm. 9301 1150 W. Medical Center Dr. Ann Arbor, MI 48109-5648, USA
| | - Kelly G Lunghamer
- Kresge Hearing Research Institute, Otolaryngology - Head and Neck Surgery, The University of Michigan Medical School, MSRB-3, Rm. 9301 1150 W. Medical Center Dr. Ann Arbor, MI 48109-5648, USA
| | - Takaomi Kurioka
- Kresge Hearing Research Institute, Otolaryngology - Head and Neck Surgery, The University of Michigan Medical School, MSRB-3, Rm. 9301 1150 W. Medical Center Dr. Ann Arbor, MI 48109-5648, USA
| | - Benjamin R Loomis
- Kresge Hearing Research Institute, Otolaryngology - Head and Neck Surgery, The University of Michigan Medical School, MSRB-3, Rm. 9301 1150 W. Medical Center Dr. Ann Arbor, MI 48109-5648, USA
| | - Donald L Swiderski
- Kresge Hearing Research Institute, Otolaryngology - Head and Neck Surgery, The University of Michigan Medical School, MSRB-3, Rm. 9301 1150 W. Medical Center Dr. Ann Arbor, MI 48109-5648, USA
| | - R Keith Duncan
- Kresge Hearing Research Institute, Otolaryngology - Head and Neck Surgery, The University of Michigan Medical School, MSRB-3, Rm. 9301 1150 W. Medical Center Dr. Ann Arbor, MI 48109-5648, USA
| | - Yehoash Raphael
- Kresge Hearing Research Institute, Otolaryngology - Head and Neck Surgery, The University of Michigan Medical School, MSRB-3, Rm. 9301 1150 W. Medical Center Dr. Ann Arbor, MI 48109-5648, USA
| |
Collapse
|
22
|
Dixon AR, Ramirez Y, Haengel K, Barald KF. A drop array culture for patterning adherent mouse embryonic stem cell-derived neurospheres. J Tissue Eng Regen Med 2016; 12:e379-e383. [PMID: 27943657 DOI: 10.1002/term.2389] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 10/01/2016] [Accepted: 12/06/2016] [Indexed: 01/24/2023]
Abstract
New therapeutic approaches for repairing an injured or degenerating nervous system have accelerated the development of methods to generate populations of neurons derived from various stem cell sources efficiently. Many of these methods require the generation of neurospheres. Here a simple technique is described for creating an array of adherent mouse embryonic stem cell (mESC)-derived neurospheres using a conventional plastic culture dish and a patterning template. mESC-derived neurospheres are confined to circular (4-mm diameter), gel-coated regions within an array. The adherent neurosphere arrays require 3 days to prepare from an mESC source; they can be maintained in 15 μl drops of medium, and exhibit extensive neurite elaboration after 8 days of cultivation. Additionally, the potential of treating the adherent neurospheres in selected drops of an array is demonstrated with a variety of differentiation-inducing reagents and subsequently individually analysing such neurospheres for gene expression, protein levels and morphological development. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Angela R Dixon
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Yadah Ramirez
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Kathryn Haengel
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - Kate F Barald
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, Medical School, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
23
|
Zhou Y, Hu Z. Epigenetic DNA Demethylation Causes Inner Ear Stem Cell Differentiation into Hair Cell-Like Cells. Front Cell Neurosci 2016; 10:185. [PMID: 27536218 PMCID: PMC4971107 DOI: 10.3389/fncel.2016.00185] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 07/12/2016] [Indexed: 12/17/2022] Open
Abstract
The DNA methyltransferase (DNMT) inhibitor 5-azacytidine (5-aza) causes genomic demethylation to regulate gene expression. However, it remains unclear whether 5-aza affects gene expression and cell fate determination of stem cells. In this study, 5-aza was applied to mouse utricle sensory epithelia-derived progenitor cells (MUCs) to investigate whether 5-aza stimulated MUCs to become sensory hair cells. After treatment, MUCs increased expression of hair cell genes and proteins. The DNA methylation level (indicated by percentage of 5-methylcytosine) showed a 28.57% decrease after treatment, which causes significantly repressed DNMT1 protein expression and DNMT activity. Additionally, FM1-43 permeation assays indicated that the permeability of 5-aza-treated MUCs was similar to that of sensory hair cells, which may result from mechanotransduction channels. This study not only demonstrates a possible epigenetic approach to induce tissue specific stem/progenitor cells to become sensory hair cell-like cells, but also provides a cell model to epigenetically modulate stem cell fate determination.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Otolaryngology-Head and Neck Surgery, Wayne State University School of Medicine Detroit, MI, USA
| | - Zhengqing Hu
- Department of Otolaryngology-Head and Neck Surgery, Wayne State University School of Medicine Detroit, MI, USA
| |
Collapse
|
24
|
Effects of genetic correction on the differentiation of hair cell-like cells from iPSCs with MYO15A mutation. Cell Death Differ 2016; 23:1347-57. [PMID: 26915297 DOI: 10.1038/cdd.2016.16] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 01/23/2016] [Accepted: 01/26/2016] [Indexed: 12/22/2022] Open
Abstract
Deafness or hearing loss is a major issue in human health. Inner ear hair cells are the main sensory receptors responsible for hearing. Defects in hair cells are one of the major causes of deafness. A combination of induced pluripotent stem cell (iPSC) technology with genome-editing technology may provide an attractive cell-based strategy to regenerate hair cells and treat hereditary deafness in humans. Here, we report the generation of iPSCs from members of a Chinese family carrying MYO15A c.4642G>A and c.8374G>A mutations and the induction of hair cell-like cells from those iPSCs. The compound heterozygous MYO15A mutations resulted in abnormal morphology and dysfunction of the derived hair cell-like cells. We used a CRISPR/Cas9 approach to genetically correct the MYO15A mutation in the iPSCs and rescued the morphology and function of the derived hair cell-like cells. Our data demonstrate the feasibility of generating inner ear hair cells from human iPSCs and the functional rescue of gene mutation-based deafness by using genetic correction.
Collapse
|
25
|
Azadeh J, Song Z, Laureano AS, Toro-Ramos A, Kwan K. Initiating Differentiation in Immortalized Multipotent Otic Progenitor Cells. J Vis Exp 2016. [PMID: 26780605 DOI: 10.3791/53692] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Use of human induced pluripotent stem cells (iPSC) or embryonic stem cells (ESC) for cell replacement therapies holds great promise. Several limitations including low yields and heterogeneous populations of differentiated cells hinder the progress of stem cell therapies. A fate restricted immortalized multipotent otic progenitor (iMOP) cell line was generated to facilitate efficient differentiation of large numbers of functional hair cells and spiral ganglion neurons (SGN) for inner ear cell replacement therapies. Starting from dissociated cultures of single iMOP cells, protocols that promote cell cycle exit and differentiation by basic fibroblast growth factor (bFGF) withdrawal were described. A significant decrease in proliferating cells after bFGF withdrawal was confirmed using an EdU cell proliferation assay. Concomitant with a decrease in proliferation, successful differentiation resulted in expression of molecular markers and morphological changes. Immunostaining of Cdkn1b (p27(KIP)) and Cdh1 (E-cadherin) in iMOP-derived otospheres was used as an indicator for differentiation into inner ear sensory epithelia while immunostaining of Cdkn1b and Tubb3 (neuronal β-tubulin) was used to identify iMOP-derived neurons. Use of iMOP cells provides an important tool for understanding cell fate decisions made by inner ear neurosensory progenitors and will help develop protocols for generating large numbers of iPSC or ESC-derived hair cells and SGNs. These methods will accelerate efforts for generating otic cells for replacement therapies.
Collapse
Affiliation(s)
| | | | | | | | - Kelvin Kwan
- Cell Biology & Neuroscience, Rutgers University;
| |
Collapse
|
26
|
Zhou Y, Hu Z. Genome-wide demethylation by 5-aza-2'-deoxycytidine alters the cell fate of stem/progenitor cells. Stem Cell Rev Rep 2015; 11:87-95. [PMID: 25096638 DOI: 10.1007/s12015-014-9542-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
DNA methyltransferase (DNMT) inhibitor 5-aza-2'-deoxycytidine (5-aza-CdR) is able to cause DNA demethylation in the genome and induce the expression of silenced genes. Whether DNA demethylation can affect the gene expression of stem/progenitor cells has not been understood. Mouse utricle epithelia-derived progenitor cells (MUCs), which possess stem cell features as previously described, exhibit a potential DNA methylation status in the genome. In this study, MUCs were treated with 5-aza-CdR to determine whether DNMT inhibitor is able to induce the differentiation of MUCs. With 5-aza-CdR treatment for 72 hr, MUCs expressed epithelial genes including Cdh1, Krt8, Krt18, and Dsp. Further, hair cell genes Myo7a and Myo6 increased their expressions in response to 5-aza-CdR treatment. The decrease in the global methylated DNA values after 5-aza-CdR treatment indicated a significant DNA demethylation in the genome of MUCs, which may contribute to remarkably increased expression of epithelial genes and hair cell genes. The progenitor MUCs then turned into an epithelial-like hair cell fate with the expression of both epithelial and hair cell genes. This study suggests that stem cell differentiation can be stimulated by DNA demethylation, which may open avenues for studying stem cell fate induction using epigenetic approaches.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Otolaryngology-HNS, Wayne State University School of Medicine, 550 E Canfield St. 258, Lande Detroit, MI, 48201, USA
| | | |
Collapse
|
27
|
Gillespie LN, Richardson RT, Nayagam BA, Wise AK. Treating hearing disorders with cell and gene therapy. J Neural Eng 2015; 11:065001. [PMID: 25420002 DOI: 10.1088/1741-2560/11/6/065001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Hearing loss is an increasing problem for a substantial number of people and, with an aging population, the incidence and severity of hearing loss will become more significant over time. There are very few therapies currently available to treat hearing loss, and so the development of new therapeutic strategies for hearing impaired individuals is of paramount importance to address this unmet clinical need. Most forms of hearing loss are progressive in nature and therefore an opportunity exists to develop novel therapeutic approaches to slow or halt hearing loss progression, or even repair or replace lost hearing function. Numerous emerging technologies have potential as therapeutic options. This paper details the potential of cell- and gene-based therapies to provide therapeutic agents to protect sensory and neural cells from various insults known to cause hearing loss; explores the potential of replacing lost sensory and nerve cells using gene and stem cell therapy; and describes the considerations for clinical translation and the challenges that need to be overcome.
Collapse
|
28
|
Park YH. Stem Cell Therapy for Sensorineural Hearing Loss, Still Alive? J Audiol Otol 2015; 19:63-7. [PMID: 26413570 PMCID: PMC4582452 DOI: 10.7874/jao.2015.19.2.63] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 08/15/2015] [Accepted: 08/18/2015] [Indexed: 12/24/2022] Open
Abstract
In mammals, the auditory system, which includes the cochlea, has a very complex structure harboring many types of cells performing different functions. Among these cells are the auditory hair cells (HCs), which are terminally and well differentiated unique cells which have lost their regenerative potential after development. The auditory HCs are easily damaged by aging as well as during episodes of ototoxicity and acoustic trauma. HCs damages typically occur in the early stage of injury and can result a permanent hearing loss. Recently, there have been tremendous developments from stem cells (SCs) research involving sensorineural hearing loss, but several limitations and obstacles persist in allowing these developments from continuing onto clinical applications. This review discusses the recent advances in SC research in sensorineural hearing loss with the subsequent sections discussing the possible hurdles and limitations that currently preclude their clinical application.
Collapse
Affiliation(s)
- Yong-Ho Park
- Department of Otolaryngology-Head and Neck Surgery, Brain Research Institute, College of Medicine, Chungnam National University, Daejeon, Korea
| |
Collapse
|
29
|
Almeida-Branco MS, Cabrera S, Lopez-Escamez JA. Perspectivas para el tratamiento de la hipoacusia neurosensorial mediante regeneración celular del oído interno. ACTA OTORRINOLARINGOLOGICA ESPANOLA 2015; 66:286-95. [DOI: 10.1016/j.otorri.2014.07.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 07/30/2014] [Indexed: 12/16/2022]
|
30
|
Perspectives for the Treatment of Sensorineural Hearing Loss by Cellular Regeneration of the Inner Ear. ACTA OTORRINOLARINGOLOGICA ESPANOLA 2015. [DOI: 10.1016/j.otoeng.2015.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
31
|
Abstract
BACKGROUND Tissue engineering using biocompatible scaffolds, with or without cells, can permit surgeons to restore structure and function following tissue resection or in cases of congenital abnormality. Tracheal regeneration has emerged as a spearhead application of these technologies, whilst regenerative therapies are now being developed to treat most other diseases within otolaryngology. METHODS AND RESULTS A systematic review of the literature was performed using Ovid Medline and Ovid Embase, from database inception to 15 November 2014. A total of 561 papers matched the search criteria, with 76 fulfilling inclusion criteria. Articles were predominantly pre-clinical animal studies, reflecting the current status of research in this field. Several key human research articles were identified and discussed. CONCLUSION The main issues facing research in regenerative surgery are translation of animal model work into human models, increasing stem cell availability so it can be used to further research, and development of better facilities to enable implementation of these advances.
Collapse
|
32
|
Abstract
Hearing loss is the most common form of sensory impairment in humans and affects more than 40 million people in the United States alone. No drug-based therapy has been approved by the Food and Drug Administration, and treatment mostly relies on devices such as hearing aids and cochlear implants. Over recent years, more than 100 genetic loci have been linked to hearing loss and many of the affected genes have been identified. This understanding of the genetic pathways that regulate auditory function has revealed new targets for pharmacological treatment of the disease. Moreover, approaches that are based on stem cells and gene therapy, which may have the potential to restore or maintain auditory function, are beginning to emerge.
Collapse
Affiliation(s)
- Ulrich Müller
- Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, San Diego, California 92037, USA
| | - Peter G Barr-Gillespie
- Oregon Hearing Research Center, Vollum Institute, Oregon Health &Science University, 3181 South West Sam Jackson Park Road, Portland, Oregon 97239, USA
| |
Collapse
|
33
|
Adjudin protects rodent cochlear hair cells against gentamicin ototoxicity via the SIRT3-ROS pathway. Sci Rep 2015; 5:8181. [PMID: 25640330 PMCID: PMC4313083 DOI: 10.1038/srep08181] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 12/18/2014] [Indexed: 12/15/2022] Open
Abstract
Hearing loss resulting from hair cell degeneration is a common disease that affects millions of people worldwide. Strategies to overcome the apparent irreversible hair cell loss in mammals become paramount for hearing protection. Here we reported that, by using a well-established gentamicin-induced hair cell loss model in vitro, adjudin, a multi-functional small molecule drug, protected cochlear hair cells from gentamicin damage. Immunohistochemistry, Western blotting and quantitative RT-PCR analyses revealed that adjudin exerted its otoprotective effects by up-regulating the level of Sirt3, a member of Sirtuin family protein located in mitochondria, which regulates reactive oxygen species (ROS) production in cochlear cells and inhibits the production of ROS and apoptotic cells induced by gentamicin. Sirt3 silencing experiments confirmed that Sirt3-ROS signaling axis mediated hair cell protection against gentamicin by adjudin, at least in part. Furthermore, adjudin's otoprotection effects were also observed in an in vivo gentamicin-injured animal model. Taken together, these findings identify adjudin as a novel otoprotective small molecule via elevating Sirt3 levels and Sirt3 may be of therapeutic value in hair cell protection from ototoxic insults.
Collapse
|
34
|
Lou X, Xie J, Wang X, Yang L, Zhang Y. Comparison of sphere-forming capabilities of the cochlear stem cells derived from apical, middle and basal turns of murine organ of Corti. Neurosci Lett 2014; 579:1-6. [DOI: 10.1016/j.neulet.2014.06.060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 06/11/2014] [Accepted: 06/30/2014] [Indexed: 10/25/2022]
|
35
|
Lush ME, Piotrowski T. Sensory hair cell regeneration in the zebrafish lateral line. Dev Dyn 2014; 243:1187-202. [PMID: 25045019 DOI: 10.1002/dvdy.24167] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 07/12/2014] [Accepted: 07/14/2014] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Damage or destruction of sensory hair cells in the inner ear leads to hearing or balance deficits that can be debilitating, especially in older adults. Unfortunately, the damage is permanent, as regeneration of the inner ear sensory epithelia does not occur in mammals. RESULTS Zebrafish and other non-mammalian vertebrates have the remarkable ability to regenerate sensory hair cells and understanding the molecular and cellular basis for this regenerative ability will hopefully aid us in designing therapies to induce regeneration in mammals. Zebrafish not only possess hair cells in the ear but also in the sensory lateral line system. Hair cells in both organs are functionally analogous to hair cells in the inner ear of mammals. The lateral line is a mechanosensory system found in most aquatic vertebrates that detects water motion and aids in predator avoidance, prey capture, schooling, and mating. Although hair cell regeneration occurs in both the ear and lateral line, most research to date has focused on the lateral line due to its relatively simple structure and accessibility. CONCLUSIONS Here we review the recent discoveries made during the characterization of hair cell regeneration in zebrafish.
Collapse
Affiliation(s)
- Mark E Lush
- Stowers Institute for Medical Research, Kansas City, Missouri
| | | |
Collapse
|
36
|
Hu Z, Wang J. Histone deacetylase inhibitor induces the expression of select epithelial genes in mouse utricle sensory epithelia-derived progenitor cells. Cell Reprogram 2014; 16:266-75. [PMID: 24945595 DOI: 10.1089/cell.2013.0086] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Mouse utricle sensory epithelial cell-derived progenitor cells (MUCs), which have hair cell progenitor and mesenchymal features via epithelial-to-mesenchymal transition (EMT) as previously described, provide a potential approach for hair cell regeneration via cell transplantation. In this study, we treated MUCs with trichostatin A (TSA) to determine whether histone deacetylase inhibitor is able to stimulate the expression of epithelial genes in MUCs, an essential step for guiding mesenchymal-like MUCs to become sensory epithelial cells. After 72 h of TSA treatment, MUCs acquired epithelial-like features, which were indicated by increased expression of epithelial markers such as Cdh1, Krt18, and Dsp. Additionally, TSA decreased the expression of mesenchymal markers, including Zeb1, Zeb2, Snai1, and Snai2, and prosensory genes Lfng, Six1, and Dlx5. Moreover, the expression of the hair cell genes Atoh1 and Myo6 was increased in TSA-treated MUCs. We also observed significantly decreased expression of Hdac2 and Hdac3 in TSA-treated MUCs. However, no remarkable change was detected in protein expression using immunofluorescence, indicating that TSA-induced HDAC inhibition may contribute to the initial stage of the mesenchymal-to-epithelial phenotypic change. In the future, more work is needed to induce hair cell regeneration using inner ear tissue-derived progenitors to achieve an entire mesenchymal-to-epithelial transition.
Collapse
Affiliation(s)
- Zhengqing Hu
- Department of Otolaryngology-HNS, Wayne State University School of Medicine , Detroit, MI, 48201
| | | |
Collapse
|
37
|
Liu Q, Chen P, Wang J. Molecular mechanisms and potentials for differentiating inner ear stem cells into sensory hair cells. Dev Biol 2014; 390:93-101. [PMID: 24680894 DOI: 10.1016/j.ydbio.2014.03.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 03/15/2014] [Accepted: 03/18/2014] [Indexed: 12/31/2022]
Abstract
In mammals, hair cells may be damaged or lost due to genetic mutation, infectious disease, chemical ototoxicity, noise and other factors, causing permanent sensorineural deafness. Regeneration of hair cells is a basic pre-requisite for recovery of hearing in deaf animals. The inner ear stem cells in the organ of Corti and vestibular utricle are the most ideal precursors for regeneration of inner ear hair cells. This review highlights some recent findings concerning the proliferation and differentiation of inner ear stem cells. The differentiation of inner ear stem cells into hair cells involves a series of signaling pathways and regulatory factors. This paper offers a comprehensive analysis of the related studies.
Collapse
Affiliation(s)
- Quanwen Liu
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Ping Chen
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou 310058, PR China; Department of Cell Biology and Otolaryngology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Jinfu Wang
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou 310058, PR China.
| |
Collapse
|
38
|
|
39
|
Hu Z, Ulfendahl M. The potential of stem cells for the restoration of auditory function in humans. Regen Med 2014; 8:309-18. [PMID: 23627825 DOI: 10.2217/rme.13.32] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hearing loss is one of the most common disabilities, affecting approximately 10% of the population. Hair cells and spiral ganglion neurons are usually damaged in most cases of hearing loss. Currently, there is virtually no biological approach to replace damaged hearing cells. Recent developments in stem cell technology provide new opportunities for the treatment of deafness. Two major strategies have been investigated: differentiation of endogenous stem cells into new hair cells; and introduction of exogenous cells into the inner ear to substitute injured hearing neurons. Although there is still a learning curve in stem cell-based replacement, the probability exists to utilize personalized stem cells to eventually provide a novel intervention for patients with deafness in future clinical research trials.
Collapse
Affiliation(s)
- Zhengqing Hu
- Department of Otolaryngology-HNS, Wayne State University, MI, USA.
| | | |
Collapse
|
40
|
Ciorba A, Martini A. Regeneration in the mammalian inner ear: A glimpse into the future. HEARING BALANCE AND COMMUNICATION 2014. [DOI: 10.3109/21695717.2013.872835] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
41
|
Jiang H, Wang L, Beier KT, Cepko CL, Fekete DM, Brigande JV. Lineage analysis of the late otocyst stage mouse inner ear by transuterine microinjection of a retroviral vector encoding alkaline phosphatase and an oligonucleotide library. PLoS One 2013; 8:e69314. [PMID: 23935981 PMCID: PMC3723842 DOI: 10.1371/journal.pone.0069314] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 06/12/2013] [Indexed: 12/23/2022] Open
Abstract
The mammalian inner ear subserves the special senses of hearing and balance. The auditory and vestibular sensory epithelia consist of mechanically sensitive hair cells and associated supporting cells. Hearing loss and balance dysfunction are most frequently caused by compromise of hair cells and/or their innervating neurons. The development of gene- and cell-based therapeutics will benefit from a thorough understanding of the molecular basis of patterning and cell fate specification in the mammalian inner ear. This includes analyses of cell lineages and cell dispersals across anatomical boundaries (such as sensory versus nonsensory territories). The goal of this study was to conduct retroviral lineage analysis of the embryonic day 11.5(E11.5) mouse otic vesicle. A replication-defective retrovirus encoding human placental alkaline phosphatase (PLAP) and a variable 24-bp oligonucleotide tag was microinjected into the E11.5 mouse otocyst. PLAP-positive cells were microdissected from cryostat sections of the postnatal inner ear and subjected to nested PCR. PLAP-positive cells sharing the same sequence tag were assumed to have arisen from a common progenitor and are clonally related. Thirty five multicellular clones consisting of an average of 3.4 cells per clone were identified in the auditory and vestibular sensory epithelia, ganglia, spiral limbus, and stria vascularis. Vestibular hair cells in the posterior crista were related to one another, their supporting cells, and nonsensory epithelial cells lining the ampulla. In the organ of Corti, outer hair cells were related to a supporting cell type and were tightly clustered. By contrast, spiral ganglion neurons, interdental cells, and Claudius' cells were related to cells of the same type and could be dispersed over hundreds of microns. These data contribute new information about the developmental potential of mammalian otic precursors in vivo.
Collapse
Affiliation(s)
- Han Jiang
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
- Jilin Province Key Laboratory of Animal Embryo Engineering, College of Animal Science and Veterinary Medicine, Jilin University, Changchun, China
| | - Lingyan Wang
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Kevin T. Beier
- Department of Genetics, Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Constance L. Cepko
- Department of Genetics, Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Donna M. Fekete
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - John V. Brigande
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
- * E-mail:
| |
Collapse
|
42
|
Hu Z, Luo X, Zhang L, Lu F, Dong F, Monsell E, Jiang H. Generation of human inner ear prosensory-like cells via epithelial-to-mesenchymal transition. Regen Med 2013; 7:663-73. [PMID: 22954437 DOI: 10.2217/rme.12.53] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIM To identify human hair cell progenitors from adult inner ear sensory epithelium. MATERIALS & METHODS We collected discarded utricles from translabyrinthine surgery and isolated human utricular sensory epithelial cells (HUCs) to explore whether they can proliferate and obtain features of stem/progenitor cells in vitro using reverse transcription PCR and immunofluorescence. RESULTS When cultured in vitro, HUCs expressed genes and proteins that are usually present in prosensory cells and stem cells. Additionally, dissociated HUCs expanded on the substrates and presented properties of mesenchymal cells via epithelial-to-mesenchymal transition. CONCLUSION The results reveal that sensory epithelial cells from the adult human inner ear can re-enter the cell cycle and adopt a stem/progenitor cell fate. The outcomes of this study may open avenues for human hair cell progenitor generation, which could potentially provide a novel stem cell-based replacement for hearing loss and other inner ear disorders.
Collapse
Affiliation(s)
- Zhengqing Hu
- Department of Otolaryngology, Wayne State University School of Medicine, Detroit, MI, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Spiral ganglion neurons (SGNs) play a key role in hearing by rapidly and faithfully transmitting signals from the cochlea to the brain. Identification of the transcriptional networks that ensure the proper specification and wiring of SGNs during development will lay the foundation for efforts to rewire a damaged cochlea. Here, we show that the transcription factor Gata3, which is expressed in SGNs throughout their development, is essential for formation of the intricately patterned connections in the cochlea. We generated conditional knock-out mice in which Gata3 is deleted after SGNs are specified. Cochlear wiring is severely disrupted in these animals, with premature extension of neurites that follow highly abnormal trajectories toward their targets, as shown using in vitro neurite outgrowth assays together with time-lapse imaging of whole embryonic cochleae. Expression profiling of mutant neurons revealed a broad shift in gene expression toward a more differentiated state, concomitant with minor changes in SGN identity. Thus, Gata3 appears to serve as an "intermediate regulator" that guides SGNs through differentiation and preserves the auditory fate. As the first auditory-specific regulator of SGN development, Gata3 provides a useful molecular entry point for efforts to engineer SGNs for the restoration of hearing.
Collapse
|
44
|
Rubel EW, Furrer SA, Stone JS. A brief history of hair cell regeneration research and speculations on the future. Hear Res 2013; 297:42-51. [PMID: 23321648 DOI: 10.1016/j.heares.2012.12.014] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 12/18/2012] [Accepted: 12/19/2012] [Indexed: 12/24/2022]
Abstract
Millions of people worldwide suffer from hearing and balance disorders caused by loss of the sensory hair cells that convert sound vibrations and head movements into electrical signals that are conveyed to the brain. In mammals, the great majority of hair cells are produced during embryogenesis. Hair cells that are lost after birth are virtually irreplaceable, leading to permanent disability. Other vertebrates, such as fish and amphibians, produce hair cells throughout life. However, hair cell replacement after damage to the mature inner ear was either not investigated or assumed to be impossible until studies in the late 1980s proved this to be false. Adult birds were shown to regenerate lost hair cells in the auditory sensory epithelium after noise- and ototoxic drug-induced damage. Since then, the field of hair cell regeneration has continued to investigate the capacity of the auditory and vestibular epithelia in vertebrates (fishes, birds, reptiles, and mammals) to regenerate hair cells and to recover function, the molecular mechanisms governing these regenerative capabilities, and the prospect of designing biologically-based treatments for hearing loss and balance disorders. Here, we review the major findings of the field during the past 25 years and speculate how future inner ear repair may one day be achieved.
Collapse
Affiliation(s)
- Edwin W Rubel
- Virginia Merrill Bloedel Hearing Research Center and Department of Otolaryngology and Head & Neck Surgery, University of Washington, Seattle, WA 98195, USA.
| | | | | |
Collapse
|
45
|
Coticchia JM, Cohen D, Sachdeva L. Grand challenges in pediatric otolaryngology. Front Pediatr 2013; 1:10. [PMID: 24400256 PMCID: PMC3860887 DOI: 10.3389/fped.2013.00010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 05/01/2013] [Indexed: 11/18/2022] Open
Affiliation(s)
- James M Coticchia
- Division of Pediatric Otolaryngology, Department of Otolaryngology - Head and Neck Surgery, School of Medicine, Wayne State University Detroit, MI, USA
| | - David Cohen
- Department of Otolaryngology - Head and Neck Surgery, School of Medicine, Wayne State University Detroit, MI, USA
| | - Livjot Sachdeva
- Division of Pediatric Otolaryngology, Department of Otolaryngology - Head and Neck Surgery, School of Medicine, Wayne State University Detroit, MI, USA
| |
Collapse
|
46
|
Garland CB, Pomerantz JH. Regenerative strategies for craniofacial disorders. Front Physiol 2012; 3:453. [PMID: 23248598 PMCID: PMC3521957 DOI: 10.3389/fphys.2012.00453] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 11/12/2012] [Indexed: 01/26/2023] Open
Abstract
Craniofacial disorders present markedly complicated problems in reconstruction because of the complex interactions of the multiple, simultaneously affected tissues. Regenerative medicine holds promise for new strategies to improve treatment of these disorders. This review addresses current areas of unmet need in craniofacial reconstruction and emphasizes how craniofacial tissues differ from their analogs elsewhere in the body. We present a problem-based approach to illustrate current treatment strategies for various craniofacial disorders, to highlight areas of need, and to suggest regenerative strategies for craniofacial bone, fat, muscle, nerve, and skin. For some tissues, current approaches offer excellent reconstructive solutions using autologous tissue or prosthetic materials. Thus, new “regenerative” approaches would need to offer major advantages in order to be adopted. In other tissues, the unmet need is great, and we suggest the greatest regenerative need is for muscle, skin, and nerve. The advent of composite facial tissue transplantation and the development of regenerative medicine are each likely to add important new paradigms to our treatment of craniofacial disorders.
Collapse
Affiliation(s)
- Catharine B Garland
- Department of Surgery, Division of Plastic and Reconstructive Surgery, University of California San Francisco San Francisco, CA, USA ; Craniofacial and Mesenchymal Biology Program, University of California San Francisco San Francisco, CA, USA
| | | |
Collapse
|