1
|
Li XJ, Morgan C, Li L, Zhang WY, Chrysostomou E, Doetzlhofer A. The Notch ligand Jagged1 plays a dual role in cochlear hair cell regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.02.640998. [PMID: 40093047 PMCID: PMC11908178 DOI: 10.1101/2025.03.02.640998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Hair cells (HCs) within the inner ear cochlea are specialized mechanoreceptors required for hearing. Cochlear HCs are not regenerated in mammals, and their loss is a leading cause of deafness in humans. Cochlear supporting cells (SCs) in newborn mice have the capacity to regenerate HCs, but persistent Notch signaling, presumably activated by SC-specific Notch ligand Jagged1 (JAG1), prevents SCs from converting into HCs. Here, employing an organoid platform, we show that while JAG1 participates in HC-fate repression, JAG1's primary function is to preserve the "progenitor-like characteristics" of cochlear SCs. Transcriptomic and mechanistic studies reveal that JAG1/Notch signaling maintains the expression of progenitor and metabolic genes in cochlear SCs and sustains pro-growth pathways, including PI3K-Akt-mTOR signaling, a function that is mediated by Notch1 and Notch2. Finally, we show that JAG1/Notch signaling stimulation with JAG1-Fc peptide enhances the HC-forming capacity of cochlear SCs undergoing maturation in cochlear explants and in vivo .
Collapse
|
2
|
Deletion of Notch1 during Cochlear Maturation Leads to Rapid Supporting Cell Death and Profound Deafness. J Neurosci 2023; 43:199-210. [PMID: 36418183 PMCID: PMC9838715 DOI: 10.1523/jneurosci.1090-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/14/2022] [Accepted: 11/08/2022] [Indexed: 11/25/2022] Open
Abstract
The sensory region of the mammalian hearing organ contains two main cell types-hair cells and supporting cells. During development, Notch signaling plays an important role in whether a cell becomes either a hair cell or supporting cell by mediating lateral inhibition. However, once the cell fate decisions have been determined, little is understood about the role Notch plays in cochlear maturation. Here, we report that deletion of Notch1 from the early postnatal mouse cochlea in both male and female animals resulted in profound deafness at 6 weeks of age. Histologic analyses at 6 weeks revealed significant hair cell and supporting cell loss throughout the Notch1-deficient cochlea. Early analyses revealed a reduction in supporting cells in the outer hair cell region between postnatal day (P) 2 and P6, without a comparable increase in outer hair cell number, suggesting a mechanism other than lateral inhibition. Consistent with this, we found apoptotic cells in the outer supporting cell region of the cochlea at P1 and P2, indicating that Notch1 is required for outer supporting cell survival during early cochlear maturation. Interestingly, inner supporting cell types were not lost after Notch1 deletion. Surprisingly, we do not detect outer hair cell loss in Notch1 mutants until after the onset of hearing, around P14, suggesting that hair cell loss is caused by loss of the supporting cells. Together, these results demonstrate that Notch1 is required for supporting cell survival during early maturation and that loss of these cells causes later loss of the hair cells and cochlear dysfunction.SIGNIFICANCE STATEMENT During development, Notch signaling has been shown to be critical in regulating the cell fate choices between hair cells and supporting cells. However, little is known about how Notch functions after those cell fate choices are made. Here, we examine the role of Notch1 in the maturing cochlea. We demonstrate that deletion of Notch1 results in profound deafness by 6 weeks of age. Histologic analyses revealed rapid supporting cell death shortly after Notch1 deletion, followed by eventual loss of the hair cells. These results reveal an unexpected role for Notch in supporting cell survival during cochlear maturation.
Collapse
|
3
|
Liu SS, Yang R. Inner Ear Drug Delivery for Sensorineural Hearing Loss: Current Challenges and Opportunities. Front Neurosci 2022; 16:867453. [PMID: 35685768 PMCID: PMC9170894 DOI: 10.3389/fnins.2022.867453] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/02/2022] [Indexed: 12/20/2022] Open
Abstract
Most therapies for treating sensorineural hearing loss are challenged by the delivery across multiple tissue barriers to the hard-to-access anatomical location of the inner ear. In this review, we will provide a recent update on various pharmacotherapy, gene therapy, and cell therapy approaches used in clinical and preclinical studies for the treatment of sensorineural hearing loss and approaches taken to overcome the drug delivery barriers in the ear. Small-molecule drugs for pharmacotherapy can be delivered via systemic or local delivery, where the blood-labyrinth barrier hinders the former and tissue barriers including the tympanic membrane, the round window membrane, and/or the oval window hinder the latter. Meanwhile, gene and cell therapies often require targeted delivery to the cochlea, which is currently achieved via intra-cochlear or intra-labyrinthine injection. To improve the stability of the biomacromolecules during treatment, e.g., RNAs, DNAs, proteins, additional packing vehicles are often required. To address the diverse range of biological barriers involved in inner ear drug delivery, each class of therapy and the intended therapeutic cargoes will be discussed in this review, in the context of delivery routes commonly used, delivery vehicles if required (e.g., viral and non-viral nanocarriers), and other strategies to improve drug permeation and sustained release (e.g., hydrogel, nanocarriers, permeation enhancers, and microfluidic systems). Overall, this review aims to capture the important advancements and key steps in the development of inner ear therapies and delivery strategies over the past two decades for the treatment and prophylaxis of sensorineural hearing loss.
Collapse
Affiliation(s)
- Sophie S. Liu
- Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, United States
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - Rong Yang
- Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, United States
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| |
Collapse
|
4
|
Chen Z, Liu Y, Liang R, Cui C, Zhu Y, Zhang F, Zhang J, Chen X. Comparative transcriptome analysis provides insights into the molecular mechanisms of high-frequency hearing differences between the sexes of Odorrana tormota. BMC Genomics 2022; 23:296. [PMID: 35410120 PMCID: PMC9004125 DOI: 10.1186/s12864-022-08536-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 04/07/2022] [Indexed: 11/15/2022] Open
Abstract
Background Acoustic communication is important for the survival and reproduction of anurans and masking background noise is a critical factor for their effective acoustic communication. Males of the concave-eared frog (Odorrana tormota) have evolved an ultrasonic communication capacity to avoid masking by the widespread background noise of local fast-flowing streams, whereas females exhibit no ultrasonic sensitivity. However, the molecular mechanisms underlying the high-frequency hearing differences between the sexes of O. tormota are still poorly understood. Results In this study, we sequenced the brain transcriptomes of male and female O. tormota, and compared their differential gene expression. A total of 4,605 differentially expressed genes (DEGs) between the sexes of O. tormota were identified and eleven of them were related to auditory based on the annotation and enrichment analysis. Most of these DEGs in males showed a higher expression trend than females in both quantity and expression quantity. The highly expressed genes in males were relatively concentrated in neurogenesis, signal transduction, ion transport and energy metabolism, whereas the up-expressed genes in females were mainly related to the growth and development regulation of specific auditory cells. Conclusions The transcriptome of male and female O. tormota has been sequenced and de novo assembled, which will provide gene reference for further genomic studies. In addition, this is the first research to reveal the molecular mechanisms of sex differences in ultrasonic hearing between the sexes of O. tormota and will provide new insights into the genetic basis of the auditory adaptation in amphibians during their transition from water to land. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08536-2.
Collapse
Affiliation(s)
- Zhuo Chen
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China.,The Observation and Research Field Station of Taihang Mountain Forest Ecosystems of Henan Province, Xinxiang, 453007, China
| | - Yao Liu
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| | - Rui Liang
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| | - Chong Cui
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| | - Yanjun Zhu
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| | - Fang Zhang
- College of Life Sciences, Anhui Normal University, Wuhu, 241000, China
| | - Jie Zhang
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China.
| | - Xiaohong Chen
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China. .,The Observation and Research Field Station of Taihang Mountain Forest Ecosystems of Henan Province, Xinxiang, 453007, China.
| |
Collapse
|
5
|
Wang S, Lee MP, Jones S, Liu J, Waldhaus J. Mapping the regulatory landscape of auditory hair cells from single-cell multi-omics data. Genome Res 2021; 31:1885-1899. [PMID: 33837132 DOI: 10.1101/gr.271080.120] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 03/23/2021] [Indexed: 11/25/2022]
Abstract
Auditory hair cells transduce sound to the brain and in mammals these cells reside together with supporting cells in the sensory epithelium of the cochlea, called the organ of Corti. To establish the organ's delicate function during development and differentiation, spatiotemporal gene expression is strictly controlled by chromatin accessibility and cell type-specific transcription factors, jointly representing the regulatory landscape. Bulk-sequencing technology and cellular heterogeneity obscured investigations on the interplay between transcription factors and chromatin accessibility in inner ear development. To study the formation of the regulatory landscape in hair cells, we collected single-cell chromatin accessibility profiles accompanied by single-cell RNA data from genetically labeled murine hair cells and supporting cells after birth. Using an integrative approach, we predicted cell type-specific activating and repressing functions of developmental transcription factors. Furthermore, by integrating gene expression and chromatin accessibility datasets, we reconstructed gene regulatory networks. Then, using a comparative approach, 20 hair cell-specific activators and repressors, including putative downstream target genes, were identified. Clustering of target genes resolved groups of related transcription factors and was utilized to infer their developmental functions. Finally, the heterogeneity in the single-cell data allowed us to spatially reconstruct transcriptional as well as chromatin accessibility trajectories, indicating that gradual changes in the chromatin accessibility landscape were lagging behind the transcriptional identity of hair cells along the organ's longitudinal axis. Overall, this study provides a strategy to spatially reconstruct the formation of a lineage specific regulatory landscape using a single-cell multi-omics approach.
Collapse
Affiliation(s)
- Shuze Wang
- University of Michigan, Kresge Hearing Research Institute
| | - Mary P Lee
- University of Michigan, Kresge Hearing Research Institute
| | - Scott Jones
- University of Michigan, Kresge Hearing Research Institute
| | | | - Joerg Waldhaus
- University of Michigan, Kresge Hearing Research Institute;
| |
Collapse
|
6
|
Toxic Effects of 3,3'-Iminodipropionitrile on Vestibular System in Adult C57BL/6J Mice In Vivo. Neural Plast 2020; 2020:1823454. [PMID: 32714382 PMCID: PMC7354661 DOI: 10.1155/2020/1823454] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 06/01/2020] [Accepted: 06/11/2020] [Indexed: 11/18/2022] Open
Abstract
The utricle is one of the five sensory organs in the mammalian vestibular system, and while the utricle has a limited ability to repair itself, this is not sufficient for the recovery of vestibular function after hair cell (HC) loss induced by ototoxic drugs. In order to further explore the possible self-recovery mechanism of the adult mouse vestibular system, we established a reliable utricle epithelium injury model for studying the regeneration of HCs and examined the toxic effects of 3,3'-iminodiproprionitrile (IDPN) on the utricle in vivo in C57BL/6J mice, which is one of the most commonly used strains in inner ear research. This work focused on the epithelial cell loss, vestibular dysfunction, and spontaneous cell regeneration after IDPN administration. HC loss and supporting cell (SC) loss after IDPN treatment was dose-dependent and resulted in dysfunction of the vestibular system, as indicated by the swim test and the rotating vestibular ocular reflex (VOR) test. EdU-positive SCs were observed only in severely injured utricles wherein above 47% SCs were dead. No EdU-positive HCs were observed in either control or injured utricles. RT-qPCR showed transient upregulation of Hes5 and Hey1 and fluctuating upregulation of Axin2 and β-catenin after IDPN administration. We conclude that a single intraperitoneal injection of IDPN is a practical way to establish an injured utricle model in adult C57BL/6J mice in vivo. We observed activation of Notch and Wnt signaling during the limited spontaneous HC regeneration after vestibular sensory epithelium damage, and such signaling might act as the promoting factors for tissue self-repair in the inner ear.
Collapse
|
7
|
Ogier JM, Lockhart PJ, Burt RA. Intravenously delivered aminoglycoside antibiotics, tobramycin and amikacin, are not ototoxic in mice. Hear Res 2020; 386:107870. [PMID: 31864009 DOI: 10.1016/j.heares.2019.107870] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/24/2019] [Accepted: 12/09/2019] [Indexed: 12/20/2022]
Abstract
Many drugs on the World Health Organization's list of critical medicines are ototoxic, destroying sensory hair cells within the ear. These drugs preserve life, but patients can experience side effects including permanent hearing loss and vestibular dysfunction. Aminoglycoside ototoxicity was first recognised 80 years ago. However, no preventative treatments have been developed. In order to develop such treatments, we must identify the factors driving hair cell death. In vivo, studies of cell death are typically conducted using mouse models. However, a robust model of aminoglycoside ototoxicity does not exist. Previous studies testing aminoglycoside delivery via intraperitoneal or subcutaneous injection have produced variable ototoxic effects in the mouse. As a result, surgical drug delivery to the rodent ear is often used to achieve ototoxicity. However, this technique does not accurately model clinical practice. In the clinic, aminoglycosides are administered to humans intravenously (i.v.). However, repeated i.v. delivery has not been reported in the mouse. This study evaluated whether repeated i.v. administration of amikacin or tobramycin would induce hearing loss. Daily i.v. injections over a two-week period were well tolerated and transient low frequency hearing loss was observed in the aminoglycoside treatment groups. However, the hearing changes observed did not mimic the high frequency patterns of hearing loss observed in humans. Our results indicate that the i.v. delivery of tobramycin or amikacin is not an effective technique for inducing ototoxicity in mice. This result is consistent with previously published reports indicating that the mouse cochlea is resistant to systemically delivered aminoglycoside ototoxicity.
Collapse
Affiliation(s)
- Jacqueline M Ogier
- Bruce Lefroy Centre, Murdoch Children's Research Institute, 50 Flemington Road, Parkville, VIC, 3052, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Paul J Lockhart
- Bruce Lefroy Centre, Murdoch Children's Research Institute, 50 Flemington Road, Parkville, VIC, 3052, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Rachel A Burt
- Bruce Lefroy Centre, Murdoch Children's Research Institute, 50 Flemington Road, Parkville, VIC, 3052, Australia; School of Biosciences, University of Melbourne, Parkville, VIC, 3010, Australia
| |
Collapse
|
8
|
Notch Signalling: The Multitask Manager of Inner Ear Development and Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1218:129-157. [DOI: 10.1007/978-3-030-34436-8_8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
9
|
Novel insights into inner ear development and regeneration for targeted hearing loss therapies. Hear Res 2019; 397:107859. [PMID: 31810596 DOI: 10.1016/j.heares.2019.107859] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/06/2019] [Accepted: 11/25/2019] [Indexed: 02/06/2023]
Abstract
Sensorineural hearing loss is the most common sensory deficit in humans. Despite the global scale of the problem, only limited treatment options are available today. The mammalian inner ear is a highly specialized postmitotic organ, which lacks proliferative or regenerative capacity. Since the discovery of hair cell regeneration in non-mammalian species however, much attention has been placed on identifying possible strategies to reactivate similar responses in humans. The development of successful regenerative approaches for hearing loss strongly depends on a detailed understanding of the mechanisms that control human inner ear cellular specification, differentiation and function, as well as on the development of robust in vitro cellular assays, based on human inner ear cells, to study these processes and optimize therapeutic interventions. We summarize here some aspects of inner ear development and strategies to induce regeneration that have been investigated in rodents. Moreover, we discuss recent findings in human inner ear development and compare the results with findings from animal models. Finally, we provide an overview of strategies for in vitro generation of human sensory cells from pluripotent and somatic progenitors that may provide a platform for drug development and validation of therapeutic strategies in vitro.
Collapse
|
10
|
Roccio M, Edge ASB. Inner ear organoids: new tools to understand neurosensory cell development, degeneration and regeneration. Development 2019; 146:146/17/dev177188. [PMID: 31477580 DOI: 10.1242/dev.177188] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The development of therapeutic interventions for hearing loss requires fundamental knowledge about the signaling pathways controlling tissue development as well as the establishment of human cell-based assays to validate therapeutic strategies ex vivo Recent advances in the field of stem cell biology and organoid culture systems allow the expansion and differentiation of tissue-specific progenitors and pluripotent stem cells in vitro into functional hair cells and otic-like neurons. We discuss how inner ear organoids have been developed and how they offer for the first time the opportunity to validate drug-based therapies, gene-targeting approaches and cell replacement strategies.
Collapse
Affiliation(s)
- Marta Roccio
- Inner Ear Research Laboratory, Department of Biomedical Research (DBMR), University of Bern, Bern 3008, Switzerland .,Department of Otorhinolaryngology, Head & Neck Surgery, Inselspital, Bern University Hospital, University of Bern, Bern 3010, Switzerland
| | - Albert S B Edge
- Department of Otolaryngology, Harvard Medical School, Boston, MA 02115, USA.,Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA.,Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
11
|
Horvath L, Bächinger D, Honegger T, Bodmer D, Naldi AM. Functional and morphological analysis of different aminoglycoside treatment regimens inducing hearing loss in mice. Exp Ther Med 2019; 18:1123-1130. [PMID: 31316607 PMCID: PMC6601143 DOI: 10.3892/etm.2019.7687] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 05/23/2019] [Indexed: 01/05/2023] Open
Abstract
Aminoglycoside ototoxicity is common in clinical practice but reliable protective agents currently do not exist. Aminoglycoside regimens causing ototoxicity in different laboratory animals are under investigation. The assessment method used most commonly to determine auditory effects is the auditory brainstem response (ABR). Distortion product otoacoustic emissions (DPOAE) have been used less frequently. A precise recommendation on the specific method to assess peripheral auditory function before and after aminoglycoside toxicity in mice does not exist. In order to evaluate various mouse models for ototoxic injury caused by various aminoglycoside regimens, there is a need for performing preliminary tests in small cohorts before large experiments. The aim of our study was to investigate different aminoglycoside regimens that cause substantial ototoxic damage in vivo. Aminoglycosides are safe and produce a detectable hearing threshold shift in a small cohort of mice that can be used as a model for preliminary tests. Different ototoxic regimens were assessed by ABR and DPOAE measurements pre- and post-treatment. Further, the sensory cell loss was quantified by counting hair cells in the cochlea. It was revealed that an ototoxic regimen with kanamycin twice daily for 15 consecutive days is safe, well tolerated and produces an early significant hearing threshold shift detected by DPOAE in a small cohort of mice. The study compared ABR and DPOAE in mentioned regimens for the first time and illustrated that DPOAE is well suited for detecting hearing threshold shifts in high frequencies before ABR threshold shifts occur in accordance with predominating outer hair cell damage mainly in the basal turn of the cochlea.
Collapse
Affiliation(s)
- Lukas Horvath
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland.,Department of Biomedicine and The Clinic for Otorhinolaryngology, University Hospital Basel, 4031 Basel, Switzerland
| | - David Bächinger
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Tim Honegger
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Daniel Bodmer
- Department of Biomedicine and The Clinic for Otorhinolaryngology, University Hospital Basel, 4031 Basel, Switzerland
| | - Arianne Monge Naldi
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
12
|
P2X 2 Receptor Deficiency in Mouse Vestibular End Organs Attenuates Vestibular Function. Neuroscience 2018; 386:41-50. [PMID: 29944897 DOI: 10.1016/j.neuroscience.2018.06.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 06/13/2018] [Accepted: 06/18/2018] [Indexed: 02/02/2023]
Abstract
P2X2 receptors are ligand-gated cation channels activated by extracellular ATP that modulate neural transmission in various neuronal systems. Although the function and distribution of P2X2 receptors in the cochlea portion of the inner ear are well established, their physiological role in the vestibular portion is still not understood. Therefore, we investigated P2X2 receptor localization in the peripheral vestibular portion, and assessed their physiological function in vivo using P2X2 receptor knock out (P2X2-KO) mice. Histological analysis revealed that P2X2 receptors were localized on the epithelial surface of supporting and transitional cells of the vestibular end organs. To examine vestibular function in P2X2-KO mice, we conducted behavioral tests and tested the vestibulo-ocular reflex (VOR) during sinusoidal rotations. P2X2-KO mice exhibited significant motor balance impairment in the balance beam test. VOR gain in P2X2-KO mice was significantly reduced, with no decrease in the optokinetic response. In conclusion, we showed that P2X2 receptors are mainly localized in the supporting cells of the vestibular inner ear, and the loss of P2X2 receptors causes mild vestibular dysfunction. Taken together, our findings suggest that the P2X2 receptor plays a modulatory role in vestibular function.
Collapse
|
13
|
Zhou W, Du J, Jiang D, Wang X, Chen K, Tang H, Zhang X, Cao H, Zong L, Dong C, Jiang H. microRNA‑183 is involved in the differentiation and regeneration of Notch signaling‑prohibited hair cells from mouse cochlea. Mol Med Rep 2018; 18:1253-1262. [PMID: 29901127 PMCID: PMC6072138 DOI: 10.3892/mmr.2018.9127] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 12/05/2017] [Indexed: 01/20/2023] Open
Abstract
Auditory hair cell regeneration following injury is critical to hearing restoration. The Notch signaling pathway participates in the regulation of inner ear development and cell differentiation. Recent evidence suggests that microRNA (miR)-183 has a similar role in the inner ear. However, it is unclear how Notch signaling functions in hair cell regeneration in mammals and if there is cross-talk between Notch signaling and miR-183. The present study used a gentamicin-induced cochlear injury mouse model. Gentamicin-induced damage of the hair cells activated the Notch signaling pathway and downregulated miR-183 expression. Notch signaling inhibition by the γ-secretase inhibitor, 24-diamino-5-phenylthiazole (DAPT), attenuated gentamicin-induced hair cell loss and reversed the downregulation of miR-183 expression. Further investigation revealed that the novel hair cells produced, induced by DAPT, were derived from transdifferentiated supporting cells. Additionally, myosin VI-positive hair cell numbers were increased by Notch signaling inhibition in in vitro experiments with cultured neonatal mouse inner ear precursor cells. This effect was reversed by miR-183 inhibition. These findings indicate that the Notch signaling pathway served a repressing role during the regeneration of hair cells. Inhibiting this signal improved hair cell regeneration in the gentamicin-damaged cochlear model. miR-183 was demonstrated to be involved in hair cell differentiation and regeneration, and was required for the differentiation of the Notch-inhibited hair cells.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Otolaryngology, The First Affiliated Hospital and Institute of Otorhinolaryngology, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jintao Du
- Department of Otolaryngology, The First Affiliated Hospital and Institute of Otorhinolaryngology, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Di Jiang
- Department of Otolaryngology, The First Affiliated Hospital and Institute of Otorhinolaryngology, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xianren Wang
- Department of Otolaryngology, The First Affiliated Hospital and Institute of Otorhinolaryngology, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Kaitian Chen
- Department of Otolaryngology, The First Affiliated Hospital and Institute of Otorhinolaryngology, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Haocheng Tang
- Department of Otolaryngology, The First Affiliated Hospital and Institute of Otorhinolaryngology, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xuemei Zhang
- Department of Otolaryngology, The First Affiliated Hospital and Institute of Otorhinolaryngology, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Hui Cao
- Department of Otolaryngology, The First Affiliated Hospital and Institute of Otorhinolaryngology, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Ling Zong
- Department of Otolaryngology, The First Affiliated Hospital and Institute of Otorhinolaryngology, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Chang Dong
- Department of Otolaryngology, The First Affiliated Hospital and Institute of Otorhinolaryngology, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Hongyan Jiang
- Department of Otolaryngology, The First Affiliated Hospital and Institute of Otorhinolaryngology, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
14
|
Stone JS, Wisner SR, Bucks SA, Mellado Lagarde MM, Cox BC. Characterization of Adult Vestibular Organs in 11 CreER Mouse Lines. J Assoc Res Otolaryngol 2018; 19:381-399. [PMID: 29869046 DOI: 10.1007/s10162-018-0676-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/07/2018] [Indexed: 10/14/2022] Open
Abstract
Utricles are vestibular sense organs that encode linear head movements. They are composed of a sensory epithelium with type I and type II hair cells and supporting cells, sitting atop connective tissue, through which vestibular nerves project. We characterized utricular Cre expression in 11 murine CreER lines using the ROSA26tdTomato reporter line and tamoxifen induction at 6 weeks of age. This characterization included Calbindin2CreERT2, Fgfr3-iCreERT2, GFAP-A-CreER™, GFAP-B-CreER™, GLAST-CreERT2, Id2CreERT2, OtoferlinCreERT2, ParvalbuminCreERT2, Prox1CreERT2, Sox2CreERT2, and Sox9-CreERT2. OtoferlinCreERT2 mice had inducible Cre activity specific to hair cells. GLAST-CreERT2, Id2CreERT2, and Sox9-CreERT2 had inducible Cre activity specific to supporting cells. Sox2CreERT2 had inducible Cre activity in supporting cells and most type II hair cells. ParvalbuminCreERT2 mice had small numbers of labeled vestibular nerve afferents. Calbindin2CreERT2 mice had labeling of most type II hair cells and some type I hair cells and supporting cells. Only rare (or no) tdTomato-positive cells were detected in utricles of Fgfr3-iCreERT2, GFAP-A-CreER™, GFAP-B-CreER™, and Prox1CreERT2 mice. No Cre leakiness (tdTomato expression in the absence of tamoxifen) was observed in OtoferlinCreERT2 mice. A small degree of leakiness was seen in GLAST-CreERT2, Id2CreERT2, Sox2CreERT2, and Sox9-CreERT2 lines. Calbindin2CreERT2 mice had similar tdTomato expression with or without tamoxifen, indicating lack of inducible control under the conditions tested. In conclusion, 5 lines-GLAST-CreERT2, Id2CreERT2, OtoferlinCreERT2, Sox2CreERT2, and Sox9-CreERT2-showed cell-selective, inducible Cre activity with little leakiness, providing new genetic tools for researchers studying the vestibular periphery.
Collapse
Affiliation(s)
- Jennifer S Stone
- Department of Otolaryngology-Head and Neck Surgery, Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, WA, USA
| | - Serena R Wisner
- Department of Otolaryngology-Head and Neck Surgery, Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, WA, USA
| | - Stephanie A Bucks
- Department of Otolaryngology-Head and Neck Surgery, Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, WA, USA
| | - Marcia M Mellado Lagarde
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Brandon C Cox
- Departments of Pharmacology and Surgery, Division of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, USA.
| |
Collapse
|
15
|
McGovern MM, Zhou L, Randle MR, Cox BC. Spontaneous Hair Cell Regeneration Is Prevented by Increased Notch Signaling in Supporting Cells. Front Cell Neurosci 2018; 12:120. [PMID: 29780306 PMCID: PMC5945818 DOI: 10.3389/fncel.2018.00120] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/13/2018] [Indexed: 11/13/2022] Open
Abstract
During embryonic development, differentiation of cochlear progenitor cells into hair cells (HCs) or supporting cells (SCs) is partially controlled through Notch signaling. Many studies have shown that inhibition of Notch signaling allows SCs to convert into HCs in both normal and drug damaged neonatal mouse cochleae. This mechanism is also implicated during HC regeneration in non-mammalian vertebrates; however, the mechanism of spontaneous HC regeneration in the neonatal mouse cochlea is less understood. While inhibition of Notch signaling can force SCs to convert into HCs and increase the number of regenerated HCs, it is currently unknown whether this pathway is involved in spontaneous HC regeneration observed in vivo. Therefore, we investigated the role of Notch signaling during the spontaneous HC regeneration process using Atoh1-CreERTM::Rosa26loxP-stop-loxP-DTA/+ mice injected with tamoxifen at postnatal day (P) 0 and P1 to ablate HCs and stimulate spontaneous HC regeneration. Expression changes of genes in the Notch pathway were measured using immunostaining and in situ hybridization, with most changes observed in the apical one-third of the cochlea where the majority of HC regeneration occurs. Expression of the Notch target genes Hes1, Hes5, Hey1, HeyL, and Jagged1 were decreased. To investigate whether reduction of Notch signaling is involved in the spontaneous HC regeneration process, we overexpressed the Notch1 intracellular fragment (N1ICD) in cochlear SCs and other non-sensory epithelial cells in the context of HC damage. Specifically, Atoh1-CreERTM::Rosa26loxP-stop-loxP-DTA/+::Sox10rtTA::TetO-LacZ::TetO-N1ICD mice were injected with tamoxifen at P0/P1 to stimulate spontaneous HC regeneration and given doxycycline from P0-P7 to induce expression of N1ICD as well as LacZ for fate-mapping. We observed a 92% reduction in the number of fate-mapped regenerated HCs in mice with N1ICD overexpression compared to controls with HC damage but no manipulation of Notch signaling. Therefore, we conclude that increased Notch signaling prevents spontaneous HC regeneration from occurring in the neonatal mouse cochlea. Understanding which components of the Notch pathway regulates regenerative plasticity in the neonatal mouse cochlea will inform investigations focused on stimulating HC regeneration in mature cochlea and eventually in humans to treat hearing loss.
Collapse
Affiliation(s)
- Melissa M. McGovern
- Department of Pharmacology, School of Medicine, Southern Illinois University, Springfield, IL, United States
| | - Luyi Zhou
- Department of Pharmacology, School of Medicine, Southern Illinois University, Springfield, IL, United States
| | - Michelle R. Randle
- Department of Pharmacology, School of Medicine, Southern Illinois University, Springfield, IL, United States
| | - Brandon C. Cox
- Department of Pharmacology, School of Medicine, Southern Illinois University, Springfield, IL, United States
- Department of Surgery, Division of Otolaryngology, School of Medicine, Southern Illinois University, Springfield, IL, United States
| |
Collapse
|
16
|
Atkinson PJ, Dong Y, Gu S, Liu W, Najarro EH, Udagawa T, Cheng AG. Sox2 haploinsufficiency primes regeneration and Wnt responsiveness in the mouse cochlea. J Clin Invest 2018; 128:1641-1656. [PMID: 29553487 DOI: 10.1172/jci97248] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 02/01/2018] [Indexed: 12/31/2022] Open
Abstract
During development, Sox2 is indispensable for cell division and differentiation, yet its roles in regenerating tissues are less clear. Here, we used combinations of transgenic mouse models to reveal that Sox2 haploinsufficiency (Sox2haplo) increases rather than impairs cochlear regeneration in vivo. Sox2haplo cochleae had delayed terminal mitosis and ectopic sensory cells, yet normal auditory function. Sox2haplo amplified and expanded domains of damage-induced Atoh1+ transitional cell formation in neonatal cochlea. Wnt activation via β-catenin stabilization (β-cateninGOF) alone failed to induce proliferation or transitional cell formation. By contrast, β-cateninGOF caused proliferation when either Sox2haplo or damage was present, and transitional cell formation when both were present in neonatal, but not mature, cochlea. Mechanistically, Sox2haplo or damaged neonatal cochleae showed lower levels of Sox2 and Hes5, but not of Wnt target genes. Together, our study unveils an interplay between Sox2 and damage in directing tissue regeneration and Wnt responsiveness and thus provides a foundation for potential combinatorial therapies aimed at stimulating mammalian cochlear regeneration to reverse hearing loss in humans.
Collapse
Affiliation(s)
- Patrick J Atkinson
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Yaodong Dong
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, USA.,Department of Otology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shuping Gu
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Wenwen Liu
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Elvis Huarcaya Najarro
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Tomokatsu Udagawa
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Alan G Cheng
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
17
|
Ramku E, Ramku R, Spanca D, Zhjeqi V. Functional Pattern of Increasing Concentrations of Brain-Derived Neurotrophic Factor in Spiral Ganglion: Implications for Research on Cochlear Implants. Open Access Maced J Med Sci 2017; 5:121-125. [PMID: 28507614 PMCID: PMC5420760 DOI: 10.3889/oamjms.2017.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 02/22/2017] [Accepted: 02/25/2017] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND: As previously various studies have suggested application of brain-derived neurotrophic factor (BDNF) may be considered as a promising future therapy for hearing deficits, in particular for the improvement of cochlear neurone loss during cochlear implantation. AIM: The present study’s aim was to establish the upper threshold of the concentration of BDNF in Naval Medical Research Institute (NMRI) mice spiral ganglion outgrowth. METHODS: Spiral ganglion explants were prepared from post-natal day 4 (p4) (NMRI) mice of both sexes under the approval and guidelines of the regional council of Hearing Research Institute Tubingen. RESULTS: Spiral ganglion explants were cultured at postnatal days 4 in the presence of different concentrations of BDNF as described under methods. We chose an age of postnatal day (P4) and concentrations of BDNF 0; 6; 12.5; 25 and 50 ƞg/ml. Averaged neurite outgrowth is measured in 4 different cultures that were treated with different concentrations. Results show that with increasing concentrations of BDNF, the neurite density increases. CONCLUSION: The present finding show evidence that BDNF has a clear incremental effect on the number of neurites of spiral ganglia in the prehearing organ, but less on the neurite length. The upper threshold of exogenous BNDF concentration on spiral ganglion explant is 25 ƞg/ml. This means that concentration beyond this level has no further incremental impact. Therefore our suggestion for hydrogel concentration in NMRA mice in future research should be 25 ƞg/ml.
Collapse
Affiliation(s)
- Emina Ramku
- University Clinical Center, Prishtina, Kosovo
| | - Refik Ramku
- Private Polyclinic OTOKIRURGJIA, Prishtina, Kosovo
| | | | | |
Collapse
|
18
|
Role of Wnt and Notch signaling in regulating hair cell regeneration in the cochlea. Front Med 2016; 10:237-49. [PMID: 27527363 DOI: 10.1007/s11684-016-0464-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 06/12/2016] [Indexed: 01/22/2023]
Abstract
Sensory hair cells in the inner ear are responsible for sound recognition. Damage to hair cells in adult mammals causes permanent hearing impairment because these cells cannot regenerate. By contrast, newborn mammals possess limited regenerative capacity because of the active participation of various signaling pathways, including Wnt and Notch signaling. The Wnt and Notch pathways are highly sophisticated and conserved signaling pathways that control multiple cellular events necessary for the formation of sensory hair cells. Both signaling pathways allow resident supporting cells to regenerate hair cells in the neonatal cochlea. In this regard, Wnt and Notch signaling has gained increased research attention in hair cell regeneration. This review presents the current understanding of the Wnt and Notch signaling pathways in the auditory portion of the inner ear and discusses the possibilities of controlling these pathways with the hair cell fate determiner Atoh1 to regulate hair cell regeneration in the mammalian cochlea.
Collapse
|
19
|
Basch ML, Brown RM, Jen H, Groves AK. Where hearing starts: the development of the mammalian cochlea. J Anat 2016; 228:233-54. [PMID: 26052920 PMCID: PMC4718162 DOI: 10.1111/joa.12314] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2015] [Indexed: 12/11/2022] Open
Abstract
The mammalian cochlea is a remarkable sensory organ, capable of perceiving sound over a range of 10(12) in pressure, and discriminating both infrasonic and ultrasonic frequencies in different species. The sensory hair cells of the mammalian cochlea are exquisitely sensitive, responding to atomic-level deflections at speeds on the order of tens of microseconds. The number and placement of hair cells are precisely determined during inner ear development, and a large number of developmental processes sculpt the shape, size and morphology of these cells along the length of the cochlear duct to make them optimally responsive to different sound frequencies. In this review, we briefly discuss the evolutionary origins of the mammalian cochlea, and then describe the successive developmental processes that lead to its induction, cell cycle exit, cellular patterning and the establishment of topologically distinct frequency responses along its length.
Collapse
Affiliation(s)
- Martin L. Basch
- Department of NeuroscienceBaylor College of MedicineHoustonTXUSA
| | - Rogers M. Brown
- Program in Developmental BiologyBaylor College of MedicineHoustonTXUSA
| | - Hsin‐I Jen
- Program in Developmental BiologyBaylor College of MedicineHoustonTXUSA
| | - Andrew K. Groves
- Department of NeuroscienceBaylor College of MedicineHoustonTXUSA
- Program in Developmental BiologyBaylor College of MedicineHoustonTXUSA
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonTXUSA
| |
Collapse
|
20
|
A central to peripheral progression of cell cycle exit and hair cell differentiation in the developing mouse cristae. Dev Biol 2016; 411:1-14. [PMID: 26826497 DOI: 10.1016/j.ydbio.2016.01.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 01/26/2016] [Indexed: 01/15/2023]
Abstract
The inner ear contains six distinct sensory organs that each maintains some ability to regenerate hair cells into adulthood. In the postnatal cochlea, there appears to be a relationship between the developmental maturity of a region and its ability to regenerate as postnatal regeneration largely occurs in the apical turn, which is the last region to differentiate and mature during development. In the mature cristae there are also regional differences in regenerative ability, which led us to hypothesize that there may be a general relationship between the relative maturity of a region and the regenerative competence of that region in all of the inner ear sensory organs. By analyzing adult mouse cristae labeled embryonically with BrdU, we found that hair cell birth starts in the central region and progresses to the periphery with age. Since the peripheral region of the adult cristae also maintains active Notch signaling and some regenerative competence, these results are consistent with the hypothesis that the last regions to develop retain some of their regenerative ability into adulthood. Further, by analyzing embryonic day 14.5 inner ears we provide evidence for a wave of hair cell birth along the longitudinal axis of the cristae from the central regions to the outer edges. Together with the data from the adult inner ears labeled with BrdU as embryos, these results suggest that hair cell differentiation closely follows cell cycle exit in the cristae, unlike in the cochlea where they are uncoupled.
Collapse
|
21
|
Żak M, Klis SFL, Grolman W. The Wnt and Notch signalling pathways in the developing cochlea: Formation of hair cells and induction of regenerative potential. Int J Dev Neurosci 2015; 47:247-58. [PMID: 26471908 DOI: 10.1016/j.ijdevneu.2015.09.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 09/25/2015] [Accepted: 09/26/2015] [Indexed: 12/21/2022] Open
Abstract
The Wnt and Notch signalling pathways control proliferation, specification, and cell fate choices during embryonic development and in adult life. Hence, there is much interest in both signalling pathways in the context of stem cell biology and tissue regeneration. In the developing ear, the Wnt and Notch signalling pathways specify otic cells and refine the ventral boundary of the otic placode. Since both signalling pathways control events essential for the formation of sensory cells, such as proliferation and hair cell differentiation, these pathways could hold promise for the regeneration of hair cells in adult mammalian cochlea. Indeed, modulating either the Wnt or Notch pathways can trigger the regenerative potential of supporting cells. In the neonatal mouse cochlea, Notch-mediated regeneration of hair cells partially depends on Wnt signalling, which implies an interaction between the pathways. This review presents how the Wnt and Notch signalling pathways regulate the formation of sensory hair cells and how modulating their activity induces regenerative potential in the mammalian cochlea.
Collapse
Affiliation(s)
- Magdalena Żak
- Department of Otorhinolaryngology and Head & Neck Surgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Room G.02.531, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands.
| | - Sjaak F L Klis
- Department of Otorhinolaryngology and Head & Neck Surgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Room G.02.531, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands
| | - Wilko Grolman
- Department of Otorhinolaryngology and Head & Neck Surgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Room G.02.531, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands
| |
Collapse
|
22
|
Abstract
UNLABELLED The mammalian cochlea exhibit minimal spontaneous regeneration, and loss of sensory hair cells (HCs) results in permanent hearing loss. In nonmammalian vertebrates, spontaneous HC regeneration occurs through both proliferation and differentiation of surrounding supporting cells (SCs). HC regeneration in postnatal mammalian cochleae in vivo remains limited by the small HC number and subsequent death of regenerated HCs. Here, we describe in vivo generation of 10-fold more new HCs in the mouse cochlea than previously reported, most of which survive to adulthood. We achieved this by combining the expression of a constitutively active form of β-catenin (a canonical Wnt activator) with ectopic expression of Atoh1 (a HC fate determination factor) in neonatal Lgr5+ cells (the presumed SC and HC progenitors of the postnatal mouse cochlea), and discovered synergistic increases in proliferation and differentiation. The new HCs were predominantly located near the endogenous inner HCs, expressed early HC differentiation markers, and were innervated despite incomplete alignment of presynaptic and postsynaptic markers. Surprisingly, genetic tracing revealed that only a subset of Lgr5+ cells that lie medial to the inner HCs respond to this combination, highlighting a previously unknown heterogeneity that exists among Lgr5+ cells. Together, our data indicate that β-catenin and Atoh1 mediate synergistic effects on both proliferation and differentiation of a subset of neonatal cochlear Lgr5+ cells, thus overcoming major limitations of HC regeneration in postnatal mouse cochleae in vivo. These results provide a basis for combinatorial therapeutics for hearing restoration. SIGNIFICANCE STATEMENT Hearing loss in humans from aging, noise exposure, or ototoxic drugs (i.e., cisplatin or some antibiotics) is permanent and affects every segments of the population, worldwide. However, birds, frog, and fish have the ability to recover hearing, and recent studies have focused on understanding and applying what we have learned from them for restoring hearing in humans. However, studies have been hampered by low efficiency, limited cell numbers, and subsequent death of these newly generated auditory cells. Here, we describe a combinatorial approach, which results in the generation of auditory cells in greater numbers than previously reported, with most of them surviving to adult ages in vivo. These results provide a basis for combinatorial therapeutics for hearing restoration efforts.
Collapse
|
23
|
Abstract
Hair cells of the inner ear are essential for hearing and balance. As a consequence, pathogenic variants in genes specifically expressed in hair cells often cause hereditary deafness. Hair cells are few in number and not easily isolated from the adjacent supporting cells, so the biochemistry and molecular biology of hair cells can be difficult to study. To study gene expression in hair cells, we developed a protocol for hair cell isolation by FACS. With nearly pure hair cells and surrounding cells, from cochlea and utricle and from E16 to P7, we performed a comprehensive cell type-specific RNA-Seq study of gene expression during mouse inner ear development. Expression profiling revealed new hair cell genes with distinct expression patterns: some are specific for vestibular hair cells, others for cochlear hair cells, and some are expressed just before or after maturation of mechanosensitivity. We found that many of the known hereditary deafness genes are much more highly expressed in hair cells than surrounding cells, suggesting that genes preferentially expressed in hair cells are good candidates for unknown deafness genes.
Collapse
|
24
|
Maass JC, Gu R, Basch ML, Waldhaus J, Lopez EM, Xia A, Oghalai JS, Heller S, Groves AK. Changes in the regulation of the Notch signaling pathway are temporally correlated with regenerative failure in the mouse cochlea. Front Cell Neurosci 2015; 9:110. [PMID: 25873862 PMCID: PMC4379755 DOI: 10.3389/fncel.2015.00110] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/10/2015] [Indexed: 12/20/2022] Open
Abstract
Sensorineural hearing loss is most commonly caused by the death of hair cells in the organ of Corti, and once lost, mammalian hair cells do not regenerate. In contrast, other vertebrates such as birds can regenerate hair cells by stimulating division and differentiation of neighboring supporting cells. We currently know little of the genetic networks which become active in supporting cells when hair cells die and that are activated in experimental models of hair cell regeneration. Several studies have shown that neonatal mammalian cochlear supporting cells are able to trans-differentiate into hair cells when cultured in conditions in which the Notch signaling pathway is blocked. We now show that the ability of cochlear supporting cells to trans-differentiate declines precipitously after birth, such that supporting cells from six-day-old mouse cochlea are entirely unresponsive to a blockade of the Notch pathway. We show that this trend is seen regardless of whether the Notch pathway is blocked with gamma secretase inhibitors, or by antibodies against the Notch1 receptor, suggesting that the action of gamma secretase inhibitors on neonatal supporting cells is likely to be by inhibiting Notch receptor cleavage. The loss of responsiveness to inhibition of the Notch pathway in the first postnatal week is due in part to a down-regulation of Notch receptors and ligands, and we show that this down-regulation persists in the adult animal, even under conditions of noise damage. Our data suggest that the Notch pathway is used to establish the repeating pattern of hair cells and supporting cells in the organ of Corti, but is not required to maintain this cellular mosaic once the production of hair cells and supporting cells is completed. Our results have implications for the proposed used of Notch pathway inhibitors in hearing restoration therapies.
Collapse
Affiliation(s)
- Juan C Maass
- Department of Neuroscience, Baylor College of Medicine Houston, TX, USA ; Department of Otolaryngology, Hospital Clínico Universidad de Chile Santiago, Chile ; Interdisciplinary Program of Physiology and Biophysics, ICBM Universidad de Chile Santiago, Chile ; Department of Otolaryngology, Clínica Alemana de Santiago, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo Santiago, Chile
| | - Rende Gu
- Department of Neuroscience, Baylor College of Medicine Houston, TX, USA
| | - Martin L Basch
- Department of Neuroscience, Baylor College of Medicine Houston, TX, USA
| | - Joerg Waldhaus
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine Palo Alto, CA, USA
| | | | - Anping Xia
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine Palo Alto, CA, USA
| | - John S Oghalai
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine Palo Alto, CA, USA
| | - Stefan Heller
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine Palo Alto, CA, USA
| | - Andrew K Groves
- Department of Neuroscience, Baylor College of Medicine Houston, TX, USA ; Department of Molecular and Human Genetics, Baylor College of Medicine Houston, TX, USA ; Program in Developmental Biology, Baylor College of Medicine Houston, TX, USA
| |
Collapse
|
25
|
Fujioka M, Okano H, Edge ASB. Manipulating cell fate in the cochlea: a feasible therapy for hearing loss. Trends Neurosci 2015; 38:139-44. [PMID: 25593106 DOI: 10.1016/j.tins.2014.12.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 11/23/2014] [Accepted: 12/02/2014] [Indexed: 02/06/2023]
Abstract
Mammalian auditory hair cells do not spontaneously regenerate, unlike hair cells in lower vertebrates, including fish and birds. In mammals, hearing loss due to the loss of hair cells is permanent and intractable. Recent studies in the mouse have demonstrated spontaneous hair cell regeneration during a short postnatal period, but this regenerative capacity is lost in the adult cochlea. Reduced regeneration coincides with a transition that results in a decreased pool of progenitor cells in the cochlear sensory epithelium. Here, we review the signaling cascades involved in hair cell formation and morphogenesis of the organ of Corti in developing mammals, the changing status of progenitor cells in the cochlea, and the regeneration of auditory hair cells in adult mammals.
Collapse
Affiliation(s)
- Masato Fujioka
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Albert S B Edge
- Eaton-Peabody Laboratory, Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Boston, MA, USA; Department of Otology and Laryngology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
26
|
Radosevic M, Fargas L, Alsina B. The role of her4 in inner ear development and its relationship with proneural genes and Notch signalling. PLoS One 2014; 9:e109860. [PMID: 25299450 PMCID: PMC4192589 DOI: 10.1371/journal.pone.0109860] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 09/08/2014] [Indexed: 01/13/2023] Open
Abstract
The generation of sensory neurons and hair cells of the inner ear is under tight control. Different members of the Hairy and Enhancer of Split genes (HES) are expressed in the inner ear, their full array of functions still not being disclosed. We have previously shown that zebrafish her9 acts as a patterning gene to restrict otic neurogenesis to an anterior domain. Here, we disclose the role of another her gene, her4, a zebrafish ortholog of Hes5 that is expressed in the neurogenic and sensory domains of the inner ear. The expression of her4 is highly dynamic and spatiotemporally regulated. We demonstrate by loss of function experiments that in the neurogenic domain her4 expression is under the regulation of neurogenin1 (neurog1) and the Notch pathway. Moreover, her4 participates in lateral inhibition during otic neurogenesis since her4 knockdown results in overproduction of the number of neurog1 and deltaB-positive otic neurons. In contrast, during sensorigenesis her4 is initially Notch-independent and induced by atoh1b in a broad prosensory domain. At later stages her4 expression becomes Notch-dependent in the future sensory domains but loss of her4 does not result in hair cell overproduction, suggesting that there other her genes can compensate its function.
Collapse
Affiliation(s)
- Marija Radosevic
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona, Barcelona, Spain
| | - Laura Fargas
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona, Barcelona, Spain
| | - Berta Alsina
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona, Barcelona, Spain
- * E-mail:
| |
Collapse
|
27
|
Abstract
HYPOTHESIS How to induce most efficiently severe sensorineural hearing loss in mice using a single coadministration of an aminoglycoside antibiotic and a loop diuretic? BACKGROUND The coadministration of aminoglycosides and a loop diuretic has been widely used to induce hair cell and spiral ganglion cell loss in guinea pigs. However, the development of new treatment strategies against sensorineural hearing loss, such as tissue engineering techniques, requires the use of mouse models. Previous attempts to induce hearing loss in mice have rendered inconsistent results because of resistance to aminoglycoside-induced ototoxicity. Especially inner hair cells seem to be resistant to aminoglycoside-induced ototoxicity. METHODS In the present study, we aim to optimize hearing loss in mice, using a single high-dose kanamycin (700 and 1,000 mg/kg) injection followed by a furosemide (100 mg/kg) administration. Although previous studies used intraperitoneal furosemide injections 30 minutes after kanamycin administration, we used intravenous furosemide injections administered within 5 minutes after kanamycin treatment. RESULTS Auditory brain stem responses illustrated severe threshold shifts, and histologic analysis showed marked outer hair cell destruction as well as spiral ganglion cell loss. The present protocol results in more severe inner hair cell loss when compared with the results of previous researches. CONCLUSION We conclude that severe sensorineural hearing loss can be induced in mice. Moreover, we found that this mouse model can be augmented via the use of rapid intravenous furosemide administrations to maximize inner hair cell loss.
Collapse
|
28
|
Fernandes VT, Lin VYW. Development of an ototoxicity model in the adult CBA/CaJ mouse and determination of a golden window of corticosteroid intervention for otoprotection. J Otolaryngol Head Neck Surg 2014; 43:12. [PMID: 24762042 PMCID: PMC4029804 DOI: 10.1186/1916-0216-43-12] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 04/11/2014] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE To investigate the effect of timing of dexamethasone administration on auditory hair cell survival following an ototoxic insult with kanamycin and furosemide. STUDY DESIGN Controlled experimental study. SETTING Translational science experimental laboratory. METHODS 5-6 week old CBA/CaJ mice, divided into 6 groups, were injected with kanamycin (1 mg/g SC) followed by furosemide (0.5 mg/g IP). Dexamethasone (0.1 mg/g IP) was injected at either 1 hour prior to insult, +1 hr, +6 hr, +12 hr, or +72 hr post insult. Temporal bones harvested on day 7 underwent Organ of Corti dissection. Immunohistochemical staining was performed using antibodies to myosin 7a, phalloidin, and TO-PRO. RESULTS Hair cell counts demonstrate a uniform ototoxicity model with total loss of outer hair cells (OHCs) and near-total loss of inner hair cells (IHCs). The group pre-treated with dexamethasone showed a statistically significant improvement in counts compared to controls (p = 0.004). Counts from the other experimental groups given dexamethasone after the insult were highly variable but demonstrated some apical and middle turn inner hair cell survival. CONCLUSION Treatment of systemic dexamethasone prior to ototoxic insult attenuates hair cell loss in a reliable, novel, ototoxicity model using kanamycin and furosemide in CBA/CaJ mice. Dosing with dexamethasone following ototoxic insult shows promising yet variable response in hair cell survival.
Collapse
Affiliation(s)
| | - Vincent Y W Lin
- Department of Otolaryngolgy - Head and Neck Surgery, Sunnybrook Health Sciences Centre, University of Toronto, 2075 Bayview Ave, Suite M1-102, Toronto, ON M4N 3M5, Canada.
| |
Collapse
|
29
|
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: 239] [Impact Index Per Article: 21.7] [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
|
30
|
Hair cell generation by notch inhibition in the adult mammalian cristae. J Assoc Res Otolaryngol 2013; 14:813-28. [PMID: 23989618 DOI: 10.1007/s10162-013-0414-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 08/12/2013] [Indexed: 10/26/2022] Open
Abstract
Balance disorders caused by hair cell loss in the sensory organs of the vestibular system pose a significant health problem worldwide, particularly in the elderly. Currently, this hair cell loss is permanent as there is no effective treatment. This is in stark contrast to nonmammalian vertebrates who robustly regenerate hair cells after damage. This disparity in regenerative potential highlights the need for further manipulation in order to stimulate more robust hair cell regeneration in mammals. In the utricle, Notch signaling is required for maintaining the striolar support cell phenotype into the second postnatal week. Notch signaling has further been implicated in hair cell regeneration after damage in the mature utricle. Here, we investigate the role of Notch signaling in the mature mammalian cristae in order to characterize the Notch-mediated regenerative potential of these sensory organs. For these studies, we used the γ-secretase inhibitor, N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT), in conjunction with a method we developed to culture cristae in vitro. In postnatal and adult cristae, we found that 5 days of DAPT treatment resulted in a downregulation of the Notch effectors Hes1 and Hes5 and also an increase in the total number of Gfi1(+) hair cells. Hes5, as reported by Hes5-GFP, was downregulated specifically in peripheral support cells. Using lineage tracing with proteolipid protein (PLP)/CreER;mTmG mice, we found that these hair cells arose through transdifferentiation of support cells in cristae explanted from mice up to 10 weeks of age. These transdifferentiated cells arose without proliferation and were capable of taking on a hair cell morphology, migrating to the correct cell layer, and assembling what appears to be a stereocilia bundle with a long kinocilium. Overall, these data show that Notch signaling is active in the mature cristae and suggest that it may be important in maintaining the support cell fate in a subset of peripheral support cells.
Collapse
|
31
|
Du X, Li W, Gao X, West MB, Saltzman WM, Cheng CJ, Stewart C, Zheng J, Cheng W, Kopke RD. Regeneration of mammalian cochlear and vestibular hair cells through Hes1/Hes5 modulation with siRNA. Hear Res 2013; 304:91-110. [PMID: 23850665 DOI: 10.1016/j.heares.2013.06.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 05/16/2013] [Accepted: 06/27/2013] [Indexed: 12/31/2022]
Abstract
The Notch pathway is a cell signaling pathway determining initial specification and subsequent cell fate in the inner ear. Previous studies have suggested that new hair cells (HCs) can be regenerated in the inner ear by manipulating the Notch pathway. In the present study, delivery of siRNA to Hes1 and Hes5 using a transfection reagent or siRNA to Hes1 encapsulated within poly(lactide-co-glycolide acid) (PLGA) nanoparticles increased HC numbers in non-toxin treated organotypic cultures of cochleae and maculae of postnatal day 3 mouse pups. An increase in HCs was also observed in cultured cochleae and maculae of mouse pups pre-conditioned with a HC toxin (4-hydroxy-2-nonenal or neomycin) and then treated with the various siRNA formulations. Treating cochleae with siRNA to Hes1 associated with a transfection reagent or siRNA to Hes1 delivered by PLGA nanoparticles decreased Hes1 mRNA and up-regulated Atoh1 mRNA expression allowing supporting cells (SCs) to acquire a HC fate. Experiments using cochleae and maculae of p27(kip1)/-GFP transgenic mouse pups demonstrated that newly generated HCs trans-differentiated from SCs. Furthermore, PLGA nanoparticles are non-toxic to inner ear tissue, readily taken up by cells within the tissue of interest, and present a synthetic delivery system that is a safe alternative to viral vectors. These results indicate that when delivered using a suitable vehicle, Hes siRNAs are potential therapeutic molecules that may have the capacity to regenerate new HCs in the inner ear and possibly restore human hearing and balance function.
Collapse
Affiliation(s)
- Xiaoping Du
- Hough Ear Institute, P.O. Box 23206, Oklahoma City, OK 73112, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Liu Z, Liu Z, Walters BJ, Owen T, Kopan R, Zuo J. In vivo visualization of Notch1 proteolysis reveals the heterogeneity of Notch1 signaling activity in the mouse cochlea. PLoS One 2013; 8:e64903. [PMID: 23741415 PMCID: PMC3669271 DOI: 10.1371/journal.pone.0064903] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 04/19/2013] [Indexed: 11/18/2022] Open
Abstract
Mechanosensory hair cells (HCs) and surrounding supporting cells (SCs) in the mouse cochlea are important for hearing and are derived from the same prosensory progenitors. Notch1 signaling plays dual but contrasting and age-dependent roles in mouse cochlear development: early lateral induction and subsequent lateral inhibition. However, it has been difficult to directly visualize mouse cochlear cells experiencing various levels of Notch1 activity at single cell resolution. Here, we characterized two knock-in mouse lines, Notch1Cre (Low)/+ and Notch1Cre (High)/+, with different Cre recombinase activities, that can detect Notch1 receptor proteolysis or Notch1 activity at high and low thresholds, respectively. Using both lines together with a highly sensitive Cre reporter line, we showed that Notch1 activity is nearly undetectable during lateral induction but increases to medium and high levels during lateral inhibition. Furthermore, we found that within the neonatal organ of Corti, the vast majority of cells that experience Notch1 activity were SCs not HCs, suggesting that HCs kept undetectable Notch1 activity during the entire lineage development. Furthermore, among SC subtypes, ∼85–99% of Deiters’ and outer pillar cells but only ∼19–38% of inner pillar cells experience medium and high levels of Notch1 activity. Our results demonstrate that Notch1 activity is highly heterogeneous: 1) between lateral induction and inhibition; 2) between HC and SC lineages; 3) among different SC subtypes; 4) among different cells within each SC subtype. Such heterogeneity should elucidate how the development of the cochclear sensory epithelium is precisely controlled and how HC regeneration can be best achieved in postnatal cochleae.
Collapse
Affiliation(s)
- Zhiyong Liu
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- Integrated Program in Biomedical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Zhenyi Liu
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Division of Dermatology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Bradley J. Walters
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Thomas Owen
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- University of Bath, Bath, United Kingdom
| | - Raphael Kopan
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Division of Dermatology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Jian Zuo
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
33
|
Mizutari K, Fujioka M, Hosoya M, Bramhall N, Okano HJ, Okano H, Edge ASB. Notch inhibition induces cochlear hair cell regeneration and recovery of hearing after acoustic trauma. Neuron 2013; 77:58-69. [PMID: 23312516 DOI: 10.1016/j.neuron.2012.10.032] [Citation(s) in RCA: 309] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2012] [Indexed: 02/07/2023]
Abstract
Hearing loss due to damage to auditory hair cells is normally irreversible because mammalian hair cells do not regenerate. Here, we show that new hair cells can be induced and can cause partial recovery of hearing in ears damaged by noise trauma, when Notch signaling is inhibited by a γ-secretase inhibitor selected for potency in stimulating hair cell differentiation from inner ear stem cells in vitro. Hair cell generation resulted from an increase in the level of bHLH transcription factor Atoh1 in response to inhibition of Notch signaling. In vivo prospective labeling of Sox2-expressing cells with a Cre-lox system unambiguously demonstrated that hair cell generation resulted from transdifferentiation of supporting cells. Manipulating cell fate of cochlear sensory cells in vivo by pharmacological inhibition of Notch signaling is thus a potential therapeutic approach to the treatment of deafness.
Collapse
Affiliation(s)
- Kunio Mizutari
- Department of Otology and Laryngology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Jung JY, Avenarius MR, Adamsky S, Alpert E, Feinstein E, Raphael Y. siRNA targeting Hes5 augments hair cell regeneration in aminoglycoside-damaged mouse utricle. Mol Ther 2013; 21:834-41. [PMID: 23439501 DOI: 10.1038/mt.2013.18] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Notch signaling is active during the development of mosaic epithelial sheets and during their turnover and regeneration. After the loss of hair cells in the mosaic sheet of the vestibular sensory epithelium, new hair cells can be spontaneously generated by transdifferentiation of supporting cells. This regenerative process involves downregulation of the Hes5 gene and is known to be limited and incomplete, especially when the lesion is severe. Here, we test whether further downregulation of Hes5 gene accomplished by the use of siRNA after a severe lesion induced by an aminoglycoside in the mouse utricle can enhance the transdifferentiation of supporting cells and lead to the increased production of new hair cells. We demonstrate that Hes5 levels in the utricle decreased after the application of siRNA and that the number of hair cells in these utricles was significantly larger than following control treatment. The data suggest that siRNA technology may be useful for inducing repair and regeneration in the inner ear and that the Notch signaling pathway is a potentially useful target for specific gene expression inhibition.
Collapse
Affiliation(s)
- Jae Yun Jung
- KHRI, Otolaryngology, Head & Neck Surgery, The University of Michigan, Ann Arbor, Michigan 48109-5648, USA
| | | | | | | | | | | |
Collapse
|
35
|
Slowik AD, Bermingham-McDonogh O. Notch signaling in mammalian hair cell regeneration. TRENDS IN DEVELOPMENTAL BIOLOGY 2013; 7:73-89. [PMID: 25328289 PMCID: PMC4199338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
In the inner ear, Notch signaling has been shown to have two key developmental roles. The first occurs early in otic development and defines the prosensory domains that will develop into the six sensory organs of the inner ear. The second role occurs later in development and establishes the mosaic-like pattern of the mechanosensory hair cells and their surrounding support cells through the more well-characterized process of lateral inhibition. These dual developmental roles have inspired several different strategies to regenerate hair cells in the mature inner ear organs. These strategies include (1) modulation of Notch signaling in inner ear stem cells in order to increase hair cell yield, (2) activation of Notch signaling in order to promote the formation of ectopic sensory regions in normally non-sensory regions within the inner ear, and (3) inhibition of Notch signaling to disrupt lateral inhibition and allow support cells to transdifferentiate into hair cells. In this review, we summarize some of the promising studies that have used these various strategies for hair cell regeneration through modulation of Notch signaling and some of the challenges that remain in developing therapies based on hair cell regeneration.
Collapse
Affiliation(s)
- Amber D. Slowik
- Department of Biological Structure, University of Washington, Seattle, Washington 98195
- Graduate Program in Neurobiology and Behavior, University of Washington, Seattle, Washington 98195
| | - Olivia Bermingham-McDonogh
- Department of Biological Structure, University of Washington, Seattle, Washington 98195
- Institute for Stem Cells and Regenerative Medicine, University of Washington, Seattle, Washington 98109, USA
| |
Collapse
|
36
|
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: 2.9] [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
|
37
|
Son EJ, Wu L, Yoon H, Kim S, Choi JY, Bok J. Developmental gene expression profiling along the tonotopic axis of the mouse cochlea. PLoS One 2012; 7:e40735. [PMID: 22808246 PMCID: PMC3395647 DOI: 10.1371/journal.pone.0040735] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 06/12/2012] [Indexed: 12/13/2022] Open
Abstract
The mammalian cochlear duct is tonotopically organized such that the basal cochlea is tuned to high frequency sounds and the apical cochlea to low frequency sounds. In an effort to understand how this tonotopic organization is established, we searched for genes that are differentially expressed along the tonotopic axis during neonatal development. Cochlear tissues dissected from P0 and P8 mice were divided into three equal pieces, representing the base, middle and apex, and gene expression profiles were determined using the microarray technique. The gene expression profiles were grouped according to changes in expression levels along the tonotopic axis as well as changes during neonatal development. The classified groups were further analyzed by functional annotation clustering analysis to determine whether genes associated with specific biological function or processes are particularly enriched in each group. These analyses identified several candidate genes that may be involved in cochlear development and acquisition of tonotopy. We examined the expression domains for a few candidate genes in the developing mouse cochlea. Tnc (tenacin C) and Nov (nephroblastoma overexpressed gene) are expressed in the basilar membrane, with increased expression toward the apex, which may contribute to graded changes in the structure of the basilar membrane along the tonotopic axis. In addition, Fst (Follistatin), an antagonist of TGF-β/BMP signaling, is expressed in the lesser epithelial ridge and at gradually higher levels towards the apex. The graded expression pattern of Fst is established at the time of cochlear specification and maintained throughout embryonic and postnatal development, suggesting its possible role in the organization of tonotopy. Our data will provide a good resource for investigating the developmental mechanisms of the mammalian cochlea including the acquisition of tonotopy.
Collapse
Affiliation(s)
- Eun Jin Son
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
- * E-mail: (JB); (EJS)
| | - Ling Wu
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
- BK21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Heejei Yoon
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
| | - Sunhee Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Young Choi
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
- BK21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Jinwoong Bok
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
- BK21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
- * E-mail: (JB); (EJS)
| |
Collapse
|
38
|
Wnt signaling induces proliferation of sensory precursors in the postnatal mouse cochlea. Proc Natl Acad Sci U S A 2012; 109:8167-72. [PMID: 22562792 DOI: 10.1073/pnas.1202774109] [Citation(s) in RCA: 233] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Inner ear hair cells are specialized sensory cells essential for auditory function. Previous studies have shown that the sensory epithelium is postmitotic, but it harbors cells that can behave as progenitor cells in vitro, including the ability to form new hair cells. Lgr5, a Wnt target gene, marks distinct supporting cell types in the neonatal cochlea. Here, we tested the hypothesis that Lgr5(+) cells are Wnt-responsive sensory precursor cells. In contrast to their quiescent in vivo behavior, Lgr5(+) cells isolated by flow cytometry from neonatal Lgr5(EGFP-CreERT2/+) mice proliferated and formed clonal colonies. After 10 d in culture, new sensory cells formed and displayed specific hair cell markers (myo7a, calretinin, parvalbumin, myo6) and stereocilia-like structures expressing F-actin and espin. In comparison with other supporting cells, Lgr5(+) cells were enriched precursors to myo7a(+) cells, most of which formed without mitotic division. Treatment with Wnt agonists increased proliferation and colony-formation capacity. Conversely, small-molecule inhibitors of Wnt signaling suppressed proliferation without compromising the myo7a(+) cells formed by direct differentiation. In vivo lineage tracing supported the idea that Lgr5(+) cells give rise to myo7a(+) hair cells in the neonatal Lgr5(EGFP-CreERT2/+) cochlea. In addition, overexpression of β-catenin initiated proliferation and led to transient expansion of Lgr5(+) cells within the cochlear sensory epithelium. These results suggest that Lgr5 marks sensory precursors and that Wnt signaling can promote their proliferation and provide mechanistic insights into Wnt-responsive progenitor cells during sensory organ development.
Collapse
|
39
|
Ronaghi M, Nasr M, Heller S. Concise review: Inner ear stem cells--an oxymoron, but why? Stem Cells 2012; 30:69-74. [PMID: 22102534 DOI: 10.1002/stem.785] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Hearing loss, caused by irreversible loss of cochlear sensory hair cells, affects millions of patients worldwide. In this concise review, we examine the conundrum of inner ear stem cells, which obviously are present in the inner ear sensory epithelia of nonmammalian vertebrates, giving these ears the ability to functionally recover even from repetitive ototoxic insults. Despite the inability of the mammalian inner ear to regenerate lost hair cells, there is evidence for cells with regenerative capacity because stem cells can be isolated from vestibular sensory epithelia and from the neonatal cochlea. Challenges and recent progress toward identification of the intrinsic and extrinsic signaling pathways that could be used to re-establish stemness in the mammalian organ of Corti are discussed.
Collapse
Affiliation(s)
- Mohammad Ronaghi
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California 94305-5739, USA
| | | | | |
Collapse
|
40
|
Liu Z, Owen T, Fang J, Srinivasan RS, Zuo J. In vivo Notch reactivation in differentiating cochlear hair cells induces Sox2 and Prox1 expression but does not disrupt hair cell maturation. Dev Dyn 2012; 241:684-96. [PMID: 22354878 DOI: 10.1002/dvdy.23754] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2012] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Notch signaling is active in mouse cochlear prosensory progenitors but declines in differentiating sensory hair cells (HCs). Overactivation of the Notch1 intracellular domain (NICD) in progenitors blocks HC fate commitment and/or differentiation. However, it is not known whether reactivation of NICD in differentiating HCs also interrupts their developmental program and reactivates its downstream targets. RESULTS By analyzing Atoh1(CreER+) ; Rosa26-NICD(loxp/+) or Atoh1(CreER+) ; Rosa26-NICD(loxp/+) ; RBP-J(loxp/loxp) mice, we demonstrated that ectopic NICD in differentiating HCs caused reactivation of Sox2 and Prox1 in an RBP-J-dependent manner. Interestingly, Prox1 reactivation was exclusive to outer HCs (OHCs). In addition, lineage tracing analysis of Prox1(CreER/+) ; Rosa26-EYFP(loxp/+) and Prox1(CreEGFP/+) ; Rosa26-EYFP(loxp/+) mice showed that nearly all HCs experiencing Prox1 expression were OHCs. Surprisingly, these HCs still matured normally with expression of prestin, wild-type-like morphology, and uptake of FM4-64FX dye at adult ages. CONCLUSIONS Our results suggest that the developmental program of cochlear differentiating HCs is refractory to Notch reactivation and that Notch is an upstream regulator of Sox2 and Prox1 in cochlear development. In addition, our results support that Sox2 and Prox1 should not be the main blockers for terminal differentiation of HCs newly regenerated from postnatal cochlear SCs that still maintain Sox2 and Prox1 expression.
Collapse
Affiliation(s)
- Zhiyong Liu
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | | | | | | | | |
Collapse
|
41
|
Inhibition of Notch activity promotes nonmitotic regeneration of hair cells in the adult mouse utricles. J Neurosci 2011; 31:15329-39. [PMID: 22031879 DOI: 10.1523/jneurosci.2057-11.2011] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The capacity of adult mammals to regenerate sensory hair cells is not well defined. To explore early steps in this process, we examined reactivation of a transiently expressed developmental gene, Atoh1, in adult mouse utricles after neomycin-induced hair cell death in culture. Using an adenoviral reporter for Atoh1 enhancer, we found that Atoh1 transcription is activated in some hair cell progenitors (supporting cells) 3 d after neomycin treatment. By 18 d after neomycin, the number of cells with Atoh1 transcriptional activity increased significantly, but few cells acquired hair cell features (i.e., accumulated ATOH1 or myosin VIIa protein or developed stereocilia). Treatment with DAPT, an inhibitor of γ-secretase, reduced notch pathway activity, enhanced Atoh1 transcriptional activity, and dramatically increased the number of Atoh1-expressing cells with hair cell features, but only in the striolar/juxtastriolar region. Similar effects were seen with TAPI-1, an inhibitor of another enzyme required for notch activity (TACE). Division of supporting cells was rare in any control or DAPT-treated utricles. This study shows that mature mammals have a natural capacity to initiate vestibular hair cell regeneration and suggests that regional notch activity is a significant inhibitor of direct transdifferentiation of supporting cells into hair cells following damage.
Collapse
|
42
|
Effects of DAPT and Atoh1 overexpression on hair cell production and hair bundle orientation in cultured Organ of Corti from neonatal rats. PLoS One 2011; 6:e23729. [PMID: 22028767 PMCID: PMC3197578 DOI: 10.1371/journal.pone.0023729] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2011] [Accepted: 07/26/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND In mammals, hair cells do not undergo spontaneous regeneration when they are damaged and result in permanent hearing loss. Previous studies in cultured Organ of Corti dissected from neonatal animals have shown that both DAPT (r-secretase inhibitor in the Notch signal pathway) treatment and Atoh1 overexpression can induce supernumerary hair cells. The effects of simultaneous DAPT treatment and Atoh1 over expression in the cells of cultured Organ of Corti from neonatal rats are still obscure. PRINCIPAL FINDINGS In this study, we set out to investigate the interaction of DAPT treatment and Atoh1 overexpression as well as culture time and the location of basilar fragment isolated form neonatal rat inner ear. Our results showed that DAPT treatment induced more hair cells in the apical turn, while Atoh1 overexpression induced more extra hair cells in the middle turn of the cultured Organ of Corti. When used together, their effects are additive but not synergistic. In addition, the induction of supernumerary hair cells by both DAPT and Atoh1 overexpression is dependent on the treatment time and the location of the cochlear tissue. Moreover, DAPT treatment causes dramatic changes in the orientation of the stereociliary bundles of hair cells, whereas Atoh1 overexpression didn't induce drastic change of the polarity of stereociliary bundles. CONCLUSIONS/SIGNIFICANCE Taken together, these results suggest that DAPT treatment are much more potent in inducing supernumerary hair cells than Atoh1 overexpression and that the new hair cells mainly come from the trans-differentiation of supporting cells around hair cells. The orientation change of stereociliary bundle of hair cells may be attributed to the insertion of the newly formed hair cells. The immature hair bundles on the newly formed hair cells may also contribute to the overall chaos of the stereociliary bundle of the sensory epithelia.
Collapse
|
43
|
Bermingham-McDonogh O, Reh TA. Regulated reprogramming in the regeneration of sensory receptor cells. Neuron 2011; 71:389-405. [PMID: 21835338 DOI: 10.1016/j.neuron.2011.07.015] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2011] [Indexed: 12/15/2022]
Abstract
Vision, olfaction, hearing, and balance are mediated by receptors that reside in specialized sensory epithelial organs. Age-related degeneration of the photoreceptors in the retina and the hair cells in the cochlea, caused by macular degeneration and sensorineural hearing loss, respectively, affect a growing number of individuals. Although sensory receptor cells in the mammalian retina and inner ear show only limited or no regeneration, in many nonmammalian vertebrates, these sensory epithelia show remarkable regenerative potential. We summarize the current state of knowledge of regeneration in the specialized sense organs in both nonmammalian vertebrates and mammals and discuss possible areas where new advances in regenerative medicine might provide approaches to successfully stimulate sensory receptor cell regeneration. The field of regenerative medicine is still in its infancy, but new approaches using stem cells and reprogramming suggest ways in which the potential for regeneration may be restored in individuals suffering from sensory loss.
Collapse
Affiliation(s)
- Olivia Bermingham-McDonogh
- Department of Biological Structure, Institute for Stem Cells and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA.
| | | |
Collapse
|
44
|
Beyer LA, Galano MM, Nair TS, Kommareddi PK, Sha SH, Raphael Y, Carey TE. Age-related changes in expression of CTL2/SLC44A2 and its isoforms in the mouse inner ear. Hear Res 2011; 282:63-8. [PMID: 21986210 DOI: 10.1016/j.heares.2011.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 08/26/2011] [Accepted: 09/19/2011] [Indexed: 11/19/2022]
Abstract
The membrane glycoprotein CTL2/SLC44A2 is expressed by supporting cells in the inner ear and has been identified as a target of antibodies that may induce auto-immune hearing loss. To determine if CTL2/SLC44A2 also has roles in inner ear development and to distinguish between isoform-specific roles, we assessed age-related changes in expression of CTL2/SLC44A2 isoforms and protein in the developing murine inner ear. We determined that both isoform p1 and isoform p2 (named for the upstream p1 and proximal p2 promoters that control alternate exons 1a and 1b) were robustly expressed as early as E14 and persisted during embryonic development, but after birth the p1 isoform fell to barely detectable levels while isoform p2 levels were maintained. This trend continued and became even more apparent later in post-natal development and remained in mature ears until at least 6 weeks of age. In aged (18 mo old) mice, the level of isoform p1 transcripts rose again to levels similar to the p2 isoform like that seen early in development. At the earliest stage examined, CTL2/SLC44A2 protein was expressed in both immature supporting cells and immature sensory cells, but after birth expression in the sensory cells declined in both the utricle and cochlea and by day P1 expression of CTL2/SLC44A2 was restricted to supporting cells. The changes we observed in isoform distribution are indicative of differential developmental roles and age related changes between the two isoforms of CTL2/SLC44A2 in the inner ear.
Collapse
Affiliation(s)
- Lisa A Beyer
- Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Parker MA, Jiang K, Kempfle JS, Mizutari K, Simmons CL, Bieber R, Adams J, Edge ASB. TAK1 expression in the cochlea: a specific marker for adult supporting cells. J Assoc Res Otolaryngol 2011; 12:471-83. [PMID: 21472480 PMCID: PMC3123448 DOI: 10.1007/s10162-011-0265-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Accepted: 03/08/2011] [Indexed: 10/18/2022] Open
Abstract
Transforming growth factor-β-activated kinase-1 (TAK1) is a mitogen activated protein kinase kinase kinase that is involved in diverse biological roles across species. Functioning downstream of TGF-β and BMP signaling, TAK1 mediates the activation of the c-Jun N-terminal kinase signaling pathway, serves as the target of pro-inflammatory cytokines, such as TNF-α, mediates NF-κβ activation, and plays a role in Wnt/Fz signaling in mesenchymal stem cells. Expression of TAK1 in the cochlea has not been defined. Data mining of previously published murine cochlear gene expression databases indicated that TAK1, along with TAK1 interacting proteins 1 (TAB1), and 2 (TAB2), is expressed in the developing and adult cochlea. The expression of TAK1 in the developing cochlea was confirmed using RT-PCR and immunohistochemistry. Immunolabeling of TAK1 in embryonic, neonatal, and mature cochleas via DAB chromogenic and fluorescent immunohistochemistry indicated that TAK1 is broadly expressed in both the developing otocyst and periotic mesenchyme at E12.5 but becomes more restricted to specific types of supporting cells as the organ of Corti matures. By P1, TAK1 immunolabeling is found in cells of the stria vascularis, hair cells, supporting cells, and Kölliker's organ. By P16, TAK1 labeling is limited to cochlear supporting cells. In the adult cochlea, TAK1 immunostaining is only present in the cytoplasm of Deiters' cells, pillar cells, inner phalangeal cells, and inner border cells, with no expression in any other cochlear cell types. While the role of TAK1 in the inner ear is unclear, TAK1 expression may be used as a novel marker for specific sub-populations of supporting cells.
Collapse
Affiliation(s)
- Mark A Parker
- Department of Communication Sciences and Disorders, Emerson College, Boston, MA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Sox2 up-regulation and glial cell proliferation following degeneration of spiral ganglion neurons in the adult mouse inner ear. J Assoc Res Otolaryngol 2011; 12:151-71. [PMID: 21061038 DOI: 10.1007/s10162-010-0244-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Accepted: 10/19/2010] [Indexed: 10/18/2022] Open
Abstract
In the present study, glial cell responses to spiral ganglion neuron (SGN) degeneration were evaluated using a murine model of auditory neuropathy. Ouabain, a well-known Na,K-ATPase inhibitor, has been shown to induce SGN degeneration while sparing hair cell function. In addition to selectively removing type I SGNs, ouabain leads to hyperplasia and hypertrophy of glia-like cells in the injured auditory nerves. As the transcription factor Sox2 is predominantly expressed in proliferating and undifferentiated neural precursors during neurogenesis,we sought to examine Sox2 expression patterns following SGN injury by ouabain. Real-time RT-PCR and Western blot analyses of cochlea indicated a significant increase in Sox2 expression by 3 days posttreatment with ouabain. Cells incorporating bromodeoxyuridine(BrdU) and expressing Sox2 were counted in the auditory nerves of control and ouabain-treated ears. The glial phenotype of Sox2+cells was identified by two neural glial markers: S100 and Sox10. The number of Sox2+ glial cells significantly increased at 3 days post-treatment and reached its maximum level at 7 days post-treatment. Similarly,the number of BrdU+ cells increased at 3 and 7 days post-treatment in the injured nerves. Quantitative analysis with dual-immunostaining procedures indicated that about 70% of BrdU+ cells in the injured nerves were Sox2+ glial cells. These results demonstrate that up-regulation of Sox2 expression is associated with increased cell proliferation in the auditory nerve after injury.
Collapse
|
47
|
Staecker H, Praetorius M, Brough DE. Development of gene therapy for inner ear disease: Using bilateral vestibular hypofunction as a vehicle for translational research. Hear Res 2011; 276:44-51. [PMID: 21251965 PMCID: PMC3109124 DOI: 10.1016/j.heares.2011.01.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2010] [Revised: 01/10/2011] [Accepted: 01/11/2011] [Indexed: 12/16/2022]
Abstract
Despite the significant impact of hearing and balance disorders on the general population there are currently no dedicated pharmaceuticals that target the inner ear. Advances in molecular biology and neuroscience have improved our understanding of the inner ear allowing the development of a range of molecular targets that have the potential to treat both hearing and balance disorders. One of the principal advantages of the inner ear is that it is accessible through a variety of approaches that would allow a potential to be delivered locally rather than systemically. This significantly broadens the potential medications that can be developed and opens the possibility of local gene delivery as a therapeutic intervention. Several potential clinical targets have been identified including delivery of neurotrophin expressing genes as an adjunct to cochlear implantation, delivery of protective genes to prevent trauma and the development of strategies for regenerating inner ear sensory cells. In order to translate these potential therapeutics into humans we will want to optimize the gene delivery methodology, dosing and activity of the drug for therapeutic value. To this end we have developed a series of adenovectors that efficiently transduce the inner ear. The use of these gene delivery approaches are attractive for the potential of hair cell regeneration after loss induced by trauma or ototoxins. This approach is particularly suited for the development of molecular therapies targeted at the vestibular system given that no device based therapeutic such a cochlear implant available for vestibular loss.
Collapse
Affiliation(s)
- Hinrich Staecker
- Dept. Otolaryngology Head and Neck Surgery, University of Kansas School of Medicine, MS 3010, 3901 Rainbow Blvd, Kansas City, KS 66209, USA.
| | | | | |
Collapse
|
48
|
Chai R, Xia A, Wang T, Jan TA, Hayashi T, Bermingham-McDonogh O, Cheng AGL. Dynamic expression of Lgr5, a Wnt target gene, in the developing and mature mouse cochlea. J Assoc Res Otolaryngol 2011; 12:455-69. [PMID: 21472479 DOI: 10.1007/s10162-011-0267-2] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Accepted: 03/17/2011] [Indexed: 12/11/2022] Open
Abstract
The Wnt signaling pathway is a recurring theme in tissue development and homeostasis. Its specific roles during inner ear development are just emerging, but few studies have characterized Wnt target genes. Lgr5, a member of the G protein-coupled receptor family, is a Wnt target in the gastrointestinal and integumentary systems. Although its function is unknown, its deficiency leads to perinatal lethality due to gastrointestinal distension. In this study, we used a knock-in reporter mouse to examine the spatiotemporal expression of Lgr5 in the cochlear duct during embryonic and postnatal periods. In the embryonic day 15.5 (E15.5) cochlear duct, Lgr5-EGFP is expressed in the floor epithelium and overlapped with the prosensory markers Sox2, Jagged1, and p27(Kip1). Nascent hair cells and supporting cells in the apical turn of the E18.5 cochlear duct express Lgr5-EGFP, which becomes downregulated in hair cells and subsets of supporting cells in more mature stages. In situ hybridization experiments validated the reporter expression, which gradually decreases until the second postnatal week. Only the third row of Deiters' cells expresses Lgr5-EGFP in the mature organ of Corti. Normal cochlear development was observed in Lgr5(EGFP/EGFP) and Lgr5(EGFP/+) mice, which exhibited normal auditory thresholds. The expression pattern of Lgr5 contrasts with another Wnt target gene, Axin2, a feedback inhibitor of the Wnt pathway. Robust Axin2 expression was found in cells surrounding the embryonic cochlear duct and becomes restricted to tympanic border cells below the basilar membrane in the postnatal cochlea. Both Lgr5 and Axin2 act as Wnt targets in the cochlea because purified Wnt3a promoted and Wnt antagonist suppressed their expression. Their differential expression among cell populations highlights the dynamic but complex distribution of Wnt-activated cells in and around the embryonic and postnatal cochlea.
Collapse
Affiliation(s)
- Renjie Chai
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, CA 94305, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Tateya T, Imayoshi I, Tateya I, Ito J, Kageyama R. Cooperative functions of Hes/Hey genes in auditory hair cell and supporting cell development. Dev Biol 2011; 352:329-40. [PMID: 21300049 DOI: 10.1016/j.ydbio.2011.01.038] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Revised: 12/16/2010] [Accepted: 01/28/2011] [Indexed: 01/08/2023]
Abstract
Notch-mediated lateral inhibition has been reported to regulate auditory hair cell and supporting cell development from common precursors. While the Notch effector genes Hes1, Hes5 and Hey1 are expressed in the developing cochlea, inactivation of either of them causes only mild abnormality, suggesting their functional redundancy. To explore the roles of Hes/Hey genes in cochlear development, we examined compound heterozygous or homozygous mutant mice that lacked Hes1, Hes5 and Hey1 alleles. We found that a reduction in Hes/Hey gene dosage led to graded increase of hair cell formation. However, if at least one allele of Hes1, Hes5 or Hey1 was intact, excessive hair cells were accompanied by overproduction of supporting cells, suggesting that the hair cell increase does not occur at the expense of supporting cells, and that each Hes/Hey gene functions to induce supporting cells. By contrast, when all alleles of Hes1, Hes5 and Hey1 were inactivated, the number of hair cells increased more drastically, whereas that of supporting cells was unchanged compared with control, suggesting that supporting cell formation was balanced by their overproduction and fate conversion into hair cells. The increase of the cell numbers seemed to occur after the prosensory domain formation in the mutants because the proliferation state and the size of the prosensory domain were not affected. Thus, Hes1, Hes5 and Hey1 cooperatively inhibit hair cell formation, and one allele of Hes1, Hes5 or Hey1 is sufficient for supporting cell production probably by lateral inhibition in the sensory epithelium. Strikingly, Hes/Hey mutations lead to disorganized cell alignment and polarity and to hearing loss despite hair cell overproduction. These results suggest that Hes/Hey gene dosage is essential not only for generation of appropriate numbers of hair cells and supporting cells by controlling cell proliferation and lateral inhibition but also for the hearing ability by regulating the cell alignment and polarity.
Collapse
Affiliation(s)
- Tomoko Tateya
- Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan
| | | | | | | | | |
Collapse
|
50
|
Gajecka M, Saitta SC, Gentles AJ, Campbell L, Ciprero K, Geiger E, Catherwood A, Rosenfeld JA, Shaikh T, Shaffer LG. Recurrent interstitial 1p36 deletions: Evidence for germline mosaicism and complex rearrangement breakpoints. Am J Med Genet A 2010; 152A:3074-83. [PMID: 21108392 PMCID: PMC3058890 DOI: 10.1002/ajmg.a.33733] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Deletions of chromosome 1p36 are one of the most frequently encountered subtelomeric alterations. Clinical features of monosomy 1p36 include neurocognitive impairment, hearing loss, seizures, cardiac defects, and characteristic facial features. The majority of cases have occurred sporadically, implying that genomic instability plays a role in the prevalence of the syndrome. Here, we report two siblings with mild phenotypic features of the deletion syndrome, including developmental delay, hearing loss, and left ventricular non-compaction (LVNC). Microarray analysis using bacterial artificial chromosome and oligonucleotide microarrays indicated the deletions were identical, suggesting germline mosaicism. Parental phenotypes were normal, and analysis by fluorescence in situ hybridization (FISH) did not show mosaicism. These small interstitial deletions were not detectable by conventional subtelomeric FISH analysis. To investigate the mechanism of deletion further, the breakpoints were cloned and sequenced, demonstrating the presence of a complex rearrangement. Sequence analysis of genes in the deletion interval did not reveal any mutations on the intact homologue that may have contributed to the LVNC seen in both children. This is the first report of apparent germline mosaicism for this disorder. Thus, our findings have important implications for diagnostic approaches and for recurrence risk counseling in families with a child with monosomy 1p36. In addition, our results further refine the minimal critical region for LVNC and hearing loss.
Collapse
Affiliation(s)
- Marzena Gajecka
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Sulagna C. Saitta
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | | | - Lindsey Campbell
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Karen Ciprero
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Elizabeth Geiger
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Anne Catherwood
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Tamim Shaikh
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | | |
Collapse
|