1
|
Wijesinghe P, Sastry A, Hui E, Cogan TA, Zheng B, Ho G, Kakal J, Nunez DA. Adult porcine (Sus scrofa) derived inner ear cells: Characteristics in in-vitro cultures. Anat Rec (Hoboken) 2023. [PMID: 36598271 DOI: 10.1002/ar.25149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/21/2022] [Accepted: 12/10/2022] [Indexed: 01/05/2023]
Abstract
There is a need for an animal model that closely parallels human cochlea gestational development. This study aims to document porcine inner ear anatomy, and in vitro porcine derived inner ear cell culture characteristics. Twenty-four temporal bone were harvested from 12 adult pigs (Sus scrofa). Six were formalin fixed and their maximal diameters were measured. The cochlea duct length was determined by the insertion length of a Nucleus 22 cochlear implant in two bones. Four formalin fixed bones were sectioned for histology. Cochlear and vestibular tissues were harvested from non-fixed bones, cultured and characterized at different passages (P). Gene and protein expression of multipotent stem/progenitor (Nestin and Sox2), inner ear hair (Myosin VIIa, Prestin) and supporting (Cytokeratin 18 and Vimentin) cell markers were determined. The porcine cochlea was a 3.5 turn spiral. There was a separate vestibular compartment. The cochlear mean maximal diameter and height was 7.99 and 3.77 mm, respectively. Sphere forming cells were identified on phase-contrast microscopy. The relative mRNA expression levels of KRT18, MYO7A and SLC26A5 were significantly positively correlated in cochlear cultures; and MYO7A and SLC26A5; SOX2 and KRT18; NES and SLC26A5 genes were positively correlated in vestibular cultures (p = .037, Spearman correlation [τ] = .900). Inner ear sensory and stem cell characteristics persist in passaged porcine inner ear cells. Further work is required to establish the usefulness of porcine inner ear cell cultures to the study of human inner ear disorders.
Collapse
Affiliation(s)
- Printha Wijesinghe
- Division of Otolaryngology, Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
| | - Anand Sastry
- Bristol Veterinary School, University of Bristol, Bristol, UK
| | - Elizabeth Hui
- Division of Otolaryngology, Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
| | - Tristan A Cogan
- Bristol Veterinary School, University of Bristol, Bristol, UK
| | - Boyuan Zheng
- Division of Otolaryngology, Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
| | - Germain Ho
- Division of Otolaryngology, Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
| | - Juzer Kakal
- Division of Otolaryngology, Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
| | - Desmond A Nunez
- Division of Otolaryngology, Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada
| |
Collapse
|
2
|
Emerging Roles of RNA-Binding Proteins in Inner Ear Hair Cell Development and Regeneration. Int J Mol Sci 2022; 23:ijms232012393. [PMID: 36293251 PMCID: PMC9604452 DOI: 10.3390/ijms232012393] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/07/2022] [Accepted: 10/14/2022] [Indexed: 11/05/2022] Open
Abstract
RNA-binding proteins (RBPs) regulate gene expression at the post-transcriptional level. They play major roles in the tissue- and stage-specific expression of protein isoforms as well as in the maintenance of protein homeostasis. The inner ear is a bi-functional organ, with the cochlea and the vestibular system required for hearing and for maintaining balance, respectively. It is relatively well documented that transcription factors and signaling pathways are critically involved in the formation of inner ear structures and in the development of hair cells. Accumulating evidence highlights emerging functions of RBPs in the post-transcriptional regulation of inner ear development and hair cell function. Importantly, mutations of splicing factors of the RBP family and defective alternative splicing, which result in inappropriate expression of protein isoforms, lead to deafness in both animal models and humans. Because RBPs are critical regulators of cell proliferation and differentiation, they present the potential to promote hair cell regeneration following noise- or ototoxin-induced damage through mitotic and non-mitotic mechanisms. Therefore, deciphering RBP-regulated events during inner ear development and hair cell regeneration can help define therapeutic strategies for treatment of hearing loss. In this review, we outline our evolving understanding of the implications of RBPs in hair cell formation and hearing disease with the aim of promoting future research in this field.
Collapse
|
3
|
He L, Guo JY, Liu K, Wang GP, Gong SS. Research progress on flat epithelium of the inner ear. Physiol Res 2020; 69:775-785. [PMID: 32901490 DOI: 10.33549/physiolres.934447] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Sensorineural hearing loss and vertigo, resulting from lesions in the sensory epithelium of the inner ear, have a high incidence worldwide. The sensory epithelium of the inner ear may exhibit extreme degeneration and is transformed to flat epithelium (FE) in humans and mice with profound sensorineural hearing loss and/or vertigo. Various factors, including ototoxic drugs, noise exposure, aging, and genetic defects, can induce FE. Both hair cells and supporting cells are severely damaged in FE, and the normal cytoarchitecture of the sensory epithelium is replaced by a monolayer of very thin, flat cells of irregular contour. The pathophysiologic mechanism of FE is unclear but involves robust cell division. The cellular origin of flat cells in FE is heterogeneous; they may be transformed from supporting cells that have lost some features of supporting cells (dedifferentiation) or may have migrated from the flanking region. The epithelial-mesenchymal transition may play an important role in this process. The treatment of FE is challenging given the severe degeneration and loss of both hair cells and supporting cells. Cochlear implant or vestibular prosthesis implantation, gene therapy, and stem cell therapy show promise for the treatment of FE, although many challenges remain to be overcome.
Collapse
Affiliation(s)
- L He
- Department of Otolaryngology-Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China. ,
| | | | | | | | | |
Collapse
|
4
|
Van De Water TR. Historical Aspects of Gene Therapy and Stem Cell Therapy in the Treatment of Hearing and Balance Disorder. Anat Rec (Hoboken) 2020; 303:390-407. [DOI: 10.1002/ar.24332] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/18/2019] [Accepted: 11/22/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Thomas R. Van De Water
- Cochlear Implant Research Program, Department of Otolaryngology, University of Miami Ear InstituteUniversity of Miami Miller School of Medicine Miami Florida
| |
Collapse
|
5
|
Xia M, Ma J, Sun S, Li W, Li H. The biological strategies for hearing re-establishment based on the stem/progenitor cells. Neurosci Lett 2019; 711:134406. [PMID: 31377244 DOI: 10.1016/j.neulet.2019.134406] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/18/2019] [Accepted: 07/27/2019] [Indexed: 01/04/2023]
Abstract
The cochlea is the essential organ for hearing and includes both auditory sensory hair cells and spiral ganglion neurons. The discovery of inner ear stem cell brings hope to the regeneration of hair cell and spiral ganglion neuron as well as the followed hearing re-establishment. Thus the investigation on characteristics of inner ear stem/progenitor cells and related regulating clue is important to make such regeneration a reality. In addition, attempts have also been made to transplant exogenous stem cells into the inner ear to restore hearing function. In this review, we describe recent advances in the characterization of mammalian inner ear progenitor/stem cells and the mechanisms of regulating their proliferation and differentiation, and summarize studies that have used exogenous stem cells to repair damaged hair cells and neurons in the inner ear.
Collapse
Affiliation(s)
- Mingyu Xia
- ENT Institute and Otorhinolaryngology Department of the Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Jiaoyao Ma
- ENT Institute and Otorhinolaryngology Department of the Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Shan Sun
- ENT Institute and Otorhinolaryngology Department of the Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Wenyan Li
- ENT Institute and Otorhinolaryngology Department of the Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| | - Huawei Li
- ENT Institute and Otorhinolaryngology Department of the Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China; NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031, China; Shanghai Engineering Research Centre of Cochlear Implant, Shanghai, 200031, China; The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
6
|
Zhong C, Fu Y, Pan W, Yu J, Wang J. Atoh1 and other related key regulators in the development of auditory sensory epithelium in the mammalian inner ear: function and interplay. Dev Biol 2019; 446:133-141. [DOI: 10.1016/j.ydbio.2018.12.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/30/2018] [Accepted: 12/30/2018] [Indexed: 01/08/2023]
|
7
|
Powles-Glover N, Maconochie M. Prenatal and postnatal development of the mammalian ear. Birth Defects Res 2017; 110:228-245. [DOI: 10.1002/bdr2.1167] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/16/2017] [Accepted: 10/28/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Nicola Powles-Glover
- AstraZeneca, Innovative Medicines and Early Development; Drug Safety and Metabolism; Hertfordshire SG8 6HB United Kingdom
| | - Mark Maconochie
- Queen Mary University of London; London E1 4NS United Kingdom
| |
Collapse
|
8
|
Massucci-Bissoli M, Lezirovitz K, Oiticica J, Bento RF. Evidence of progenitor cells in the adult human cochlea: sphere formation and identification of ABCG2. Clinics (Sao Paulo) 2017; 72:714-717. [PMID: 29236919 PMCID: PMC5707191 DOI: 10.6061/clinics/2017(11)11] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 07/12/2017] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES The aim of this study was to search for evidence of stem or progenitor cells in the adult human cochlea by testing for sphere formation capacity and the presence of the stem cell marker ABCG2. METHODS Cochleas removed from patients undergoing vestibular schwannoma resection (n=2) and from brain-dead organ donors (n=4) were dissociated for either flow cytometry analysis for the stem cell marker ABCG2 or a sphere formation assay that is widely used to test the sphere-forming capacity of cells from mouse inner ear tissue. RESULTS Spheres were identified after 2-5 days in vitro, and the stem cell marker ABCG2 was detected using flow cytometric analysis after cochlear dissociation. CONCLUSIONS Evidence suggests that there may be progenitor cells in the adult human cochlea, although further studies are required.
Collapse
Affiliation(s)
- Milene Massucci-Bissoli
- Departamento de Otorrinolaringologia, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, BR
- *Corresponding authors. E-mails: /
| | - Karina Lezirovitz
- Departamento de Otorrinolaringologia, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, BR
- *Corresponding authors. E-mails: /
| | - Jeanne Oiticica
- Departamento de Otorrinolaringologia, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Ricardo Ferreira Bento
- Departamento de Otorrinolaringologia, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, BR
| |
Collapse
|
9
|
Wakasaki T, Niiro H, Jabbarzadeh-Tabrizi S, Ohashi M, Kimitsuki T, Nakagawa T, Komune S, Akashi K. Musashi-1 is the candidate of the regulator of hair cell progenitors during inner ear regeneration. BMC Neurosci 2017; 18:64. [PMID: 28814279 PMCID: PMC5559865 DOI: 10.1186/s12868-017-0382-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 08/10/2017] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Hair cell loss in the cochlea is caused by ototoxic drugs, aging, and environmental stresses and could potentially lead to devastating pathophysiological effects. In adult mammals, hair cell loss is irreversible and may result in hearing and balance deficits. In contrast, nonmammalian vertebrates, including birds, can regenerate hair cells through differentiation of supporting cells and restore inner ear function, suggesting that hair cell progenitors are present in the population of supporting cells. RESULTS In the present study, we aimed to identify novel genes related to regeneration in the chicken utricle by gene expression profiling of supporting cell and hair cell populations obtained by laser capture microdissection. The volcano plot identified 408 differentially expressed genes (twofold change, p = 0.05, Benjamini-Hochberg multiple testing correction), 175 of which were well annotated. Among these genes, we focused on Musashi-1 (MSI1), a marker of neural stem cells involved in Notch signaling, and the downstream genes in the Notch pathway. Higher expression of these genes in supporting cells compared with that in hair cells was confirmed by quantitative reverse transcription polymerase chain reaction. Immunohistochemistry analysis demonstrated that MSI1 was mainly localized at the basal side of the supporting cell layer in normal chick utricles. During the regeneration period following aminoglycoside antibiotic-induced damage of chicken utricles, the expression levels of MSI1, hairy and enhancer of split-5, and cyclin D1 were increased, and BrdU labeling indicated that cell proliferation was enhanced. CONCLUSIONS The findings of this study suggested that MSI1 played an important role in the proliferation of supporting cells in the inner ear during normal and damaged conditions and could be a potential therapeutic target in the treatment of vestibular defects.
Collapse
Affiliation(s)
- Takahiro Wakasaki
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan. .,Department of Head and Neck Surgery, National Hospital Organization, Kyushu Cancer Center, 3-1-1 Notame, Miniami-ku, Fukuoka, 811-1395, Japan.
| | - Hiroaki Niiro
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Siamak Jabbarzadeh-Tabrizi
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mitsuru Ohashi
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takashi Kimitsuki
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takashi Nakagawa
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shizuo Komune
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
10
|
Cells Expressing Prominin-1 in Neonatal Murine Inferior Colliculus Differentiate into Neurons and Glia. Mol Neurobiol 2017; 55:4998-5005. [PMID: 28795331 PMCID: PMC5948249 DOI: 10.1007/s12035-017-0701-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 07/31/2017] [Indexed: 01/15/2023]
Abstract
Inferior colliculus (IC) is a major center for the integration and processing of acoustic information from ascending auditory pathways. Damage to the IC as well as normal aging can impair auditory function. Novel strategies such as stem cell (SC)-based regenerative therapy are required for functional recovery because mature neural cells have a minimal regenerative capacity after an injury. However, it is not known if there are neural stem cells (NSCs) in the IC. Herein, we screened for NSCs by surface marker analysis using flow cytometry. Isolated IC cells expressing prominin-1 (CD133) exhibited the cardinal NSC properties self-renewal capacity, expression of known NSC markers (SOX2 and nestin), and multipotency. Prominin-1-expressing cells from neonatal IC generated neurospheres, and culture of these neurospheres in differentiation-conditioned medium gave rise to gamma-aminobutyric acid-ergic (GABAergic) neurons, astrocytes, and oligodendrocytes. The presence of NSC-like cells in the IC has important implications for understanding IC development and for potential regenerative therapy.
Collapse
|
11
|
Rask-Andersen H, Li H, Löwenheim H, Müller M, Pfaller K, Schrott-Fischer A, Glueckert R. Supernumerary human hair cells-signs of regeneration or impaired development? A field emission scanning electron microscopy study. Ups J Med Sci 2017; 122:11-19. [PMID: 28145795 PMCID: PMC5361427 DOI: 10.1080/03009734.2016.1271843] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Current attempts to regenerate cochlear sensorineural structures motivate further inspection of the human organ of hearing. Here, we analyzed the supernumerary inner hair cell (sIHC), a possible sign of regeneration and cell replacement. METHODS Human cochleae were studied using field emission scanning electron microscopy (FESEM; maximum resolution 2 nm) obtained from individuals aged 44, 48, and 58 years with normal sensorineural pure-tone average (PTA) thresholds (PTA <20 dB). The wasted tissue was harvested during trans-cochlear approaches and immediately fixed for ultrastructural analysis. RESULTS All specimens exhibited sIHCs at all turns except at the extreme lower basal turn. In one specimen, it was possible to image and count the inner hair cells (IHCs) along the cochlea representing the 0.2 kHz-8 kHz region according to the Greenwood place/frequency scale. In a region with 2,321 IHCs, there were 120 scattered one-cell losses or 'gaps' (5%). Forty-two sIHCs were present facing the modiolus. Thirty-eight percent of the sIHCs were located near a 'gap' in the IHC row (±6 IHCs). CONCLUSIONS The prevalence of ectopic inner hair cells was higher than expected. The morphology and placement could reflect a certain ongoing regeneration. Further molecular studies are needed to verify if the regenerative capacity of the human auditory periphery might have been underestimated.
Collapse
Affiliation(s)
- Helge Rask-Andersen
- Department of Surgical Sciences, Head and Neck Surgery, Section of Otolaryngology, Uppsala University Hospital, Uppsala, Sweden
- Department of Otolaryngology, Uppsala University Hospital, Uppsala, Sweden
- CONTACT Helge Rask-Andersen Department of Surgical Sciences, Head and Neck Surgery, Section of Otolaryngology, Uppsala University Hospital, SE-751 85, Uppsala, Sweden
| | - Hao Li
- Department of Surgical Sciences, Head and Neck Surgery, Section of Otolaryngology, Uppsala University Hospital, Uppsala, Sweden
- Department of Otolaryngology, Uppsala University Hospital, Uppsala, Sweden
| | - Hubert Löwenheim
- Department of Otolaryngology, Medical University of Innsbruck, Innsbruck, Austria
- Medical Campus University of Oldenburg School of Medicine and Health Sciences, European Medical School, Oldenburg, Germany
- Research Center of Neurosensory Science, University of Oldenburg, Oldenburg, Germany
| | - Marcus Müller
- Department of Otolaryngology, Medical University of Innsbruck, Innsbruck, Austria
- Medical Campus University of Oldenburg School of Medicine and Health Sciences, European Medical School, Oldenburg, Germany
- Research Center of Neurosensory Science, University of Oldenburg, Oldenburg, Germany
| | - Kristian Pfaller
- Cluster of Excellence Hearing4all, University of Oldenburg, Oldenburg, Germany
| | - Annelies Schrott-Fischer
- Department of Histology and Molecular Cell Biology, Institute of Anatomy and Histology, Medical University of Innsbruck, Innsbruck, Austria
| | - Rudolf Glueckert
- Department of Histology and Molecular Cell Biology, Institute of Anatomy and Histology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
12
|
Santos ACM, Borghesi J, Mario LC, Anunciação ARA, Mess AM, Carreira ACO, Favaron PO, Miglino MA. Cochlear epithelial of dog fetuses: a new source of multipotent stem cells. Cytotechnology 2017; 69:179-189. [PMID: 28074389 DOI: 10.1007/s10616-016-0049-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 11/22/2016] [Indexed: 10/20/2022] Open
Abstract
Hearing loss caused by the damage of cochlea sensory cells or neurons is a common human disease, but also affects dogs and other animals. To test their progenitor nature as potential value for future therapies, we characterized cells derived from the cochlear epithelium in dog fetuses. In total, 8 fetuses of 35-40 days of gestation, derived from castration campaigns, were investigated. Cells were analysed by the MTT colorimetric assay and in regard to cell cycle, differentiation capacities, immunophenotypes and qPCR analysis. In culture, cells had a fibroblast-like morphology. Phenotypic immunocharacterization showed positive staining for mesenchymal stem cell and pluripotency markers and were negative for hematopoietic cell markers. Cells possessed differentiation capacity for the three main cell lineages: osteogenic, adipogenic and chondrogenic, altogether indicating their nature as mesenchymal stem cells. Thus, cells derived from fetal cochlear tissues indeed may provide valuable sources of progenitor cells for cell therapy of canine deafness and other diseases.
Collapse
Affiliation(s)
- Ana Carolina M Santos
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, Cidade Universitária, São Paulo, SP, 05508-270, Brazil
| | - Jéssica Borghesi
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, Cidade Universitária, São Paulo, SP, 05508-270, Brazil.
| | - Lara Carolina Mario
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, Cidade Universitária, São Paulo, SP, 05508-270, Brazil
| | - Adriana Raquel A Anunciação
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, Cidade Universitária, São Paulo, SP, 05508-270, Brazil
| | - Andrea Maria Mess
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, Cidade Universitária, São Paulo, SP, 05508-270, Brazil
| | - Ana Claudia O Carreira
- NUCEL (Cell and Molecular Therapy Center) and NETCEM (Center for Studies in Cell and Molecular Therapy), Internal Medicine Department, School of Medicine, University of Sao Paulo, São Paulo, SP, Brazil
| | - Phelipe O Favaron
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, Cidade Universitária, São Paulo, SP, 05508-270, Brazil
| | - Maria Angélica Miglino
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, Cidade Universitária, São Paulo, SP, 05508-270, Brazil
| |
Collapse
|
13
|
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: 1.8] [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
|
14
|
Abboud N, Fontbonne A, Watabe I, Tonetto A, Brezun JM, Feron F, Zine A. Culture conditions have an impact on the maturation of traceable, transplantable mouse embryonic stem cell-derived otic progenitor cells. J Tissue Eng Regen Med 2016; 11:2629-2642. [PMID: 27099197 DOI: 10.1002/term.2163] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 12/29/2015] [Accepted: 02/03/2016] [Indexed: 01/02/2023]
Abstract
The generation of replacement inner ear hair cells (HCs) remains a challenge and stem cell therapy holds the potential for developing therapeutic solutions to hearing and balance disorders. Recent developments have made significant strides in producing mouse otic progenitors using cell culture techniques to initiate HC differentiation. However, no consensus has been reached as to efficiency and therefore current methods remain unsatisfactory. In order to address these issues, we compare the generation of otic and HC progenitors from embryonic stem (ES) cells in two cell culture systems: suspension vs. adherent conditions. In the present study, an ES cell line derived from an Atoh1-green fluorescent protein (GFP) transgenic mouse was used to track the generation of otic progenitors, initial HCs and to compare these two differentiation systems. We used a two-step short-term differentiation method involving an induction period of 5 days during which ES cells were cultured in the presence of Wnt/transforming growth factor TGF-β inhibitors and insulin-like growth factor IGF-1 to suppress mesoderm and reinforce presumptive ectoderm and otic lineages. The generated embryoid bodies were then differentiated in medium containing basic fibroblast growth factor (bFGF) for an additional 5 days using either suspension or adherent culture methods. Upon completion of differentiation, quantitative polymerase chain reaction analysis and immunostaining monitored the expression of otic/HC progenitor lineage markers. The results indicate that cells differentiated in suspension cultures produced cells expressing otic progenitor/HC markers at a higher efficiency compared with the production of these cell types within adherent cultures. Furthermore, we demonstrated that a fraction of these cells can incorporate into ototoxin-injured mouse postnatal cochlea explants and express MYO7A after transplantation. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Nesrine Abboud
- Aix-Marseille Université, LNIA, CNRS, UMR 7260, Marseille, France
| | - Arnaud Fontbonne
- Aix-Marseille Université, LNIA, CNRS, UMR 7260, Marseille, France
| | - Isabelle Watabe
- Aix-Marseille Université, LNIA, CNRS, UMR 7260, Marseille, France
| | - Alain Tonetto
- Pôle PRATIM, Fédération de Chimie de Marseille. Aix-Marseille Université, Marseille, France
| | | | - François Feron
- Aix-Marseille Université, NICN, CNRS, UMR 7259, Marseille, France
| | - Azel Zine
- Aix-Marseille Université, LNIA, CNRS, UMR 7260, Marseille, France.,Laboratoire de Biophysique, Faculté de Pharmacie, Université de Montpellier, Montpellier, France
| |
Collapse
|
15
|
Induction of Functional Hair-Cell-Like Cells from Mouse Cochlear Multipotent Cells. Stem Cells Int 2015; 2016:8197279. [PMID: 27057177 PMCID: PMC4709769 DOI: 10.1155/2016/8197279] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 09/15/2015] [Accepted: 09/28/2015] [Indexed: 11/18/2022] Open
Abstract
In this paper, we developed a two-step-induction method of generating functional hair cells from inner ear multipotent cells. Multipotent cells from the inner ear were established and induced initially into progenitor cells committed to the inner ear cell lineage on the poly-L-lysine substratum. Subsequently, the committed progenitor cells were cultured on the mitotically inactivated chicken utricle stromal cells and induced into hair-cell-like cells containing characteristic stereocilia bundles. The hair-cell-like cells exhibited rapid permeation of FM1-43FX. The whole-cell patch-clamp technique was used to measure the membrane currents of cells differentiated for 7 days on chicken utricle stromal cells and analyze the biophysical properties of the hair-cell-like cells by recording membrane properties of cells. The results suggested that the hair-cell-like cells derived from inner ear multipotent cells were functional following differentiation in an enabling environment.
Collapse
|
16
|
Induction of differentiation of human embryonic stem cells into functional hair-cell-like cells in the absence of stromal cells. Int J Biochem Cell Biol 2015; 81:208-222. [PMID: 26615761 DOI: 10.1016/j.biocel.2015.11.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 10/27/2015] [Accepted: 11/20/2015] [Indexed: 02/07/2023]
Abstract
Sensorineural hearing loss and vestibular dysfunction have become the most common forms of sensory defects. Stem cell-based therapeutic strategies for curing hearing loss are being developed. Several attempts to develop hair cells by using chicken utricle stromal cells as feeder cells have resulted in phenotypic conversion of stem cells into inner ear hair-cell-like cells. Here, we induced the differentiation of human embryonic stem cells (hESCs) into otic epithelial progenitors (OEPs), and further induced the differentiation of OEPs into hair-cell-like cells using different substrates. Our results showed that OEPs cultured on the chicken utricle stromal cells with the induction medium could differentiate into hair-cell-like cells with stereociliary bundles. Co-culture with stromal cells, however, may be problematic for subsequent examination of the induced hair-cell-like cells. In order to avoid the interference from stromal cells, we cultured OEPs on laminin with different induction media and examined the effects of the induction medium on the differentiation potentials of OEPs into hair-cell-like cells. The results revealed that the culture of OEPs on laminin with the conditioned medium from chicken utricle stromal cells supplemented with EGF and all-trans retinoic acid (RA) could promote the organization of cells into epithelial clusters displaying hair-cell-like cells with stereociliary bundles. These cells also displayed the expected electrophysiological properties.
Collapse
|
17
|
Atkinson PJ, Huarcaya Najarro E, Sayyid ZN, Cheng AG. Sensory hair cell development and regeneration: similarities and differences. Development 2015; 142:1561-71. [PMID: 25922522 DOI: 10.1242/dev.114926] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Sensory hair cells are mechanoreceptors of the auditory and vestibular systems and are crucial for hearing and balance. In adult mammals, auditory hair cells are unable to regenerate, and damage to these cells results in permanent hearing loss. By contrast, hair cells in the chick cochlea and the zebrafish lateral line are able to regenerate, prompting studies into the signaling pathways, morphogen gradients and transcription factors that regulate hair cell development and regeneration in various species. Here, we review these findings and discuss how various signaling pathways and factors function to modulate sensory hair cell development and regeneration. By comparing and contrasting development and regeneration, we also highlight the utility and limitations of using defined developmental cues to drive mammalian hair cell regeneration.
Collapse
Affiliation(s)
- Patrick J Atkinson
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Elvis Huarcaya Najarro
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Zahra N Sayyid
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alan G Cheng
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
18
|
Hypoxia Induces a Metabolic Shift and Enhances the Stemness and Expansion of Cochlear Spiral Ganglion Stem/Progenitor Cells. BIOMED RESEARCH INTERNATIONAL 2015; 2015:359537. [PMID: 26236724 PMCID: PMC4506838 DOI: 10.1155/2015/359537] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 10/03/2014] [Indexed: 11/17/2022]
Abstract
Previously, we demonstrated that hypoxia (1% O2) enhances stemness markers and expands the cell numbers of cochlear stem/progenitor cells (SPCs). In this study, we further investigated the long-term effect of hypoxia on stemness and the bioenergetic status of cochlear spiral ganglion SPCs cultured at low oxygen tensions. Spiral ganglion SPCs were obtained from postnatal day 1 CBA/CaJ mouse pups. The measurement of oxygen consumption rate, extracellular acidification rate (ECAR), and intracellular adenosine triphosphate levels corresponding to 20% and 5% oxygen concentrations was determined using a Seahorse XF extracellular flux analyzer. After low oxygen tension cultivation for 21 days, the mean size of the hypoxia-expanded neurospheres was significantly increased at 5% O2; this correlated with high-level expression of hypoxia-inducible factor-1 alpha (Hif-1α), proliferating cell nuclear antigen (PCNA), cyclin D1, Abcg2, nestin, and Nanog proteins but downregulated expression of p27 compared to that in a normoxic condition. Low oxygen tension cultivation tended to increase the side population fraction, with a significant difference found at 5% O2 compared to that at 20% O2. In addition, hypoxia induced a metabolic energy shift of SPCs toward higher basal ECARs and higher maximum mitochondrial respiratory capacity but lower proton leak than under normoxia, where the SPC metabolism was switched toward glycolysis in long-term hypoxic cultivation.
Collapse
|
19
|
Defourny J, Mateo Sánchez S, Schoonaert L, Robberecht W, Davy A, Nguyen L, Malgrange B. Cochlear supporting cell transdifferentiation and integration into hair cell layers by inhibition of ephrin-B2 signalling. Nat Commun 2015; 6:7017. [DOI: 10.1038/ncomms8017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 03/25/2015] [Indexed: 01/08/2023] Open
|
20
|
Romero-Guevara R, Cencetti F, Donati C, Bruni P. Sphingosine 1-phosphate signaling pathway in inner ear biology. New therapeutic strategies for hearing loss? Front Aging Neurosci 2015; 7:60. [PMID: 25954197 PMCID: PMC4407579 DOI: 10.3389/fnagi.2015.00060] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 04/08/2015] [Indexed: 12/13/2022] Open
Abstract
Hearing loss is one of the most prevalent conditions around the world, in particular among people over 60 years old. Thus, an increase of this affection is predicted as result of the aging process in our population. In this context, it is important to further explore the function of molecular targets involved in the biology of inner ear sensory cells to better individuate new candidates for therapeutic application. One of the main causes of deafness resides into the premature death of hair cells and auditory neurons. In this regard, neurotrophins and growth factors such as insulin like growth factor are known to be beneficial by favoring the survival of these cells. An elevated number of published data in the last 20 years have individuated sphingolipids not only as structural components of biological membranes but also as critical regulators of key biological processes, including cell survival. Ceramide, formed by catabolism of sphingomyelin (SM) and other complex sphingolipids, is a strong inducer of apoptotic pathway, whereas sphingosine 1-phosphate (S1P), generated by cleavage of ceramide to sphingosine and phosphorylation catalyzed by two distinct sphingosine kinase (SK) enzymes, stimulates cell survival. Interestingly S1P, by acting as intracellular mediator or as ligand of a family of five distinct S1P receptors (S1P1–S1P5), is a very powerful bioactive sphingolipid, capable of triggering also other diverse cellular responses such as cell migration, proliferation and differentiation, and is critically involved in the development and homeostasis of several organs and tissues. Although new interesting data have become available, the information on S1P pathway and other sphingolipids in the biology of the inner ear is limited. Nonetheless, there are several lines of evidence implicating these signaling molecules during neurogenesis in other cell populations. In this review, we discuss the role of S1P during inner ear development, also as guidance for future studies.
Collapse
Affiliation(s)
- Ricardo Romero-Guevara
- Department Scienze Biomediche Sperimentali e Cliniche "Mario Serio", University of Florence Firenze, Italy
| | - Francesca Cencetti
- Department Scienze Biomediche Sperimentali e Cliniche "Mario Serio", University of Florence Firenze, Italy
| | - Chiara Donati
- Department Scienze Biomediche Sperimentali e Cliniche "Mario Serio", University of Florence Firenze, Italy
| | - Paola Bruni
- Department Scienze Biomediche Sperimentali e Cliniche "Mario Serio", University of Florence Firenze, Italy
| |
Collapse
|
21
|
Liu Q, Chen J, Gao X, Ding J, Tang Z, Zhang C, Chen J, Li L, Chen P, Wang J. Identification of stage-specific markers during differentiation of hair cells from mouse inner ear stem cells or progenitor cells in vitro. Int J Biochem Cell Biol 2015; 60:99-111. [PMID: 25582750 DOI: 10.1016/j.biocel.2014.12.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Revised: 12/10/2014] [Accepted: 12/31/2014] [Indexed: 12/11/2022]
Abstract
The induction of inner ear hair cells from stem cells or progenitor cells in the inner ear proceeds through a committed inner ear sensory progenitor cell stage prior to hair cell differentiation. To increase the efficacy of inducing inner ear hair cell differentiation from the stem cells or progenitor cells, it is essential to identify comprehensive markers for the stem cells/progenitor cells from the inner ear, the committed inner ear sensory progenitor cells and the differentiating hair cells to optimize induction conditions. Here, we report that we efficiently isolated and expanded the stem cells or progenitor cells from postnatal mouse cochleae, and induced the generation of inner ear progenitor cells and subsequent differentiation of hair cells. We profiled the gene expression of the stem cells or progenitor cells, the inner ear progenitor cells, and hair cells using aRNA microarray analysis. The pathway and gene ontology (GO) analysis of differentially expressed genes was performed. Analysis of genes exclusively detected in one particular cellular population revealed 30, 38, and 31 genes specific for inner ear stem cells, inner ear progenitor cells, and hair cells, respectively. We further examined the expression of these genes in vivo and determined that Gdf10+Ccdc121, Tmprss9+Orm1, and Chrna9+Espnl are marker genes specific for inner ear stem cells, inner ear progenitor cells, and differentiating hair cells, respectively. The identification of these marker genes will likely help the effort to increase the efficacy of hair cell induction from the stem cells or progenitor cells.
Collapse
Affiliation(s)
- Quanwen Liu
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Jiarong Chen
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Xiangli Gao
- Laboratory of Bone Marrow, The First Hospital, Zhejiang University, Hangzhou, Zhejiang 310006, PR China
| | - Jie Ding
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Zihua Tang
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Cui Zhang
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Jianling Chen
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Liang Li
- 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; Departments of Cell Biology and Otolaryngology, Emory University School of Medicine, Atlanta, GA 30322, United States
| | - Jinfu Wang
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou 310058, PR China.
| |
Collapse
|
22
|
Diensthuber M, Zecha V, Wagenblast J, Arnhold S, Edge ASB, Stöver T. Spiral ganglion stem cells can be propagated and differentiated into neurons and glia. Biores Open Access 2014; 3:88-97. [PMID: 24940560 PMCID: PMC4048968 DOI: 10.1089/biores.2014.0016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The spiral ganglion is an essential functional component of the peripheral auditory system. Most types of hearing loss are associated with spiral ganglion cell degeneration which is irreversible due to the inner ear's lack of regenerative capacity. Recent studies revealed the existence of stem cells in the postnatal spiral ganglion, which gives rise to the hope that these cells might be useful for regenerative inner ear therapies. Here, we provide an in-depth analysis of sphere-forming stem cells isolated from the spiral ganglion of postnatal mice. We show that spiral ganglion spheres have characteristics similar to neurospheres isolated from the brain. Importantly, spiral ganglion sphere cells maintain their major stem cell characteristics after repeated propagation, which enables the culture of spheres for an extended period of time. In this work, we also demonstrate that differentiated sphere-derived cell populations not only adopt the immunophenotype of mature spiral ganglion cells but also develop distinct ultrastructural features of neurons and glial cells. Thus, our work provides further evidence that self-renewing spiral ganglion stem cells might serve as a promising source for the regeneration of lost auditory neurons.
Collapse
Affiliation(s)
- Marc Diensthuber
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Frankfurt am Main , Goethe University, Frankfurt am Main, Germany . ; Department of Otology and Laryngology, Harvard Medical School , Boston, Massachusetts. ; Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary , Boston, Massachusetts
| | - Veronika Zecha
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Frankfurt am Main , Goethe University, Frankfurt am Main, Germany
| | - Jens Wagenblast
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Frankfurt am Main , Goethe University, Frankfurt am Main, Germany
| | - Stefan Arnhold
- Institute of Veterinary Anatomy, Histology, and Embryology, Justus-Liebig University Giessen , Giessen, Germany
| | - Albert S B Edge
- Department of Otology and Laryngology, Harvard Medical School , Boston, Massachusetts. ; Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary , Boston, Massachusetts. ; Program in Speech and Hearing Bioscience and Technology, Division of Health Sciences and Technology, Harvard and MIT , Cambridge, Massachusetts
| | - Timo Stöver
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Frankfurt am Main , Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
23
|
Cox BC, Chai R, Lenoir A, Liu Z, Zhang L, Nguyen DH, Chalasani K, Steigelman KA, Fang J, Rubel EW, Cheng AG, Zuo J. Spontaneous hair cell regeneration in the neonatal mouse cochlea in vivo. Development 2014; 141:816-29. [PMID: 24496619 DOI: 10.1242/dev.103036] [Citation(s) in RCA: 231] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Loss of cochlear hair cells in mammals is currently believed to be permanent, resulting in hearing impairment that affects more than 10% of the population. Here, we developed two genetic strategies to ablate neonatal mouse cochlear hair cells in vivo. Both Pou4f3(DTR/+) and Atoh1-CreER™; ROSA26(DTA/+) alleles allowed selective and inducible hair cell ablation. After hair cell loss was induced at birth, we observed spontaneous regeneration of hair cells. Fate-mapping experiments demonstrated that neighboring supporting cells acquired a hair cell fate, which increased in a basal to apical gradient, averaging over 120 regenerated hair cells per cochlea. The normally mitotically quiescent supporting cells proliferated after hair cell ablation. Concurrent fate mapping and labeling with mitotic tracers showed that regenerated hair cells were derived by both mitotic regeneration and direct transdifferentiation. Over time, regenerated hair cells followed a similar pattern of maturation to normal hair cell development, including the expression of prestin, a terminal differentiation marker of outer hair cells, although many new hair cells eventually died. Hair cell regeneration did not occur when ablation was induced at one week of age. Our findings demonstrate that the neonatal mouse cochlea is capable of spontaneous hair cell regeneration after damage in vivo. Thus, future studies on the neonatal cochlea might shed light on the competence of supporting cells to regenerate hair cells and on the factors that promote the survival of newly regenerated hair cells.
Collapse
Affiliation(s)
- Brandon C Cox
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Martone T, Giordano P, Dagna F, Carulli D, Albera R, Rossi F. Nestin expression and reactive phenomena in the mouse cochlea after kanamycin ototoxicity. Eur J Neurosci 2014; 39:1729-41. [PMID: 24689961 DOI: 10.1111/ejn.12576] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 02/28/2014] [Indexed: 11/28/2022]
Abstract
Following injury to the adult mammalian cochlea, hair cells cannot be spontaneously replaced. Nonetheless, the postnatal cochlea contains progenitor cells, distinguished by the expression of nestin, which are able to proliferate and form neurospheres in vitro. Such resident progenitors might be endowed with reparative potential. However, to date little is known about their behaviour in situ following hair cell injury. Using adult mice and ex vivo cochlear cultures, we sought to determine whether: (i) resident cochlear progenitors respond to kanamycin ototoxicity and compensate for it; and (ii) the reparative potential of cochlear progenitors can be stimulated by the addition of growth factors. Morphological changes of cochlear tissue, expression of nestin mRNA and protein and cell proliferation were investigated in these models. Our observations show that ototoxic injury has modest effects on nestin expression and cell proliferation. On the other hand, the addition of growth factors to the injured cochlear explants induced the appearance of nestin-positive cells in the supporting cell area of the organ of Corti. The vast majority of nestin-expressing cells, however, were not proliferating. Growth factors also had a robust stimulatory effect on axonal sprouting and the proliferative response, which was more pronounced in injured cochleae. On the whole, our findings indicate that nestin expression after kanamycin ototoxicity is related to tissue reactivity rather than activation of resident progenitors attempting to replace the lost receptors. In addition, administration of growth factors significantly enhances tissue remodelling, suggesting that cochlear repair may be promoted by the exogenous application of regeneration-promoting substances.
Collapse
Affiliation(s)
- Tiziana Martone
- Department of Neuroscience, Neuroscience Institute of Turin (NIT), Turin, Italy; Neuroscience Institute Cavalieri-Ottolenghi (NICO), University of Turin, Orbassano, Turin, Italy
| | | | | | | | | | | |
Collapse
|
25
|
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: 1.3] [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
|
26
|
Generation of induced pluripotent stem cells from neonatal mouse cochlear cells. Differentiation 2014; 87:127-33. [PMID: 24582575 DOI: 10.1016/j.diff.2014.02.004] [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: 11/11/2013] [Revised: 01/18/2014] [Accepted: 02/11/2014] [Indexed: 12/12/2022]
Abstract
The sensory epithelium (SE) within the mammalian cochleae has a limited capacity for regeneration, and the loss of mammalian cochlear hair cells always lead to permanent hearing loss. Previous reports show that early postnatal cochlea harbors stem/progenitor-like cells nominated otospheres which have a limited regenerative/repair capacity, while these cell populations are progressively lost during the postnatal development. Induced pluripotent stem cells (iPS cells) directly reprogrammed from non-embryonic cells have captured great attentions in the scientific community. In the present study, we determine whether Yamanaka׳s factors can induce the reprogramming of cochlear cells into iPS cells. We introduce defined factors Oct3/4, Sox2 and Klf4 into otospheres derived from postnatal day-1 (P1) mouse SE, and analyze characteristics alterations in cochlear cells. After transduction, otospheres generated colonies exhibiting a normal karyotype and morphology similar to that of mouse embryonic stem cells (ESCs). Moreover, these cochlear iPS cells also express ESC-like markers. Importantly, the cochlear iPS cells show pluripotency in vitro and in vivo, as evidenced by differentiation into three germ layers by embryoid body formation, as well as high efficient formation of teratomas containing three germ layers in immunodeficient mice. Thus, pluripotent cochlear iPS cells can be generated from cochlear cells by using three Yamanaka׳s transcription factors. These attempts represent the first step toward generating fully pluripotent iPS cells from mammalian cochleae with defined exogenous genes.
Collapse
|
27
|
Toward Translating Molecular Ear Development to Generate Hair Cells from Stem Cells. ADULT STEM CELLS 2014. [DOI: 10.1007/978-1-4614-9569-7_6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
28
|
Adherent culture conditions enrich the side population obtained from the cochlear modiolus-derived stem/progenitor cells. Int J Pediatr Otorhinolaryngol 2013; 77:779-84. [PMID: 23489885 DOI: 10.1016/j.ijporl.2013.02.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 02/07/2013] [Accepted: 02/11/2013] [Indexed: 12/28/2022]
Abstract
OBJECTIVES Previously, our group reported that sphere-forming cells derived from the organ of Corti represent the stem/progenitor cells (SPCs) of the cochlea due to their properties of self-renewal and multipotency. However, long-term propagation of sphere-forming cells under suspension culture conditions may fail to maintain the characteristic stemness of these cells. Therefore, this study investigated whether an adherent culture system would be beneficial in terms of preserving more stem-like cells for long-term manipulations in vitro. METHODS Isolated modiolus-derived SPCs were placed on poly-d-lysine-coated petri dishes to form the so-called "adherent" culture system. RESULTS Modiolus SPCs cultured under adherent conditions exhibited a significantly increased percentage of cells with the side population (SP) phenotype (18.6%) compared with cells cultured under conventional suspension culture conditions (0.8%). Even after repeated passages, modiolus SPCs cultured under adherent culture conditions preserved more SP phenotype cells. In comparison with the non-SP phenotype cells, the sorted SP cells exhibited more stem-like but less differentiated properties, with an upregulated expression of the ATP-binding cassette subfamily G member 2 (ABCG2), Nestin, Sox2, and Nanog proteins. Furthermore, Retinoic acid (RA) treatment confirmed the expression of the multipotent differentiation markers in the SP cells, including TUJ1, pancytokeratin, glial fibrillary acidic protein (GFAP), and p27(Kip1). CONCLUSION Employment of an adherent culture system, instead of a suspension culture system, resulted in the enrichment of the SP cells from SPCs while retaining their stemness and multipotency.
Collapse
|
29
|
Jongkamonwiwat N, Rivolta MN. The Development of a Stem Cell Therapy for Deafness. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
30
|
Lou XX, Nakagawa T, Ohnishi H, Nishimura K, Ito J. Otospheres derived from neonatal mouse cochleae retain the progenitor cell phenotype after ex vivo expansions. Neurosci Lett 2012; 534:18-23. [PMID: 23238450 DOI: 10.1016/j.neulet.2012.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 10/16/2012] [Accepted: 12/03/2012] [Indexed: 12/23/2022]
Abstract
Because of their limited regenerative potential, cochlear hair cell loss is one of the major causes of permanent hearing loss in mammals. However, recent studies have shown that postnatal cochlear epithelia retain the progenitor cells that form otospheres. Otospheres are capable of self-renewing and differentiating into inner ear cell lineages, thereby suggesting a promising source for hair cell regeneration. We investigated retention of the progenitor cell phenotype in otospheres after ex vivo expansion, which is crucial for transplantation approaches. Reverse transcriptase-polymerase chain reaction and immunocytochemical analyses showed that otospheres derived from neonatal mice retained expression of stem and cochlear cell markers. After in vitro differentiation, otosphere-consisting cells differentiated into hair cell phenotypes after ex vivo expansion. However, the capacity of otospheres for self-renewal weakened with subsequent generations of ex vivo expansion. Our results indicate that ex vivo expanded-otospheres are useful experimental tools for studying hair cell regeneration in transplantation approaches and that the mechanisms for retention of the progenitor cell phenotype in otospheres should be investigated.
Collapse
Affiliation(s)
- Xiang-Xin Lou
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | | | | | | | | |
Collapse
|
31
|
Postnatal development, maturation and aging in the mouse cochlea and their effects on hair cell regeneration. Hear Res 2012; 297:68-83. [PMID: 23164734 DOI: 10.1016/j.heares.2012.11.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 10/22/2012] [Accepted: 11/07/2012] [Indexed: 12/23/2022]
Abstract
The organ of Corti in the mammalian inner ear is comprised of mechanosensory hair cells (HCs) and nonsensory supporting cells (SCs), both of which are believed to be terminally post-mitotic beyond late embryonic ages. Consequently, regeneration of HCs and SCs does not occur naturally in the adult mammalian cochlea, though recent evidence suggests that these cells may not be completely or irreversibly quiescent at earlier postnatal ages. Furthermore, regenerative processes can be induced by genetic and pharmacological manipulations, but, more and more reports suggest that regenerative potential declines as the organ of Corti continues to age. In numerous mammalian systems, such effects of aging on regenerative potential are well established. However, in the cochlea, the problem of regeneration has not been traditionally viewed as one of aging. This is an important consideration as current models are unable to elicit widespread regeneration or full recovery of function at adult ages yet regenerative therapies will need to be developed specifically for adult populations. Still, the advent of gene targeting and other genetic manipulations has established mice as critically important models for the study of cochlear development and HC regeneration and suggests that auditory HC regeneration in adult mammals may indeed be possible. Thus, this review will focus on the pursuit of regeneration in the postnatal and adult mouse cochlea and highlight processes that occur during postnatal development, maturation, and aging that could contribute to an age-related decline in regenerative potential. Second, we will draw upon the wealth of knowledge pertaining to age related senescence in tissues outside of the ear to synthesize new insights and potentially guide future research aimed at promoting HC regeneration in the adult cochlea.
Collapse
|
32
|
Senthilan PR, Piepenbrock D, Ovezmyradov G, Nadrowski B, Bechstedt S, Pauls S, Winkler M, Möbius W, Howard J, Göpfert MC. Drosophila auditory organ genes and genetic hearing defects. Cell 2012; 150:1042-54. [PMID: 22939627 DOI: 10.1016/j.cell.2012.06.043] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Revised: 03/02/2012] [Accepted: 06/20/2012] [Indexed: 12/22/2022]
Abstract
The Drosophila auditory organ shares equivalent transduction mechanisms with vertebrate hair cells, and both are specified by atonal family genes. Using a whole-organ knockout strategy based on atonal, we have identified 274 Drosophila auditory organ genes. Only four of these genes had previously been associated with fly hearing, yet one in five of the genes that we identified has a human cognate that is implicated in hearing disorders. Mutant analysis of 42 genes shows that more than half of them contribute to auditory organ function, with phenotypes including hearing loss, auditory hypersusceptibility, and ringing ears. We not only discover ion channels and motors important for hearing, but also show that auditory stimulus processing involves chemoreceptor proteins as well as phototransducer components. Our findings demonstrate mechanosensory roles for ionotropic receptors and visual rhodopsins and indicate that different sensory modalities utilize common signaling cascades.
Collapse
Affiliation(s)
- Pingkalai R Senthilan
- Department of Cellular Neurobiology, University of Göttingen, Julia-Lermontowa-Weg 3, 37077 Göttingen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Taniguchi M, Yamamoto N, Nakagawa T, Ogino E, Ito J. Identification of tympanic border cells as slow-cycling cells in the cochlea. PLoS One 2012; 7:e48544. [PMID: 23119055 PMCID: PMC3485350 DOI: 10.1371/journal.pone.0048544] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Accepted: 09/27/2012] [Indexed: 01/18/2023] Open
Abstract
Mammalian cochlear sensory epithelial cells are believed to possess minimal regenerative potential because they halt proliferation during late stage of embryogenesis and never regenerate after birth. This means that sensorineural hearing loss caused by the death of cochlear sensory epithelial cells is a permanent condition. However, stem cells were recently identified in neonatal mice following dissociation of their inner ear organs. This suggests that regenerative therapy for sensorineural hearing loss may be possible. Unfortunately, dissociation distorts the microanatomy of the inner ear, making it difficult to determine the precise location of stem cells in unaltered specimens. To develop new therapeutic approaches based on sensory epithelial cell regeneration, the location of these stem cells must be elucidated. Stem cells normally proliferate at a slow rate in adult organs. In fact, so-called label-retaining cells, or slow-cycling cells, of the brain and skin are recognized as stem cells. In this study, using the exogenous proliferation marker, 5′-bromo-2′-deoxyuridine (BrdU) in combination with the endogenous proliferation marker Ki-67, we identified tympanic border cells. These cells, which are located beneath the basilar membrane in vivo, represent slow-cycling cells of the murine cochlea. Immunohistochemically, these cells stained positive for the immature cell marker Nestin. But it will be difficult to achieve regeneration of the cochlear function because these slow-cycling cells disappear in the mature murine cochlea.
Collapse
Affiliation(s)
- Mirei Taniguchi
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Norio Yamamoto
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- * E-mail:
| | - Takayuki Nakagawa
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Eriko Ogino
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Juichi Ito
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
34
|
Transcriptomic analysis of the developing and adult mouse cochlear sensory epithelia. PLoS One 2012; 7:e42987. [PMID: 22900075 DOI: 10.1371/journal.pone.0042987] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 07/16/2012] [Indexed: 01/13/2023] Open
Abstract
The adult mammalian cochlea lacks regenerative ability and the irreversible degeneration of cochlear sensory hair cells leads to permanent hearing loss. Previous data show that early postnatal cochlea harbors stem/progenitor-like cells and shows a limited regenerative/repair capacity. These properties are progressively lost later during the postnatal development. Little is known about the genes and pathways that are potentially involved in this difference of the regenerative/repair potentialities between early postnatal and adult mammalian cochlear sensory epithelia (CSE). The goal of our study is to investigate the transcriptomic profiles of these two stages. We used Mouse Genome 430 2.0 microarray to perform an extensive analysis of the genes expressed in mouse postnatal day-3 (P3) and adult CSE. Statistical analysis of microarray data was performed using SAM (Significance Analysis of Microarrays) software. We identified 5644 statistically significant differentially expressed transcripts with a fold change (FC) >2 and a False Discovery Rate (FDR) ≤0.05. The P3 CSE signature included 3,102 transcripts, among which were known genes in the cochlea, but also new transcripts such as, Hmga2 (high mobility group AT-hook 2) and Nrarp (Notch-regulated ankyrin repeat protein). The adult CSE overexpressed 2,542 transcripts including new transcripts, such as Prl (Prolactin) and Ar (Androgen receptor), that previously were not known to be expressed in the adult cochlea. Our comparative study revealed important genes and pathways differentially expressed between the developing and adult CSE. The identification of new candidate genes would be useful as potential markers of the maintenance or the loss of stem cells and regenerative/repair ability during mammalian cochlear development.
Collapse
|
35
|
Unno K, Jain M, Liao R. Cardiac side population cells: moving toward the center stage in cardiac regeneration. Circ Res 2012; 110:1355-63. [PMID: 22581921 DOI: 10.1161/circresaha.111.243014] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Over the past decade, extensive work in animal models and humans has identified the presence of adult cardiac progenitor cells, capable of cardiomyogenic differentiation and likely contributors to cardiomyocyte turnover during normal development and disease. Among cardiac progenitor cells, there is a distinct subpopulation, termed "side population" (SP) progenitor cells, identified by their unique ability to efflux DNA binding dyes through an ATP-binding cassette transporter. This review highlights the literature on the isolation, characterization, and functional relevance of cardiac SP cells. We review the initial discovery of cardiac SP cells in adult myocardium as well as their capacity for functional cardiomyogenic differentiation and role in cardiac regeneration after myocardial injury. Finally, we discuss recent advances in understanding the molecular regulators of cardiac SP cell proliferation and differentiation, as well as likely future areas of investigation required to realize the goal of effective cardiac regeneration.
Collapse
Affiliation(s)
- Kazumasa Unno
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
36
|
Ooka H, Kanda S, Okazaki H, Suzuki H, Mishima K, Saito I, Yagi M, Tomoda K, Nishiyama T. Characterization of side population (SP) cells in murine cochlear nucleus. Acta Otolaryngol 2012; 132:693-701. [PMID: 22667338 DOI: 10.3109/00016489.2012.657358] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
CONCLUSION We characterized side population (SP) cells in the cochlear nucleus (CN). Some genes of stem/progenitor markers in sorted SP cells were identified by microarray analysis and RT-PCR. Furthermore, some cells in the CN also demonstrated self-renewal and clonal expansion activities. These results suggest that tissue stem/ progenitor like cells would be identified and characterized as a slow cycling and immaturity in SP cells of CN. OBJECTIVES SP cells were sorted and characterized as regards their activity in the CN in order to identify the tissue progenitor/stem cells in the auditory nervous system. METHODS Bromodeoxyuridine (BrdU)-injected mice were prepared and the long-term BrdU-retaining cells were detected by flow cytometry. Gene expression of SP and MP cells was analyzed by microarray analysis and RT-PCR. SP cells were cultured in conditioned medium to expand stem/progenitor cells in vitro and to estimate the spheroid-forming activity of stem cells. RESULTS In all, 1% of cells in the CN were detected as BrdU-positive. SP cells were detected at a frequency of 4.4% and expressed stem/progenitor markers, Abcb1b, Abcg2, Sca1, Notch1, Notch4, Hes1, and Jag1 in microarray analysis. Expression of Abcb1b, Abcg2, Sca1,Oct3/4, and Sox2 as determined by RT-PCR was supported by the microarray data. CN cells also had sphere-forming activity in young mice, but this activity was decreased by aging.
Collapse
Affiliation(s)
- Hisashi Ooka
- Regeneration Research Center for Intractable Diseases, Kansai Medical University, Osaka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Waldhaus J, Cimerman J, Gohlke H, Ehrich M, Müller M, Löwenheim H. Stemness of the organ of Corti relates to the epigenetic status of Sox2 enhancers. PLoS One 2012; 7:e36066. [PMID: 22570694 PMCID: PMC3343037 DOI: 10.1371/journal.pone.0036066] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 03/30/2012] [Indexed: 12/11/2022] Open
Abstract
In the adult mammalian auditory epithelium, the organ of Corti, loss of sensory hair cells results in permanent hearing loss. The underlying cause for the lack of regenerative response is the depletion of otic progenitors in the cell pool of the sensory epithelium. Here, we show that an increase in the sequence-specific methylation of the otic Sox2 enhancers NOP1 and NOP2 is correlated with a reduced self-renewal potential in vivo and in vitro; additionally, the degree of methylation of NOP1 and NOP2 is correlated with the dedifferentiation potential of postmitotic supporting cells into otic stem cells. Thus, the stemness the organ of Corti is related to the epigenetic status of the otic Sox2 enhancers. These observations validate the continued exploration of treatment strategies for dedifferentiating or reprogramming of differentiated supporting cells into progenitors to regenerate the damaged organ of Corti.
Collapse
Affiliation(s)
- Jörg Waldhaus
- Department of Otorhinolaryngology, Head and Neck Surgery, Hearing Research Center Tübingen, University of Tübingen Medical Center, Tübingen, Germany
| | - Jelka Cimerman
- Department of Otorhinolaryngology, Head and Neck Surgery, Hearing Research Center Tübingen, University of Tübingen Medical Center, Tübingen, Germany
| | | | - Mathias Ehrich
- SEQUENOM Inc., San Diego, California, United States of America
| | - Marcus Müller
- Department of Otorhinolaryngology, Head and Neck Surgery, Hearing Research Center Tübingen, University of Tübingen Medical Center, Tübingen, Germany
| | - Hubert Löwenheim
- Department of Otorhinolaryngology, Head and Neck Surgery, Hearing Research Center Tübingen, University of Tübingen Medical Center, Tübingen, Germany
- * E-mail:
| |
Collapse
|
38
|
Xu F, Wang HJ, Ma D. [Epigenetics--a new perspective for the study of deafness]. YI CHUAN = HEREDITAS 2012; 34:253-9. [PMID: 22425943 DOI: 10.3724/sp.j.1005.2012.00253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Hearing loss is the most common sensory disorder in humans. The newborn morbidity is 1/1000 approximately 3/1000. In most cases, the cause comes from abnormal development of inner-ear or degeneration of the cochlear hair cells. Genetic factors make a significant contribution to hearing impairment. Some genes and chromosome locus responsible for syndromic or non-syndromic hearing loss have been identified. However, etiology of deafness still remains obscure. In addition to some hot spot mutations (GJB2, SLC26A4, mitochondrial DNA C1494T, A1555G, etc.), epigenetics may also provide a significant contribution to this sensory disease. For example, miR-96 seed region mutations can result in progressive hearing loss in humans and mice, and aberrant CpG methylation has been linked to a few inherited syndromes that can induce hearing loss, etc.. This review aims to summarize the research progress of epigenetics in the fields of hearing and deafness.
Collapse
Affiliation(s)
- Fei Xu
- Key Laboratory of Molecular Medicine, Ministry of Education, Shanghai Medical College, Fudan University, China.
| | | | | |
Collapse
|
39
|
White PM, Stone JS, Groves AK, Segil N. EGFR signaling is required for regenerative proliferation in the cochlea: conservation in birds and mammals. Dev Biol 2012; 363:191-200. [PMID: 22230616 DOI: 10.1016/j.ydbio.2011.12.035] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 12/19/2011] [Accepted: 12/22/2011] [Indexed: 11/27/2022]
Abstract
Proliferation and transdifferentiaton of supporting cells in the damaged auditory organ of birds lead to robust regeneration of sensory hair cells. In contrast, regeneration of lost auditory hair cells does not occur in deafened mammals, resulting in permanent hearing loss. In spite of this failure of regeneration in mammals, we have previously shown that the perinatal mouse supporting cells harbor a latent potential for cell division. Here we show that in a subset of supporting cells marked by p75, EGFR signaling is required for proliferation, and this requirement is conserved between birds and mammals. Purified p75+ mouse supporting cells express receptors and ligands for the EGF signaling pathway, and their proliferation in culture can be blocked with the EGFR inhibitor AG1478. Similarly, in cultured chicken basilar papillae, supporting cell proliferation in response to hair cell ablation requires EGFR signaling. In addition, we show that EGFR signaling in p75+ mouse supporting cells is required for the down-regulation of the cell cycle inhibitor p27(Kip1) (CDKN1b) to enable cell cycle re-entry. Taken together, our data suggest that a conserved mechanism involving EGFR signaling governs proliferation of auditory supporting cells in birds and mammals and may represent a target for future hair cell regeneration strategies.
Collapse
Affiliation(s)
- Patricia M White
- Division of Cell Biology and Genetics, House Research Institute, 2100 W 3rd St., Los Angeles, CA 90057, USA
| | | | | | | |
Collapse
|
40
|
Sinkkonen ST, Chai R, Jan TA, Hartman BH, Laske RD, Gahlen F, Sinkkonen W, Cheng AG, Oshima K, Heller S. Intrinsic regenerative potential of murine cochlear supporting cells. Sci Rep 2011; 1:26. [PMID: 22355545 PMCID: PMC3216513 DOI: 10.1038/srep00026] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Accepted: 06/17/2011] [Indexed: 12/17/2022] Open
Abstract
The lack of cochlear regenerative potential is the main cause for the permanence of hearing loss. Albeit quiescent in vivo, dissociated non-sensory cells from the neonatal cochlea proliferate and show ability to generate hair cell-like cells in vitro. Only a few non-sensory cell-derived colonies, however, give rise to hair cell-like cells, suggesting that sensory progenitor cells are a subpopulation of proliferating non-sensory cells. Here we purify from the neonatal mouse cochlea four different non-sensory cell populations by fluorescence-activated cell sorting (FACS). All four populations displayed proliferative potential, but only lesser epithelial ridge and supporting cells robustly gave rise to hair cell marker-positive cells. These results suggest that cochlear supporting cells and cells of the lesser epithelial ridge show robust potential to de-differentiate into prosensory cells that proliferate and undergo differentiation in similar fashion to native prosensory cells of the developing inner ear.
Collapse
Affiliation(s)
- Saku T Sinkkonen
- Department of Otolaryngology - Head & Neck Surgery, Stanford University School of Medicine, Stanford CA 94305, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
The Development of a Stem Cell Therapy for Deafness. Regen Med 2011. [DOI: 10.1007/978-90-481-9075-1_27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
42
|
Chen HC, Sytwu HK, Chang JL, Wang HW, Chen HK, Kang BH, Liu DW, Chen CH, Chao TT, Wang CH. Hypoxia enhances the stemness markers of cochlear stem/progenitor cells and expands sphere formation through activation of hypoxia-inducible factor-1 alpha. Hear Res 2010; 275:43-52. [PMID: 21147209 DOI: 10.1016/j.heares.2010.12.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Revised: 11/22/2010] [Accepted: 12/01/2010] [Indexed: 10/18/2022]
Abstract
Unlike neural stem cells that maintain populations in the adult brains of both rodents and humans, cochlear stem cells appear to diminish in number after birth and may become quiescent in adult mammalian cochleae. Hypoxia has been observed to promote an undifferentiated cell state in various stem cell populations; however, little is known about such an effect on cochlear stem/progenitor cells (SPCs). The aims of this study were to assess the effect of hypoxia on cochlear SPCs and to examine the impact of hypoxia-inducible factor-1 alpha (Hif-1a) on regulating such an effect. Our data demonstrate that hypoxic culturing for 24 h significantly increased sphere formation and viability of cochlear SPCs compared with those cultured under normoxic conditions. Concurrent with these proliferation promotion effects are changes in the expression of multiple stemness and cell-cycle quiescent associated gene targets, including Abcg2, nestin, p27(Kip1)and Vegf. Knockdown of Hif-1a expression by small-interfering RNA inhibited hypoxia-induced cochlear SPC expansion and resulted in downregulation of Vegf, Abcg2, and nestin and upregulation of p27(Kip1) gene expression. These results suggest that Hif-1a plays an important role in the stimulation of the proliferation of cochlear SPCs, which confers a great benefit of expanding cochlear SPCs via hypoxic conditions.
Collapse
Affiliation(s)
- Hsin-Chien Chen
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC; Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Oiticica J, Barboza-Junior LCM, Batissoco AC, Lezirovitz K, Mingroni-Netto RC, Haddad LA, Bento RF. Retention of progenitor cell phenotype in otospheres from guinea pig and mouse cochlea. J Transl Med 2010; 8:119. [PMID: 21087511 PMCID: PMC3001427 DOI: 10.1186/1479-5876-8-119] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2010] [Accepted: 11/18/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Culturing otospheres from dissociated organ of Corti is an appropriate starting point aiming at the development of cell therapy for hair cell loss. Although guinea pigs have been widely used as an excellent experimental model for studying the biology of the inner ear, the mouse cochlea has been more suitable for yielding otospheres in vitro. The aim of this study was to compare conditions and outcomes of otosphere suspension cultures from dissociated organ of Corti of either mouse or guinea pig at postnatal day three (P3), and to evaluate the guinea pig as a potential cochlea donor for preclinical cell therapy. METHODS Organs of Corti were surgically isolated from P3 guinea pig or mouse cochlea, dissociated and cultivated under non-adherent conditions. Cultures were maintained in serum-free DMEM:F12 medium, supplemented with epidermal growth factor (EGF) plus either basic fibroblast growth factor (bFGF) or transforming growth factor alpha (TGFα). Immunofluorescence assays were conducted for phenotype characterization. RESULTS The TGFα group presented a number of spheres significantly higher than the bFGF group. Although mouse cultures yielded more cells per sphere than guinea pig cultures, sox2 and nestin distributed similarly in otosphere cells from both organisms. We present evidence that otospheres retain properties of inner ear progenitor cells such as self-renewal, proliferation, and differentiation into hair cells or supporting cells. CONCLUSIONS Dissociated guinea pig cochlea produced otospheres in vitro, expressing sox2 and nestin similarly to mouse otospheres. Our data is supporting evidence for the presence of inner ear progenitor cells in the postnatal guinea pig. However, there is limited viability for these cells in neonatal guinea pig cochlea when compared to the differentiation potential observed for the mouse organ of Corti at the same developmental stage.
Collapse
Affiliation(s)
- Jeanne Oiticica
- Department of Otolaryngology, Medical School, University of São Paulo, São Paulo, Brasil.
| | | | | | | | | | | | | |
Collapse
|
44
|
Carricondo F, Iglesias MC, Rodríguez F, Poch-Broto J, Gil-Loyzaga P. In vitro long-term development of cultured inner ear stem cells of newborn rat. Cell Tissue Res 2010; 342:13-9. [PMID: 20838813 DOI: 10.1007/s00441-010-1039-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Accepted: 08/16/2010] [Indexed: 11/30/2022]
Abstract
The adult mammalian auditory receptor lacks any ability to repair and/or regenerate after injury. However, the late developing cochlea still contains some stem-cell-like elements that might be used to regenerate damaged neurons and/or cells of the organ of Corti. Before their use in any application, stem cell numbers need to be amplified because they are usually rare in late developing and adult tissues. The numerous re-explant cultures required for the progressive amplification process can result in a spontaneous differentiation process. This aspect has been implicated in the tumorigenicity of stem cells when transplanted into a tissue. The aim of this study has been to determine whether cochlear stem cells can proliferate and differentiate spontaneously in long-term cultures without the addition of any factor that might influence these processes. Cochlear stem cells, which express nestin protein, were cultured in monolayers and fed with DMEM containing 5% FBS. They quickly organized themselves into typical spheres exhibiting a high proliferation rate, self-renewal property, and differentiation ability. Secondary cultures of these stem cell spheres spontaneously differentiated into neuroectodermal-like cells. The expression of nestin, glial-fibrillary-acidic protein, vimentin, and neurofilaments was evaluated to identify early differentiation. Nestin expression appeared in primary and secondary cultures. Other markers were also identified in differentiating cells. Further research might demonstrate the spontaneous differentiation of cochlear stem cells and their teratogenic probability when they are used for transplantation.
Collapse
Affiliation(s)
- Francisco Carricondo
- Department of Ophtalmology and Otorhinolaryngology, Faculty of Medicine, Complutense University of Madrid, Apartado de Correos 60.075, 28080 Madrid, Spain
| | | | | | | | | |
Collapse
|
45
|
Smeti I, Savary E, Capelle V, Hugnot JP, Uziel A, Zine A. Expression of candidate markers for stem/progenitor cells in the inner ears of developing and adult GFAP and nestin promoter-GFP transgenic mice. Gene Expr Patterns 2010; 11:22-32. [PMID: 20817025 DOI: 10.1016/j.gep.2010.08.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Revised: 08/20/2010] [Accepted: 08/30/2010] [Indexed: 01/08/2023]
Abstract
Loss of hair cells in the mammalian cochlea leads to permanent sensori-neural hearing loss. Hair cells degenerate and their places are taken by phalangeal scars formed by non-sensory supporting cells. Current data indicate that early postnatal post-mitotic supporting cells can proliferate and differentiate into hair cell-like cells in culture. In this study, we used GFAP and nestin promoter-GFP transgenic mice in combination with other stem cell markers to characterize supporting cell subtypes in the postnatal day-3 (P3) and adult organs of Corti with potential stem/progenitor cell phenotype. In P3 organ of Corti, we show GFAP-GFP signal in all the supporting cell subtypes while the nestin-GFP was restricted to the supporting cells in the inner hair cell area. At this stage, GFAP and selected stem/progenitor markers displayed overlapping expression pattern in the supporting cell population. In the adult, GFAP expression is down-regulated from the supporting cells in the outer hair cell area and nestin expression is down-regulated in the supporting cells of the inner hair cell area. Sox2 and Jagged1 expression is maintained in the mature supporting cells, while Abcg2 was down-regulated in these cells. In contrast, GFAP and Abcg2 expression was up-regulated in the inner sulcus limbal cells outside the mature organ of Corti's area. Using quantitative reverse transcription-PCR, we found a decrease in transcripts for Jagged1 and Sox2 in adult cochleae. Our findings suggest that the loss of regenerative capacity of the adult organ of Corti is related to down-regulation of stem/progenitor key-markers from the mature supporting cells.
Collapse
Affiliation(s)
- Ibtihel Smeti
- INSERM U583, Institute for Neurosciences of Montpellier, University of Montpellier I, 34091 Montpellier, France
| | | | | | | | | | | |
Collapse
|
46
|
Bodson M, Breuskin I, Lefebvre P, Malgrange B. Hair cell progenitors: identification and regulatory genes. Acta Otolaryngol 2010. [DOI: 10.3109/00016480903121057] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
47
|
MicroRNAs and epigenetic regulation in the mammalian inner ear: implications for deafness. Mamm Genome 2009; 20:581-603. [PMID: 19876605 DOI: 10.1007/s00335-009-9230-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Accepted: 09/30/2009] [Indexed: 01/19/2023]
Abstract
Sensorineural hearing loss is the most common sensory disorder in humans and derives, in most cases, from inner-ear defects or degeneration of the cochlear sensory neuroepithelial hair cells. Genetic factors make a significant contribution to hearing impairment. While mutations in 51 genes have been associated with hereditary sensorineural nonsyndromic hearing loss (NSHL) in humans, the responsible mutations in many other chromosomal loci linked with NSHL have not been identified yet. Recently, mutations in a noncoding microRNA (miRNA) gene, MIR96, which is expressed specifically in the inner-ear hair cells, were linked with progressive hearing loss in humans and mice. Furthermore, additional miRNAs were found to have essential roles in the development and survival of inner-ear hair cells. Epigenetic mechanisms, in particular, DNA methylation and histone modifications, have also been implicated in human deafness, suggesting that several layers of noncoding genes that have never been studied systematically in the inner-ear sensory epithelia are required for normal hearing. This review aims to summarize the current knowledge about the roles of miRNAs and epigenetic regulatory mechanisms in the development, survival, and function of the inner ear, specifically in the sensory epithelia, tectorial membrane, and innervation, and their contribution to hearing.
Collapse
|
48
|
Chen W, Johnson SL, Marcotti W, Andrews PW, Moore HD, Rivolta MN. Human fetal auditory stem cells can be expanded in vitro and differentiate into functional auditory neurons and hair cell-like cells. Stem Cells 2009; 27:1196-204. [PMID: 19418454 DOI: 10.1002/stem.62] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In the quest to develop the tools necessary for a cell-based therapy for deafness, a critical step is to identify a suitable stem cell population. Moreover, the lack of a self-renovating model system for the study of cell fate determination in the human cochlea has impaired our understanding of the molecular events involved in normal human auditory development. We describe here the identification and isolation of a population of SOX2+OCT4+ human auditory stem cells from 9-week-old to 11-week-old fetal cochleae (hFASCs). These cells underwent long-term expansion in vitro and retained their capacity to differentiate into sensory hair cells and neurons, whose functional and electrophysiological properties closely resembled their in vivo counterparts during development. hFASCs, and the differentiating protocols defined here, could be used to study developing human cochlear neurons and hair cells, as models for drug screening and toxicity and may facilitate the development of cell-based therapies for deafness.
Collapse
Affiliation(s)
- Wei Chen
- Centre for Stem Cell Biology, University of Sheffield, Sheffield, UK
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
Hearing loss is a global health problem with profound socioeconomic impact. We contend that acquired hearing loss is mainly a modern disorder caused by man-made noise and modern drugs, among other causes. These factors, combined with increasing lifespan, have exposed a deficit in cochlear self-regeneration that was irrelevant for most of mammalian evolution. Nevertheless, the mammalian cochlea has evolved from phylogenetically older structures, which do have the capacity for self-repair. Moreover, nonmammalian vertebrates can regenerate auditory hair cells that restore sensory function. We will offer a critical perspective on recent advances in stem cell biology, gene therapy, cell cycle regulation and pharmacotherapeutics to define and validate regenerative medical interventions for mammalian hair cell loss. Although these advances are promising, we are only beginning to fully appreciate the complexity of the many challenges that lie ahead.
Collapse
Affiliation(s)
- John V Brigande
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | | |
Collapse
|
50
|
Diensthuber M, Oshima K, Heller S. Stem/progenitor cells derived from the cochlear sensory epithelium give rise to spheres with distinct morphologies and features. J Assoc Res Otolaryngol 2009; 10:173-90. [PMID: 19247714 DOI: 10.1007/s10162-009-0161-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2008] [Accepted: 02/05/2009] [Indexed: 11/25/2022] Open
Abstract
Nonmammalian vertebrates regenerate lost sensory hair cells by means of asymmetric division of supporting cells. Inner ear or lateral line supporting cells in birds, amphibians, and fish consequently serve as bona fide stem cells resulting in high regenerative capacity of hair cell-bearing organs. Hair cell regeneration does not happen in the mammalian cochlea, but cells with proliferative capacity can be isolated from the neonatal cochlea. These cells have the ability to form clonal floating colonies, so-called spheres, when cultured in nonadherent conditions. We noticed that the sphere population derived from mouse cochlear sensory epithelium cells was heterogeneous, consisting of morphologically distinct sphere types, hereby classified as solid, transitional, and hollow. Cochlear sensory epithelium-derived stem/progenitor cells initially give rise to small solid spheres, which subsequently transition into hollow spheres, a change that is accompanied by epithelial differentiation of the majority of sphere cells. Only solid spheres, and to a lesser extent, transitional spheres, appeared to harbor self-renewing stem cells, whereas hollow spheres could not be consistently propagated. Solid spheres contained significantly more rapidly cycling Pax-2-expressing presumptive otic progenitor cells than hollow spheres. Islet-1, which becomes upregulated in nascent sensory patches, was also more abundant in solid than in hollow spheres. Likewise, hair cell-like cells, characterized by the expression of multiple hair cell markers, differentiated in significantly higher numbers in cell populations derived from solid spheres. We conclude that cochlear sensory epithelium cell populations initially give rise to small solid spheres that have self-renewing capacity before they subsequently convert into hollow spheres, a process that is accompanied by loss of stemness and reduced ability to spontaneously give rise to hair cell-like cells. Solid spheres might, therefore, represent the most suitable sphere type for cell-based assays or animal model transplantation studies aimed at development of cell replacement therapies.
Collapse
Affiliation(s)
- Marc Diensthuber
- Departments of Otolaryngology-Head & Neck Surgery and Molecular & Cellular Physiology, School of Medicine, Stanford University, 801 Welch Road, Stanford, CA 94305-5739, USA
| | | | | |
Collapse
|