1
|
Heffer A, Lee C, Mayernik JP, Holt JC, Kiernan AE. Notch1 is Required to Maintain Supporting Cell Identity and Vestibular Function during Maturation of the Mammalian Balance Organs. J Neurosci 2025; 45:e1365242024. [PMID: 39779370 PMCID: PMC11867012 DOI: 10.1523/jneurosci.1365-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/27/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
The inner ear houses both hearing and balance sensory modalities. The hearing and balance organs consist of similar cell types, including sensory hair cells and associated supporting cells. Previously we showed that Notch1 is required for maintaining supporting cell survival during cochlear maturation. To understand the role of Notch during vestibular maturation, we deleted Notch1 from the vestibular organs of both male and female mice at birth. Histological analyses showed a reduction of supporting cells accompanied by an increase in type II hair cells, indicating a conversion of supporting cells to hair cells. Analysis of mature sensory organs indicate the converted hair cells survive, despite a severe reduction of supporting cells. Vestibular sensory evoked potentials (VsEPs), thought to be generated within the striola regions of the maculae, were absent, indicating that NOTCH1 is critical for striolar function. Specialized type I hair cells in the striola failed to develop the complex calyces typical of these cells. Notch1 mutants did not exhibit vestibular behaviors such as circling and head shaking but showed difficulties with tests of balance and swimming. These results indicate that, unlike the cochlea, supporting cells in balance organs retain the plasticity to convert to hair cells which can survive into adulthood. Despite hair cell survival, vestibular function is compromised likely due to the loss of supporting cells and altered innervation.
Collapse
MESH Headings
- Animals
- Receptor, Notch1/genetics
- Receptor, Notch1/physiology
- Receptor, Notch1/deficiency
- Receptor, Notch1/metabolism
- Mice
- Female
- Male
- Postural Balance/physiology
- Vestibule, Labyrinth/growth & development
- Vestibule, Labyrinth/physiology
- Vestibule, Labyrinth/cytology
- Hair Cells, Auditory/physiology
- Hair Cells, Vestibular/physiology
- Mice, Knockout
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Alison Heffer
- Flaum Eye Institute, Department of Ophthalmology, University of Rochester Medical Center, Rochester, New York 14642
| | - Choongheon Lee
- Departments of Otolaryngology, University of Rochester, Rochester, New York 14642
- Mechanical Engineering, University of Rochester, Rochester, New York 14642
| | - Joseph P Mayernik
- Flaum Eye Institute, Department of Ophthalmology, University of Rochester Medical Center, Rochester, New York 14642
| | - Joseph C Holt
- Departments of Otolaryngology, University of Rochester, Rochester, New York 14642
- Neuroscience, University of Rochester, Rochester, New York 14642
| | - Amy E Kiernan
- Flaum Eye Institute, Department of Ophthalmology, University of Rochester Medical Center, Rochester, New York 14642
| |
Collapse
|
2
|
Kasirer S, Sprinzak D. Interplay between Notch signaling and mechanical forces during developmental patterning processes. Curr Opin Cell Biol 2024; 91:102444. [PMID: 39608232 DOI: 10.1016/j.ceb.2024.102444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/03/2024] [Accepted: 10/30/2024] [Indexed: 11/30/2024]
Abstract
The coordination between biochemical signals and cell mechanics has emerged in recent years as a crucial mechanism driving developmental patterning processes across a variety of developing and homeostatic systems. An important class of such developmental processes relies on local communication between neighboring cells through Notch signaling. Here, we review how the coordination between Notch-mediated differentiation and cell mechanics can give rise to unique cellular patterns. We discuss how global and local mechanical cues can affect, and be affected by, cellular differentiation and reorganization controlled by Notch signaling. We compare recent studies of such developmental processes, including the mammalian inner ear, Drosophila ommatidia, intestinal organoids, and zebrafish myocardium, to draw shared general concepts and their broader implications in biology.
Collapse
Affiliation(s)
- Shahar Kasirer
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel; Raymond and Beverly Sackler School of Physics and Astronomy, Faculty of Exact Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - David Sprinzak
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel.
| |
Collapse
|
3
|
Heffer A, Lee C, Holt JC, Kiernan AE. Notch1 is required to maintain supporting cell identity and vestibular function during maturation of the mammalian balance organs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.21.600098. [PMID: 38948821 PMCID: PMC11212955 DOI: 10.1101/2024.06.21.600098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The inner ear houses two sensory modalities: the hearing organ, located in the cochlea, and the balance organs, located throughout the vestibular regions of the ear. Both hearing and vestibular sensory regions are composed of similar cell types, including hair cells and associated supporting cells. Recently, we showed that Notch1 is required for maintaining supporting cell survival postnatally during cochlear maturation. However, it is not known whether Notch1 plays a similar role in the balance organs of the inner ear. To characterize the role of Notch during vestibular maturation, we conditionally deleted Notch1 from Sox2-expressing cells of the vestibular organs in the mouse at P0/P1. Histological analyses showed a dramatic loss of supporting cells accompanied by an increase in type II hair cells without cell death, indicating the supporting cells are converting to hair cells in the maturing vestibular regions. Analysis of 6-week old animals indicate that the converted hair cells survive, despite the reduction of supporting cells. Interestingly, measurements of vestibular sensory evoked potentials (VsEPs), known to be generated in the striolar regions of the vestibular afferents in the maculae, failed to show a response, indicating that NOTCH1 expression is critical for striolar function postnatally. Consistent with this, we find that the specialized type I hair cells in the striola fail to develop the complex calyces typical of these cells. These defects are likely due to the reduction in supporting cells, which have previously been shown to express factors critical for the striolar region. Similar to other mutants that lack proper striolar development, Notch1 mutants do not exhibit typical vestibular behaviors such as circling and head shaking, but do show difficulties in some vestibular tests, including the balance beam and forced swim test. These results indicate that, unlike the hearing organ in which the supporting cells undergo cell death, supporting cells in the balance regions retain the ability to convert to hair cells during maturation, which survive into adulthood despite the reduction in supporting cells.
Collapse
Affiliation(s)
- Alison Heffer
- Flaum Eye Institute, Department of Ophthalmology, University of Rochester Medical Center, Rochester, New York, 14642, USA
| | - Choongheon Lee
- Department of Otolaryngology, University of Rochester, Rochester, NY, 14642, USA
| | - Joseph C. Holt
- Department of Otolaryngology, University of Rochester, Rochester, NY, 14642, USA
- Dept. of Neuroscience, University of Rochester, Rochester, New York 14642, USA
| | - Amy E. Kiernan
- Flaum Eye Institute, Department of Ophthalmology, University of Rochester Medical Center, Rochester, New York, 14642, USA
| |
Collapse
|
4
|
Hosoya M, Fujioka M, Okano H, Ozawa H. Mapping of Notch signaling in the developing organ of Corti in common marmosets. Front Neuroanat 2023; 17:1188886. [PMID: 37351521 PMCID: PMC10282542 DOI: 10.3389/fnana.2023.1188886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 05/24/2023] [Indexed: 06/24/2023] Open
Abstract
Introduction The well-regulated development of the sensory epithelium is essential for hearing. This process involves the specification of a pro-sensory epithelium containing common progenitors that differentiate into hair and supporting cells. Notch signaling is one of the most critical pathways during these processes, and its modification is thought to be a feasible approach for treating hearing loss. Despite interspecies differences between rodents and primates or humans, most of our current knowledge regarding cochlear development has been obtained from rodent models. Methods We therefore examined and mapped the expression patterns of Notch signal components in the developing cochlea of the common marmoset (Callithrix jacchus), a small monkey species native to the New World, a primate model animal. Results In contrast to the preserved expression patterns of the Notch signaling components in the hair cell differentiation between primates and rodents, we unveiled relatively large interspecies differences during the maturation of supporting cells. Discussion This improved knowledge of Notch signaling during primate cochlear development will facilitate the development of future regenerative therapies.
Collapse
Affiliation(s)
- Makoto Hosoya
- Department of Otolaryngology, Head and Neck Surgery, Keio University School of Medicine, Shinjuku City, Japan
| | - Masato Fujioka
- Department of Molecular Genetics, Kitasato University School of Medicine, Sagamihara, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Shinjuku City, Japan
- Laboratory for Marmoset Neural Architecture, Center for Brain Science, Saitama, Japan
| | - Hiroyuki Ozawa
- Department of Otolaryngology, Head and Neck Surgery, Keio University School of Medicine, Shinjuku City, Japan
| |
Collapse
|
5
|
Flores-Flores M, Muñoz-Nava LM, Rodríguez-Muñoz R, Zartman J, Nahmad M. Vestigial-dependent induction contributes to robust patterning but is not essential for wing-fate recruitment in Drosophila. Biol Open 2023; 12:bio059908. [PMID: 37199309 PMCID: PMC10214856 DOI: 10.1242/bio.059908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 03/16/2023] [Indexed: 05/19/2023] Open
Abstract
Cell recruitment is a process by which a differentiated cell induces neighboring cells to adopt its same cell fate. In Drosophila, cells expressing the protein encoded by the wing selector gene, vestigial (vg), drive a feed-forward recruitment signal that expands the Vg pattern as a wave front. However, previous studies on Vg pattern formation do not reveal these dynamics. Here, we use live imaging to show that multiple cells at the periphery of the wing disc simultaneously activate a fluorescent reporter of the recruitment signal, suggesting that cells may be recruited without the need for their contact neighbors be recruited in advance. In support of this observation, when Vg expression is inhibited either at the dorsal-ventral boundary or away from it, the activation of the recruitment signal still occurs at a distance, suggesting that Vg expression is not absolutely required to send or propagate the recruitment signal. However, the strength and extent of the recruitment signal is clearly compromised. We conclude that a feed-forward, contact-dependent cell recruitment process is not essential for Vg patterning, but it is necessary for robustness. Overall, our findings reveal a previously unidentified role of cell recruitment as a robustness-conferring cell differentiation mechanism.
Collapse
Affiliation(s)
- Marycruz Flores-Flores
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City 07360, Mexico
- Department of Chemical and Biomolecular Engineering, Notre Dame University, Notre Dame, IN 46556, USA
| | - Luis Manuel Muñoz-Nava
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City 07360, Mexico
| | - Rafael Rodríguez-Muñoz
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City 07360, Mexico
| | - Jeremiah Zartman
- Department of Chemical and Biomolecular Engineering, Notre Dame University, Notre Dame, IN 46556, USA
| | - Marcos Nahmad
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City 07360, Mexico
| |
Collapse
|
6
|
Abstract
Notch signaling is a highly conserved signaling pathway that coordinates cellular differentiation during the development and homeostasis in numerous organs and tissues across metazoans. Activation of Notch signaling relies on direct contact between neighboring cells and mechanical pulling of the Notch receptors by the Notch ligands. Notch signaling is commonly used in developmental processes to coordinate the differentiation into distinct cell fates of neighboring cells. In this Development at a Glance article, we describe the current understanding of the Notch pathway activation and the different regulatory levels that control the pathway. We then describe several developmental processes where Notch is crucial for coordinating differentiation. These examples include processes that are largely based on lateral inhibition mechanisms giving rise to alternating patterns (e.g. SOP selection, hair cell in the inner ear and neural stem cell maintenance), as well as processes where Notch activity is oscillatory (e.g. somitogenesis and neurogenesis in mammals).
Collapse
Affiliation(s)
- Oren Gozlan
- School of Neurobiology, Biochemistry, and Biophysics, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - David Sprinzak
- School of Neurobiology, Biochemistry, and Biophysics, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
7
|
Chen X, Wan H, Bai Y, Zhang Y, Hua Q. Advances in Understanding the Notch Signaling Pathway in the Cochlea. Curr Pharm Des 2023; 29:3266-3273. [PMID: 37990430 DOI: 10.2174/0113816128273532231103110910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/17/2023] [Indexed: 11/23/2023]
Abstract
The cochlear structure is highly complex and specific, and its development is regulated by multiple signaling pathways. Abnormalities in cochlear development can lead to different degrees of loss of function. Hair cells (HCs), which are difficult to regenerate in the mature mammalian cochlea, are susceptible to damage from noise and ototoxic drugs, and damage to HCs can cause hearing loss to varying degrees. Notch, a classical developmental signaling molecule, has been shown to be closely associated with embryonic cochlear development and plays an important role in HC regeneration in mammals, suggesting that the Notch signaling pathway may be a potential therapeutic target for cochlear development and hearing impairment due to HC damage. In recent years, the important role of the Notch signaling pathway in the cochlea has received increasing attention. In this paper, we review the role of Notch signaling in cochlear development and HC regeneration, with the aim of providing new research ideas for the prevention and treatment of related diseases.
Collapse
Affiliation(s)
- Xiaoying Chen
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Huanzhi Wan
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yutong Bai
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yuanyuan Zhang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Qingquan Hua
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
8
|
Deletion of the Notch ligand Jagged1 during cochlear maturation leads to inner hair cell defects and hearing loss. Cell Death Dis 2022; 13:971. [PMID: 36400760 PMCID: PMC9674855 DOI: 10.1038/s41419-022-05380-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 11/19/2022]
Abstract
The mammalian cochlea is an exceptionally well-organized epithelium composed of hair cells, supporting cells, and innervating neurons. Loss or defects in any of these cell types, particularly the specialized sensory hair cells, leads to deafness. The Notch pathway is known to play a critical role in the decision to become either a hair cell or a supporting cell during embryogenesis; however, little is known about how Notch functions later during cochlear maturation. Uniquely amongst Notch ligands, Jagged1 (JAG1) is localized to supporting cells during cell fate acquisition and continues to be expressed into adulthood. Here, we demonstrate that JAG1 in maturing cochlear supporting cells is essential for normal cochlear function. Specifically, we show that deletion of JAG1 during cochlear maturation disrupts the inner hair cell pathway and leads to a type of deafness clinically similar to auditory neuropathy. Common pathologies associated with disruptions in inner hair cell function, including loss of hair cells, synapses, or auditory neurons, were not observed in JAG1 mutant cochleae. Instead, RNA-seq analysis of JAG1-deficient cochleae identified dysregulation of the Rho GTPase pathway, known to be involved in stereocilia development and maintenance. Interestingly, the overexpression of one of the altered genes, Diaph3, is responsible for autosomal dominant auditory neuropathy-1 (AUNA1) in humans and mice, and is associated with defects in the inner hair cell stereocilia. Strikingly, ultrastructural analyses of JAG1-deleted cochleae revealed stereocilia defects in inner hair cells, including fused and elongated bundles, that were similar to those stereocilia defects reported in AUNA1 mice. Taken together, these data indicate a novel role for Notch signaling in normal hearing development through maintaining stereocilia integrity of the inner hair cells during cochlear maturation.
Collapse
|
9
|
Kelley MW. Cochlear Development; New Tools and Approaches. Front Cell Dev Biol 2022; 10:884240. [PMID: 35813214 PMCID: PMC9260282 DOI: 10.3389/fcell.2022.884240] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/19/2022] [Indexed: 12/21/2022] Open
Abstract
The sensory epithelium of the mammalian cochlea, the organ of Corti, is comprised of at least seven unique cell types including two functionally distinct types of mechanosensory hair cells. All of the cell types within the organ of Corti are believed to develop from a population of precursor cells referred to as prosensory cells. Results from previous studies have begun to identify the developmental processes, lineage restrictions and signaling networks that mediate the specification of many of these cell types, however, the small size of the organ and the limited number of each cell type has hampered progress. Recent technical advances, in particular relating to the ability to capture and characterize gene expression at the single cell level, have opened new avenues for understanding cellular specification in the organ of Corti. This review will cover our current understanding of cellular specification in the cochlea, discuss the most commonly used methods for single cell RNA sequencing and describe how results from a recent study using single cell sequencing provided new insights regarding cellular specification.
Collapse
|
10
|
Ray M, Rath SN, Sarkar S, Sable MN. Presentation of potential genes and deleterious variants associated with non-syndromic hearing loss: a computational approach. Genomics Inform 2022; 20:e5. [PMID: 35399004 PMCID: PMC9001992 DOI: 10.5808/gi.21070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 02/17/2022] [Indexed: 11/20/2022] Open
Abstract
Non-syndromic hearing loss (NSHL) is a common hereditary disorder. Both clinical and genetic heterogeneity has created many obstacles to understanding the causes of NSHL. The present study has attempted to ravel the genetic aetiology in NSHL progression and to screen out potential target genes using computational approaches. The reported NSHL target genes (2009-2020) have been studied by analyzing different biochemical and signaling pathways, interpretation of their functional association network, and discovery of important regulatory interactions with three previously established miRNAs in the human inner ear as well as in NSHL such as miR-183, miR-182, and miR-96. This study has identified SMAD4 and SNAI2 as the most putative target genes of NSHL. But pathogenic and deleterious non-synonymous single nucleotide polymorphisms discovered within SMAD4 is anticipated to have an impact on NSHL progression. Additionally, the identified deleterious variants in the functional domains of SMAD4 added a supportive clue for further study. Thus, the identified deleterious variant i.e., rs377767367 (G491V) in SMAD4 needs further clinical validation. The present outcomes would provide insights into the genetics of NSHL progression.
Collapse
Affiliation(s)
- Manisha Ray
- Department of Pathology and Lab Medicine, All India Institute of Medical Sciences, Bhubaneswar, Odisha 751019, India
| | - Surya Narayan Rath
- Department of Bioinformatics, Odisha University of Agriculture and Technology, Bhubaneswar, Odisha 751003, India
| | - Saurav Sarkar
- Department of Ear Nose Throat, All India Institute of Medical Sciences, Bhubaneswar, Odisha 751019, India
| | - Mukund Namdev Sable
- Department of Pathology and Lab Medicine, All India Institute of Medical Sciences, Bhubaneswar, Odisha 751019, India
| |
Collapse
|
11
|
Jiang P, Ma X, Han S, Ma L, Ai J, Wu L, Zhang Y, Xiao H, Tian M, Tao WA, Zhang S, Chai R. Characterization of the microRNA transcriptomes and proteomics of cochlear tissue-derived small extracellular vesicles from mice of different ages after birth. Cell Mol Life Sci 2022; 79:154. [PMID: 35218422 PMCID: PMC11072265 DOI: 10.1007/s00018-022-04164-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 12/30/2021] [Accepted: 01/23/2022] [Indexed: 12/22/2022]
Abstract
The cochlea is an important sensory organ for both balance and sound perception, and the formation of the cochlea is a complex developmental process. The development of the mouse cochlea begins on embryonic day (E)9 and continues until postnatal day (P)21 when the hearing system is considered mature. Small extracellular vesicles (sEVs), with a diameter ranging from 30 to 200 nm, have been considered a significant medium for information communication in both physiological and pathological processes. However, there are no studies exploring the role of sEVs in the development of the cochlea. Here, we isolated tissue-derived sEVs from the cochleae of FVB mice at P3, P7, P14, and P21 by ultracentrifugation. These sEVs were first characterized by transmission electron microscopy, nanoparticle tracking analysis, and western blotting. Next, we used small RNA-seq and mass spectrometry to characterize the microRNA transcriptomes and proteomes of cochlear sEVs from mice at different ages. Many microRNAs and proteins were discovered to be related to inner ear development, anatomical structure development, and auditory nervous system development. These results all suggest that sEVs exist in the cochlea and are likely to be essential for the normal development of the auditory system. Our findings provide many sEV microRNA and protein targets for future studies of the roles of cochlear sEVs.
Collapse
Affiliation(s)
- Pei Jiang
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Xiangyu Ma
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Shanying Han
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Leyao Ma
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Jingru Ai
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Leilei Wu
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Yuan Zhang
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Hairong Xiao
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Mengyao Tian
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - W Andy Tao
- Department of Chemistry, Department of Biochemistry, Purdue University, West Lafayette, Indiana, 47907, USA.
- Center for Cancer Research, Purdue University, West Lafayette, Indiana, 47907, USA.
| | - Shasha Zhang
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
| | - Renjie Chai
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing, China.
- Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
12
|
He Z, Ding Y, Mu Y, Xu X, Kong W, Chai R, Chen X. Stem Cell-Based Therapies in Hearing Loss. Front Cell Dev Biol 2021; 9:730042. [PMID: 34746126 PMCID: PMC8567027 DOI: 10.3389/fcell.2021.730042] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 10/04/2021] [Indexed: 12/19/2022] Open
Abstract
In recent years, neural stem cell transplantation has received widespread attention as a new treatment method for supplementing specific cells damaged by disease, such as neurodegenerative diseases. A number of studies have proved that the transplantation of neural stem cells in multiple organs has an important therapeutic effect on activation and regeneration of cells, and restore damaged neurons. This article describes the methods for inducing the differentiation of endogenous and exogenous stem cells, the implantation operation and regulation of exogenous stem cells after implanted into the inner ear, and it elaborates the relevant signal pathways of stem cells in the inner ear, as well as the clinical application of various new materials. At present, stem cell therapy still has limitations, but the role of this technology in the treatment of hearing diseases has been widely recognized. With the development of related research, stem cell therapy will play a greater role in the treatment of diseases related to the inner ear.
Collapse
Affiliation(s)
- Zuhong He
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yanyan Ding
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yurong Mu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxiang Xu
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Weijia Kong
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Renjie Chai
- State Key Laboratory of Bioelectronics, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, School of Life Sciences and Technology, Southeast University, Nanjing, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China.,Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Xiong Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
13
|
Erni ST, Gill JC, Palaferri C, Fernandes G, Buri M, Lazarides K, Grandgirard D, Edge ASB, Leib SL, Roccio M. Hair Cell Generation in Cochlear Culture Models Mediated by Novel γ-Secretase Inhibitors. Front Cell Dev Biol 2021; 9:710159. [PMID: 34485296 PMCID: PMC8414802 DOI: 10.3389/fcell.2021.710159] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 07/26/2021] [Indexed: 12/30/2022] Open
Abstract
Sensorineural hearing loss is prevalent within society affecting the quality of life of 460 million worldwide. In the majority of cases, this is due to insult or degeneration of mechanosensory hair cells in the cochlea. In adult mammals, hair cell loss is irreversible as sensory cells are not replaced spontaneously. Genetic inhibition of Notch signaling had been shown to induce hair cell formation by transdifferentiation of supporting cells in young postnatal rodents and provided an impetus for targeting Notch pathway with small molecule inhibitors for hearing restoration. Here, the oto-regenerative potential of different γ-secretase inhibitors (GSIs) was evaluated in complementary assay models, including cell lines, organotypic cultures of the organ of Corti and cochlear organoids to characterize two novel GSIs (CPD3 and CPD8). GSI-treatment induced hair cell gene expression in all these models and was effective in increasing hair cell numbers, in particular outer hair cells, both in baseline conditions and in response to ototoxic damage. Hair cells were generated from transdifferentiation of supporting cells. Similar findings were obtained in cochlear organoid cultures, used for the first time to probe regeneration following sisomicin-induced damage. Finally, effective absorption of a novel GSI through the round window membrane and hair cell induction was attained in a whole cochlea culture model and in vivo pharmacokinetic comparisons of transtympanic delivery of GSIs and different vehicle formulations were successfully conducted in guinea pigs. This preclinical evaluation of targeting Notch signaling with novel GSIs illustrates methods of characterization for hearing restoration molecules, enabling translation to more complex animal studies and clinical research.
Collapse
Affiliation(s)
- Silvia T Erni
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Laboratory of Inner Ear Research, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - John C Gill
- Audion Therapeutics B.V., Amsterdam, Netherlands
| | - Carlotta Palaferri
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Laboratory of Inner Ear Research, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Gabriella Fernandes
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Laboratory of Inner Ear Research, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Michelle Buri
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Laboratory of Inner Ear Research, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | | | - Denis Grandgirard
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Albert S B Edge
- Massachusetts Eye and Ear, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States.,Harvard Stem Cell Institute, Cambridge, MA, United States
| | - Stephen L Leib
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Marta Roccio
- Cluster for Regenerative Neuroscience, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Laboratory of Inner Ear Research, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich, Zurich, Switzerland.,Department of Otorhinolaryngology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
14
|
Liu H, Giffen KP, Grati M, Morrill SW, Li Y, Liu X, Briegel KJ, He DZ. Transcription co-factor LBH is necessary for the survival of cochlear hair cells. J Cell Sci 2021; 134:237781. [PMID: 33674448 DOI: 10.1242/jcs.254458] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 02/11/2021] [Indexed: 11/20/2022] Open
Abstract
Hearing loss affects ∼10% of adults worldwide. Most sensorineural hearing loss is caused by the progressive loss of mechanosensitive hair cells (HCs) in the cochlea. The molecular mechanisms underlying HC maintenance and loss remain poorly understood. LBH, a transcription co-factor implicated in development, is abundantly expressed in outer hair cells (OHCs). We used Lbh-null mice to identify its role in HCs. Surprisingly, Lbh deletion did not affect differentiation and the early development of HCs, as nascent HCs in Lbh knockout mice had normal looking stereocilia. The stereocilia bundle was mechanosensitive and OHCs exhibited the characteristic electromotility. However, Lbh-null mice displayed progressive hearing loss, with stereocilia bundle degeneration and OHC loss as early as postnatal day 12. RNA-seq analysis showed significant gene enrichment of biological processes related to transcriptional regulation, cell cycle, DNA damage/repair and autophagy in Lbh-null OHCs. In addition, Wnt and Notch pathway-related genes were found to be dysregulated in Lbh-deficient OHCs. Our study implicates, for the first time, loss of LBH function in progressive hearing loss, and demonstrates a critical requirement of LBH in promoting HC survival in adult mice.
Collapse
Affiliation(s)
- Huizhan Liu
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Kimberlee P Giffen
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - M'Hamed Grati
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Seth W Morrill
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Yi Li
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE 68178, USA.,Department of Otorhinolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, 100730 Beijing, China
| | - Xuezhong Liu
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Karoline J Briegel
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - David Z He
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE 68178, USA
| |
Collapse
|
15
|
The Notch Ligand Jagged1 Is Required for the Formation, Maintenance, and Survival of Hensen's Cells in the Mouse Cochlea. J Neurosci 2020; 40:9401-9413. [PMID: 33127852 DOI: 10.1523/jneurosci.1192-20.2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 10/16/2020] [Accepted: 10/23/2020] [Indexed: 01/09/2023] Open
Abstract
During cochlear development, the Notch ligand JAGGED 1 (JAG1) plays an important role in the specification of the prosensory region, which gives rise to sound-sensing hair cells and neighboring supporting cells (SCs). While JAG1's expression is maintained in SCs through adulthood, the function of JAG1 in SC development is unknown. Here, we demonstrate that JAG1 is essential for the formation and maintenance of Hensen's cells, a highly specialized SC subtype located at the edge of the auditory epithelium. Using Sox2 CreERT2/+::Jag1loxP/loxP mice of both genders, we show that Jag1 deletion at the onset of differentiation, at embryonic day 14.5, disrupted Hensen's cell formation. Similar loss of Hensen's cells was observed when Jag1 was deleted after Hensen's cell formation at postnatal day (P) 0/P1 and fate-mapping analysis revealed that in the absence of Jag1, some Hensen's cells die, but others convert into neighboring Claudius cells. In support of a role for JAG1 in cell survival, genes involved in mitochondrial function and protein synthesis were downregulated in the sensory epithelium of P0 cochlea lacking Jag1 Finally, using Fgfr3-iCreERT2 ::Jag1loxP/loxP mice to delete Jag1 at P0, we observed a similar loss of Hensen's cells and found that adult Jag1 mutant mice have hearing deficits at the low-frequency range.SIGNIFICANCE STATEMENT Hensen's cells play an essential role in the development and homeostasis of the cochlea. Defects in the biophysical or functional properties of Hensen's cells have been linked to auditory dysfunction and hearing loss. Despite their importance, surprisingly little is known about the molecular mechanisms that guide their development. Morphologic and fate-mapping analyses in our study revealed that, in the absence of the Notch ligand JAGGED1, Hensen's cells died or converted into Claudius cells, which are specialized epithelium-like cells outside the sensory epithelium. Confirming a link between JAGGED1 and cell survival, transcriptional profiling showed that JAGGED1 maintains genes critical for mitochondrial function and tissue homeostasis. Finally, auditory phenotyping revealed that JAGGED1's function in supporting cells is necessary for low-frequency hearing.
Collapse
|
16
|
Wall SM, Verlander JW, Romero CA. The Renal Physiology of Pendrin-Positive Intercalated Cells. Physiol Rev 2020; 100:1119-1147. [PMID: 32347156 PMCID: PMC7474261 DOI: 10.1152/physrev.00011.2019] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 11/06/2019] [Accepted: 11/12/2019] [Indexed: 12/13/2022] Open
Abstract
Intercalated cells (ICs) are found in the connecting tubule and the collecting duct. Of the three IC subtypes identified, type B intercalated cells are one of the best characterized and known to mediate Cl- absorption and HCO3- secretion, largely through the anion exchanger pendrin. This exchanger is thought to act in tandem with the Na+-dependent Cl-/HCO3- exchanger, NDCBE, to mediate net NaCl absorption. Pendrin is stimulated by angiotensin II and aldosterone administration via the angiotensin type 1a and the mineralocorticoid receptors, respectively. It is also stimulated in models of metabolic alkalosis, such as with NaHCO3 administration. In some rodent models, pendrin-mediated HCO3- secretion modulates acid-base balance. However, of probably more physiological or clinical significance is the role of these pendrin-positive ICs in blood pressure regulation, which occurs, at least in part, through pendrin-mediated renal Cl- absorption, as well as their effect on the epithelial Na+ channel, ENaC. Aldosterone stimulates ENaC directly through principal cell mineralocorticoid hormone receptor (ligand) binding and also indirectly through its effect on pendrin expression and function. In so doing, pendrin contributes to the aldosterone pressor response. Pendrin may also modulate blood pressure in part through its action in the adrenal medulla, where it modulates the release of catecholamines, or through an indirect effect on vascular contractile force. In addition to its role in Na+ and Cl- balance, pendrin affects the balance of other ions, such as K+ and I-. This review describes how aldosterone and angiotensin II-induced signaling regulate pendrin and the contribution of pendrin-positive ICs in the kidney to distal nephron function and blood pressure.
Collapse
Affiliation(s)
- Susan M Wall
- Departments of Medicine and Physiology, Emory University School of Medicine, Atlanta, Georgia; and Department of Medicine, University of Florida, Gainesville, Florida
| | - Jill W Verlander
- Departments of Medicine and Physiology, Emory University School of Medicine, Atlanta, Georgia; and Department of Medicine, University of Florida, Gainesville, Florida
| | - Cesar A Romero
- Departments of Medicine and Physiology, Emory University School of Medicine, Atlanta, Georgia; and Department of Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|
17
|
Assmus AM, Mullins JJ, Brown CM, Mullins LJ. Cellular plasticity: A mechanism for homeostasis in the kidney. Acta Physiol (Oxf) 2020; 229:e13447. [PMID: 31991057 DOI: 10.1111/apha.13447] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/15/2020] [Accepted: 01/24/2020] [Indexed: 12/30/2022]
Abstract
Cellular plasticity is a topical subject with interest spanning a wide range of fields from developmental biology to regenerative medicine. Even the nomenclature is a subject of debate, and the underlying mechanisms are still under investigation. On top of injury repair, cell plasticity is a constant physiological process in adult organisms and tissues, in response to homeostatic challenges. In this review we discuss two examples of plasticity for the maintenance of homeostasis in the renal system-namely the renin-producing juxtaglomerular cells (JG cells) and cortical collecting duct (CCD) cells. JG cells show plasticity through recruitment mechanisms, answering the demand for an increase in renin production. In the CCD, cells appear to have the ability to transdifferentiate between principal and intercalated cells to help maintain the highly regulated solute transport levels of that segment. These two cases highlight the complexity of plasticity processes and the role they can play in the kidney.
Collapse
Affiliation(s)
- Adrienne M. Assmus
- The University of Edinburgh ‐ Cardiovascular Science (CVS) Queen's Medical Research Institute Edinburgh Scotland UK
| | - John J. Mullins
- The University of Edinburgh ‐ Cardiovascular Science (CVS) Queen's Medical Research Institute Edinburgh Scotland UK
| | - Cara M. Brown
- The University of Edinburgh ‐ Cardiovascular Science (CVS) Queen's Medical Research Institute Edinburgh Scotland UK
| | - Linda J. Mullins
- The University of Edinburgh ‐ Cardiovascular Science (CVS) Queen's Medical Research Institute Edinburgh Scotland UK
| |
Collapse
|
18
|
Forge A, Jagger DJ, Gale JE. Restoring the balance: regeneration of hair cells in the vestibular system of the inner ear. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2019.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
19
|
Notch-mediated lateral induction is necessary to maintain vestibular prosensory identity during inner ear development. Dev Biol 2020; 462:74-84. [PMID: 32147304 DOI: 10.1016/j.ydbio.2020.02.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 01/24/2023]
Abstract
The five vestibular organs of the inner ear derive from patches of prosensory cells that express the transcription factor SOX2 and the Notch ligand JAG1. Previous work suggests that JAG1-mediated Notch signaling is both necessary and sufficient for prosensory formation and that the separation of developing prosensory patches is regulated by LMX1a, which antagonizes Notch signaling. We used an inner ear-specific deletion of the Rbpjκ gene in which Notch signaling is progressively lost from the inner ear to show that Notch signaling, is continuously required for the maintenance of prosensory fate. Loss of Notch signaling in prosensory patches causes them to shrink and ultimately disappear. We show this loss of prosensory fate is not due to cell death, but rather to the conversion of prosensory tissue into non-sensory tissue that expresses LMX1a. Notch signaling is therefore likely to stabilize, rather than induce prosensory fate.
Collapse
|
20
|
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
|
21
|
Tian C, Johnson KR. TBX1 is required for normal stria vascularis and semicircular canal development. Dev Biol 2019; 457:91-103. [PMID: 31550482 DOI: 10.1016/j.ydbio.2019.09.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/12/2019] [Accepted: 09/20/2019] [Indexed: 12/15/2022]
Abstract
Little is known about the role of TBX1 in post-otocyst stages of inner ear development. Here, we report on mice with a missense mutation of Tbx1 that are viable with fully developed but abnormally formed inner ears. Mutant mice are deaf due to an undeveloped stria vascularis and show vestibular dysfunction associated with abnormal semicircular canal formation. We show that TBX1 is expressed in endolymph-producing strial marginal cells and vestibular dark cells of the inner ear and is an upstream regulator of Esrrb, which previously was shown to control the developmental fate of these cells. We also show that TBX1 is expressed in sensory cells of the crista ampullaris, which may relate to the semicircular canal abnormalities observed in mutant mice. Inner ears of mutant embryos have a non-resorbed fusion plate in the posterior semicircular canal and a single ampulla connecting anterior and lateral canals. We hypothesize that the TBX1 missense mutation prevents binding with specific co-regulatory proteins. These findings reveal previously unknown functions of TBX1 during later stages of inner ear development.
Collapse
Affiliation(s)
- Cong Tian
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA
| | | |
Collapse
|
22
|
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
|
23
|
Jung JS, Zhang KD, Wang Z, McMurray M, Tkaczuk A, Ogawa Y, Hertzano R, Coate TM. Semaphorin-5B Controls Spiral Ganglion Neuron Branch Refinement during Development. J Neurosci 2019; 39:6425-6438. [PMID: 31209173 PMCID: PMC6697390 DOI: 10.1523/jneurosci.0113-19.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 05/03/2019] [Accepted: 06/10/2019] [Indexed: 01/30/2023] Open
Abstract
During nervous system development, axons often undergo elaborate changes in branching patterns before circuits have achieved their mature patterns of innervation. In the auditory system, type I spiral ganglion neurons (SGNs) project their peripheral axons into the cochlear epithelium and then undergo a process of branch refinement before forming synapses with sensory hair cells. Here, we report that Semaphorin-5B (Sema5B) acts as an important mediator of this process. During cochlear development in mouse, immature hair cells express Sema5B, whereas the SGNs express both PlexinA1 and PlexinA3, which are known Sema5B receptors. In these studies, genetic sparse labeling and three-dimensional reconstruction techniques were leveraged to determine the morphologies of individual type I SGNs after manipulations of Sema5B signaling. Treating cultured mouse cochleae with Sema5B-Fc (to activate Plexin-As) led to type I SGNs with less numerous, but longer terminal branches. Conversely, cochleae from Sema5b knock-out mice showed type I SGNs with more numerous, but shorter terminal branches. In addition, conditional loss of Plxna1 in SGNs (using Bhlhb5Cre) led to increased type I SGN branching, suggesting that PlexinA1 normally responds to Sema5B in this process. In these studies, mice of either sex were used. The data presented here suggest that Sema5B-PlexinA1 signaling limits SGN terminal branch numbers without causing axonal repulsion, which is a role that distinguishes Sema5B from other Semaphorins in cochlear development.SIGNIFICANCE STATEMENT The sensorineural components of the cochlea include hair cells, which respond mechanically to sound waves, and afferent spiral ganglion neurons (SGNs), which respond to glutamate released by hair cells and transmit auditory information into the CNS. An important component of synapse formation in the cochlea is a process of SGN "debranching" whereby SGNs lose extraneous branches before developing unramified bouton endings that contact the hair cells. In this work, we have found that the transmembrane ligand Semaphorin-5B and its receptor PlexinA1 regulate the debranching process. The results in this report provide new knowledge regarding the molecular control of cochlear afferent innervation.
Collapse
Affiliation(s)
- Johnny S Jung
- Department of Biology, Georgetown University, Washington, DC 20007, and
| | - Kaidi D Zhang
- Department of Biology, Georgetown University, Washington, DC 20007, and
| | - Zhirong Wang
- Department of Biology, Georgetown University, Washington, DC 20007, and
| | - Mark McMurray
- Departments of Otorhinolaryngology Head and Neck Surgery
| | - Andrew Tkaczuk
- Departments of Otorhinolaryngology Head and Neck Surgery
| | - Yoko Ogawa
- Departments of Otorhinolaryngology Head and Neck Surgery
| | - Ronna Hertzano
- Departments of Otorhinolaryngology Head and Neck Surgery
- Anatomy and Neurobiology, and
- Institute for Genome Sciences, University of Maryland School of Medicine, University of Maryland, Baltimore, Maryland 21201
| | - Thomas M Coate
- Department of Biology, Georgetown University, Washington, DC 20007, and
| |
Collapse
|
24
|
Samarajeewa A, Jacques BE, Dabdoub A. Therapeutic Potential of Wnt and Notch Signaling and Epigenetic Regulation in Mammalian Sensory Hair Cell Regeneration. Mol Ther 2019; 27:904-911. [PMID: 30982678 PMCID: PMC6520458 DOI: 10.1016/j.ymthe.2019.03.017] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 02/07/2023] Open
Abstract
Hearing loss is one of the most prevalent sensory deficits worldwide and can result from the death of mechanosensory hair cells that transduce auditory signals in the cochlea. The mammalian cochlea lacks the capacity to regenerate these hair cells once damaged, and currently there are no biological therapies for hearing loss. Understanding the signaling pathways responsible for hair cell development can inform regenerative strategies and identify targets for treating hearing loss. The canonical Wnt and Notch pathways are critical for cochlear development; they converge on several key molecules, such as Atoh1, to regulate prosensory specification, proliferation, hair cell differentiation, and cellular organization. Much work has focused on Wnt and Notch modulation in the neonatal mouse cochlea, where they can promote hair cell regeneration. However, this regenerative response is limited in the adult cochlea and this might be attributed to age-dependent epigenetic modifications. Indeed, the epigenetic status at key gene loci undergoes dynamic changes during cochlear development, maturation, and aging. Therefore, strategies to improve regenerative success in the adult cochlea might require the modulation of Wnt, Notch, or other pathways, as well as targeted epigenetic modifications to alter the activity of key genes critical for supporting cell proliferation or transdifferentiation.
Collapse
Affiliation(s)
- Anshula Samarajeewa
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | | | - Alain Dabdoub
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Biological Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; Department of Otolaryngology - Head & Neck Surgery, University of Toronto, Toronto, ON M5G 2C4, Canada.
| |
Collapse
|
25
|
Samarajeewa A, Lenz DR, Xie L, Chiang H, Kirchner R, Mulvaney JF, Edge ASB, Dabdoub A. Transcriptional response to Wnt activation regulates the regenerative capacity of the mammalian cochlea. Development 2018; 145:dev.166579. [PMID: 30389848 PMCID: PMC6288390 DOI: 10.1242/dev.166579] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 10/25/2018] [Indexed: 01/04/2023]
Abstract
Lack of sensory hair cell (HC) regeneration in mammalian adults is a major contributor to hearing loss. In contrast, the neonatal mouse cochlea retains a transient capacity for regeneration, and forced Wnt activation in neonatal stages promotes supporting cell (SC) proliferation and induction of ectopic HCs. We currently know little about the temporal pattern and underlying mechanism of this age-dependent regenerative response. Using an in vitro model, we show that Wnt activation promotes SC proliferation following birth, but prior to postnatal day (P) 5. This age-dependent decline in proliferation occurs despite evidence that the Wnt pathway is postnatally active and can be further enhanced by Wnt stimulators. Using an in vivo mouse model and RNA sequencing, we show that proliferation in the early neonatal cochlea is correlated with a unique transcriptional response that diminishes with age. Furthermore, we find that augmenting Wnt signaling through the neonatal stages extends the window for HC induction in response to Notch signaling inhibition. Our results suggest that the downstream transcriptional response to Wnt activation, in part, underlies the regenerative capacity of the mammalian cochlea. Summary: Canonical Wnt activation in the mammalian cochlea elicits a unique, age-dependent transcriptional response, which in part regulates the regenerative capacity of supporting cells during cochlear maturation.
Collapse
Affiliation(s)
- Anshula Samarajeewa
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto ON, M5S 1A8, Canada
| | - Danielle R Lenz
- Department of Otolaryngology, Harvard Medical School, Boston, MA 02114, USA.,Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Lihong Xie
- Biological Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto ON, M4N 3M5, Canada.,Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning Guangxi, 530021, China
| | - Hao Chiang
- Department of Otolaryngology, Harvard Medical School, Boston, MA 02114, USA.,Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Rory Kirchner
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Joanna F Mulvaney
- Biological Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto ON, M4N 3M5, Canada
| | - Albert S B Edge
- Department of Otolaryngology, Harvard Medical School, Boston, MA 02114, USA.,Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Alain Dabdoub
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto ON, M5S 1A8, Canada .,Biological Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto ON, M4N 3M5, Canada.,Department of Otolaryngology - Head & Neck Surgery, University of Toronto, Toronto ON, M5G 2C4, Canada
| |
Collapse
|
26
|
Pérez Saturnino A, Lust K, Wittbrodt J. Notch signalling patterns retinal composition by regulating atoh7 during post-embryonic growth. Development 2018; 145:dev.169698. [PMID: 30337377 PMCID: PMC6240314 DOI: 10.1242/dev.169698] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 10/09/2018] [Indexed: 01/01/2023]
Abstract
Patterning of a continuously growing naive field in the context of a life-long growing organ such as the teleost eye is of high functional relevance. Intrinsic and extrinsic signals have been proposed to regulate lineage specification in progenitors that exit the stem cell niche in the ciliary marginal zone (CMZ). The proper cell-type composition arising from those progenitors is a prerequisite for retinal function. Our findings in the teleost medaka (Oryzias latipes) uncover that the Notch-Atoh7 axis continuously patterns the CMZ. The complement of cell types originating from the two juxtaposed progenitors marked by Notch or Atoh7 activity contains all constituents of a retinal column. Modulation of Notch signalling specifically in Atoh7-expressing cells demonstrates the crucial role of this axis in generating the correct cell-type proportions. After transiently blocking Notch signalling, retinal patterning and differentiation is re-initiated de novo. Taken together, our data show that Notch activity in the CMZ continuously structures the growing retina by juxtaposing Notch and Atoh7 progenitors that give rise to distinct complementary lineages, revealing coupling of de novo patterning and cell-type specification in the respective lineages. Summary: Mutually exclusive activity of Notch and Atoh7 in the ciliary marginal zone gives rise to two distinct lineages resulting in specification of the full complement of cell types in medaka retina.
Collapse
Affiliation(s)
- Alicia Pérez Saturnino
- Centre for Organismal Studies, Heidelberg University, Heidelberg 69120, Germany.,Heidelberg Biosciences International Graduate School (HBIGS), Heidelberg 69120, Germany
| | - Katharina Lust
- Centre for Organismal Studies, Heidelberg University, Heidelberg 69120, Germany
| | - Joachim Wittbrodt
- Centre for Organismal Studies, Heidelberg University, Heidelberg 69120, Germany
| |
Collapse
|
27
|
Taylor RR, Filia A, Paredes U, Asai Y, Holt JR, Lovett M, Forge A. Regenerating hair cells in vestibular sensory epithelia from humans. eLife 2018; 7:34817. [PMID: 30019672 PMCID: PMC6078492 DOI: 10.7554/elife.34817] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 07/16/2018] [Indexed: 01/28/2023] Open
Abstract
Human vestibular sensory epithelia in explant culture were incubated in gentamicin to ablate hair cells. Subsequent transduction of supporting cells with ATOH1 using an Ad-2 viral vector resulted in generation of highly significant numbers of cells expressing the hair cell marker protein myosin VIIa. Cells expressing myosin VIIa were also generated after blocking the Notch signalling pathway with TAPI-1 but less efficiently. Transcriptomic analysis following ATOH1 transduction confirmed up-regulation of 335 putative hair cell marker genes, including several downstream targets of ATOH1. Morphological analysis revealed numerous cells bearing dense clusters of microvilli at the apical surfaces which showed some hair cell-like characteristics confirming a degree of conversion of supporting cells. However, no cells bore organised hair bundles and several expected hair cell markers genes were not expressed suggesting incomplete differentiation. Nevertheless, the results show a potential to induce conversion of supporting cells in the vestibular sensory tissues of humans. The inner ear contains our balance system (the vestibular system) and our hearing organ (the cochlea). Their sensing units, the hair cells, detect movement or sound waves. A loss of hair cells is a major cause of inner ear disorders, such as dizziness, imbalance and deafness. When hair cells die, supporting cells that surround them close the ‘wound’ to repair the tissue. In fish, amphibians, reptiles and birds, the supporting cells can replace lost hair cells, but in mammals – including humans – hair cells are unable to regenerate in the cochlea, so hearing loss is permanent. However, previous research has shown that in certain mammals, spontaneous replacement of lost hair cells in the vestibular system can occur, but not enough to lead to a full recovery. Scientists have been able to convert supporting cells in the vestibular system of mice into hair cells by using either certain chemicals, or by introducing a specific gene into the supporting cells. In the mouse embryo, this gene, called Atoh1, switches on a signalling pathway in the inner ear, through which a non-specialised precursor cell becomes a hair cell. Inducing hair cell regeneration could be a therapy for inner ear disorders. Therefore, Taylor et al. wanted to find out if such procedures would work in inner ear tissue from humans. The researchers collected intact tissue samples from the vestibular system of patients who had undergone surgery to have a tumour removed, which would normally destroy the inner ear. All existing hair cells were removed so that mainly supporting cells remained. Then, the tissue was either treated with chemicals that increased the production of hair cells or received the gene ATOH1. The results showed that the cells containing the gene were able to develop many features characteristic of hair cells. And a smaller number of hair cells treated with the chemicals also started to develop hair cell-like features. A gene analysis after the ATOH1 transfer revealed a number of active genes known to be markers of hair cells, but also several inactive ones. This suggests that additional factors are necessary for generating fully functional hair cells. Dizziness and balance disorders present a major health care burden, particularly in the elderly population. Yet, they are often disregarded and overlooked. This study suggests that hair cell regeneration could be a feasible therapy for some forms of balance disorders linked to loss of vestibular hair cells. More research is needed to identify the other factors at play to test if hair cell regeneration in the cochlea could be used to treat hearing impairment.
Collapse
Affiliation(s)
| | - Anastasia Filia
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Ursula Paredes
- UCL Ear Institute, University College London, London, United Kingdom
| | - Yukako Asai
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, United States
| | - Jeffrey R Holt
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, United States
| | - Michael Lovett
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Andrew Forge
- UCL Ear Institute, University College London, London, United Kingdom
| |
Collapse
|
28
|
Harley RJ, Murdy JP, Wang Z, Kelly MC, Ropp TJF, Park SH, Maness PF, Manis PB, Coate TM. Neuronal cell adhesion molecule (NrCAM) is expressed by sensory cells in the cochlea and is necessary for proper cochlear innervation and sensory domain patterning during development. Dev Dyn 2018; 247:934-950. [PMID: 29536590 PMCID: PMC6105381 DOI: 10.1002/dvdy.24629] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 03/06/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND In the cochlea, auditory development depends on precise patterns of innervation by afferent and efferent nerve fibers, as well as a stereotyped arrangement of hair and supporting cells. Neuronal cell adhesion molecule (NrCAM) is a homophilic cell adhesion molecule that controls diverse aspects of nervous system development, but the function of NrCAM in cochlear development is not well understood. RESULTS Throughout cochlear innervation, NrCAM is detectable on spiral ganglion neuron (SGN) afferent and olivocochlear efferent fibers, and on the membranes of developing hair and supporting cells. Neonatal Nrcam-null cochleae show errors in type II SGN fasciculation, reduced efferent innervation, and defects in the stereotyped packing of hair and supporting cells. Nrcam loss also leads to dramatic changes in the profiles of presynaptic afferent and efferent synaptic markers at the time of hearing onset. Despite these numerous developmental defects, Nrcam-null adults do not show defects in auditory acuity, and by postnatal day 21, the developmental deficits in ribbon synapse distribution and sensory domain structure appear to have been corrected. CONCLUSIONS NrCAM is expressed by several neural and sensory epithelial subtypes within the developing cochlea, and the loss of Nrcam confers numerous, but nonpermanent, developmental defects in innervation and sensory domain patterning. Developmental Dynamics 247:934-950, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Randall J. Harley
- Department of Biology, Georgetown University, 37 and O St. NW, Regents Hall 410, Washington, DC 20007, USA
| | - Joseph P. Murdy
- Department of Biology, Georgetown University, 37 and O St. NW, Regents Hall 410, Washington, DC 20007, USA
| | - Zhirong Wang
- Department of Biology, Georgetown University, 37 and O St. NW, Regents Hall 410, Washington, DC 20007, USA
| | - Michael C. Kelly
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, 35 Convent Dr., Bethesda, MD 20892, USA
| | - Tessa-Jonne F. Ropp
- Department of Otolaryngology/Head and Neck Surgery, The University of North Carolina at Chapel Hill, B251 Marsico Hall, CB#7070, 125 Mason Farm Rd., Chapel Hill, NC 27599, USA
| | - SeHoon H. Park
- Department of Biology, Georgetown University, 37 and O St. NW, Regents Hall 410, Washington, DC 20007, USA
| | - Patricia F. Maness
- Department of Biochemistry and Biophysics, The University of North Carolina School of Medicine, 120 Mason Farm Rd., suite 3020, CB#7260, Chapel Hill, NC 27599, USA
| | - Paul B. Manis
- Department of Otolaryngology/Head and Neck Surgery and Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, B027 Marsico Hall, CB#7070. 125 Mason Farm Rd., Chapel Hill, NC 27599
| | - Thomas M. Coate
- Department of Biology, Georgetown University, 37 and O St. NW, Regents Hall 410, Washington, DC 20007, USA
| |
Collapse
|
29
|
Lahlou H, Lopez-Juarez A, Fontbonne A, Nivet E, Zine A. Modeling human early otic sensory cell development with induced pluripotent stem cells. PLoS One 2018; 13:e0198954. [PMID: 29902227 PMCID: PMC6002076 DOI: 10.1371/journal.pone.0198954] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 05/24/2018] [Indexed: 11/18/2022] Open
Abstract
The inner ear represents a promising system to develop cell-based therapies from human induced pluripotent stem cells (hiPSCs). In the developing ear, Notch signaling plays multiple roles in otic region specification and for cell fate determination. Optimizing hiPSC induction for the generation of appropriate numbers of otic progenitors and derivatives, such as hair cells, may provide an unlimited supply of cells for research and cell-based therapy. In this study, we used monolayer cultures, otic-inducing agents, Notch modulation, and marker expression to track early and otic sensory lineages during hiPSC differentiation. Otic/placodal progenitors were derived from hiPSC cultures in medium supplemented with FGF3/FGF10 for 13 days. These progenitor cells were then treated for 7 days with retinoic acid (RA) and epidermal growth factor (EGF) or a Notch inhibitor. The differentiated cultures were analyzed in parallel by qPCR and immunocytochemistry. After the 13 day induction, hiPSC-derived cells displayed an upregulated expression of a panel of otic/placodal markers. Strikingly, a subset of these induced progenitor cells displayed key-otic sensory markers, the percentage of which was increased in cultures under Notch inhibition as compared to RA/EGF-treated cultures. Our results show that modulating Notch pathway during in vitro differentiation of hiPSC-derived otic/placodal progenitors is a valuable strategy to promote the expression of human otic sensory lineage genes.
Collapse
Affiliation(s)
- Hanae Lahlou
- Aix Marseille Université, CNRS, LNIA UMR 7260, Marseille, France
| | | | - Arnaud Fontbonne
- Aix Marseille Université, CNRS, LNIA UMR 7260, Marseille, France
| | - Emmanuel Nivet
- Aix Marseille Université, CNRS, NICN UMR 7259, Marseille, France
| | - Azel Zine
- Aix Marseille Université, CNRS, LNIA UMR 7260, Marseille, France
- Université de Montpellier, Faculté de Pharmacie, Montpellier, France
- * E-mail: ,
| |
Collapse
|
30
|
Zhang Y, Guo L, Lu X, Cheng C, Sun S, Li W, Zhao L, Lai C, Zhang S, Yu C, Tang M, Chen Y, Chai R, Li H. Characterization of Lgr6+ Cells as an Enriched Population of Hair Cell Progenitors Compared to Lgr5+ Cells for Hair Cell Generation in the Neonatal Mouse Cochlea. Front Mol Neurosci 2018; 11:147. [PMID: 29867341 PMCID: PMC5961437 DOI: 10.3389/fnmol.2018.00147] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 04/12/2018] [Indexed: 12/20/2022] Open
Abstract
Hair cell (HC) loss is irreversible because only very limited HC regeneration has been observed in the adult mammalian cochlea. Wnt/β-catenin signaling regulates prosensory cell proliferation and differentiation during cochlear development, and Wnt activation promotes the proliferation of Lgr5+ cochlear HC progenitors in newborn mice. Similar to Lgr5, Lgr6 is also a Wnt downstream target gene. Lgr6 is reported to be present in adult stem cells in the skin, nail, tongue, lung, and mammary gland, and this protein is very important for adult stem cell maintenance in rapidly proliferating organs. Our previous studies showed that Lgr6+ cells are a subpopulation of Lgr5+ progenitor cells and that both Lgr6+ and Lgr5+ progenitors can generate Myosin7a+ HCs in vitro. Thus we hypothesized that Lgr6+ cells are an enriched population of cochlear progenitor cells. However, the detailed distinctions between the Lgr5+ and Lgr6+ progenitors are unclear. Here, we systematically compared the proliferation, HC differentiation, and detailed transcriptome expression profiles of these two progenitor populations. We found that the same number of isolated Lgr6+ progenitors generated significantly more Myosin7a+ HCs compared to Lgr5+ progenitors; however, Lgr5+ progenitors formed more epithelial colonies and more spheres than Lgr6+ progenitors in vitro. Using RNA-Seq, we compared the transcriptome differences between Lgr5+ and Lgr6+ progenitors and identified a list of significantly differential expressed genes that might regulate the proliferation and differentiation of these HC progenitors, including 4 cell cycle genes, 9 cell signaling pathway genes, and 54 transcription factors. In conclusion, we demonstrate that Lgr6+ progenitors are an enriched population of inner ear progenitors that generate more HCs compared to Lgr5+ progenitors in the newborn mouse cochlea, and the our research provides a series of genes that might regulate the proliferation of progenitors and HC generation.
Collapse
Affiliation(s)
- Yanping Zhang
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Luo Guo
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Xiaoling Lu
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Cheng Cheng
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Shan Sun
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Wen Li
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Liping Zhao
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Chuijin Lai
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Shasha Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Chenjie Yu
- Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline Laboratory, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Mingliang Tang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Yan Chen
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China.,Jiangsu Province Hi-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Huawei Li
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine, National Health and Family Planning Commission (NHFPC), Shanghai, China.,Shanghai Engineering Research Center of Cochlear Implant, Shanghai, China.,The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
31
|
Li Y, Jia S, Liu H, Tateya T, Guo W, Yang S, Beisel KW, He DZZ. Characterization of Hair Cell-Like Cells Converted From Supporting Cells After Notch Inhibition in Cultures of the Organ of Corti From Neonatal Gerbils. Front Cell Neurosci 2018; 12:73. [PMID: 29662441 PMCID: PMC5890164 DOI: 10.3389/fncel.2018.00073] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 03/02/2018] [Indexed: 12/11/2022] Open
Abstract
The senses of hearing and balance depend upon hair cells, the sensory receptors of the inner ear. Hair cells transduce mechanical stimuli into electrical activity. Loss of hair cells as a result of aging or exposure to noise and ototoxic drugs is the major cause of noncongenital hearing and balance deficits. In the ear of non-mammals, lost hair cells can spontaneously be replaced by production of new hair cells from conversion of supporting cells. Although supporting cells in adult mammals have lost that capability, neonatal supporting cells are able to convert to hair cells after inhibition of Notch signaling. We questioned whether Notch inhibition is sufficient to convert supporting cells to functional hair cells using electrophysiology and electron microscopy. We showed that pharmacological inhibition of the canonical Notch pathway in the cultured organ of Corti prepared from neonatal gerbils induced stereocilia formation in supporting cells (defined as hair cell-like cells or HCLCs) and supernumerary stereocilia in hair cells. The newly emerged stereocilia bundles of HCLCs were functional, i.e., able to respond to mechanical stimulation with mechanotransduction (MET) current. Transmission electron microscopy (TEM) showed that HCLCs converted from pillar cells maintained the pillar cell shape and that subsurface cisternae, normally observed underneath the cytoskeleton in outer hair cells (OHCs), was not present in Deiters’ cells-derived HCLCs. Voltage-clamp recordings showed that whole-cell currents from Deiters’ cells-derived HCLCs retained the same kinetics and magnitude seen in normal Deiters’ cells and that nonlinear capacitance (NLC), an electrical hallmark of OHC electromotility, was not detected from any HCLCs measured. Taken together, these results suggest that while Notch inhibition is sufficient for promoting stereocilia bundle formation, it is insufficient to convert neonatal supporting cells to mature hair cells. The fact that Notch inhibition led to stereocilia formation in supporting cells and supernumerary stereocilia in existing hair cells appears to suggest that Notch signaling may regulate stereocilia formation and stability during development.
Collapse
Affiliation(s)
- Yi Li
- Department of Otorhinolaryngology, Beijing Tongren Hospital, Beijing Capital Medical University, Beijing, China.,Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| | - Shuping Jia
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| | - Huizhan Liu
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| | - Tomoko Tateya
- Institute for Virus Research, Kyoto University, Kyoto, Japan
| | - Weiwei Guo
- Department of Otorhinolaryngology, PLA General Hospital, Beijing, China
| | - Shiming Yang
- Department of Otorhinolaryngology, PLA General Hospital, Beijing, China
| | - Kirk W Beisel
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| | - David Z Z He
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, United States
| |
Collapse
|
32
|
Notch pathway signaling in the skin antagonizes Merkel cell development. Dev Biol 2018; 434:207-214. [DOI: 10.1016/j.ydbio.2017.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/08/2017] [Accepted: 12/09/2017] [Indexed: 01/16/2023]
|
33
|
Du J, Wang X, Zhang X, Zhang X, Jiang H. DNER modulates the length, polarity and synaptogenesis of spiral ganglion neurons via the Notch signaling pathway. Mol Med Rep 2017; 17:2357-2365. [PMID: 29207144 PMCID: PMC5783477 DOI: 10.3892/mmr.2017.8115] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 02/16/2017] [Indexed: 02/05/2023] Open
Abstract
The Delta/Notch-like epidermal growth factor-related receptor (DNER) serves an important role in the developing central nervous system. However, the actions of DNER in the development of the spiral ganglion in the inner ear have yet to be elucidated. Wild-type C57BL/6 mice were housed and time-mated for use in the present study. Primary neuronal cultures were prepared using spiral ganglion progenitors isolated from the modiolus of postnatal day 1 (P1) mice. DNER recombinant lentiviral vectors were constructed and transfected into the cultured primary neurons. The relative proportion of differentiated neurons and the length of their neurites were evaluated using microscopy. The results of the present study demonstrated that DNER was expressed in spiral ganglion neurons (SGNs) that exhibited significant polarity in the early differentiation stages; DNER expression gradually decreased until the polarity was lost on week 35. The in vitro expression of DNER was revealed to be similar to that in vivo. When DNER expression was silenced using RNA interference, the polarity of the differentiated neurons was altered and they exhibited significantly reduced dendritic length. In addition, the proportion of bipolar neurons was decreased compared with the control group. Furthermore, the expression of α-synuclein and the GluR2/3 subunits of the α-amin-o-3-hydroxy-5-methyl-4-isoxazolepropionic acid glutamate receptor were also reduced in cultured neurons in which DNER was silenced. Notch1 was co-expressed with DNER in SGNs isolated from P1 mice. The indirect Notch inhibitor N-[N-(3,5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester also affected the polarity and the formation of protrusions, and reduced the expression of DNER and glial fibrillary acidic protein in SGNs. In conclusion, the present study demonstrated that DNER was expressed in SGNs and appeared to be involved in the mechanisms underlying neuronal polarity and neuritogenesis, via a Notch-dependent signaling pathway.
Collapse
Affiliation(s)
- Jintao Du
- Department of Otorhinolaryngology Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xianren Wang
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xiaobo Zhang
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xuemei Zhang
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Hongyan Jiang
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
34
|
Sjöqvist M, Andersson ER. Do as I say, Not(ch) as I do: Lateral control of cell fate. Dev Biol 2017; 447:58-70. [PMID: 28969930 DOI: 10.1016/j.ydbio.2017.09.032] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 09/15/2017] [Accepted: 09/26/2017] [Indexed: 01/19/2023]
Abstract
Breaking symmetry in populations of uniform cells, to induce adoption of an alternative cell fate, is an essential developmental mechanism. Similarly, domain and boundary establishment are crucial steps to forming organs during development. Notch signaling is a pathway ideally suited to mediating precise patterning cues, as both receptors and ligands are membrane-bound and can thus act as a precise switch to toggle cell fates on or off. Fine-tuning of signaling by positive or negative feedback mechanisms dictate whether signaling results in lateral induction or lateral inhibition, respectively, allowing Notch to either induce entire regions of cell specification, or dictate binary fate choices. Furthermore, pathway activity is modulated by Fringe modification of receptors or ligands, co-expression of receptors with ligands, mode of ligand presentation, and cell surface area in contact. In this review, we describe how Notch signaling is fine-tuned to mediate lateral induction or lateral inhibition cues, and discuss examples from C.elegans, D. melanogaster and M. musculus. Identifying the cellular machinery dictating the choice between lateral induction and lateral inhibition highlights the versatility of the Notch signaling pathway in development.
Collapse
Affiliation(s)
- Marika Sjöqvist
- Department of Biosciences and Nutrition, Karolinska Institutet, Sweden
| | - Emma R Andersson
- Department of Biosciences and Nutrition, Karolinska Institutet, Sweden.
| |
Collapse
|
35
|
Driver EC, Northrop A, Kelley MW. Cell migration, intercalation and growth regulate mammalian cochlear extension. Development 2017; 144:3766-3776. [PMID: 28870992 DOI: 10.1242/dev.151761] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 08/24/2017] [Indexed: 01/01/2023]
Abstract
Developmental remodeling of the sensory epithelium of the cochlea is required for the formation of an elongated, tonotopically organized auditory organ, but the cellular processes that mediate these events are largely unknown. We used both morphological assessments of cellular rearrangements and time-lapse imaging to visualize cochlear remodeling in mouse. Analysis of cell redistribution showed that the cochlea extends through a combination of radial intercalation and cell growth. Live imaging demonstrated that concomitant cellular intercalation results in a brief period of epithelial convergence, although subsequent changes in cell size lead to medial-lateral spreading. Supporting cells, which retain contact with the basement membrane, exhibit biased protrusive activity and directed movement along the axis of extension. By contrast, hair cells lose contact with the basement membrane, but contribute to continued outgrowth through increased cell size. Regulation of cellular protrusions, movement and intercalation within the cochlea all require myosin II. These results establish, for the first time, many of the cellular processes that drive the distribution of sensory cells along the tonotopic axis of the cochlea.
Collapse
Affiliation(s)
- Elizabeth Carroll Driver
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, USA
| | - Amy Northrop
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, USA
| | - Matthew W Kelley
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
36
|
Warchol ME, Stone J, Barton M, Ku J, Veile R, Daudet N, Lovett M. ADAM10 and γ-secretase regulate sensory regeneration in the avian vestibular organs. Dev Biol 2017; 428:39-51. [PMID: 28526588 PMCID: PMC5873298 DOI: 10.1016/j.ydbio.2017.05.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 05/15/2017] [Accepted: 05/15/2017] [Indexed: 11/19/2022]
Abstract
The loss of sensory hair cells from the inner ear is a leading cause of hearing and balance disorders. The mammalian ear has a very limited ability to replace lost hair cells, but the inner ears of non-mammalian vertebrates can spontaneously regenerate hair cells after injury. Prior studies have shown that replacement hair cells are derived from epithelial supporting cells and that the differentiation of new hair cells is regulated by the Notch signaling pathway. The present study examined molecular influences on regeneration in the avian utricle, which has a particularly robust regenerative ability. Chicken utricles were placed in organotypic culture and hair cells were lesioned by application of the ototoxic antibiotic streptomycin. Cultures were then allowed to regenerate in vitro for seven days. Some specimens were treated with small molecule inhibitors of γ-secretase or ADAM10, proteases which are essential for transmission of Notch signaling. As expected, treatment with both inhibitors led to increased numbers of replacement hair cells. However, we also found that inhibition of both proteases resulted in increased regenerative proliferation. Subsequent experiments showed that inhibition of γ-secretase or ADAM10 could also trigger proliferation in undamaged utricles. To better understand these phenomena, we used RNA-Seq profiling to characterize changes in gene expression following γ-secretase inhibition. We observed expression patterns that were consistent with Notch pathway inhibition, but we also found that the utricular sensory epithelium contains numerous γ-secretase substrates that might regulate cell cycle entry and possibly supporting cell-to-hair cell conversion. Together, our data suggest multiple roles for γ-secretase and ADAM10 in vestibular hair cell regeneration.
Collapse
Affiliation(s)
- Mark E Warchol
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Jennifer Stone
- The Virginia Merrill Bloedel Hearing Research Center and Department of Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Seattle, WA 98195, United States
| | - Matthew Barton
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Jeffrey Ku
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Rose Veile
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO 63110, United States; Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Nicolas Daudet
- Center for Auditory Research, University College London, London, United Kingdom
| | - Michael Lovett
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, United States; NHLI, Imperial College, London, United Kingdom
| |
Collapse
|
37
|
Zhang S, Zhang Y, Yu P, Hu Y, Zhou H, Guo L, Xu X, Zhu X, Waqas M, Qi J, Zhang X, Liu Y, Chen F, Tang M, Qian X, Shi H, Gao X, Chai R. Characterization of Lgr5+ Progenitor Cell Transcriptomes after Neomycin Injury in the Neonatal Mouse Cochlea. Front Mol Neurosci 2017; 10:213. [PMID: 28725177 PMCID: PMC5496572 DOI: 10.3389/fnmol.2017.00213] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/16/2017] [Indexed: 12/17/2022] Open
Abstract
Lgr5+ supporting cells (SCs) are enriched hair cell (HC) progenitors in the cochlea. Both in vitro and in vivo studies have shown that HC injury can spontaneously activate Lgr5+ progenitors to regenerate HCs in the neonatal mouse cochlea. Promoting HC regeneration requires the understanding of the mechanism of HC regeneration, and this requires knowledge of the key genes involved in HC injury-induced self-repair responses that promote the proliferation and differentiation of Lgr5+ progenitors. Here, as expected, we found that neomycin-treated Lgr5+ progenitors (NLPs) had significantly greater HC regeneration ability, and greater but not significant proliferation ability compared to untreated Lgr5+ progenitors (ULPs) in response to neomycin exposure. Next, we used RNA-seq analysis to determine the differences in the gene-expression profiles between the transcriptomes of NLPs and ULPs from the neonatal mouse cochlea. We first analyzed the genes that were enriched and differentially expressed in NLPs and ULPs and then analyzed the cell cycle genes, the transcription factors, and the signaling pathway genes that might regulate the proliferation and differentiation of Lgr5+ progenitors. We found 9 cell cycle genes, 88 transcription factors, 8 microRNAs, and 16 cell-signaling pathway genes that were significantly upregulated or downregulated after neomycin injury in NLPs. Lastly, we constructed a protein-protein interaction network to show the interaction and connections of genes that are differentially expressed in NLPs and ULPs. This study has identified the genes that might regulate the proliferation and HC regeneration of Lgr5+ progenitors after neomycin injury, and investigations into the roles and mechanisms of these genes in the cochlea should be performed in the future to identify potential therapeutic targets for HC regeneration.
Collapse
Affiliation(s)
- Shasha Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China.,Research Institute of OtolaryngologyNanjing, China.,Co-innovation Center of Neuroregeneration, Nantong UniversityNantong, China
| | - Yuan Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China
| | - Pengfei Yu
- Bioinformatics Department, Admera Health LLCSouth Plainfield, NJ, United States
| | - Yao Hu
- School of Pharmacy, Institute for Stem Cell and Neural Regeneration, Nanjing Medical UniversityNanjing, China
| | - Han Zhou
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing, China
| | - Lingna Guo
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China
| | - Xiaochen Xu
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China
| | - Xiaocheng Zhu
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing, China
| | - Muhammad Waqas
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China.,Department of Biotechnology, Federal Urdu University of Arts, Science and TechnologyKarachi, Pakistan
| | - Jieyu Qi
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China
| | - Xiaoli Zhang
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing, China
| | - Yan Liu
- School of Pharmacy, Institute for Stem Cell and Neural Regeneration, Nanjing Medical UniversityNanjing, China
| | - Fangyi Chen
- Department of Biomedical Engineering, Southern University of Science and TechnologyShenzhen, China
| | - Mingliang Tang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China
| | - Xiaoyun Qian
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing, China
| | - Haibo Shi
- Department of Otorhinolaryngology Head and Neck Surgery, The Sixth People's Hospital Affiliated to Shanghai Jiao Tong UniversityShanghai, China
| | - Xia Gao
- Research Institute of OtolaryngologyNanjing, China.,Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing, China
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China.,Research Institute of OtolaryngologyNanjing, China.,Co-innovation Center of Neuroregeneration, Nantong UniversityNantong, China
| |
Collapse
|
38
|
Revuelta M, Santaolalla F, Arteaga O, Alvarez A, Sánchez-del-Rey A, Hilario E. Recent advances in cochlear hair cell regeneration-A promising opportunity for the treatment of age-related hearing loss. Ageing Res Rev 2017; 36:149-155. [PMID: 28414155 DOI: 10.1016/j.arr.2017.04.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 04/05/2017] [Accepted: 04/10/2017] [Indexed: 01/22/2023]
Abstract
The objective of this paper is to review current information regarding the treatment of age-related hearing loss by using cochlear hair cell regeneration. Recent advances in the regeneration of the inner ear, including the usefulness of stem cells, are also presented. Based on the current literature, cochlear cell regeneration may well be possible in the short term and cochlear gene therapy may also be useful for the treatment of hearing loss associated with ageing. The present review provide further insight into the pathogenesis of Inner Ear senescence and aged-related hearing loss and facilitate the development of therapeutic strategies to repair hair cells damaged by ageing. More research will be needed in order to translate them into an effective treatment for deafness linked to cochlear senescence in humans.
Collapse
|
39
|
Chen Q, Quan Y, Wang N, Xie C, Ji Z, He H, Chai R, Li H, Yin S, Chin YE, Wei X, Gao WQ. Inactivation of STAT3 Signaling Impairs Hair Cell Differentiation in the Developing Mouse Cochlea. Stem Cell Reports 2017; 9:231-246. [PMID: 28669599 PMCID: PMC5511372 DOI: 10.1016/j.stemcr.2017.05.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Revised: 05/24/2017] [Accepted: 05/24/2017] [Indexed: 11/19/2022] Open
Abstract
Although STAT3 signaling is demonstrated to regulate sensory cell differentiation and regeneration in the zebrafish, its exact role is still unclear in mammalian cochleae. Here, we report that STAT3 and its activated form are specifically expressed in hair cells during mouse cochlear development. Importantly, conditional cochlear deletion of Stat3 leads to an inhibition on hair cell differentiation in mice in vivo and in vitro. By cell fate analysis, inactivation of STAT3 signaling shifts the cell division modes from asymmetric to symmetric divisions from supporting cells. Moreover, inhibition of Notch signaling stimulates STAT3 phosphorylation, and inactivation of STAT3 signaling attenuates production of supernumerary hair cells induced by a Notch pathway inhibitor. Our findings highlight an important role of the STAT3 signaling during mouse cochlear hair cell differentiation and may have clinical implications for the recovery of hair cell loss-induced hearing impairment.
Collapse
Affiliation(s)
- Qianqian Chen
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; Med-X Research Institute & School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yizhou Quan
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; Med-X Research Institute & School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Naitao Wang
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Chengying Xie
- Med-X Research Institute & School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Zhongzhong Ji
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; Med-X Research Institute & School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Hao He
- Med-X Research Institute & School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; The Affiliated Six People's Hospital, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Renjie Chai
- MOE Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Huawei Li
- Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China
| | - Shankai Yin
- The Affiliated Six People's Hospital, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Y Eugene Chin
- China Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences-Shanghai Jiao Tong University School of Medicine, Shanghai 200031, China
| | - Xunbin Wei
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; Med-X Research Institute & School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; Med-X Research Institute & School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; The Affiliated Six People's Hospital, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|
40
|
Werth M, Schmidt-Ott KM, Leete T, Qiu A, Hinze C, Viltard M, Paragas N, Shawber CJ, Yu W, Lee P, Chen X, Sarkar A, Mu W, Rittenberg A, Lin CS, Kitajewski J, Al-Awqati Q, Barasch J. Transcription factor TFCP2L1 patterns cells in the mouse kidney collecting ducts. eLife 2017; 6. [PMID: 28577314 PMCID: PMC5484618 DOI: 10.7554/elife.24265] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 06/03/2017] [Indexed: 12/19/2022] Open
Abstract
Although most nephron segments contain one type of epithelial cell, the collecting ducts consists of at least two: intercalated (IC) and principal (PC) cells, which regulate acid-base and salt-water homeostasis, respectively. In adult kidneys, these cells are organized in rosettes suggesting functional interactions. Genetic studies in mouse revealed that transcription factor Tfcp2l1 coordinates IC and PC development. Tfcp2l1 induces the expression of IC specific genes, including specific H+-ATPase subunits and Jag1. Jag1 in turn, initiates Notch signaling in PCs but inhibits Notch signaling in ICs. Tfcp2l1 inactivation deletes ICs, whereas Jag1 inactivation results in the forfeiture of discrete IC and PC identities. Thus, Tfcp2l1 is a critical regulator of IC-PC patterning, acting cell-autonomously in ICs, and non-cell-autonomously in PCs. As a result, Tfcp2l1 regulates the diversification of cell types which is the central characteristic of 'salt and pepper' epithelia and distinguishes the collecting duct from all other nephron segments. DOI:http://dx.doi.org/10.7554/eLife.24265.001
Collapse
Affiliation(s)
- Max Werth
- Columbia University, New York, United States
| | - Kai M Schmidt-Ott
- Columbia University, New York, United States.,Max Delbruck Center for Molecular Medicine, Berlin, Germany.,Department of Nephrology and Intensive Care Medicine, Charité - Universitaetsmedizin Berlin, Berlin, Germany
| | | | - Andong Qiu
- Columbia University, New York, United States.,Tongji University, Shanghai, China
| | | | - Melanie Viltard
- Columbia University, New York, United States.,Institute for European Expertise in Physiology, Paris, France
| | - Neal Paragas
- Columbia University, New York, United States.,University of Washington, Seattle, United States
| | | | - Wenqiang Yu
- Columbia University, New York, United States.,Fudan University, Shanghai, China
| | - Peter Lee
- Columbia University, New York, United States
| | - Xia Chen
- Columbia University, New York, United States
| | - Abby Sarkar
- Columbia University, New York, United States
| | - Weiyi Mu
- Columbia University, New York, United States
| | | | | | - Jan Kitajewski
- Columbia University, New York, United States.,University of Illinois at Chicago, Chicago, United States
| | | | | |
Collapse
|
41
|
Corson F, Couturier L, Rouault H, Mazouni K, Schweisguth F. Self-organized Notch dynamics generate stereotyped sensory organ patterns in Drosophila. Science 2017; 356:science.aai7407. [PMID: 28386027 DOI: 10.1126/science.aai7407] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 03/20/2017] [Indexed: 12/26/2022]
Abstract
The emergence of spatial patterns in developing multicellular organisms relies on positional cues and cell-cell communication. Drosophila sensory organs have informed a paradigm in which these operate in two distinct steps: Prepattern factors drive localized proneural activity, then Notch-mediated lateral inhibition singles out neural precursors. Here we show that self-organization through Notch signaling also establishes the proneural stripes that resolve into rows of sensory bristles on the fly thorax. Patterning, initiated by a gradient of Delta ligand expression, progresses through inhibitory signaling between and within stripes. Thus, Notch signaling can support self-organized tissue patterning as a prepattern is transduced by cell-cell interactions into a refined arrangement of cellular fates.
Collapse
Affiliation(s)
- Francis Corson
- Laboratoire de Physique Statistique, Ecole Normale Supérieure, CNRS, Université Pierre et Marie Curie, Université Paris Diderot, 75005 Paris, France.
| | - Lydie Couturier
- Department of Developmental and Stem Cell Biology, Institut Pasteur, 75015 Paris, France.,CNRS, UMR3738, 75015 Paris, France
| | - Hervé Rouault
- Department of Developmental and Stem Cell Biology, Institut Pasteur, 75015 Paris, France.,CNRS, UMR3738, 75015 Paris, France
| | - Khalil Mazouni
- Department of Developmental and Stem Cell Biology, Institut Pasteur, 75015 Paris, France.,CNRS, UMR3738, 75015 Paris, France
| | - François Schweisguth
- Department of Developmental and Stem Cell Biology, Institut Pasteur, 75015 Paris, France. .,CNRS, UMR3738, 75015 Paris, France
| |
Collapse
|
42
|
Mittal R, Debs LH, Nguyen D, Patel AP, Grati M, Mittal J, Yan D, Eshraghi AA, Liu XZ. Signaling in the Auditory System: Implications in Hair Cell Regeneration and Hearing Function. J Cell Physiol 2017; 232:2710-2721. [DOI: 10.1002/jcp.25695] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 11/18/2016] [Indexed: 02/06/2023]
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - Luca H. Debs
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - Desiree Nguyen
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - Amit P. Patel
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - M'hamed Grati
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - Jeenu Mittal
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - Denise Yan
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - Adrien A. Eshraghi
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| | - Xue Zhong Liu
- Department of Otolaryngology; University of Miami Miller School of Medicine; Miami Florida
| |
Collapse
|
43
|
Lee S, Jeong HS, Cho HH. Atoh1 as a Coordinator of Sensory Hair Cell Development and Regeneration in the Cochlea. Chonnam Med J 2017; 53:37-46. [PMID: 28184337 PMCID: PMC5299128 DOI: 10.4068/cmj.2017.53.1.37] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/22/2016] [Accepted: 09/23/2016] [Indexed: 12/18/2022] Open
Abstract
Cochlear sensory hair cells (HCs) are crucial for hearing as mechanoreceptors of the auditory systems. Clarification of transcriptional regulation for the cochlear sensory HC development is crucial for the improvement of cell replacement therapies for hearing loss. Transcription factor Atoh1 is the key player during HC development and regeneration. In this review, we will focus on Atoh1 and its related signaling pathways (Notch, fibroblast growth factor, and Wnt/β-catenin signaling) involved in the development of cochlear sensory HCs. We will also discuss the potential applicability of these signals for the induction of HC regeneration.
Collapse
Affiliation(s)
- Sungsu Lee
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Korea.; Research Institute of Medical Sciences, Chonnam National University, Gwangju, Korea
| | - Han-Seong Jeong
- Research Institute of Medical Sciences, Chonnam National University, Gwangju, Korea.; Department of Physiology, Chonnam National University Medical School, Gwangju, Korea
| | - Hyong-Ho Cho
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Korea.; Research Institute of Medical Sciences, Chonnam National University, Gwangju, Korea
| |
Collapse
|
44
|
Inomata H. Scaling of pattern formations and morphogen gradients. Dev Growth Differ 2017; 59:41-51. [PMID: 28097650 DOI: 10.1111/dgd.12337] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 12/08/2016] [Accepted: 12/08/2016] [Indexed: 12/31/2022]
Abstract
The concentration gradient of morphogens provides positional information for an embryo and plays a pivotal role in pattern formation of tissues during the developmental processes. Morphogen-dependent pattern formations show robustness despite various perturbations. Although tissues usually grow and dynamically change their size during histogenesis, proper patterns are formed without the influence of size variations. Furthermore, even when the blastula embryo of Xenopus laevis is bisected into dorsal and ventral halves, the dorsal half of the embryo leads to proportionally patterned half-sized embryos. This robustness of pattern formation despite size variations is termed as scaling. In this review, I focused on the morphogen-dependent dorsal-ventral axis formation in Xenopus and described how morphogens form a proper gradient shape according to the embryo size.
Collapse
Affiliation(s)
- Hidehiko Inomata
- Axial Pattern Dynamics Team, Center for Developmental Biology, RIKEN, Kobe, Japan
| |
Collapse
|
45
|
Basch ML, Brown RM, Jen HI, Semerci F, Depreux F, Edlund RK, Zhang H, Norton CR, Gridley T, Cole SE, Doetzlhofer A, Maletic-Savatic M, Segil N, Groves AK. Fine-tuning of Notch signaling sets the boundary of the organ of Corti and establishes sensory cell fates. eLife 2016; 5:19921. [PMID: 27966429 PMCID: PMC5215100 DOI: 10.7554/elife.19921] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 12/12/2016] [Indexed: 01/08/2023] Open
Abstract
The signals that induce the organ of Corti and define its boundaries in the cochlea are poorly understood. We show that two Notch modifiers, Lfng and Mfng, are transiently expressed precisely at the neural boundary of the organ of Corti. Cre-Lox fate mapping shows this region gives rise to inner hair cells and their associated inner phalangeal cells. Mutation of Lfng and Mfng disrupts this boundary, producing unexpected duplications of inner hair cells and inner phalangeal cells. This phenotype is mimicked by other mouse mutants or pharmacological treatments that lower but not abolish Notch signaling. However, strong disruption of Notch signaling causes a very different result, generating many ectopic hair cells at the expense of inner phalangeal cells. Our results show that Notch signaling is finely calibrated in the cochlea to produce precisely tuned levels of signaling that first set the boundary of the organ of Corti and later regulate hair cell development. DOI:http://dx.doi.org/10.7554/eLife.19921.001
Collapse
Affiliation(s)
- Martin L Basch
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | - Rogers M Brown
- Program in Developmental Biology, Baylor College of Medicine, Houston, United States
| | - Hsin-I Jen
- Program in Developmental Biology, Baylor College of Medicine, Houston, United States
| | - Fatih Semerci
- Program in Developmental Biology, Baylor College of Medicine, Houston, United States
| | - Frederic Depreux
- Department of Cell Biology and Anatomy, Rosalind Franklin University of Medicine and Science, Chicago, United States
| | - Renée K Edlund
- Program in Developmental Biology, Baylor College of Medicine, Houston, United States
| | - Hongyuan Zhang
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | | | - Thomas Gridley
- Maine Medical Center Research Institute, Scarborough, United States
| | - Susan E Cole
- Department of Molecular Genetics, The Ohio State University, Columbus, United States
| | - Angelika Doetzlhofer
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, School of Medicine, Baltimore, United States
| | - Mirjana Maletic-Savatic
- Department of Neuroscience, Baylor College of Medicine, Houston, United States.,Program in Developmental Biology, Baylor College of Medicine, Houston, United States.,Department of Pediatrics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Neil Segil
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, United States
| | - Andrew K Groves
- Department of Neuroscience, Baylor College of Medicine, Houston, United States.,Program in Developmental Biology, Baylor College of Medicine, Houston, United States.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
| |
Collapse
|
46
|
Maass JC, Gu R, Cai T, Wan YW, Cantellano SC, Asprer JST, Zhang H, Jen HI, Edlund RK, Liu Z, Groves AK. Transcriptomic Analysis of Mouse Cochlear Supporting Cell Maturation Reveals Large-Scale Changes in Notch Responsiveness Prior to the Onset of Hearing. PLoS One 2016; 11:e0167286. [PMID: 27918591 PMCID: PMC5137903 DOI: 10.1371/journal.pone.0167286] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 11/13/2016] [Indexed: 01/22/2023] Open
Abstract
Neonatal mouse cochlear supporting cells have a limited ability to divide and trans-differentiate into hair cells, but this ability declines rapidly in the two weeks after birth. This decline is concomitant with the morphological and functional maturation of the organ of Corti prior to the onset of hearing. However, despite this association between maturation and loss of regenerative potential, little is known of the molecular changes that underlie these events. To identify these changes, we used RNA-seq to generate transcriptional profiles of purified cochlear supporting cells from 1- and 6-day-old mice. We found many significant changes in gene expression during this period, many of which were related to regulation of proliferation, differentiation of inner ear components and the maturation of the organ of Corti prior to the onset of hearing. One example of a change in regenerative potential of supporting cells is their robust production of hair cells in response to a blockade of the Notch signaling pathway at the time of birth, but a complete lack of response to such blockade just a few days later. By comparing our supporting cell transcriptomes to those of supporting cells cultured in the presence of Notch pathway inhibitors, we show that the transcriptional response to Notch blockade disappears almost completely in the first postnatal week. Our results offer some of the first molecular insights into the failure of hair cell regeneration in the mammalian cochlea.
Collapse
Affiliation(s)
- Juan C. Maass
- Department of Otolaryngology, Hospital Clínico Universidad de Chile and 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, 1 Baylor Plaza, Houston, TX, United States of America
| | - Tiantian Cai
- Department of Neuroscience, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, United States of America
| | - Ying-Wooi Wan
- Department of Pediatrics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, United States of America
- The Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States of America
| | - Silvia C. Cantellano
- Department of Otolaryngology, Hospital Clínico Universidad de Chile and Interdisciplinary Program of Physiology and Biophysics ICBM Universidad de Chile, Santiago, Chile
| | - Joanna S. T. Asprer
- Department of Neuroscience, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, United States of America
| | - Hongyuan Zhang
- Department of Neuroscience, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, United States of America
| | - Hsin-I Jen
- Program in Developmental Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, United States of America
| | - Renée K. Edlund
- Department of Neuroscience, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, United States of America
| | - Zhandong Liu
- Department of Pediatrics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, United States of America
- The Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States of America
| | - Andrew K. Groves
- Department of Neuroscience, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, United States of America
- Program in Developmental Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, United States of America
- * E-mail:
| |
Collapse
|
47
|
Figueiredo M, Silva JC, Santos AS, Proa V, Alcobia I, Zilhão R, Cidadão A, Neves H. Notch and Hedgehog in the thymus/parathyroid common primordium: Crosstalk in organ formation. Dev Biol 2016; 418:268-82. [DOI: 10.1016/j.ydbio.2016.08.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 08/12/2016] [Accepted: 08/13/2016] [Indexed: 12/30/2022]
|
48
|
Munnamalai V, Fekete DM. Notch-Wnt-Bmp crosstalk regulates radial patterning in the mouse cochlea in a spatiotemporal manner. Development 2016; 143:4003-4015. [PMID: 27633988 DOI: 10.1242/dev.139469] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 09/01/2016] [Indexed: 01/04/2023]
Abstract
The sensory cells of the mammalian organ of Corti assume a precise mosaic arrangement during embryonic development. Manipulation of Wnt signaling can modulate the proliferation of cochlear progenitors, but whether Wnts are responsible for patterning compartments, or specific hair cells within them, is unclear. To address how the precise timing of Wnt signaling impacts patterning across the radial axis, mouse cochlear cultures were initiated at embryonic day 12.5 and subjected to pharmacological treatments at different stages. Early changes in major patterning genes were assessed to understand the mechanisms underlying alterations of compartments. Results show that Wnt activation can promote medial cell fates by regulating medially expressed Notch genes in a spatiotemporal manner. Wnts can also suppress lateral cell fates by antagonizing Bmp4 expression. Perturbation of the Notch and Bmp pathways revealed which secondary effects were linked to these pathways. Importantly, these effects on cochlear development are dependent on the timing of drug delivery. In conclusion, Wnt signaling in the cochlea influences patterning through complex crosstalk with the Notch and Bmp pathways at several stages of embryonic development.
Collapse
Affiliation(s)
- Vidhya Munnamalai
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA.,Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
| | - Donna M Fekete
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA .,Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA.,Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
49
|
Lineage tracing of Sox2-expressing progenitor cells in the mouse inner ear reveals a broad contribution to non-sensory tissues and insights into the origin of the organ of Corti. Dev Biol 2016; 414:72-84. [PMID: 27090805 DOI: 10.1016/j.ydbio.2016.03.027] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 03/09/2016] [Accepted: 03/26/2016] [Indexed: 11/22/2022]
Abstract
The transcription factor Sox2 is both necessary and sufficient for the generation of sensory regions of the inner ear. It regulates expression of the Notch ligand Jag1 in prosensory progenitors, which signal to neighboring cells to up-regulate Sox2 and sustain prosensory identity. However, the expression pattern of Sox2 in the early inner ear is very broad, suggesting that Sox2-expressing progenitors form a wide variety of cell types in addition to generating the sensory regions of the ear. We used Sox2-CreER mice to follow the fates of Sox2-expressing cells at different stages in ear development. We find that Sox2-expressing cells in the early otocyst give rise to large numbers of non-sensory structures throughout the inner ear, and that Sox2 only becomes a truly prosensory marker at embryonic day (E)11.5. Our fate map reveals the organ of Corti derives from a central domain on the medial side of the otocyst and shows that a significant amount of the organ of Corti derives from a Sox2-negative population in this region.
Collapse
|
50
|
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
|