1
|
Ueda Y, Nakamura T, Nie J, Solivais AJ, Hoffman JR, Daye BJ, Hashino E. Defining developmental trajectories of prosensory cells in human inner ear organoids at single-cell resolution. Development 2023; 150:dev201071. [PMID: 37381908 PMCID: PMC10323240 DOI: 10.1242/dev.201071] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 05/24/2023] [Indexed: 06/29/2023]
Abstract
The inner ear sensory epithelia contain mechanosensitive hair cells and supporting cells. Both cell types arise from SOX2-expressing prosensory cells, but the mechanisms underlying the diversification of these cell lineages remain unclear. To determine the transcriptional trajectory of prosensory cells, we established a SOX2-2A-ntdTomato human embryonic stem cell line using CRISPR/Cas9, and performed single-cell RNA-sequencing analyses with SOX2-positive cells isolated from inner ear organoids at various time points between differentiation days 20 and 60. Our pseudotime analysis suggests that vestibular type II hair cells arise primarily from supporting cells, rather than bi-fated prosensory cells in organoids. Moreover, ion channel- and ion-transporter-related gene sets were enriched in supporting cells versus prosensory cells, whereas Wnt signaling-related gene sets were enriched in hair cells versus supporting cells. These findings provide valuable insights into how prosensory cells give rise to hair cells and supporting cells during human inner ear development, and may provide a clue to promote hair cell regeneration from resident supporting cells in individuals with hearing loss or balance disorders.
Collapse
Affiliation(s)
- Yoshitomo Ueda
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Takashi Nakamura
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Jing Nie
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alexander J. Solivais
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - John R. Hoffman
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Becca J. Daye
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Eri Hashino
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
2
|
Guo L, Wang W, Song W, Cao H, Tian H, Wang Z, Ren J, Ning F, Zhang D, Duan H. Genome-wide DNA methylation analysis of middle-aged and elderly monozygotic twins with age-related hearing loss in Qingdao, China. Gene 2022; 849:146918. [PMID: 36179964 DOI: 10.1016/j.gene.2022.146918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To explore the differences in DNA methylation associated with age-related hearing loss in a study of 57 twin pairs from China. DESIGN Monozygotic twins were identified through the Qingdao Twin Registration system. The median age of participants was >50 years. Their hearing thresholds were measured using a multilevel pure-tone audiometry assessment. The pure-tone audiometry was calculated at low frequencies (0.5, 1.0, and 2.0 kHz), speech frequencies (0.5, 1.0, 2.0, and 4.0kHz), and high frequencies (4.0 and 8 kHz). The CpG sites were tested using a linear mixed-effects model, and the function of the cis-regulatory regions and ontological enrichments were predicted using the online Genomic Regions Enrichment of Annotations Tool. The differentially methylated regions were identified using a comb-p python library approach. RESULTS In each of the PTA categories (low-, speech-, high-frequency), age-related hearing loss was detected in 25.9%, 19.3%, and 52.8% of participants. In the low-, speech- and high-frequency categories we identified 18, 42, and 12 individual CpG sites and 6, 11, and 6 differentially methylated regions. The CpG site located near DUSP4 had the strongest association with low- and speech-frequency, while the strongest association with high-frequency was near C21orf58. We identified associations of ALG10 with high-frequency hearing, C3 and LCK with low- and speech-frequency hearing, and GBX2 with low-frequency hearing. Top pathways that may be related to hearing, such as the Notch signaling pathway, were also identified. CONCLUSION Our study is the first of its kind to identify these genes and their associated with DNA methylation may play essential roles in the hearing process. The results of our epigenome-wide association study on twins clarify the complex mechanisms underlying age-related hearing loss.
Collapse
Affiliation(s)
- Longzi Guo
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| | - Weijing Wang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| | - Wanxue Song
- Qingdao Maternal and Child Health and Family Planning Service Center, Qingdao, China
| | - Hainan Cao
- Department of Otorhinolaryngology, Qingdao Municipal Hospital, Qingdao, China
| | - Huimin Tian
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| | - Zhaoguo Wang
- Qingdao Municipal Center for Disease Control and Prevention, Qingdao, China
| | - Jifeng Ren
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| | - Feng Ning
- Qingdao Municipal Center for Disease Control and Prevention, Qingdao, China
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, China
| | - Haiping Duan
- Qingdao Municipal Center for Disease Control and Prevention, Qingdao, China.
| |
Collapse
|
3
|
Knockout of mafba Causes Inner-Ear Developmental Defects in Zebrafish via the Impairment of Proliferation and Differentiation of Ionocyte Progenitor Cells. Biomedicines 2021; 9:biomedicines9111699. [PMID: 34829928 PMCID: PMC8616026 DOI: 10.3390/biomedicines9111699] [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: 08/11/2021] [Revised: 10/18/2021] [Accepted: 10/28/2021] [Indexed: 12/03/2022] Open
Abstract
Zebrafish is an excellent model for exploring the development of the inner ear. Its inner ear has similar functions to that of humans, specifically in the maintenance of hearing and balance. Mafba is a component of the Maf transcription factor family. It participates in multiple biological processes, but its role in inner-ear development remains poorly understood. In this study, we constructed a mafba knockout (mafba−/−) zebrafish model using CRISPR/Cas9 technology. The mafba−/− mutant inner ear displayed severe impairments, such as enlarged otocysts, smaller or absent otoliths, and insensitivity to sound stimulation. The proliferation of p63+ epidermal stem cells and dlc+ ionocyte progenitors was inhibited in mafba−/− mutants. Moreover, the results showed that mafba deletion induces the apoptosis of differentiated K+-ATPase-rich (NR) cells and H+-ATPase-rich (HR) cells. The activation of p53 apoptosis and G0/G1 cell cycle arrest resulted from DNA damage in the inner-ear region, providing a mechanism to account for the inner ear deficiencies. The loss of homeostasis resulting from disorders of ionocyte progenitors resulted in structural defects in the inner ear and, consequently, loss of hearing. In conclusion, the present study elucidated the function of ionic channel homeostasis and inner-ear development using a zebrafish Mafba model and clarified the possible physiological roles.
Collapse
|
4
|
Mackowetzky K, Yoon KH, Mackowetzky EJ, Waskiewicz AJ. Development and evolution of the vestibular apparatuses of the inner ear. J Anat 2021; 239:801-828. [PMID: 34047378 PMCID: PMC8450482 DOI: 10.1111/joa.13459] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/07/2021] [Accepted: 05/06/2021] [Indexed: 12/16/2022] Open
Abstract
The vertebrate inner ear is a labyrinthine sensory organ responsible for perceiving sound and body motion. While a great deal of research has been invested in understanding the auditory system, a growing body of work has begun to delineate the complex developmental program behind the apparatuses of the inner ear involved with vestibular function. These animal studies have helped identify genes involved in inner ear development and model syndromes known to include vestibular dysfunction, paving the way for generating treatments for people suffering from these disorders. This review will provide an overview of known inner ear anatomy and function and summarize the exciting discoveries behind inner ear development and the evolution of its vestibular apparatuses.
Collapse
Affiliation(s)
- Kacey Mackowetzky
- Department of Biological SciencesUniversity of AlbertaEdmontonAlbertaCanada
| | - Kevin H. Yoon
- Department of Biological SciencesUniversity of AlbertaEdmontonAlbertaCanada
| | | | - Andrew J. Waskiewicz
- Department of Biological SciencesUniversity of AlbertaEdmontonAlbertaCanada
- Women & Children’s Health Research InstituteUniversity of AlbertaEdmontonAlbertaCanada
| |
Collapse
|
5
|
Abstract
The cochlea, a coiled structure located in the ventral region of the inner ear, acts as the primary structure for the perception of sound. Along the length of the cochlear spiral is the organ of Corti, a highly derived and rigorously patterned sensory epithelium that acts to convert auditory stimuli into neural impulses. The development of the organ of Corti requires a series of inductive events that specify unique cellular characteristics and axial identities along its three major axes. Here, we review recent studies of the cellular and molecular processes regulating several aspects of cochlear development, such as axial patterning, cochlear outgrowth and cellular differentiation. We highlight how the precise coordination of multiple signaling pathways is required for the successful formation of a complete organ of Corti.
Collapse
Affiliation(s)
- Elizabeth Carroll Driver
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthew W Kelley
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
6
|
Cheng C, Wang Y, Guo L, Lu X, Zhu W, Muhammad W, Zhang L, Lu L, Gao J, Tang M, Chen F, Gao X, Li H, Chai R. Age-related transcriptome changes in Sox2+ supporting cells in the mouse cochlea. Stem Cell Res Ther 2019; 10:365. [PMID: 31791390 PMCID: PMC6889721 DOI: 10.1186/s13287-019-1437-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/29/2019] [Accepted: 10/01/2019] [Indexed: 12/17/2022] Open
Abstract
Background Inner ear supporting cells (SCs) in the neonatal mouse cochlea are a potential source for hair cell (HC) regeneration, but several studies have shown that the regeneration ability of SCs decreases dramatically as mice age and that lost HCs cannot be regenerated in adult mice. To better understand how SCs might be better used to regenerate HCs, it is important to understand how the gene expression profile changes in SCs at different ages. Methods Here, we used Sox2GFP/+ mice to isolate the Sox2+ SCs at postnatal day (P)3, P7, P14, and P30 via flow cytometry. Next, we used RNA-seq to determine the transcriptome expression profiles of P3, P7, P14, and P30 SCs. To further analyze the relationships between these age-related and differentially expressed genes in Sox2+ SCs, we performed gene ontology (GO) analysis. Results Consistent with previous reports, we also found that the proliferation and HC regeneration ability of isolated Sox2+ SCs significantly decreased as mice aged. We identified numerous genes that are enriched and differentially expressed in Sox2+ SCs at four different postnatal ages, including cell cycle genes, signaling pathway genes, and transcription factors that might be involved in regulating the proliferation and HC differentiation ability of SCs. We thus present a set of genes that might regulate the proliferation and HC regeneration ability of SCs, and these might serve as potential new therapeutic targets for HC regeneration. Conclusions In our research, we found several genes that might play an important role in regulating the proliferation and HC regeneration ability of SCs. These datasets are expected to serve as a resource to provide potential new therapeutic targets for regulating the ability of SCs to regenerate HCs in postnatal mammals.
Collapse
Affiliation(s)
- Cheng Cheng
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008, China.,Research Institute of Otolaryngology, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Yunfeng Wang
- Shanghai Fenyang Vision & Audition Center, Shanghai, China.,ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Room 611, Building 9, No. 83, Fenyang Road, Xuhui District, Shanghai, 200031, China
| | - Luo Guo
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Room 611, Building 9, No. 83, Fenyang Road, Xuhui District, Shanghai, 200031, China
| | - Xiaoling Lu
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Room 611, Building 9, No. 83, Fenyang Road, Xuhui District, Shanghai, 200031, China
| | - Weijie Zhu
- MOE Key Laboratory for Developmental Genes and Human Disease, State Key Laboratory of Bioelectronics, Co-Innovation Center of Neuroregeneration, Institute of Life Sciences, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Waqas Muhammad
- MOE Key Laboratory for Developmental Genes and Human Disease, State Key Laboratory of Bioelectronics, Co-Innovation Center of Neuroregeneration, Institute of Life Sciences, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.,Department of Biotechnology, Federal Urdu University of Arts, Science and Technology, Gulshan-e-Iqbal Campus, Karachi, Pakistan
| | - Liyan Zhang
- MOE Key Laboratory for Developmental Genes and Human Disease, State Key Laboratory of Bioelectronics, Co-Innovation Center of Neuroregeneration, Institute of Life Sciences, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Ling Lu
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Junyan Gao
- Jiangsu Rehabilitation Research Center for Hearing and Speech Impairment, Nanjing, 210004, Jiangsu, China
| | - Mingliang Tang
- MOE Key Laboratory for Developmental Genes and Human Disease, State Key Laboratory of Bioelectronics, Co-Innovation Center of Neuroregeneration, Institute of Life Sciences, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Fangyi Chen
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Xia Gao
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008, China.
| | - Huawei Li
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Room 611, Building 9, No. 83, Fenyang Road, Xuhui District, Shanghai, 200031, China.
| | - Renjie Chai
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Room 611, Building 9, No. 83, Fenyang Road, Xuhui District, Shanghai, 200031, China. .,MOE Key Laboratory for Developmental Genes and Human Disease, State Key Laboratory of Bioelectronics, Co-Innovation Center of Neuroregeneration, Institute of Life Sciences, 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. .,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
|
7
|
Michalski N, Petit C. Genes Involved in the Development and Physiology of Both the Peripheral and Central Auditory Systems. Annu Rev Neurosci 2019; 42:67-86. [DOI: 10.1146/annurev-neuro-070918-050428] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The genetic approach, based on the study of inherited forms of deafness, has proven to be particularly effective for deciphering the molecular mechanisms underlying the development of the peripheral auditory system, the cochlea and its afferent auditory neurons, and how this system extracts the physical parameters of sound. Although this genetic dissection has provided little information about the central auditory system, scattered data suggest that some genes may have a critical role in both the peripheral and central auditory systems. Here, we review the genes controlling the development and function of the peripheral and central auditory systems, focusing on those with demonstrated intrinsic roles in both systems and highlighting the current underappreciation of these genes. Their encoded products are diverse, from transcription factors to ion channels, as are their roles in the central auditory system, mostly evaluated in brainstem nuclei. We examine the ontogenetic and evolutionary mechanisms that may underlie their expression at different sites.
Collapse
Affiliation(s)
- Nicolas Michalski
- Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, 75015 Paris, France;,
- Institut National de la Santé et de la Recherche Médicale, UMRS 1120, 75015 Paris, France
- Sorbonne Universités, 75005 Paris, France
| | - Christine Petit
- Unité de Génétique et Physiologie de l'Audition, Institut Pasteur, 75015 Paris, France;,
- Institut National de la Santé et de la Recherche Médicale, UMRS 1120, 75015 Paris, France
- Sorbonne Universités, 75005 Paris, France
- Syndrome de Usher et Autres Atteintes Rétino-Cochléaires, Institut de la Vision, 75012 Paris, France
- Collège de France, 75005 Paris, France
| |
Collapse
|
8
|
Sato Y, Tsukaguchi H, Morita H, Higasa K, Tran MTN, Hamada M, Usui T, Morito N, Horita S, Hayashi T, Takagi J, Yamaguchi I, Nguyen HT, Harada M, Inui K, Maruta Y, Inoue Y, Koiwa F, Sato H, Matsuda F, Ayabe S, Mizuno S, Sugiyama F, Takahashi S, Yoshimura A. A mutation in transcription factor MAFB causes Focal Segmental Glomerulosclerosis with Duane Retraction Syndrome. Kidney Int 2018; 94:396-407. [PMID: 29779709 DOI: 10.1016/j.kint.2018.02.025] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 01/24/2018] [Accepted: 02/08/2018] [Indexed: 01/02/2023]
Abstract
Focal segmental glomerulosclerosis (FSGS) is a leading cause of end-stage renal disease in children and adults. Genetic factors significantly contribute to early-onset FSGS, but the etiologies of most adult cases remain unknown. Genetic studies of monogenic syndromic FSGS exhibiting extra-renal manifestations have uncovered an unexpected biological role for genes in the development of both podocytes and other cellular lineages. To help define these roles, we studied two unrelated families with FSGS associated with Duane Retraction Syndrome, characterized by impaired horizontal eye movement due to cranial nerve malformation. All four affected individuals developed FSGS and Duane Retraction Syndrome in their first to second decade of life, manifested as restricted abduction together with globe retraction and narrowed palpebral fissure on attempted adduction. Hypoplasia of the abducens nerves and hearing impairment occurred in severely affected individuals. Genetic analyses revealed that affected individuals harbor a rare heterozygous substitution (p.Leu239Pro) in MAFB, a leucine zipper transcription factor. Luciferase assays with cultured monocytes indicated that the substitution significantly reduced transactivation of the F4/80 promoter, the known MAFB recognition element. Additionally, immunohistochemistry indicated reduced MAFB expression in the podocytes of patients. Structural modeling suggested that the p.Leu239Pro substitution in the DNA-binding domain possibly interferes with the stability of the adjacent zinc finger. Lastly, podocytes in neonatal mice with p.Leu239Pro displayed impaired differentiation. Thus, MAFB mutations impair development and/or maintenance of podocytes, abducens neurons and the inner ear. The interactions between MAFB and regulatory elements in these developing organs are likely highly specific based on spatiotemporal requirements.
Collapse
Affiliation(s)
- Yoshinori Sato
- Division of Nephrology, Department of Medicine, Showa University Fujigaoka Hospital, Fujigaoka, Japan
| | - Hiroyasu Tsukaguchi
- Second Department of Internal Medicine, Kansai Medical University, Hirakata, Japan.
| | - Hiroyuki Morita
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Koichiro Higasa
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mai Thi Nhu Tran
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Michito Hamada
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Toshiaki Usui
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan; Department of Nephrology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Naoki Morito
- Department of Nephrology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Shoichiro Horita
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takao Hayashi
- Department of Ophthalmology, School of Medicine, Teikyo University, Tokyo, Japan
| | - Junko Takagi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Izumi Yamaguchi
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Huan Thanh Nguyen
- Second Department of Internal Medicine, Kansai Medical University, Hirakata, Japan
| | - Masayo Harada
- Department of Clinical Anatomy, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kiyoko Inui
- Division of Nephrology, Department of Medicine, Showa University Fujigaoka Hospital, Fujigaoka, Japan
| | - Yuichi Maruta
- Division of Nephrology, Department of Medicine, Showa University Fujigaoka Hospital, Fujigaoka, Japan
| | - Yoshihiko Inoue
- Division of Nephrology, Department of Medicine, Showa University Fujigaoka Hospital, Fujigaoka, Japan
| | - Fumihiko Koiwa
- Division of Nephrology, Department of Medicine, Showa University Fujigaoka Hospital, Fujigaoka, Japan
| | - Hiroshi Sato
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shinya Ayabe
- Experimental Animal Division, RIKEN BioResource Center, Tsukuba, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Fumihiro Sugiyama
- Laboratory Animal Resource Center, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan; Laboratory Animal Resource Center, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan; International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan; Life Science Center of Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Japan
| | - Ashio Yoshimura
- Division of Nephrology, Department of Medicine, Showa University Fujigaoka Hospital, Fujigaoka, Japan
| |
Collapse
|
9
|
Reciprocal Negative Regulation Between Lmx1a and Lmo4 Is Required for Inner Ear Formation. J Neurosci 2018; 38:5429-5440. [PMID: 29769265 PMCID: PMC5990987 DOI: 10.1523/jneurosci.2484-17.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 02/07/2018] [Accepted: 02/11/2018] [Indexed: 02/07/2023] Open
Abstract
LIM-domain containing transcription factors (LIM-TFs) are conserved factors important for embryogenesis. The specificity of these factors in transcriptional regulation is conferred by the complexes that they form with other proteins such as LIM-domain-binding (Ldb) proteins and LIM-domain only (LMO) proteins. Unlike LIM-TFs, these proteins do not bind DNA directly. LMO proteins are negative regulators of LIM-TFs and function by competing with LIM-TFs for binding to Ldb's. Although the LIM-TF Lmx1a is expressed in the developing mouse hindbrain, which provides many of the extrinsic signals for inner ear formation, conditional knock-out embryos of both sexes show that the inner ear source of Lmx1a is the major contributor of ear patterning. In addition, we have found that the reciprocal interaction between Lmx1a and Lmo4 (a LMO protein within the inner ear) mediates the formation of both vestibular and auditory structures. Lmo4 negatively regulates Lmx1a to form the three sensory cristae, the anterior semicircular canal, and the shape of the utricle in the vestibule. Furthermore, this negative regulation blocks ectopic sensory formation in the cochlea. In contrast, Lmx1a negatively regulates Lmo4 in mediating epithelial resorption of the canal pouch, which gives rise to the anterior and posterior semicircular canals. We also found that Lmx1a is independently required for the formation of the endolymphatic duct and hair cells in the basal cochlear region. SIGNIFICANCE STATEMENT The mammalian inner ear is a structurally complex organ responsible for detecting sound and maintaining balance. Failure to form the intricate 3D structure of this organ properly during development most likely will result in sensory deficits on some level. Here, we provide genetic evidence that a transcription factor, Lmx1a, interacts with its negative regulator, Lmo4, to pattern various vestibular and auditory components of the mammalian inner ear. Identifying these key molecules that mediate formation of this important sensory organ will be helpful for designing strategies and therapeutics to alleviate hearing loss and balance disorders.
Collapse
|
10
|
Sculpting the labyrinth: Morphogenesis of the developing inner ear. Semin Cell Dev Biol 2017; 65:47-59. [DOI: 10.1016/j.semcdb.2016.09.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 07/26/2016] [Accepted: 09/25/2016] [Indexed: 01/23/2023]
|
11
|
Johnson Chacko L, Pechriggl EJ, Fritsch H, Rask-Andersen H, Blumer MJF, Schrott-Fischer A, Glueckert R. Neurosensory Differentiation and Innervation Patterning in the Human Fetal Vestibular End Organs between the Gestational Weeks 8-12. Front Neuroanat 2016; 10:111. [PMID: 27895556 PMCID: PMC5108762 DOI: 10.3389/fnana.2016.00111] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 10/28/2016] [Indexed: 11/13/2022] Open
Abstract
Balance orientation depends on the precise operation of the vestibular end organs and the vestibular ganglion neurons. Previous research on the assemblage of the neuronal network in the developing fetal vestibular organ has been limited to data from animal models. Insights into the molecular expression profiles and signaling moieties involved in embryological development of the human fetal inner ear have been limited. We present an investigation of the cells of the vestibular end organs with specific focus on the hair cell differentiation and innervation pattern using an uninterrupted series of unique specimens from gestational weeks 8-12. Nerve fibers positive for peripherin innervate the entire fetal crista and utricle. While in rodents only the peripheral regions of the cristae and the extra-striolar region of the statolithic organs are stained. At week 9, transcription factors PAX2 and PAX8 were observed in the hair cells whereas PAX6 was observed for the first time among the supporting cells of the cristae and the satellite glial cells of the vestibular ganglia. Glutamine synthetase, a regulator of the neurotransmitter glutamate, is strongly expressed among satellite glia cells, transitional zones of the utricle and supporting cells in the sensory epithelium. At gestational week 11, electron microscopic examination reveals bouton contacts at hair cells and first signs of the formation of a protocalyx at type I hair cells. Our study provides first-hand insight into the fetal development of the vestibular end organs as well as their pattern of innervation by means of immunohistochemical and EM techniques, with the aim of contributing toward our understanding of balance development.
Collapse
Affiliation(s)
- Lejo Johnson Chacko
- Department of Otolaryngology, Medical University of Innsbruck Innsbruck, Austria
| | - Elisabeth J Pechriggl
- Department of Anatomy, Histology, and Embryology, Division of Clinical and Functional Anatomy, Medical University of Innsbruck Innsbruck, Austria
| | - Helga Fritsch
- Department of Anatomy, Histology, and Embryology, Division of Clinical and Functional Anatomy, Medical University of Innsbruck Innsbruck, Austria
| | | | - Michael J F Blumer
- Department of Anatomy, Histology, and Embryology, Division of Clinical and Functional Anatomy, Medical University of Innsbruck Innsbruck, Austria
| | | | - Rudolf Glueckert
- Department of Otolaryngology, Medical University of InnsbruckInnsbruck, Austria; University Clinics Innsbruck, Tirol KlinikenInnsbruck, Austria
| |
Collapse
|
12
|
Abstract
OTX proteins, homologs of the Drosophila orthodenticle (Otd), are important for the morphogenesis of the neuroectoderm, and for the central nervous system formation. OTX1 and OTX2 are important for the cochlea and macula development, indeed when OTX1 is knocked down, these organs undergo developmental failure. Moreover OTX2 transfection revert this effect in OTX1−/− mice. The TA isoform of TP63, involved in Notch regulation pathway, has a critical function in the cochlear neuroepithelium differentiation. TAp63 positively regulates Hes5 and Atoh1 transcription. This pathway has been also demonstrated in p63−/− mice, and in patients p63 mutated, affected by Ectodermal Dysplasia (ED, OMIM 129810). These patients are affected by mild sensorineural deafness, most likely related to the mutation in p63 gene impairing the Notch pathway. We demonstrated the role of OTX2 on TAp63 regulation necessary for the correct formation of macular neuroepithelium and we confirmed the impairment of vestibular function caused by p63 mutations. Although the abnormalities found in our patient were still at a subclinical extent, aging could exacerbate this impairment and cause a decrease in quality of life.
Collapse
|
13
|
Olaya-Sánchez D, Sánchez-Guardado LÓ, Ohta S, Chapman SC, Schoenwolf GC, Puelles L, Hidalgo-Sánchez M. Fgf3 and Fgf16 expression patterns define spatial and temporal domains in the developing chick inner ear. Brain Struct Funct 2016; 222:131-149. [PMID: 26995070 DOI: 10.1007/s00429-016-1205-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 02/19/2016] [Indexed: 11/26/2022]
Abstract
The inner ear is a morphologically complex sensory structure with auditory and vestibular functions. The developing otic epithelium gives rise to neurosensory and non-sensory elements of the adult membranous labyrinth. Extrinsic and intrinsic signals manage the patterning and cell specification of the developing otic epithelium by establishing lineage-restricted compartments defined in turn by differential expression of regulatory genes. FGF3 and FGF16 are excellent candidates to govern these developmental events. Using the chick inner ear, we show that Fgf3 expression is present in the borders of all developing cristae. Strong Fgf16 expression was detected in a portion of the developing vertical and horizontal pouches, whereas the cristae show weaker or undetected Fgf16 expression at different developmental stages. Concerning the rest of the vestibular sensory elements, both the utricular and saccular maculae were Fgf3 positive. Interestingly, strong Fgf16 expression delimited these Fgf16-negative sensory patches. The Fgf3-negative macula neglecta and the Fgf3-positive macula lagena were included within weakly Fgf16-expressing areas. Therefore, different FGF-mediated mechanisms might regulate the specification of the anterior (utricular and saccular) and posterior (neglecta and lagena) maculae. In the developing cochlear duct, dynamic Fgf3 and Fgf16 expression suggests their cooperation in the early specification and later cell differentiation in the hearing system. The requirement of Fgf3 and Fgf16 genes in endolymphatic apparatus development and neurogenesis are discussed. Based on these observations, FGF3 and FGF16 seem to be key signaling pathways that control the inner ear plan by defining epithelial identities within the developing otic epithelium.
Collapse
Affiliation(s)
- Daniel Olaya-Sánchez
- Department of Cell Biology, School of Science, University of Extremadura, Avda. de Elvas s/n, 06071, Badajoz, Spain
| | - Luis Óscar Sánchez-Guardado
- Department of Cell Biology, School of Science, University of Extremadura, Avda. de Elvas s/n, 06071, Badajoz, Spain
| | - Sho Ohta
- Department of Neurobiology and Anatomy, University of Utah, 2R066 School of Medicine, 30 N. 1900 E., Salt Lake City, UT, 84132-3401, USA
| | - Susan C Chapman
- Department of Biological Sciences, Clemson University, 340 Long Hall, Clemson, SC, 29634, USA
| | - Gary C Schoenwolf
- Department of Neurobiology and Anatomy, University of Utah, 2R066 School of Medicine, 30 N. 1900 E., Salt Lake City, UT, 84132-3401, USA
| | - Luis Puelles
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia, 30100, Murcia, Spain
| | - Matías Hidalgo-Sánchez
- Department of Cell Biology, School of Science, University of Extremadura, Avda. de Elvas s/n, 06071, Badajoz, Spain.
| |
Collapse
|
14
|
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
|
15
|
Vendrell V, López-Hernández I, Durán Alonso MB, Feijoo-Redondo A, Abello G, Gálvez H, Giráldez F, Lamonerie T, Schimmang T. Otx2 is a target of N-myc and acts as a suppressor of sensory development in the mammalian cochlea. Development 2015; 142:2792-2800. [PMID: 26160903 DOI: 10.1242/dev.122465] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 06/29/2015] [Indexed: 12/30/2022]
Abstract
Transcriptional regulatory networks are essential during the formation and differentiation of organs. The transcription factor N-myc is required for proper morphogenesis of the cochlea and to control correct patterning of the organ of Corti. We show here that the Otx2 gene, a mammalian ortholog of the Drosophila orthodenticle homeobox gene, is a crucial target of N-myc during inner ear development. Otx2 expression is lost in N-myc mouse mutants, and N-myc misexpression in the chick inner ear leads to ectopic expression of Otx2. Furthermore, Otx2 enhancer activity is increased by N-myc misexpression, indicating that N-myc may directly regulate Otx2. Inactivation of Otx2 in the mouse inner ear leads to ectopic expression of prosensory markers in non-sensory regions of the cochlear duct. Upon further differentiation, these domains give rise to an ectopic organ of Corti, together with the re-specification of non-sensory areas into sensory epithelia, and the loss of Reissner's membrane. Therefore, the Otx2-positive domain of the cochlear duct shows a striking competence to develop into a mirror-image copy of the organ of Corti. Taken together, these data show that Otx2 acts downstream of N-myc and is essential for patterning and spatial restriction of the sensory domain of the mammalian cochlea.
Collapse
Affiliation(s)
- Victor Vendrell
- Instituto de Biología y Genética Molecular, Universidad de Valladolid y Consejo Superior de Investigaciones Científicas, C/Sanz y Forés 3, Valladolid E-47003, Spain
| | - Iris López-Hernández
- Instituto de Biología y Genética Molecular, Universidad de Valladolid y Consejo Superior de Investigaciones Científicas, C/Sanz y Forés 3, Valladolid E-47003, Spain
| | - María Beatriz Durán Alonso
- Instituto de Biología y Genética Molecular, Universidad de Valladolid y Consejo Superior de Investigaciones Científicas, C/Sanz y Forés 3, Valladolid E-47003, Spain
| | - Ana Feijoo-Redondo
- Instituto de Biología y Genética Molecular, Universidad de Valladolid y Consejo Superior de Investigaciones Científicas, C/Sanz y Forés 3, Valladolid E-47003, Spain
| | - Gina Abello
- CEXS, Universitat Pompeu Fabra, Parc de Recerca Biomédica de Barcelona, Barcelona E-08003, Spain
| | - Héctor Gálvez
- CEXS, Universitat Pompeu Fabra, Parc de Recerca Biomédica de Barcelona, Barcelona E-08003, Spain
| | - Fernando Giráldez
- CEXS, Universitat Pompeu Fabra, Parc de Recerca Biomédica de Barcelona, Barcelona E-08003, Spain
| | - Thomas Lamonerie
- Institut de Biologie Valrose, University of Nice Sophia Antipolis, UMR UNS/CNRS 7277/INSERM 1091, Nice F-06108, France
| | - Thomas Schimmang
- Instituto de Biología y Genética Molecular, Universidad de Valladolid y Consejo Superior de Investigaciones Científicas, C/Sanz y Forés 3, Valladolid E-47003, Spain
| |
Collapse
|
16
|
Pechriggl EJ, Bitsche M, Glueckert R, Rask‐Andersen H, Blumer MJF, Schrott‐Fischer A, Fritsch H. Development of the innervation of the human inner ear. Dev Neurobiol 2014; 75:683-702. [DOI: 10.1002/dneu.22242] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 10/28/2014] [Accepted: 10/28/2014] [Indexed: 01/04/2023]
Affiliation(s)
- Elisabeth J. Pechriggl
- Department of Anatomy, Histology, and Embryology, Division of Clinical and Functional AnatomyMedical University of InnsbruckMüllerstrasse 596020Innsbruck Austria
| | - Mario Bitsche
- Department of Anatomy, Histology, and Embryology, Division of Clinical and Functional AnatomyMedical University of InnsbruckMüllerstrasse 596020Innsbruck Austria
| | - Rudolf Glueckert
- Department of OtolaryngologyMedical University of InnsbruckAnichstrasse 356020Innsbruck Austria
- University Clinics InnsbruckTiroler LandeskrankenanstaltenInnsbruck Austria
| | - Helge Rask‐Andersen
- Departments of OtolaryngologyUppsala University Hospital751 85Uppsala Sweden
| | - Michael J. F. Blumer
- Department of Anatomy, Histology, and Embryology, Division of Clinical and Functional AnatomyMedical University of InnsbruckMüllerstrasse 596020Innsbruck Austria
| | | | - Helga Fritsch
- Department of Anatomy, Histology, and Embryology, Division of Clinical and Functional AnatomyMedical University of InnsbruckMüllerstrasse 596020Innsbruck Austria
| |
Collapse
|
17
|
Murashima A, Kishigami S, Thomson A, Yamada G. Androgens and mammalian male reproductive tract development. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:163-70. [PMID: 24875095 DOI: 10.1016/j.bbagrm.2014.05.020] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 04/28/2014] [Accepted: 05/19/2014] [Indexed: 12/31/2022]
Abstract
One of the main functions of androgen is in the sexually dimorphic development of the male reproductive tissues. During embryogenesis, androgen determines the morphogenesis of male specific organs, such as the epididymis, seminal vesicle, prostate and penis. Despite the critical function of androgens in masculinization, the downstream molecular mechanisms of androgen signaling are poorly understood. Tissue recombination experiments and tissue specific androgen receptor (AR) knockout mouse studies have revealed epithelial or mesenchymal specific androgen-AR signaling functions. These findings also indicate that epithelial-mesenchymal interactions are a key feature of AR specific activity, and paracrine growth factor action may mediate some of the effects of androgens. This review focuses on mouse models showing the interactions of androgen and growth factor pathways that promote the sexual differentiation of reproductive organs. Recent studies investigating context dependent AR target genes are also discussed. This article is part of a Special Issue entitled: Nuclear receptors in animal development.
Collapse
Affiliation(s)
- Aki Murashima
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Kimiidera 811-1, Wakayama 641-8509, Wakayama, Japan
| | - Satoshi Kishigami
- Faculty of Biology-Oriented Science and Technology, Kinki University, Kinokawa 649-6493, Wakayama, Japan
| | - Axel Thomson
- Department of Urology, McGill University Health Centre, 1650 Cedar Av, Montreal, Québec, H3A 1A4, Canada
| | - Gen Yamada
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Kimiidera 811-1, Wakayama 641-8509, Wakayama, Japan.
| |
Collapse
|
18
|
Abstract
A forward genetic screen of N-ethyl-N-nitrosourea mutagenized Xenopus tropicalis has identified an inner ear mutant named eclipse (ecl). Mutants developed enlarged otic vesicles and various defects of otoconia development; they also showed abnormal circular and inverted swimming patterns. Positional cloning identified specificity protein 8 (sp8), which was previously found to regulate limb and brain development. Two different loss-of-function approaches using transcription activator-like effector nucleases and morpholino oligonucleotides confirmed that the ecl mutant phenotype is caused by down-regulation of sp8. Depletion of sp8 resulted in otic dysmorphogenesis, such as uncompartmentalized and enlarged otic vesicles, epithelial dilation with abnormal sensory end organs. When overexpressed, sp8 was sufficient to induce ectopic otic vesicles possessing sensory hair cells, neurofilament innervation in a thickened sensory epithelium, and otoconia, all of which are found in the endogenous otic vesicle. We propose that sp8 is an important factor for initiation and elaboration of inner ear development.
Collapse
|
19
|
Raft S, Andrade LR, Shao D, Akiyama H, Henkemeyer M, Wu DK. Ephrin-B2 governs morphogenesis of endolymphatic sac and duct epithelia in the mouse inner ear. Dev Biol 2014; 390:51-67. [PMID: 24583262 DOI: 10.1016/j.ydbio.2014.02.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 02/19/2014] [Indexed: 02/03/2023]
Abstract
Control over ionic composition and volume of the inner ear luminal fluid endolymph is essential for normal hearing and balance. Mice deficient in either the EphB2 receptor tyrosine kinase or the cognate transmembrane ligand ephrin-B2 (Efnb2) exhibit background strain-specific vestibular-behavioral dysfunction and signs of abnormal endolymph homeostasis. Using various loss-of-function mouse models, we found that Efnb2 is required for growth and morphogenesis of the embryonic endolymphatic epithelium, a precursor of the endolymphatic sac (ES) and duct (ED), which mediate endolymph homeostasis. Conditional inactivation of Efnb2 in early-stage embryonic ear tissues disrupted cell proliferation, cell survival, and epithelial folding at the origin of the endolymphatic epithelium. This correlated with apparent absence of an ED, mis-localization of ES ion transport cells relative to inner ear sensory organs, dysplasia of the endolymph fluid space, and abnormally formed otoconia (extracellular calcite-protein composites) at later stages of embryonic development. A comparison of Efnb2 and Notch signaling-deficient mutant phenotypes indicated that these two signaling systems have distinct and non-overlapping roles in ES/ED development. Homozygous deletion of the Efnb2 C-terminus caused abnormalities similar to those found in the conditional Efnb2 null homozygote. Analyses of fetal Efnb2 C-terminus deletion heterozygotes found mis-localized ES ion transport cells only in the genetic background exhibiting vestibular dysfunction. We propose that developmental dysplasias described here are a gene dose-sensitive cause of the vestibular dysfunction observed in EphB-Efnb2 signaling-deficient mice.
Collapse
Affiliation(s)
- Steven Raft
- Section on Sensory Cell Regeneration and Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Leonardo R Andrade
- Laboratory of Biomineralization, Institute of Biomedical Sciences, CCS, Universidade Federal do Rio de Janeiro, RJ 21941-902, Brazil
| | - Dongmei Shao
- Department of Otolaryngology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Haruhiko Akiyama
- Department of Orthopedics, Gifu University, Gifu City 501-1194, Japan
| | - Mark Henkemeyer
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Doris K Wu
- Section on Sensory Cell Regeneration and Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
20
|
Yu WM, Appler JM, Kim YH, Nishitani AM, Holt JR, Goodrich LV. A Gata3-Mafb transcriptional network directs post-synaptic differentiation in synapses specialized for hearing. eLife 2013; 2:e01341. [PMID: 24327562 PMCID: PMC3851837 DOI: 10.7554/elife.01341] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 10/29/2013] [Indexed: 01/12/2023] Open
Abstract
Information flow through neural circuits is determined by the nature of the synapses linking the subtypes of neurons. How neurons acquire features distinct to each synapse remains unknown. We show that the transcription factor Mafb drives the formation of auditory ribbon synapses, which are specialized for rapid transmission from hair cells to spiral ganglion neurons (SGNs). Mafb acts in SGNs to drive differentiation of the large postsynaptic density (PSD) characteristic of the ribbon synapse. In Mafb mutant mice, SGNs fail to develop normal PSDs, leading to reduced synapse number and impaired auditory responses. Conversely, increased Mafb accelerates synaptogenesis. Moreover, Mafb is responsible for executing one branch of the SGN differentiation program orchestrated by the Gata3 transcriptional network. Remarkably, restoration of Mafb rescues the synapse defect in Gata3 mutants. Hence, Mafb is a powerful regulator of cell-type specific features of auditory synaptogenesis that offers a new entry point for treating hearing loss. DOI: http://dx.doi.org/10.7554/eLife.01341.001.
Collapse
Affiliation(s)
- Wei-Ming Yu
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Jessica M Appler
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Ye-Hyun Kim
- Department of Otolaryngology and FM Kirby Neurobiology Center, Children’s Hospital Boston, Harvard Medical School, Boston, United States
| | | | - Jeffrey R Holt
- Department of Otolaryngology and FM Kirby Neurobiology Center, Children’s Hospital Boston, Harvard Medical School, Boston, United States
| | - Lisa V Goodrich
- Department of Neurobiology, Harvard Medical School, Boston, United States
| |
Collapse
|
21
|
Zhang Y, Ross AC. Retinoic acid and the transcription factor MafB act together and differentially to regulate aggrecan and matrix metalloproteinase gene expression in neonatal chondrocytes. J Cell Biochem 2013; 114:471-9. [PMID: 22961837 DOI: 10.1002/jcb.24387] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 08/30/2012] [Indexed: 11/11/2022]
Abstract
Vitamin A (VA) and its active form, retinoic acid (RA), are regulators of skeletal development and chondrogenesis. MafB, a transcription factor, has been identified as an important mediator in monocyte and osteoclast differentiation. However, the presence and function of MafB in chondrocytes is not clear. In this study, MafB gene expression was regulated by both the VA status of the mother (VA-marginal, adequate, and supplemented diets) and by direct oral supplementation of the neonates with VARA (VA mixed with 10% RA). Expression was highest in neonates of VA-supplemented versus VA-marginal dams (P < 0.05), and in VARA-treated versus placebo-treated neonates across all diet groups (P < 0.05). To examine cellular changes, primary chondrocytes derived from neonatal rat ribs were cultured in the presence of RA for up to 48 h. MafB mRNA exhibited a time- and dose-dependent increase in response to RA, while the induction of MafB mRNA was attenuated by BMS-493, a pan-RAR inverse agonist, implicating RAR signaling in the regulation of MafB. The genetic knockdown of MafB in chondrocytes using siRNA (MafB(SI) chondrocytes) abrogated the RA-induced increase in MafB expression. MafB(SI) chondrocytes expressed higher levels of aggrecan mRNA. Additionally, the increased matrix metalloproteinase (MMP)3 and MMP13 gene expression due to RA was attenuated in MafB(SI) chondrocytes, while total extracellular matrix staining was increased. These results support a role for MafB as a regulator of chondrocyte gene expression and matrix formation via control of aggrecan, MMP3 and MMP13 expression, and indicate an important role for RA in the regulation of MafB.
Collapse
Affiliation(s)
- Yao Zhang
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA 16802, USA
| | | |
Collapse
|
22
|
Abstract
The inner ear is a structurally complex vertebrate organ built to encode sound, motion, and orientation in space. Given its complexity, it is not surprising that inner ear dysfunction is a relatively common consequence of human genetic mutation. Studies in model organisms suggest that many genes currently known to be associated with human hearing impairment are active during embryogenesis. Hence, the study of inner ear development provides a rich context for understanding the functions of genes implicated in hearing loss. This chapter focuses on molecular mechanisms of inner ear development derived from studies of model organisms.
Collapse
Affiliation(s)
- Doris K Wu
- National Institute on Deafness and Other Communication Disorders, Rockville, Maryland 20850, USA.
| | | |
Collapse
|
23
|
Groves AK, Fekete DM. Shaping sound in space: the regulation of inner ear patterning. Development 2012; 139:245-57. [PMID: 22186725 DOI: 10.1242/dev.067074] [Citation(s) in RCA: 189] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The inner ear is one of the most morphologically elaborate tissues in vertebrates, containing a group of mechanosensitive sensory organs that mediate hearing and balance. These organs are arranged precisely in space and contain intricately patterned sensory epithelia. Here, we review recent studies of inner ear development and patterning which reveal that multiple stages of ear development - ranging from its early induction from the embryonic ectoderm to the establishment of the three cardinal axes and the fine-grained arrangement of sensory cells - are orchestrated by gradients of signaling molecules.
Collapse
Affiliation(s)
- Andrew K Groves
- Departments of Neuroscience and Molecular and Human Genetics, BCM295, Baylor College of Medicine, Houston, TX 77030, USA.
| | | |
Collapse
|
24
|
Tsukiyama T, Yamaguchi TP. Mice lacking Wnt2b are viable and display a postnatal olfactory bulb phenotype. Neurosci Lett 2012; 512:48-52. [PMID: 22326927 DOI: 10.1016/j.neulet.2012.01.062] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 12/07/2011] [Accepted: 01/25/2012] [Indexed: 11/25/2022]
Abstract
Wnts are secreted glycoproteins that play important roles in embryonic development. Wnt2b is transiently expressed in the primitive streak (PS) during gastrulation and in several organs during organogenesis. To determine the biological function of Wnt2b during mouse development, we established a conditional null allele of Wnt2b. Mice lacking Wnt2b were viable, fertile, and displayed a normal life span, however, the olfactory bulb in adult Wnt2b mutant mice was significantly reduced in length. Our results suggest that Wnt2b primarily plays a supportive role in gastrulation and organogenesis, functioning redundantly with canonical Wnts, such as Wnt2, in numerous tissues.
Collapse
Affiliation(s)
- Tadasuke Tsukiyama
- Center for Cancer Research, Cancer and Developmental Biology Laboratory, Cell Signaling in Vertebrate Development Section, NCI-Frederick, NIH, Frederick, MD 21702, USA.
| | | |
Collapse
|
25
|
Chatterjee S, Lufkin T. The sound of silence: mouse models for hearing loss. GENETICS RESEARCH INTERNATIONAL 2011; 2011:416450. [PMID: 22567353 PMCID: PMC3335620 DOI: 10.4061/2011/416450] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 08/10/2011] [Indexed: 01/27/2023]
Abstract
Sensorineural hearing loss is one of the most common disabilities in humans. It is estimated that about 278 million people worldwide have slight to extreme hearing loss in both ears, which results in an economic loss for the country and personal loss for the individual. It is thus critical to have a deeper understanding of the causes for hearing loss to better manage and treat the affected individuals. The mouse serves as an excellent model to study and recapitulate some of these phenotypes, identify new genes which cause deafness, and to study their roles in vivo and in detail. Mutant mice have been instrumental in elucidating the function and mechanisms of the inner ear. The development and morphogenesis of the inner ear from an ectodermal layer into distinct auditory and vestibular components depends on well-coordinated gene expression and well-orchestrated signaling cascades within the otic vesicle and interactions with surrounding layers of tissues. Any disruption in these pathways can lead to hearing impairment. This review takes a look at some of the genes and their corresponding mice mutants that have shed light on the mechanism governing hearing impairment (HI) in humans.
Collapse
Affiliation(s)
- Sumantra Chatterjee
- Stem Cell and Developmental Biology, Genome Institute of Singapore, 60 Biopolis Street, Singapore 138672
| | | |
Collapse
|
26
|
Kopecky B, Fritzsch B. Regeneration of Hair Cells: Making Sense of All the Noise. Pharmaceuticals (Basel) 2011; 4:848-879. [PMID: 21966254 PMCID: PMC3180915 DOI: 10.3390/ph4060848] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 06/04/2011] [Accepted: 06/08/2011] [Indexed: 12/17/2022] Open
Abstract
Hearing loss affects hundreds of millions of people worldwide by dampening or cutting off their auditory connection to the world. Current treatments for sensorineural hearing loss (SNHL) with cochlear implants are not perfect, leaving regenerative medicine as the logical avenue to a perfect cure. Multiple routes to regeneration of damaged hair cells have been proposed and are actively pursued. Each route not only requires a keen understanding of the molecular basis of ear development but also faces the practical limitations of stem cell regulation in the delicate inner ear where topology of cell distribution is essential. Improvements in our molecular understanding of the minimal essential genes necessary for hair cell formation and recent advances in stem cell manipulation, such as seen with inducible pluripotent stem cells (iPSCs) and epidermal neural crest stem cells (EPI-NCSCs), have opened new possibilities to advance research in translational stem cell therapies for individuals with hearing loss. Despite this, more detailed network maps of gene expression are needed, including an appreciation for the roles of microRNAs (miRs), key regulators of transcriptional gene networks. To harness the true potential of stem cells for hair cell regeneration, basic science and clinical medicine must work together to expedite the transition from bench to bedside by elucidating the full mechanisms of inner ear hair cell development, including a focus on the role of miRs, and adapting this knowledge safely and efficiently to stem cell technologies.
Collapse
Affiliation(s)
- Benjamin Kopecky
- Department of Biology, University of Iowa, Iowa City, IA, 52242, USA
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, IA, 52242, USA
| |
Collapse
|
27
|
Sánchez-Guardado LÓ, Ferran JL, Rodríguez-Gallardo L, Puelles L, Hidalgo-Sánchez M. Meis gene expression patterns in the developing chicken inner ear. J Comp Neurol 2011; 519:125-47. [PMID: 21120931 DOI: 10.1002/cne.22508] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We are interested in stable gene network activities operating sequentially during inner ear specification. The implementation of this patterning process is a key event in the generation of functional subdivisions of the otic vesicle during early embryonic development. The vertebrate inner ear is a complex sensory structure that is a good model system for characterization of developmental mechanisms controlling patterning and specification. Meis genes, belonging to the TALE family, encode homodomain-containing transcription factors remarkably conserved during evolution, which play a role in normal and neoplastic development. To gain understanding of the possible role of homeobox Meis genes in the developing chick inner ear, we comprehensively analyzed their spatiotemporal expression patterns from early otic specification stages onwards. In the invaginating otic placode, Meis1/2 transcripts were observed in the borders of the otic cup, being absent in the portion of otic epithelium closest to the hindbrain. As development proceeds, Meis1 and Meis2 expressions became restricted to the dorsomedial otic epithelium. Both genes were strongly expressed in the entire presumptive domain of the semicircular canals, and more weakly in all associated cristae. The endolymphatic apparatus was labeled in part by Meis1/2. Meis1 was also expressed in the lateral wall of the growing cochlear duct, while Meis2 expression was detected in a few cells of the developing acoustic-vestibular ganglion. Our results suggest a possible role of Meis assigning regional identity in the morphogenesis, patterning, and specification of the developing inner ear.
Collapse
|
28
|
Chatterjee S, Kraus P, Lufkin T. A symphony of inner ear developmental control genes. BMC Genet 2010; 11:68. [PMID: 20637105 PMCID: PMC2915946 DOI: 10.1186/1471-2156-11-68] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Accepted: 07/16/2010] [Indexed: 01/21/2023] Open
Abstract
The inner ear is one of the most complex and detailed organs in the vertebrate body and provides us with the priceless ability to hear and perceive linear and angular acceleration (hence maintain balance). The development and morphogenesis of the inner ear from an ectodermal thickening into distinct auditory and vestibular components depends upon precise temporally and spatially coordinated gene expression patterns and well orchestrated signaling cascades within the otic vesicle and upon cellular movements and interactions with surrounding tissues. Gene loss of function analysis in mice has identified homeobox genes along with other transcription and secreted factors as crucial regulators of inner ear morphogenesis and development. While otic induction seems dependent upon fibroblast growth factors, morphogenesis of the otic vesicle into the distinct vestibular and auditory components appears to be clearly dependent upon the activities of a number of homeobox transcription factors. The Pax2 paired-homeobox gene is crucial for the specification of the ventral otic vesicle derived auditory structures and the Dlx5 and Dlx6 homeobox genes play a major role in specification of the dorsally derived vestibular structures. Some Micro RNAs have also been recently identified which play a crucial role in the inner ear formation.
Collapse
Affiliation(s)
- Sumantra Chatterjee
- Stem Cell and Developmental Biology, Genome Institute of Singapore, 60 Biopolis Street, 138672 Singapore
| | | | | |
Collapse
|
29
|
Koo SK, Hill JK, Hwang CH, Lin ZS, Millen KJ, Wu DK. Lmx1a maintains proper neurogenic, sensory, and non-sensory domains in the mammalian inner ear. Dev Biol 2009; 333:14-25. [PMID: 19540218 PMCID: PMC3400700 DOI: 10.1016/j.ydbio.2009.06.016] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Revised: 06/10/2009] [Accepted: 06/11/2009] [Indexed: 12/24/2022]
Abstract
Lmx1a is a LIM homeodomain-containing transcription factor, which is required for the formation of multiple organs. Lmx1a is broadly expressed in early stages of the developing inner ear, but its expression is soon restricted to the non-sensory regions of the developing ear. In an Lmx1a functional null mutant, dreher (dr(J)/dr(J)), the inner ears lack a non-sensory structure, the endolymphatic duct, and the membranous labyrinth is poorly developed. These phenotypes are consistent with Lmx1a's role as a selector gene. More importantly, while all three primary fates of the inner ear - neural, sensory, and non-sensory - are specified in dr(J)/dr(J), normal boundaries among these tissues are often violated. For example, the neurogenic domain of the ear epithelium, from which cells delaminate to form the cochleovestibular ganglion, is expanded. Within the neurogenic domain, the demarcation between the vestibular and auditory neurogenic domains is most likely disrupted as well, based on the increased numbers of vestibular neuroblasts and ectopic expression of Fgf3, which normally is associated specifically with the vestibular neurogenic region. Furthermore, aberrant and ectopic sensory organs are observed; most striking among these is vestibular-like hair cells located in the cochlear duct.
Collapse
Affiliation(s)
- Soo Kyung Koo
- National Institute on Deafness and Other Communication Disorders, 5 Research Court, Rm 2B34, Rockville, Rockville, MD 20850, USA
| | | | | | | | | | | |
Collapse
|
30
|
Vázquez-Echeverría C, Dominguez-Frutos E, Charnay P, Schimmang T, Pujades C. Analysis of mouse kreisler mutants reveals new roles of hindbrain-derived signals in the establishment of the otic neurogenic domain. Dev Biol 2008; 322:167-78. [PMID: 18703040 DOI: 10.1016/j.ydbio.2008.07.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2008] [Revised: 07/17/2008] [Accepted: 07/17/2008] [Indexed: 10/21/2022]
Abstract
The inner ear, the sensory organ responsible for hearing and balance, contains specialized sensory and non-sensory epithelia arranged in a highly complex three-dimensional structure. To achieve this complexity, a tight coordination between morphogenesis and cell fate specification is essential during otic development. Tissues surrounding the otic primordium, and more particularly the adjacent segmented hindbrain, have been implicated in specifying structures along the anteroposterior and dorsoventral axes of the inner ear. In this work we have first characterized the generation and axial specification of the otic neurogenic domain, and second, we have investigated the effects of the mutation of kreisler/MafB--a gene transiently expressed in rhombomeres 5 and 6 of the developing hindbrain--in early otic patterning and cell specification. We show that kr/kr embryos display an expansion of the otic neurogenic domain, due to defects in otic patterning. Although many reports have pointed to the role of FGF3 in otic regionalisation, we provide evidence that FGF3 is not sufficient to govern this process. Neither Krox20 nor Fgf3 mutant embryos, characterized by a downregulation or absence of Fgf3 in r5 and r6, display ectopic neuroblasts in the otic primordium. However, Fgf3-/-Fgf10-/- double mutants show a phenotype very similar to kr/kr embryos: they present ectopic neuroblasts along the AP and DV otic axes. Finally, partial rescue of the kr/kr phenotype is obtained when Fgf3 or Fgf10 are ectopically expressed in the hindbrain of kr/kr embryos. These results highlight the importance of hindbrain-derived signals in the regulation of otic neurogenesis.
Collapse
Affiliation(s)
- Citlali Vázquez-Echeverría
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, C/ Dr. Aiguader 88, 08003 Barcelona, Spain
| | | | | | | | | |
Collapse
|
31
|
Nishida H, Miyagawa S, Matsumaru D, Wada Y, Satoh Y, Ogino Y, Fukuda S, Iguchi T, Yamada G. Gene expression analyses on embryonic external genitalia: identification of regulatory genes possibly involved in masculinization processes. Congenit Anom (Kyoto) 2008; 48:63-7. [PMID: 18452486 DOI: 10.1111/j.1741-4520.2008.00180.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Androgen plays a crucial role in initiating and maintaining the expression of male sexual characteristics in mammals. In humans and mice, any defects along the pathway of androgen functions result in congenital urogenital abnormalities. The genital tubercle (GT), an anlage of the external genitalia, differentiates into a penis in males and a clitoris in females. Although masculinization of the external genitalia is androgen-dependent, the molecular pathway of its potential downstream genes is largely unclear. To identify the genes involved in mouse GT masculinization, we performed gene expression analyses, such as real-time quantitative polymerase chain reaction and section in situ hybridization analysis. From our studies we have identified candidate genes, Cyp1b1, Fkbp51 and MafB as potential androgen targets during mouse GT masculinization.
Collapse
Affiliation(s)
- Hisayo Nishida
- Center for Animal Resources and Development (CARD), Graduate School of Medical and Pharmaceutical Sciences, and Global COE 'Cell Fate Regulation Research and Education Unit', Kumamoto University, Kumamoto, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Adams ME, Hurd EA, Beyer LA, Swiderski DL, Raphael Y, Martin DM. Defects in vestibular sensory epithelia and innervation in mice with loss of Chd7 function: implications for human CHARGE syndrome. J Comp Neurol 2007; 504:519-32. [PMID: 17701983 DOI: 10.1002/cne.21460] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
CHD7 is a chromodomain gene mutated in CHARGE syndrome, a multiple anomaly condition characterized by ocular coloboma, heart defects, atresia of the choanae, retarded growth and development, genital hypoplasia, and ear defects including deafness and semicircular canal dysgenesis. Mice with heterozygous Chd7 deficiency have circling behavior and semicircular canal defects and are an excellent animal model for exploring the pathogenesis of CHARGE features. Inner ear vestibular defects have been characterized in heterozygous Chd7-deficient embryos and early postnatal mice, but it is not known whether vestibular defects persist throughout adulthood in Chd7-deficient mice or whether the vestibular sensory epithelia and their associated innervation and function are intact. Here we describe a detailed analysis of inner ear vestibular structures in mature mice that are heterozygous for a Chd7-deficient, gene-trapped allele (Chd7(Gt/+)). Chd7(Gt/+) mice display variable asymmetric lateral and posterior semicircular canal malformations, as well as defects in vestibular sensory epithelial innervation despite the presence of intact hair cells in the target organs. These observations have important functional implications for understanding the clinical manifestations of CHD7 mutations in humans and for designing therapies to treat inner ear vestibular dysfunction.
Collapse
MESH Headings
- Abnormalities, Multiple/genetics
- Abnormalities, Multiple/pathology
- Animals
- Choanal Atresia/complications
- Choanal Atresia/genetics
- Choanal Atresia/pathology
- DNA-Binding Proteins/deficiency
- Denervation
- Disease Models, Animal
- Epithelium/pathology
- Eye Abnormalities/complications
- Eye Abnormalities/genetics
- Eye Abnormalities/pathology
- Heart Defects, Congenital/complications
- Heart Defects, Congenital/genetics
- Heart Defects, Congenital/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microscopy, Electron, Scanning/methods
- Mutation
- Semicircular Canals/pathology
- Semicircular Canals/ultrastructure
- Stereotyped Behavior
- Syndrome
- Vestibule, Labyrinth/pathology
- Vestibule, Labyrinth/ultrastructure
Collapse
Affiliation(s)
- Meredith E Adams
- Department of Otolaryngology, The University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | |
Collapse
|
33
|
Ranea JAG, Yeats C, Grant A, Orengo CA. Predicting protein function with hierarchical phylogenetic profiles: the Gene3D Phylo-Tuner method applied to eukaryotic genomes. PLoS Comput Biol 2007; 3:e237. [PMID: 18052542 PMCID: PMC2098864 DOI: 10.1371/journal.pcbi.0030237] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2007] [Accepted: 10/17/2007] [Indexed: 11/17/2022] Open
Abstract
“Phylogenetic profiling” is based on the hypothesis that during evolution functionally or physically interacting genes are likely to be inherited or eliminated in a codependent manner. Creating presence–absence profiles of orthologous genes is now a common and powerful way of identifying functionally associated genes. In this approach, correctly determining orthology, as a means of identifying functional equivalence between two genes, is a critical and nontrivial step and largely explains why previous work in this area has mainly focused on using presence–absence profiles in prokaryotic species. Here, we demonstrate that eukaryotic genomes have a high proportion of multigene families whose phylogenetic profile distributions are poor in presence–absence information content. This feature makes them prone to orthology mis-assignment and unsuited to standard profile-based prediction methods. Using CATH structural domain assignments from the Gene3D database for 13 complete eukaryotic genomes, we have developed a novel modification of the phylogenetic profiling method that uses genome copy number of each domain superfamily to predict functional relationships. In our approach, superfamilies are subclustered at ten levels of sequence identity—from 30% to 100%—and phylogenetic profiles built at each level. All the profiles are compared using normalised Euclidean distances to identify those with correlated changes in their domain copy number. We demonstrate that two protein families will “auto-tune” with strong co-evolutionary signals when their profiles are compared at the similarity levels that capture their functional relationship. Our method finds functional relationships that are not detectable by the conventional presence–absence profile comparisons, and it does not require a priori any fixed criteria to define orthologous genes. The vast number of protein sequences being determined by the international genomics projects means that it is not possible to functionally characterise all the proteins through direct experimentation. One of the more successful electronic methods for detecting functionally associated genes has been through the comparison of genes' phylogenetic profiles. This method is based on the hypothesis that two functionally related genes will show very similar presence–absence profile patterns throughout different organisms. Whilst these methods have grown increasingly sophisticated, they have largely been based on detecting functionally homologous genes in different species (technically known as orthologous genes) and thus better suited to prokaryotic genomes, where this can be done more easily. We have developed a new type of hierarchical phylogenetic profile by subdividing protein families into subclusters in different sequence identity levels. This new approach encapsulates a more realistic model of the functional variation that uneven natural selection pressure produces on different protein families and organisms, and it can detect functional relationships between protein families without the initial application of rigid sequence similarity thresholds or complex protocols for orthology assignment. These advantages are especially useful in eukaryotes since the larger average size of eukaryotic multigene families makes them more prone to orthology mis-assignment than in prokaryotes.
Collapse
Affiliation(s)
- Juan A G Ranea
- Department of Biochemistry and Molecular Biology, University College London, London, United Kingdom.
| | | | | | | |
Collapse
|
34
|
Hatch EP, Noyes CA, Wang X, Wright TJ, Mansour SL. Fgf3 is required for dorsal patterning and morphogenesis of the inner ear epithelium. Development 2007; 134:3615-25. [PMID: 17855431 PMCID: PMC2366212 DOI: 10.1242/dev.006627] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The inner ear, which contains sensory organs specialized for hearing and balance, develops from an ectodermal placode that invaginates lateral to hindbrain rhombomeres (r) 5-6 to form the otic vesicle. Under the influence of signals from intra- and extraotic sources, the vesicle is molecularly patterned and undergoes morphogenesis and cell-type differentiation to acquire its distinct functional compartments. We show in mouse that Fgf3, which is expressed in the hindbrain from otic induction through endolymphatic duct outgrowth, and in the prospective neurosensory domain of the otic epithelium as morphogenesis initiates, is required for both auditory and vestibular function. We provide new morphologic data on otic dysmorphogenesis in Fgf3 mutants, which show a range of malformations similar to those of Mafb (Kreisler), Hoxa1 and Gbx2 mutants, the most common phenotype being failure of endolymphatic duct and common crus formation, accompanied by epithelial dilatation and reduced cochlear coiling. The malformations have close parallels with those seen in hearing-impaired patients. The morphologic data, together with an analysis of changes in the molecular patterning of Fgf3 mutant otic vesicles, and comparisons with other mutations affecting otic morphogenesis, allow placement of Fgf3 between hindbrain-expressed Hoxa1 and Mafb, and otic vesicle-expressed Gbx2, in the genetic cascade initiated by WNT signaling that leads to dorsal otic patterning and endolymphatic duct formation. Finally, we show that Fgf3 prevents ventral expansion of r5-6 neurectodermal Wnt3a, serving to focus inductive WNT signals on the dorsal otic vesicle and highlighting a new example of cross-talk between the two signaling systems.
Collapse
Affiliation(s)
- Ekaterina P Hatch
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112-5330, USA
| | | | | | | | | |
Collapse
|
35
|
Choo D. The role of the hindbrain in patterning of the otocyst. Dev Biol 2007; 308:257-65. [PMID: 17601528 PMCID: PMC1986645 DOI: 10.1016/j.ydbio.2007.05.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2007] [Revised: 05/22/2007] [Accepted: 05/24/2007] [Indexed: 11/17/2022]
Affiliation(s)
- Daniel Choo
- Ear and Hearing Center, Univeristy of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, Ohio 45229-3039, USA.
| |
Collapse
|
36
|
Lecaudey V, Ulloa E, Anselme I, Stedman A, Schneider-Maunoury S, Pujades C. Role of the hindbrain in patterning the otic vesicle: a study of the zebrafish vhnf1 mutant. Dev Biol 2006; 303:134-43. [PMID: 17137573 DOI: 10.1016/j.ydbio.2006.10.041] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2006] [Revised: 10/27/2006] [Accepted: 10/27/2006] [Indexed: 10/24/2022]
Abstract
The vertebrate inner ear develops from an ectodermal placode adjacent to rhombomeres 4 to 6 of the segmented hindbrain. The placode then transforms into a vesicle and becomes regionalised along its anteroposterior, dorsoventral and mediolateral axes. To investigate the role of hindbrain signals in instructing otic vesicle regionalisation, we analysed ear development in zebrafish mutants for vhnf1, a gene expressed in the caudal hindbrain during otic induction and regionalisation. We show that, in vhnf1 homozygous embryos, the patterning of the otic vesicle is affected along both the anteroposterior and dorsoventral axes. First, anterior gene expression domains are either expanded along the whole anteroposterior axis of the vesicle or duplicated in the posterior region. Second, the dorsal domain is severely reduced, and cell groups normally located ventrally are shifted dorsally, sometimes forming a single dorsal patch along the whole AP extent of the otic vesicle. Third, and probably as a consequence, the size and organization of the sensory and neurogenic epithelia are disturbed. These results demonstrate that, in zebrafish, signals from the hindbrain control the patterning of the otic vesicle, not only along the anteroposterior axis, but also, as in amniotes, along the dorsoventral axis. They suggest that, despite the evolution of inner ear structure and function, some of the mechanisms underlying the regionalisation of the otic vesicle in fish and amniotes have been conserved.
Collapse
Affiliation(s)
- Virginie Lecaudey
- Unité de Biologie du Développement, CNRS UMR 7622, Université Pierre et Marie Curie, Paris, France
| | | | | | | | | | | |
Collapse
|
37
|
Robledo RF, Lufkin T. Dlx5 and Dlx6 homeobox genes are required for specification of the mammalian vestibular apparatus. Genesis 2006; 44:425-37. [PMID: 16900517 DOI: 10.1002/dvg.20233] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The mammalian inner ear is a complex organ that develops from a surface ectoderm into distinct auditory and vestibular components. Congenital malformation of these two components resulting from single or multiple gene defects is a common clinical occurrence and is observed in patients with split hand/split foot malformation, a malformation which is phenocopied by Dlx5/6 null mice. Analysis of mice lacking Dlx5 and Dlx6 homeobox genes identified their restricted and combined expression in the otic epithelium as a crucial regulator of vestibular cell fates. Otic induction initiates without incident in Dlx5/6(-/-) embryos, but dorsal otic derivatives including the semicircular ducts, utricle, saccule, and endolymphatic duct fail to form. Dlx5 and Dlx6 seem to influence vestibular cell fates by restricting Pax2 and activating Gbx2 and Bmp4 expression domains. Given their proximity to the disease locus and the observed phenotype in Dlx5/6 null mice, Dlx5/6 are likely candidates to mediate the inner ear defects observed in patients with split hand/split foot malformation.
Collapse
|
38
|
Yerukhimovich MV, Bai L, Chen DHC, Miller RH, Alagramam KN. Identification and characterization of mouse cochlear stem cells. Dev Neurosci 2006; 29:251-60. [PMID: 17047322 DOI: 10.1159/000096415] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2006] [Accepted: 06/09/2006] [Indexed: 11/19/2022] Open
Abstract
Genetic, noise- and drug-induced loss of hair cells in the mouse and human cochlea leads to permanent hearing loss due to lack of regeneration of hair cells, which may be due to reduced numbers or loss of the regenerative ability of stem cells in the adult cochlea. We hypothesized that the mouse neonate cochlea harbors stem cells capable of differentiating into hair cells. Cells from the primary neonate cochlear culture began to proliferate and formed floating spheres after 14 days in vitro (DIV). By comparison, spheres from the primary culture of the cortex were observed after 7 DIV. Cochlear sphere cells could be passaged and the new spheres were observed after 7 DIV. Cochlear sphere cells were capable of differentiating into astrocytes and oligodendrocytes, but not neurons under the conditions tested. Cochlear sphere cells expressed Sox2 and Myo7a, but failed to show markers that are expressed exclusively in mature cochlear tissue, while cells from cortex spheres expressed Sox2 and Otx2, but not Myo7a. Our results show that cochleae from neonatal mice harbor cells capable of forming spheres and cells from these spheres appear to be better endowed to become hair cells.
Collapse
Affiliation(s)
- Michael V Yerukhimovich
- Department of Otolaryngology, Head and Neck Surgery, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | | | | | |
Collapse
|
39
|
Xu H, Viola A, Zhang Z, Gerken CP, Lindsay-Illingworth EA, Baldini A. Tbx1 regulates population, proliferation and cell fate determination of otic epithelial cells. Dev Biol 2006; 302:670-82. [PMID: 17074316 PMCID: PMC1850623 DOI: 10.1016/j.ydbio.2006.10.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2006] [Revised: 09/30/2006] [Accepted: 10/03/2006] [Indexed: 11/21/2022]
Abstract
The T-box transcription factor Tbx1 is required for inner ear morphogenesis. Tbx1 null mutants have a small otocyst that fails to grow and remodel and does not give rise to the vestibular and cochlear apparata. Here we show that Tbx1 expression-driven cell tracing identifies a population of otic epithelial cells that contributes to most of the otocyst. Tbx1 is essential for the contribution of this population to the inner ear. Ablation of Tbx1 after this cell population has established itself in the otocyst, restores marker expression lost in germ line mutants, but causes severe reduction in mitotic activity, cell autonomously. Furthermore, timed cell fate mapping demonstrates that loss of Tbx1 switches the fate of some members of the Tbx1-dependent cell population, from non-neurogenic to neurogenic, an event associated with activation of the Delta-Notch pathway. Finally, tissue-specific ablation of Tbx1 demonstrates that, while the abovementioned phenotypic abnormalities are due to loss of epithelial expression of Tbx1, cochlear morphogenesis requires mesodermal Tbx1 expression. We conclude that the main functions of Tbx1 in the inner ear are to control, cell-autonomously, contribution, size and fate of a large population of otic epithelial cells, and, cell non-autonomously, cochlear morphogenesis.
Collapse
Affiliation(s)
- Huansheng Xu
- Institute of Biosciences and Technology, Texas A&M University System, Houston, TX, 77030
- Program in Cardiovascular Sciences, Baylor College of Medicine, Houston, TX, 77030
| | - Antonella Viola
- Department of Biochemistry, University Federico II, and Tigem, Via P. Castellino Naples, Italy
| | - Zhen Zhang
- Program in Cardiovascular Sciences, Baylor College of Medicine, Houston, TX, 77030
| | - Claudia P. Gerken
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030
| | - Elizabeth A. Lindsay-Illingworth
- Institute of Biosciences and Technology, Texas A&M University System, Houston, TX, 77030
- Department of Biochemistry, University Federico II, and Tigem, Via P. Castellino Naples, Italy
| | - Antonio Baldini
- Institute of Biosciences and Technology, Texas A&M University System, Houston, TX, 77030
- Department of Biochemistry, University Federico II, and Tigem, Via P. Castellino Naples, Italy
- Program in Cardiovascular Sciences, Baylor College of Medicine, Houston, TX, 77030
| |
Collapse
|