1
|
Norton B, Quirk A, Matsuoka AJ. Unraveling the Mechanisms of Vestibular Neuron Formation from Human Induced Pluripotent Stem Cells. Tissue Eng Part A 2024; 30:131-143. [PMID: 37917115 DOI: 10.1089/ten.tea.2023.0166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023] Open
Abstract
The development of in vitro models that accurately recapitulate the complex cellular and molecular interactions of the inner ear is crucial for understanding inner ear development, function, and disease. In this study, we utilized a customized microfluidic platform to generate human induced pluripotent stem cell (hiPSC)-derived three-dimensional otic sensory neurons (OSNs). hiPSC-derived otic neuronal progenitors (ONPs) were cultured in hydrogel-embedded microfluidic channels over a 40-day period. Careful modulation of Wnt and Shh signaling pathways was used to influence dorsoventral patterning and direct differentiation toward a vestibular neuron lineage. After validating the microfluidic platform, OSN spheroid transcription factor and protein expression were assessed using real-time quantitative polymerase chain reaction (RT-qPCR), immunocytochemistry, and flow cytometry. The results demonstrated the successful differentiation of hiPSCs into ONPs and subsequent divergent differentiation into vestibular neuronal lineages, as evidenced by the expression of characteristic markers. Overall, our microfluidic platform provides a physiologically relevant environment for the culture and differentiation of hiPSCs, offering a valuable tool for studying inner ear development, disease and drug screening, and regenerative medicine applications.
Collapse
Affiliation(s)
- Benjamin Norton
- Department of Otolaryngology and Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Analia Quirk
- Department of Otolaryngology and Head and Neck Surgery, University of California San Diego, La Jolla, California, USA
| | - Akihiro J Matsuoka
- Department of Otolaryngology and Head and Neck Surgery, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
2
|
Bañón A, Alsina B. Pioneer statoacoustic neurons guide neuroblast behaviour during otic ganglion assembly. Development 2023; 150:dev201824. [PMID: 37938828 PMCID: PMC10651105 DOI: 10.1242/dev.201824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 09/07/2023] [Indexed: 11/10/2023]
Abstract
Cranial ganglia are aggregates of sensory neurons that mediate distinct types of sensation. The statoacoustic ganglion (SAG) develops into several lobes that are spatially arranged to connect appropriately with hair cells of the inner ear. To investigate the cellular behaviours involved in the 3D organization of the SAG, we use high-resolution confocal imaging of single-cell, labelled zebrafish neuroblasts (NBs), photoconversion, photoablation, and genetic perturbations. We show that otic NBs delaminate out of the otic epithelium in an epithelial-mesenchymal transition-like manner, rearranging apical polarity and primary cilia proteins. We also show that, once delaminated, NBs require RhoGTPases in order to perform active migration. Furthermore, tracking of recently delaminated NBs revealed their directed migration and coalescence around a small population of pioneer SAG neurons. These pioneer SAG neurons, not from otic placode origin, populate the coalescence region before otic neurogenesis begins and their ablation disrupts delaminated NB migratory pathways, consequentially affecting SAG shape. Altogether, this work shows for the first time the role of pioneer SAG neurons in orchestrating SAG development.
Collapse
Affiliation(s)
- Aitor Bañón
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra-Parc de Recerca Biomèdica de Barcelona, Dr Aiguader 88, 08003 Barcelona, Spain
| | - Berta Alsina
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra-Parc de Recerca Biomèdica de Barcelona, Dr Aiguader 88, 08003 Barcelona, Spain
| |
Collapse
|
3
|
Matern MS, Durruthy-Durruthy R, Birol O, Darmanis S, Scheibinger M, Groves AK, Heller S. Transcriptional dynamics of delaminating neuroblasts in the mouse otic vesicle. Cell Rep 2023; 42:112545. [PMID: 37227818 PMCID: PMC10592509 DOI: 10.1016/j.celrep.2023.112545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 02/23/2023] [Accepted: 05/04/2023] [Indexed: 05/27/2023] Open
Abstract
An abundance of research has recently highlighted the susceptibility of cochleovestibular ganglion (CVG) neurons to noise damage and aging in the adult cochlea, resulting in hearing deficits. Furthering our understanding of the transcriptional cascades that contribute to CVG development may provide insight into how these cells can be regenerated to treat inner ear dysfunction. Here we perform a high-depth single-cell RNA sequencing analysis of the E10.5 otic vesicle and its surrounding tissues, including CVG precursor neuroblasts and emerging CVG neurons. Clustering and trajectory analysis of otic-lineage cells reveals otic markers and the changes in gene expression that occur from neuroblast delamination toward the development of the CVG. This dataset provides a valuable resource for further identifying the mechanisms associated with CVG development from neurosensory competent cells within the otic vesicle.
Collapse
Affiliation(s)
- Maggie S Matern
- Department of Otolaryngology Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Robert Durruthy-Durruthy
- Department of Otolaryngology Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Onur Birol
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Spyros Darmanis
- Departments of Bioengineering and Applied Physics and Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Mirko Scheibinger
- Department of Otolaryngology Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Andrew K Groves
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Stefan Heller
- Department of Otolaryngology Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
4
|
Origin of Neuroblasts in the Avian Otic Placode and Their Distributions in the Acoustic and Vestibular Ganglia. BIOLOGY 2023; 12:biology12030453. [PMID: 36979145 PMCID: PMC10045822 DOI: 10.3390/biology12030453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/09/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023]
Abstract
The inner ear is a complex three-dimensional sensorial structure with auditory and vestibular functions. This intricate sensory organ originates from the otic placode, which generates the sensory elements of the membranous labyrinth, as well as all the ganglionic neuronal precursors. How auditory and vestibular neurons establish their fate identities remains to be determined. Their topological origin in the incipient otic placode could provide positional information before they migrate, to later segregate in specific portions of the acoustic and vestibular ganglia. To address this question, transplants of small portions of the avian otic placode were performed according to our previous fate map study, using the quail/chick chimeric graft model. All grafts taking small areas of the neurogenic placodal domain contributed neuroblasts to both acoustic and vestibular ganglia. A differential distribution of otic neurons in the anterior and posterior lobes of the vestibular ganglion, as well as in the proximal, intermediate, and distal portions of the acoustic ganglion, was found. Our results clearly show that, in birds, there does not seem to be a strict segregation of acoustic and vestibular neurons in the incipient otic placode.
Collapse
|
5
|
Single-cell transcriptomic landscapes of the otic neuronal lineage at multiple early embryonic ages. Cell Rep 2022; 38:110542. [PMID: 35320729 DOI: 10.1016/j.celrep.2022.110542] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/03/2021] [Accepted: 02/25/2022] [Indexed: 11/20/2022] Open
Abstract
Inner ear vestibular and spiral ganglion neurons (VGNs and SGNs) are known to play pivotal roles in balance control and sound detection. However, the molecular mechanisms underlying otic neurogenesis at early embryonic ages have remained unclear. Here, we use single-cell RNA sequencing to reveal the transcriptomes of mouse otic tissues at three embryonic ages, embryonic day 9.5 (E9.5), E11.5, and E13.5, covering proliferating and undifferentiated otic neuroblasts and differentiating VGNs and SGNs. We validate the high quality of our studies by using multiple assays, including genetic fate mapping analysis, and we uncover several genes upregulated in neuroblasts or differentiating VGNs and SGNs, such as Shox2, Myt1, Casz1, and Sall3. Notably, our findings suggest a general cascaded differentiation trajectory during early otic neurogenesis. The comprehensive understanding of early otic neurogenesis provided by our study holds critical implications for both basic and translational research.
Collapse
|
6
|
Riley BB. Comparative assessment of Fgf's diverse roles in inner ear development: A zebrafish perspective. Dev Dyn 2021; 250:1524-1551. [PMID: 33830554 DOI: 10.1002/dvdy.343] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/26/2021] [Accepted: 03/26/2021] [Indexed: 01/21/2023] Open
Abstract
Progress in understanding mechanisms of inner ear development has been remarkably rapid in recent years. The research community has benefited from the availability of several diverse model organisms, including zebrafish, chick, and mouse. The complexity of the inner ear has proven to be a challenge, and the complexity of the mammalian cochlea in particular has been the subject of intense scrutiny. Zebrafish lack a cochlea and exhibit a number of other differences from amniote species, hence they are sometimes seen as less relevant for inner ear studies. However, accumulating evidence shows that underlying cellular and molecular mechanisms are often highly conserved. As a case in point, consideration of the diverse functions of Fgf and its downstream effectors reveals many similarities between vertebrate species, allowing meaningful comparisons the can benefit the entire research community. In this review, I will discuss mechanisms by which Fgf controls key events in early otic development in zebrafish and provide direct comparisons with chick and mouse.
Collapse
Affiliation(s)
- Bruce B Riley
- Biology Department, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
7
|
Almasoudi SH, Schlosser G. Otic Neurogenesis in Xenopus laevis: Proliferation, Differentiation, and the Role of Eya1. Front Neuroanat 2021; 15:722374. [PMID: 34616280 PMCID: PMC8488300 DOI: 10.3389/fnana.2021.722374] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/27/2021] [Indexed: 11/15/2022] Open
Abstract
Using immunostaining and confocal microscopy, we here provide the first detailed description of otic neurogenesis in Xenopus laevis. We show that the otic vesicle comprises a pseudostratified epithelium with apicobasal polarity (apical enrichment of Par3, aPKC, phosphorylated Myosin light chain, N-cadherin) and interkinetic nuclear migration (apical localization of mitotic, pH3-positive cells). A Sox3-immunopositive neurosensory area in the ventromedial otic vesicle gives rise to neuroblasts, which delaminate through breaches in the basal lamina between stages 26/27 and 39. Delaminated cells congregate to form the vestibulocochlear ganglion, whose peripheral cells continue to proliferate (as judged by EdU incorporation), while central cells differentiate into Islet1/2-immunopositive neurons from stage 29 on and send out neurites at stage 31. The central part of the neurosensory area retains Sox3 but stops proliferating from stage 33, forming the first sensory areas (utricular/saccular maculae). The phosphatase and transcriptional coactivator Eya1 has previously been shown to play a central role for otic neurogenesis but the underlying mechanism is poorly understood. Using an antibody specifically raised against Xenopus Eya1, we characterize the subcellular localization of Eya1 proteins, their levels of expression as well as their distribution in relation to progenitor and neuronal differentiation markers during otic neurogenesis. We show that Eya1 protein localizes to both nuclei and cytoplasm in the otic epithelium, with levels of nuclear Eya1 declining in differentiating (Islet1/2+) vestibulocochlear ganglion neurons and in the developing sensory areas. Morpholino-based knockdown of Eya1 leads to reduction of proliferating, Sox3- and Islet1/2-immunopositive cells, redistribution of cell polarity proteins and loss of N-cadherin suggesting that Eya1 is required for maintenance of epithelial cells with apicobasal polarity, progenitor proliferation and neuronal differentiation during otic neurogenesis.
Collapse
Affiliation(s)
| | - Gerhard Schlosser
- School of Natural Sciences, National University of Galway, Galway, Ireland
| |
Collapse
|
8
|
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
|
9
|
León Y, Magariños M, Varela-Nieto I. Ceramide Kinase Inhibition Blocks IGF-1-Mediated Survival of Otic Neurosensory Progenitors by Impairing AKT Phosphorylation. Front Cell Dev Biol 2021; 9:678760. [PMID: 34179008 PMCID: PMC8220815 DOI: 10.3389/fcell.2021.678760] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/12/2021] [Indexed: 11/29/2022] Open
Abstract
Sphingolipids are bioactive lipid components of cell membranes with important signal transduction functions in health and disease. Ceramide is the central building block for sphingolipid biosynthesis and is processed to form structurally and functionally distinct sphingolipids. Ceramide can be phosphorylated by ceramide kinase (CERK) to generate ceramide-1-phosphate, a cytoprotective signaling molecule that has been widely studied in multiple tissues and organs, including the developing otocyst. However, little is known about ceramide kinase regulation during inner ear development. Using chicken otocysts, we show that genes for CERK and other enzymes of ceramide metabolism are expressed during the early stages of inner ear development and that CERK is developmentally regulated at the otic vesicle stage. To explore its role in inner ear morphogenesis, we blocked CERK activity in organotypic cultures of otic vesicles with a specific inhibitor. Inhibition of CERK activity impaired proliferation and promoted apoptosis of epithelial otic progenitors. CERK inhibition also compromised neurogenesis of the acoustic-vestibular ganglion. Insulin-like growth factor-1 (IGF-1) is a key factor for proliferation, survival and differentiation in the chicken otocyst. CERK inhibition decreased IGF-1-induced AKT phosphorylation and blocked IGF-1-induced cell survival. Overall, our data suggest that CERK is activated as a central element in the network of anti-apoptotic pro-survival pathways elicited by IGF-1 during early inner ear development.
Collapse
Affiliation(s)
- Yolanda León
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Madrid, Spain.,Departamento de Biología, Universidad Autónoma de Madrid, Madrid, Spain
| | - Marta Magariños
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Madrid, Spain.,Departamento de Biología, Universidad Autónoma de Madrid, Madrid, Spain.,CIBERER, Unit 761, CIBER, ISCIII, Madrid, Spain
| | - Isabel Varela-Nieto
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Madrid, Spain.,CIBERER, Unit 761, CIBER, ISCIII, Madrid, Spain
| |
Collapse
|
10
|
Karimi-Boroujeni M, Zahedi-Amiri A, Coombs KM. Embryonic Origins of Virus-Induced Hearing Loss: Overview of Molecular Etiology. Viruses 2021; 13:71. [PMID: 33419104 PMCID: PMC7825458 DOI: 10.3390/v13010071] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/30/2020] [Accepted: 12/31/2020] [Indexed: 12/19/2022] Open
Abstract
Hearing loss, one of the most prevalent chronic health conditions, affects around half a billion people worldwide, including 34 million children. The World Health Organization estimates that the prevalence of disabling hearing loss will increase to over 900 million people by 2050. Many cases of congenital hearing loss are triggered by viral infections during different stages of pregnancy. However, the molecular mechanisms by which viruses induce hearing loss are not sufficiently explored, especially cases that are of embryonic origins. The present review first describes the cellular and molecular characteristics of the auditory system development at early stages of embryogenesis. These developmental hallmarks, which initiate upon axial specification of the otic placode as the primary root of the inner ear morphogenesis, involve the stage-specific regulation of several molecules and pathways, such as retinoic acid signaling, Sonic hedgehog, and Wnt. Different RNA and DNA viruses contributing to congenital and acquired hearing loss are then discussed in terms of their potential effects on the expression of molecules that control the formation of the auditory and vestibular compartments following otic vesicle differentiation. Among these viruses, cytomegalovirus and herpes simplex virus appear to have the most effect upon initial molecular determinants of inner ear development. Moreover, of the molecules governing the inner ear development at initial stages, SOX2, FGFR3, and CDKN1B are more affected by viruses causing either congenital or acquired hearing loss. Abnormalities in the function or expression of these molecules influence processes like cochlear development and production of inner ear hair and supporting cells. Nevertheless, because most of such virus-host interactions were studied in unrelated tissues, further validations are needed to confirm whether these viruses can mediate the same effects in physiologically relevant models simulating otic vesicle specification and growth.
Collapse
Affiliation(s)
- Maryam Karimi-Boroujeni
- School of Rehabilitation Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1H 8M5, Canada;
| | - Ali Zahedi-Amiri
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
- Manitoba Centre for Proteomics and Systems Biology, Winnipeg, MB R3E 3P4, Canada
| | - Kevin M. Coombs
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
- Manitoba Centre for Proteomics and Systems Biology, Winnipeg, MB R3E 3P4, Canada
- Children’s Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| |
Collapse
|
11
|
Cui L, Zheng J, Zhao Q, Chen JR, Liu H, Peng G, Wu Y, Chen C, He Q, Shi H, Yin S, Friedman RA, Chen Y, Guan MX. Mutations of MAP1B encoding a microtubule-associated phosphoprotein cause sensorineural hearing loss. JCI Insight 2020; 5:136046. [PMID: 33268592 PMCID: PMC7714412 DOI: 10.1172/jci.insight.136046] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 10/28/2020] [Indexed: 12/20/2022] Open
Abstract
The pathophysiology underlying spiral ganglion cell defect–induced deafness remains elusive. Using the whole exome sequencing approach, in combination with functional assays and a mouse disease model, we identified the potentially novel deafness-causative MAP1B gene encoding a highly conserved microtubule-associated protein. Three novel heterozygous MAP1B mutations (c.4198A>G, p.1400S>G; c.2768T>C, p.923I>T; c.5512T>C, p.1838F>L) were cosegregated with autosomal dominant inheritance of nonsyndromic sensorineural hearing loss in 3 unrelated Chinese families. Here, we show that MAP1B is highly expressed in the spiral ganglion neurons in the mouse cochlea. Using otic sensory neuron–like cells, generated by pluripotent stem cells from patients carrying the MAP1B mutation and control subject, we demonstrated that the p.1400S>G mutation caused the reduced levels and deficient phosphorylation of MAP1B, which are involved in the microtubule stability and dynamics. Strikingly, otic sensory neuron–like cells exhibited disturbed dynamics of microtubules, axonal elongation, and defects in electrophysiological properties. Dysfunctions of these derived otic sensory neuron–like cells were rescued by genetically correcting MAP1B mutation using CRISPR/Cas9 technology. Involvement of MAP1B in hearing was confirmed by audiometric evaluation of Map1b heterozygous KO mice. These mutant mice displayed late-onset progressive sensorineural hearing loss that was more pronounced in the high frequencies. The spiral ganglion neurons isolated from Map1b mutant mice exhibited the deficient phosphorylation and disturbed dynamics of microtubules. Map1b deficiency yielded defects in the morphology and electrophysiology of spiral ganglion neurons, but it did not affect the morphologies of cochlea in mice. Therefore, our data demonstrate that dysfunctions of spiral ganglion neurons induced by MAP1B deficiency caused hearing loss. Dysfunctions of spiral ganglion neurons caused by Map1b deficiency leads to sensorineural hearing loss.
Collapse
Affiliation(s)
- Limei Cui
- Division of Medical Genetics and Genomics, The Children's Hospital.,Institute of Genetics and.,Department of Human Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jing Zheng
- Division of Medical Genetics and Genomics, The Children's Hospital
| | - Qiong Zhao
- Division of Medical Genetics and Genomics, The Children's Hospital.,Institute of Genetics and.,Department of Human Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jia-Rong Chen
- Division of Medical Genetics and Genomics, The Children's Hospital.,Institute of Genetics and
| | | | - Guanghua Peng
- Deaprtment of Otorhinolaryngology, the Affiliated Hospital, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yue Wu
- Division of Medical Genetics and Genomics, The Children's Hospital
| | - Chao Chen
- Division of Medical Genetics and Genomics, The Children's Hospital.,Institute of Genetics and
| | | | - Haosong Shi
- Department of Otorhinolaryngology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Shankai Yin
- Department of Otorhinolaryngology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Rick A Friedman
- Division of Otolaryngology, University of California at San Diego School of Medicine, La Jolla California, USA
| | - Ye Chen
- Division of Medical Genetics and Genomics, The Children's Hospital.,Institute of Genetics and.,Department of Human Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Min-Xin Guan
- Division of Medical Genetics and Genomics, The Children's Hospital.,Institute of Genetics and.,Department of Human Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Division of Otolaryngology, University of California at San Diego School of Medicine, La Jolla California, USA.,Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorders, Hangzhou, Zhejiang, China.,Joint Institute of Genetics and Genomic Medicine between Zhejiang University and University of Toronto, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
12
|
Law S, Stout M, Rensch A, Rowsell JM. Expression of MYOSIN VIIA in developing mouse cochleovestibular ganglion neurons. Gene Expr Patterns 2020; 35:119092. [PMID: 31918020 DOI: 10.1016/j.gep.2019.119092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 12/23/2019] [Accepted: 12/24/2019] [Indexed: 10/25/2022]
Abstract
Myosins make up a large super family of motor proteins responsible for actin-based motility in most eukaryotic cells. Myosin VIIA is essential for the development and function of sensory hair cells in the inner ear. The role of Myosin VIIA in the development of cochleovestibular ganglion (CVG) neurons in the mouse is largely unknown. Neurons of the CVG innervate sensory hair cells of the cochlea and vestibular organs to transmit hearing and balance information respectively to the brain. The aim of this study was to characterize the expression of MYOSIN VIIA in the CVG of mouse embryos. Spatiotemporal expression of MYOSIN VIIA was characterized in embryonic (E) mouse inner ear neurons from E9.5 to postnatal (P) day 0. At early stages, when otic neurons begin to delaminate to form the CVG, MYOSIN VIIA was co-expressed with TuJ1, ISLET1 and NEUROD in the otic epithelium and CVG. When CVG neurons were migrating and exiting mitosis, MYSOSIN VIIA was downregulated in a subset of neurons, which were NEUROD-negative and GATA3-positive. After segregation of the CVG, MYOSIN VIIA was observed in a subset of vestibular neurons marked by TUJ1 and absent in cochlear neurons, marked by GATA3. The differential expression of MYOSIN VIIA may indicate a role in inner ear neuron migration and specific labeling of vestibular neurons.
Collapse
Affiliation(s)
- Sarah Law
- Department of Biology, Saint Mary's College, Notre Dame, IN, 46556, USA.
| | - Molly Stout
- Department of Biology, Saint Mary's College, Notre Dame, IN, 46556, USA.
| | - Amanda Rensch
- Department of Biology, Saint Mary's College, Notre Dame, IN, 46556, USA.
| | - Jennifer M Rowsell
- Department of Biology, Saint Mary's College, Notre Dame, IN, 46556, USA.
| |
Collapse
|
13
|
Sánchez-Guardado LÓ, Puelles L, Hidalgo-Sánchez M. Origin of acoustic-vestibular ganglionic neuroblasts in chick embryos and their sensory connections. Brain Struct Funct 2019; 224:2757-2774. [PMID: 31396696 DOI: 10.1007/s00429-019-01934-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 07/31/2019] [Indexed: 01/03/2023]
Abstract
The inner ear is a complex three-dimensional sensory structure with auditory and vestibular functions. It originates from the otic placode, which generates the sensory elements of the membranous labyrinth and all the ganglionic neuronal precursors. Neuroblast specification is the first cell differentiation event. In the chick, it takes place over a long embryonic period from the early otic cup stage to at least stage HH25. The differentiating ganglionic neurons attain a precise innervation pattern with sensory patches, a process presumably governed by a network of dendritic guidance cues which vary with the local micro-environment. To study the otic neurogenesis and topographically-ordered innervation pattern in birds, a quail-chick chimaeric graft technique was used in accordance with a previously determined fate-map of the otic placode. Each type of graft containing the presumptive domain of topologically-arranged placodal sensory areas was shown to generate neuroblasts. The differentiated grafted neuroblasts established dendritic contacts with a variety of sensory patches. These results strongly suggest that, rather than reverse-pathfinding, the relevant role in otic dendritic process guidance is played by long-range diffusing molecules.
Collapse
Affiliation(s)
| | - Luis Puelles
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia, E30100, Murcia, Spain.,Instituto Murciano de Investigaciones Biosanitarias (IMIB-Arrixaca), E30100, Murcia, Spain
| | - Matías Hidalgo-Sánchez
- Department of Cell Biology, School of Science, University of Extremadura, E06071, Badajoz, Spain.
| |
Collapse
|
14
|
Noda T, Meas SJ, Nogami J, Amemiya Y, Uchi R, Ohkawa Y, Nishimura K, Dabdoub A. Direct Reprogramming of Spiral Ganglion Non-neuronal Cells into Neurons: Toward Ameliorating Sensorineural Hearing Loss by Gene Therapy. Front Cell Dev Biol 2018; 6:16. [PMID: 29492404 PMCID: PMC5817057 DOI: 10.3389/fcell.2018.00016] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 01/31/2018] [Indexed: 01/22/2023] Open
Abstract
Primary auditory neurons (PANs) play a critical role in hearing by transmitting sound information from the inner ear to the brain. Their progressive degeneration is associated with excessive noise, disease and aging. The loss of PANs leads to permanent hearing impairment since they are incapable of regenerating. Spiral ganglion non-neuronal cells (SGNNCs), comprised mainly of glia, are resident within the modiolus and continue to survive after PAN loss. These attributes make SGNNCs an excellent target for replacing damaged PANs through cellular reprogramming. We used the neurogenic pioneer transcription factor Ascl1 and the auditory neuron differentiation factor NeuroD1 to reprogram SGNNCs into induced neurons (iNs). The overexpression of both Ascl1 and NeuroD1 in vitro generated iNs at high efficiency. Transcriptome analyses revealed that iNs displayed a transcriptome profile resembling that of endogenous PANs, including expression of several key markers of neuronal identity: Tubb3, Map2, Prph, Snap25, and Prox1. Pathway analyses indicated that essential pathways in neuronal growth and maturation were activated in cells upon neuronal induction. Furthermore, iNs extended projections toward cochlear hair cells and cochlear nucleus neurons when cultured with each respective tissue. Taken together, our study demonstrates that PAN-like neurons can be generated from endogenous SGNNCs. This work suggests that gene therapy can be a viable strategy to treat sensorineural hearing loss caused by degeneration of PANs.
Collapse
Affiliation(s)
- Teppei Noda
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Otolaryngology - Head and Neck Surgery, Kyushu University, Fukuoka, Japan
| | - Steven J Meas
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Jumpei Nogami
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yutaka Amemiya
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Ryutaro Uchi
- Department of Otolaryngology - Head and Neck Surgery, Kyushu University, Fukuoka, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Koji Nishimura
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada.,Hearing Communication Medical Center, Shiga Medical Center Research Institute, Moriyama, Japan
| | - Alain Dabdoub
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Department of Otolaryngology - Head & Neck Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
15
|
Panaliappan TK, Wittmann W, Jidigam VK, Mercurio S, Bertolini JA, Sghari S, Bose R, Patthey C, Nicolis SK, Gunhaga L. Sox2 is required for olfactory pit formation and olfactory neurogenesis through BMP restriction and Hes5 upregulation. Development 2018; 145:145/2/dev153791. [PMID: 29352015 PMCID: PMC5825848 DOI: 10.1242/dev.153791] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 12/11/2017] [Indexed: 12/26/2022]
Abstract
The transcription factor Sox2 is necessary to maintain pluripotency of embryonic stem cells, and to regulate neural development. Neurogenesis in the vertebrate olfactory epithelium persists from embryonic stages through adulthood. The role Sox2 plays for the development of the olfactory epithelium and neurogenesis within has, however, not been determined. Here, by analysing Sox2 conditional knockout mouse embryos and chick embryos deprived of Sox2 in the olfactory epithelium using CRISPR-Cas9, we show that Sox2 activity is crucial for the induction of the neural progenitor gene Hes5 and for subsequent differentiation of the neuronal lineage. Our results also suggest that Sox2 activity promotes the neurogenic domain in the nasal epithelium by restricting Bmp4 expression. The Sox2-deficient olfactory epithelium displays diminished cell cycle progression and proliferation, a dramatic increase in apoptosis and finally olfactory pit atrophy. Moreover, chromatin immunoprecipitation data show that Sox2 directly binds to the Hes5 promoter in both the PNS and CNS. Taken together, our results indicate that Sox2 is essential to establish, maintain and expand the neuronal progenitor pool by suppressing Bmp4 and upregulating Hes5 expression. Summary: Analysis of Sox2 mutant mouse and Sox2 CRISPR-targeted chick embryos reveals that Sox2 controls the establishment of sensory progenitors in the olfactory epithelium by suppressing Bmp4 and upregulating Hes5 expression.
Collapse
Affiliation(s)
| | - Walter Wittmann
- Umeå Centre for Molecular Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Vijay K Jidigam
- Umeå Centre for Molecular Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Sara Mercurio
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
| | - Jessica A Bertolini
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
| | - Soufien Sghari
- Umeå Centre for Molecular Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Raj Bose
- Umeå Centre for Molecular Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Cedric Patthey
- Umeå Centre for Molecular Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Silvia K Nicolis
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
| | - Lena Gunhaga
- Umeå Centre for Molecular Medicine, Umeå University, 901 87 Umeå, Sweden
| |
Collapse
|
16
|
Gálvez H, Tena JJ, Giraldez F, Abelló G. The Repression of Atoh1 by Neurogenin1 during Inner Ear Development. Front Mol Neurosci 2017; 10:321. [PMID: 29104531 PMCID: PMC5655970 DOI: 10.3389/fnmol.2017.00321] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 09/21/2017] [Indexed: 01/01/2023] Open
Abstract
Atonal homolog 1 (Atoh1) and Neurogenin1 (Neurog1) are basic Helix-Loop-Helix (bHLH) transcription factors crucial for the generation of hair cells (HCs) and neurons in the inner ear. Both genes are induced early in development, but the expression of Atoh1 is counteracted by Neurog1. As a result, HC development is prevented during neurogenesis. This work aimed at understanding the molecular basis of this interaction. Atoh1 regulation depends on a 3'Atoh1-enhancer that is the site for Atoh1 autoregulation. Reporter assays on chick embryos and P19 cells show that Neurog1 hampers the autoactivation of Atoh1, the effect being cell autonomous and independent on Notch activity. Assay for Transposase-Accessible Chromatin with high throughput sequencing (ATAC-Seq) analysis shows that the region B of the 3'Atoh1-enhancer is accessible during development and sufficient for both activation and repression. Neurog1 requires the regions flanking the class A E-box to show its repressor effect, however, it does not require binding to DNA for Atoh1 repression. This depends on the dimerization domains Helix-1 and Helix-2 and the reduction of Atoh1 protein levels. The results point towards the acceleration of Atoh1 mRNA degradation as the potential mechanism for the reduction of Atoh1 levels. Such a mechanism dissociates the prevention of Atoh1 expression in neurosensory progenitors from the unfolding of the neurogenic program.
Collapse
Affiliation(s)
- Héctor Gálvez
- DCEXS, Universitat Pompeu Fabra (UPF) - Parc de Recerca Biomèdica de Barcelona (PRBB), Barcelona, Spain
| | - Juan J Tena
- Centro Andaluz de Biología del Desarrollo, Consejo Superior de Investigaciones Científicas (CSIC), Sevilla, Spain
| | - Fernando Giraldez
- DCEXS, Universitat Pompeu Fabra (UPF) - Parc de Recerca Biomèdica de Barcelona (PRBB), Barcelona, Spain
| | - Gina Abelló
- DCEXS, Universitat Pompeu Fabra (UPF) - Parc de Recerca Biomèdica de Barcelona (PRBB), Barcelona, Spain
| |
Collapse
|
17
|
Abstract
Neurons of the cochleovestibular ganglion (CVG) transmit hearing and balance information to the brain. During development, a select population of early otic progenitors express NEUROG1, delaminate from the otocyst, and coalesce to form the neurons that innervate all inner ear sensory regions. At present, the selection process that determines which otic progenitors activate NEUROG1 and adopt a neuroblast fate is incompletely understood. The transcription factor SOX2 has been implicated in otic neurogenesis, but its requirement in the specification of the CVG neurons has not been established. Here we tested SOX2's requirement during inner ear neuronal specification using a conditional deletion paradigm in the mouse. SOX2 deficiency at otocyst stages caused a near-absence of NEUROG1-expressing neuroblasts, increased cell death in the neurosensory epithelium, and significantly reduced the CVG volume. Interestingly, a milder decrease in neurogenesis was observed in heterozygotes, indicating SOX2 levels are important. Moreover, fate-mapping experiments revealed that the timing of SOX2 expression did not parallel the established vestibular-then-auditory sequence. These results demonstrate that SOX2 is required for the initial events in otic neuronal specification including expression of NEUROG1, although fate-mapping results suggest SOX2 may be required as a competence factor rather than a direct initiator of the neural fate.
Collapse
Affiliation(s)
- Aleta R Steevens
- Flaum Eye Institute, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Jenna C Glatzer
- Flaum Eye Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Amy E Kiernan
- Flaum Eye Institute, University of Rochester Medical Center, Rochester, NY, USA. .,Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
18
|
Hoijman E, Fargas L, Blader P, Alsina B. Pioneer neurog1 expressing cells ingress into the otic epithelium and instruct neuronal specification. eLife 2017; 6. [PMID: 28537554 PMCID: PMC5476427 DOI: 10.7554/elife.25543] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Accepted: 05/23/2017] [Indexed: 11/30/2022] Open
Abstract
Neural patterning involves regionalised cell specification. Recent studies indicate that cell dynamics play instrumental roles in neural pattern refinement and progression, but the impact of cell behaviour and morphogenesis on neural specification is not understood. Here we combine 4D analysis of cell behaviours with dynamic quantification of proneural expression to uncover the construction of the zebrafish otic neurogenic domain. We identify pioneer cells expressing neurog1 outside the otic epithelium that migrate and ingress into the epithelialising placode to become the first otic neuronal progenitors. Subsequently, neighbouring cells express neurog1 inside the placode, and apical symmetric divisions amplify the specified pool. Interestingly, pioneer cells delaminate shortly after ingression. Ablation experiments reveal that pioneer cells promote neurog1 expression in other otic cells. Finally, ingression relies on the epithelialisation timing controlled by FGF activity. We propose a novel view for otic neurogenesis integrating cell dynamics whereby ingression of pioneer cells instructs neuronal specification. DOI:http://dx.doi.org/10.7554/eLife.25543.001 The inner ear is responsible for our senses of hearing and balance, and is made up of a series of fluid-filled cavities. Sounds, and movements of the head, cause the fluid within these cavities to move. This activates neurons that line the cavities, causing them to increase their firing rates and pass on information about the sounds or head movements to the brain. Damage to these neurons can result in deafness or vertigo. But where do the neurons themselves come from? It is generally assumed that all inner ear neurons develop inside an area of the embryo called the inner ear epithelium. Cells in this region are thought to switch on a gene called neurog1, triggering a series of changes that turn them into inner ear neurons. However, using advanced microscopy techniques in zebrafish embryos, Hoijman, Fargas et al. now show that this is not the whole story. While zebrafish do not have external ears, they do possess fluid-filled structures for balance and hearing that are similar to those of other vertebrates. Zebrafish embryos are also transparent, which means that activation of genes can be visualized directly. By imaging zebrafish embryos in real time, Hoijman, Fargas et al. show that the first cells to switch on neurog1 do so outside the inner ear epithelium. These pioneer cells then migrate into the inner ear epithelium and switch on neurog1 in their new neighbors. A substance called fibroblast growth factor tells the inner ear epithelium to let the pioneers enter, and thereby controls the final number of inner ear neurons. The work of Hoijman, Fargas et al. reveals how coordinated activation of genes and movement of cells gives rise to inner ear neurons. This should provide insights into the mechanisms that generate other types of sensory tissue. In the long term, the advances made in this study may lead to new strategies for repairing damaged sensory nerves. DOI:http://dx.doi.org/10.7554/eLife.25543.002
Collapse
Affiliation(s)
- Esteban Hoijman
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - L Fargas
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Patrick Blader
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Berta Alsina
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
19
|
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]
|
20
|
Modrell MS, Lyne M, Carr AR, Zakon HH, Buckley D, Campbell AS, Davis MC, Micklem G, Baker CV. Insights into electrosensory organ development, physiology and evolution from a lateral line-enriched transcriptome. eLife 2017; 6. [PMID: 28346141 PMCID: PMC5429088 DOI: 10.7554/elife.24197] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 03/23/2017] [Indexed: 01/22/2023] Open
Abstract
The anamniote lateral line system, comprising mechanosensory neuromasts and electrosensory ampullary organs, is a useful model for investigating the developmental and evolutionary diversification of different organs and cell types. Zebrafish neuromast development is increasingly well understood, but neither zebrafish nor Xenopus is electroreceptive and our molecular understanding of ampullary organ development is rudimentary. We have used RNA-seq to generate a lateral line-enriched gene-set from late-larval paddlefish (Polyodon spathula). Validation of a subset reveals expression in developing ampullary organs of transcription factor genes critical for hair cell development, and genes essential for glutamate release at hair cell ribbon synapses, suggesting close developmental, physiological and evolutionary links between non-teleost electroreceptors and hair cells. We identify an ampullary organ-specific proneural transcription factor, and candidates for the voltage-sensing L-type Cav channel and rectifying Kv channel predicted from skate (cartilaginous fish) ampullary organ electrophysiology. Overall, our results illuminate ampullary organ development, physiology and evolution.
Collapse
Affiliation(s)
- Melinda S Modrell
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Mike Lyne
- Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom.,Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Adrian R Carr
- Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom.,Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Harold H Zakon
- Department of Neuroscience, The University of Texas at Austin, Austin, United States.,Department of Integrative Biology, The University of Texas at Austin, Austin, United States
| | - David Buckley
- Departmento de Biodiversidad y Biología Evolutiva, Museo Nacional de Ciencias Naturales-MNCN-CSIC, Madrid, Spain.,Department of Natural Sciences, Saint Louis University - Madrid Campus, Madrid, Spain
| | - Alexander S Campbell
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Marcus C Davis
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, United States
| | - Gos Micklem
- Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom.,Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Clare Vh Baker
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
21
|
Gálvez H, Abelló G, Giraldez F. Signaling and Transcription Factors during Inner Ear Development: The Generation of Hair Cells and Otic Neurons. Front Cell Dev Biol 2017; 5:21. [PMID: 28393066 PMCID: PMC5364141 DOI: 10.3389/fcell.2017.00021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 03/02/2017] [Indexed: 12/21/2022] Open
Abstract
Integration between cell signals and bHLH transcription factors plays a prominent role during the development of hair cells of the inner ear. Hair cells are the sensory receptors of the inner ear, responsible for the mechano-transduction of sound waves into electrical signals. They derive from multipotent progenitors that reside in the otic placode. Progenitor commitment is the result of cell signaling from the surrounding tissues that result in the restricted expression of SoxB1 transcription factors, Sox2 and Sox3. In turn, they induce the expression of Neurog1 and Atoh1, two bHLH factors that specify neuronal and hair cell fates, respectively. Neuronal and hair cell development, however, do not occur simultaneously. Hair cell development is prevented during neurogenesis and prosensory stages, resulting in the delay of hair cell development with respect to neuron production. Negative interactions between Neurog1 and Atoh1, and of Atoh1 with other bHLH factors driven by Notch signaling, like Hey1 and Hes5, account for this delay. In summary, the regulation of Atoh1 and hair cell development relies on interactions between cell signaling and bHLH transcription factors that dictate cell fate and timing decisions during development. Interestingly, these mechanisms operate as well during hair cell regeneration after damage and during stem cell directed differentiation, making developmental studies instrumental for improving therapies for hearing impairment.
Collapse
Affiliation(s)
- Héctor Gálvez
- Developmental Biology, CEXS, Parc de Recerca Biomèdica de Barcelona, Universitat Pompeu Fabra Barcelona, Spain
| | - Gina Abelló
- Developmental Biology, CEXS, Parc de Recerca Biomèdica de Barcelona, Universitat Pompeu Fabra Barcelona, Spain
| | - Fernando Giraldez
- Developmental Biology, CEXS, Parc de Recerca Biomèdica de Barcelona, Universitat Pompeu Fabra Barcelona, Spain
| |
Collapse
|
22
|
Deng X, Wu DK. Temporal coupling between specifications of neuronal and macular fates of the inner ear. Dev Biol 2016; 414:21-33. [PMID: 27083418 DOI: 10.1016/j.ydbio.2016.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 04/08/2016] [Accepted: 04/11/2016] [Indexed: 12/21/2022]
Abstract
The inner ear is a complex organ comprised of various specialized sensory organs for detecting sound and head movements. The timing of specification for these sensory organs, however, is not clear. Previous fate mapping results of the inner ear indicate that vestibular and auditory ganglia and two of the vestibular sensory organs, the utricular macula (UM) and saccular macula (SM), are lineage related. Based on the medial-lateral relationship where respective auditory and vestibular neuroblasts exit from the otic epithelium and the subsequent formation of the medial SM and lateral UM in these regions, we hypothesized that specification of the two lateral structures, the vestibular ganglion and the UM are coupled and likewise for the two medial structures, the auditory ganglion and the SM. We tested this hypothesis by surgically inverting the primary axes of the otic cup in ovo and investigating the fate of the vestibular neurogenic region, which had been spotted with a lipophilic dye. Our results showed that the laterally-positioned, dye-associated, vestibular ganglion and UM were largely normal in transplanted ears, whereas both auditory ganglion and SM showed abnormalities suggesting the lateral but not the medial-derived structures were mostly specified at the time of transplantation. Both of these results are consistent with a temporal coupling between neuronal and macular fate specifications.
Collapse
Affiliation(s)
- Xiaohong Deng
- National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, United States
| | - Doris K Wu
- National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, United States.
| |
Collapse
|
23
|
Evsen L, Doetzlhofer A. Gene Transfer into the Chicken Auditory Organ by In Ovo Micro-electroporation. J Vis Exp 2016. [PMID: 27167684 DOI: 10.3791/53864] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Chicken embryos are ideal model systems for studying embryonic development as manipulations of gene function can be conducted with relative ease in ovo. The inner ear auditory sensory organ is critical for our ability to hear. It houses a highly specialized sensory epithelium that consists of mechano-transducing hair cells (HCs) and surrounding glial-like supporting cells (SCs). Despite structural differences in the auditory organs, molecular mechanisms regulating the development of the auditory organ are evolutionarily conserved between mammals and aves. In ovo electroporation is largely limited to early stages at E1 - E3. Due to the relative late development of the auditory organ at E5, manipulations of the auditory organ by in ovo electroporation past E3 are difficult due to the advanced development of the chicken embryo at later stages. The method presented here is a transient gene transfer method for targeting genes of interest at stage E4 - E4.5 in the developing chicken auditory sensory organ via in ovo micro-electroporation. This method is applicable for gain- and loss-of-functions with conventional plasmid DNA-based expression vectors and can be combined with in ovo cell proliferation assay by adding EdU (5-ethynyl-2´-deoxyuridine) to the whole embryo at the time of electroporation. The use of green or red fluorescent protein (GFP or RFP) expression plasmids allows the experimenter to quickly determine whether the electroporation successfully targeted the auditory portion of the developing inner ear. In this method paper, representative examples of GFP electroporated specimens are illustrated; embryos were harvested 18 - 96 hr after electroporation and targeting of GFP to the pro-sensory area of the auditory organ was confirmed by RNA in situ hybridization. The method paper also provides an optimized protocol for the use of the thymidine analog EdU to analyze cell proliferation; an example of an EdU based cell proliferation assay that combines immuno-labeling and click EdU chemistry is provided.
Collapse
Affiliation(s)
- Lale Evsen
- The Solomon H. Snyder Department of Neuroscience, The Center for Sensory Biology, The Johns Hopkins University, School of Medicine
| | - Angelika Doetzlhofer
- The Solomon H. Snyder Department of Neuroscience, The Center for Sensory Biology, The Johns Hopkins University, School of Medicine;
| |
Collapse
|
24
|
Ohlemiller KK, Kiener AL, Gagnon PM. QTL Mapping of Endocochlear Potential Differences between C57BL/6J and BALB/cJ mice. J Assoc Res Otolaryngol 2016; 17:173-94. [PMID: 26980469 DOI: 10.1007/s10162-016-0558-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 02/25/2016] [Indexed: 12/18/2022] Open
Abstract
We reported earlier that the endocochlear potential (EP) differs between C57BL/6J (B6) and BALB/cJ (BALB) mice, being lower in BALBs by about 10 mV (Ohlemiller et al. Hear Res 220: 10-26, 2006). This difference corresponds to strain differences with respect to the density of marginal cells in cochlear stria vascularis. After about 1 year of age, BALB mice also tend toward EP reduction that correlates with further marginal cell loss. We therefore suggested that early sub-clinical features of the BALB stria vascularis may predispose these mice to a condition modeling Schuknecht's strial presbycusis. We further reported (Ohlemiller et al. J Assoc Res Otolaryngol 12: 45-58, 2011) that the acute effects of a 2-h 110 dB SPL noise exposure differ between B6 and BALB mice, such that the EP remains unchanged in B6 mice, but is reduced by 40-50 mV in BALBs. In about 25 % of BALBs, the EP does not completely recover, so that permanent EP reduction may contribute to noise-induced permanent threshold shifts in BALBs. To identify genes and alleles that may promote natural EP variation as well as noise-related EP reduction in BALB mice, we have mapped related quantitative trait loci (QTLs) using 12 recombinant inbred (RI) strains formed from B6 and BALB (CxB1-CxB12). EP and strial marginal cell density were measured in B6 mice, BALB mice, their F1 hybrids, and RI mice without noise exposure, and 1-3 h after broadband noise (4-45 kHz, 110 dB SPL, 2 h). For unexposed mice, the strain distribution patterns for EP and marginal cell density were used to generate preliminary QTL maps for both EP and marginal cell density. Six QTL regions were at least statistically suggestive, including a significant QTL for marginal cell density on chromosome 12 that overlapped a weak QTL for EP variation. This region, termed Maced (Marginal cell density QTL) supports the notion of marginal cell density as a genetically influenced contributor to natural EP variation. Candidate genes for Maced notably include Foxg1, Foxa1, Akap6, Nkx2-1, and Pax9. Noise exposure produced significant EP reductions in two RI strains as well as significant EP increases in two RI strains. QTL mapping of the EP in noise-exposed RI mice yielded four suggestive regions. Two of these overlapped with QTL regions we previously identified for noise-related EP reduction in CBA/J mice (Ohlemiller et al. Hear Res 260: 47-53, 2010) on chromosomes 5 and 18 (Nirep). The present map may narrow the Nirep interval to a ~10-Mb region of proximal Chr. 18 that includes Zeb1, Arhgap12, Mpp7, and Gjd4. This study marks the first exploration of natural gene variants that modulate the EP. Their orthologs may underlie some human hearing loss that originates in the lateral wall.
Collapse
Affiliation(s)
- Kevin K Ohlemiller
- Department of Otolaryngology, Central Institute for the Deaf at Washington University School of Medicine, Fay and Carl Simons Center for Hearing and Deafness, Washington University School of Medicine, 660 S. Euclid, St. Louis, MO, 63110, USA.
| | - Anna L Kiener
- Department of Speech and Hearing Science, Ohio State University, Columbus, OH, USA
| | - Patricia M Gagnon
- Department of Otolaryngology, Central Institute for the Deaf at Washington University School of Medicine, Fay and Carl Simons Center for Hearing and Deafness, Washington University School of Medicine, 660 S. Euclid, St. Louis, MO, 63110, USA
| |
Collapse
|
25
|
Goodrich LV. Early Development of the Spiral Ganglion. THE PRIMARY AUDITORY NEURONS OF THE MAMMALIAN COCHLEA 2016. [DOI: 10.1007/978-1-4939-3031-9_2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
26
|
Cochlear afferent innervation development. Hear Res 2015; 330:157-69. [DOI: 10.1016/j.heares.2015.07.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 06/02/2015] [Accepted: 07/21/2015] [Indexed: 01/11/2023]
|
27
|
Expression and Misexpression of the miR-183 Family in the Developing Hearing Organ of the Chicken. PLoS One 2015; 10:e0132796. [PMID: 26176784 PMCID: PMC4503353 DOI: 10.1371/journal.pone.0132796] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 06/19/2015] [Indexed: 12/31/2022] Open
Abstract
The miR-183 family consists of 3 related microRNAs (miR-183, miR-96, miR-182) that are required to complete maturation of primary sensory cells in the mammalian inner ear. Because the level of these microRNAs is not uniform across hair cell subtypes in the murine cochlea, the question arises as to whether hair cell phenotypes are influenced by microRNA expression levels. To address this, we used the chicken embryo to study expression and misexpression of this gene family. By in situ hybridization, expression of all 3 microRNAs is robust in immature hair cells of both auditory and vestibular organs and is present in the statoacoustic ganglion. The auditory organ, called the basilar papilla, shows a weak radial gradient (highest on the neural side) in prosensory cells near the base on embryonic day 7. About nine days later, the basilar papilla also displays a longitudinal gradient (highest in apical hair cells) for the 3 microRNAs. Tol2-mediated gene delivery was used to ask whether cell phenotypes are malleable when the prosensory epithelium was forced to overexpress the miR-183 family. The expression plasmid included EGFP as a reporter located upstream of an intron carrying the microRNA genes. The vectors were electroporated into the otic cup/vesicle, resulting in strong co-expression of EGFP and the miR-183 family that persisted for at least 2 weeks. This manipulation did not generate ectopic hair cells in non-sensory territories of the cochlear duct, although within the basilar papilla, hair cells were over-represented relative to supporting cells. There was no evidence for a change in hair cell phenotypes, such as short-to-tall, or basal-to-apical hair cell features. Therefore, while increasing expression of the miR-183 family was sufficient to influence cell lineage decisions, it did not redirect the differentiation of hair cells towards alternative radial or longitudinal phenotypes.
Collapse
|
28
|
Willaredt MA, Schlüter T, Nothwang HG. The gene regulatory networks underlying formation of the auditory hindbrain. Cell Mol Life Sci 2015; 72:519-535. [PMID: 25332098 PMCID: PMC11113740 DOI: 10.1007/s00018-014-1759-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 09/24/2014] [Accepted: 10/09/2014] [Indexed: 01/28/2023]
Abstract
Development and evolution of auditory hindbrain nuclei are two major unsolved issues in hearing research. Recent characterization of transgenic mice identified the rhombomeric origins of mammalian auditory nuclei and unraveled genes involved in their formation. Here, we provide an overview on these data by assembling them into rhombomere-specific gene regulatory networks (GRNs), as they underlie developmental and evolutionary processes. To explore evolutionary mechanisms, we compare the GRNs operating in the mammalian auditory hindbrain with data available from the inner ear and other vertebrate groups. Finally, we propose that the availability of genomic sequences from all major vertebrate taxa and novel genetic techniques for non-model organisms provide an unprecedented opportunity to investigate development and evolution of the auditory hindbrain by comparative molecular approaches. The dissection of the molecular mechanisms leading to auditory structures will also provide an important framework for auditory processing disorders, a clinical problem difficult to tackle so far. These data will, therefore, foster basic and clinical hearing research alike.
Collapse
Affiliation(s)
- Marc A Willaredt
- Neurogenetics group, Center of Excellence Hearing4All, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26111, Oldenburg, Germany.
| | - Tina Schlüter
- Neurogenetics group, Center of Excellence Hearing4All, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26111, Oldenburg, Germany
| | - Hans Gerd Nothwang
- Neurogenetics group, Center of Excellence Hearing4All, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26111, Oldenburg, Germany.
- Research Center for Neurosensory Science, Carl von Ossietzky University Oldenburg, 26111, Oldenburg, Germany.
| |
Collapse
|
29
|
Su YX, Hou CC, Yang WX. Control of hair cell development by molecular pathways involving Atoh1, Hes1 and Hes5. Gene 2014; 558:6-24. [PMID: 25550047 DOI: 10.1016/j.gene.2014.12.054] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 11/23/2014] [Accepted: 12/25/2014] [Indexed: 01/14/2023]
Abstract
Atoh1, Hes1 and Hes5 are crucial for normal inner ear hair cell development. They regulate the expression of each other in a complex network, while they also interact with many other genes and pathways, such as Notch, FGF, SHH, WNT, BMP and RA. This paper summarized molecular pathways that involve Atoh1, Hes1, and Hes5. Some of the pathways and gene regulation mechanisms discussed here were studied in other tissues, yet they might inspire studies in inner ear hair cell development. Thereby, we presented a complex regulatory network involving these three genes, which might be crucial for proliferation and differentiation of inner ear hair cells.
Collapse
Affiliation(s)
- Yi-Xun Su
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Cong-Cong Hou
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
30
|
Petrovic J, Gálvez H, Neves J, Abelló G, Giraldez F. Differential regulation of Hes/Hey genes during inner ear development. Dev Neurobiol 2014; 75:703-20. [PMID: 25363712 DOI: 10.1002/dneu.22243] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 10/28/2014] [Indexed: 11/09/2022]
Abstract
Notch signaling plays a crucial role during inner ear development and regeneration. Hes/Hey genes encode for bHLH transcription factors identified as Notch targets. We have studied the expression and regulation of Hes/Hey genes during inner ear development in the chicken embryo. Among several Hes/Hey genes examined, only Hey1 and Hes5 map to the sensory regions, although with salient differences. Hey1 expression follows Jag1 expression except at early prosensory stages while Hes5 expression corresponds well to Dl1 expression throughout otic development. Although Hey1 and Hes5 are direct Notch downstream targets, they differ in the level of Notch required for activation. Moreover, they also differ in mRNA stability, showing different temporal decays after Notch blockade. In addition, Bmp, Wnt and Fgf pathways also modify Hey1 and Hes5 expression in the inner ear. Particularly, the Wnt pathway modulates Hey1 and Jag1 expression. Finally, gain of function experiments show that Hey1 and Hes5 cross-regulate each other in a complex manner. Both Hey1 and Hes5 repress Dl1 and Hes5 expression, suggesting that they prevent the transition to differentiation stages, probably by preventing Atoh1 expression. In spite of its association with Jag1, Hey1 does not seem to be instrumental for lateral induction as it does not promote Jag1 expression. We suggest that, besides being both targets of Notch, Hey1 and Hes5 are subject to a rather complex regulation that includes the stability of their transcripts, cross regulation and other signaling pathways.
Collapse
Affiliation(s)
- Jelena Petrovic
- Developmental Biology Unit, CEXS, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona (PRBB), Barcelona, Spain
| | - Hector Gálvez
- Developmental Biology Unit, CEXS, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona (PRBB), Barcelona, Spain
| | - Joana Neves
- Developmental Biology Unit, CEXS, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona (PRBB), Barcelona, Spain
| | - Gina Abelló
- Developmental Biology Unit, CEXS, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona (PRBB), Barcelona, Spain
| | - Fernando Giraldez
- Developmental Biology Unit, CEXS, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona (PRBB), Barcelona, Spain
| |
Collapse
|
31
|
Kim HJ, Ryu J, Woo HM, Cho SS, Sung MK, Kim SC, Park MH, Park T, Koo SK. Patterns of gene expression associated with Pten deficiency in the developing inner ear. PLoS One 2014; 9:e97544. [PMID: 24893171 PMCID: PMC4043736 DOI: 10.1371/journal.pone.0097544] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 04/19/2014] [Indexed: 12/26/2022] Open
Abstract
In inner ear development, phosphatase and tensin homolog (PTEN) is necessary for neuronal maintenance, such as neuronal survival and accurate nerve innervations of hair cells. We previously reported that Pten conditional knockout (cKO) mice exhibited disorganized fasciculus with neuronal apoptosis in spiral ganglion neurons (SGNs). To better understand the genes and signaling networks related to auditory neuron maintenance, we compared the profiles of differentially expressed genes (DEGs) using microarray analysis of the inner ear in E14.5 Pten cKO and wild-type mice. We identified 46 statistically significant transcripts using significance analysis of microarrays, with the false-discovery rate set at 0%. Among the DEGs, expression levels of candidate genes and expression domains were validated by quantitative real-time RT-PCR and in situ hybridization, respectively. Ingenuity pathway analysis using DEGs identified significant signaling networks associated with apoptosis, cellular movement, and axon guidance (i.e., secreted phosphoprotein 1 (Spp1)-mediated cellular movement and regulator of G-protein signaling 4 (Rgs4)-mediated axon guidance). This result was consistent with the phenotypic defects of SGNs in Pten cKO mice (e.g., neuronal apoptosis, abnormal migration, and irregular nerve fiber patterns of SGNs). From this study, we suggest two key regulatory signaling networks mediated by Spp1 and Rgs4, which may play potential roles in neuronal differentiation of developing auditory neurons.
Collapse
Affiliation(s)
- Hyung Jin Kim
- Division of Intractable Diseases, Center for Biomedical Sciences, National Institute of Health, Chungcheongbuk-do, South Korea
| | - Jihee Ryu
- Division of Intractable Diseases, Center for Biomedical Sciences, National Institute of Health, Chungcheongbuk-do, South Korea
| | - Hae-Mi Woo
- Division of Intractable Diseases, Center for Biomedical Sciences, National Institute of Health, Chungcheongbuk-do, South Korea
| | - Samuel Sunghwan Cho
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, South Korea
| | - Min Kyung Sung
- Korean BioInformation Center (KOBIC), Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Sang Cheol Kim
- Korean BioInformation Center (KOBIC), Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Mi-Hyun Park
- Division of Intractable Diseases, Center for Biomedical Sciences, National Institute of Health, Chungcheongbuk-do, South Korea
| | - Taesung Park
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, South Korea
- Department of Statistics, Seoul National University, Seoul, South Korea
| | - Soo Kyung Koo
- Division of Intractable Diseases, Center for Biomedical Sciences, National Institute of Health, Chungcheongbuk-do, South Korea
- * E-mail:
| |
Collapse
|
32
|
Sánchez-Guardado LÓ, Puelles L, Hidalgo-Sánchez M. Fate map of the chicken otic placode. Development 2014; 141:2302-12. [PMID: 24821982 DOI: 10.1242/dev.101667] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The inner ear is an intricate three-dimensional sensory organ that arises from a flat, thickened portion of the ectoderm termed the otic placode. There is evidence that the ontogenetic steps involved in the progressive specification of the highly specialized inner ear of vertebrates involve the concerted actions of diverse patterning signals that originate from nearby tissues, providing positional identity and instructive context. The topology of the prospective inner ear portions at placode stages when such patterning begins has remained largely unknown. The chick-quail model was used to perform a comprehensive fate mapping study of the chick otic placode, shedding light on the precise topological position of each presumptive inner ear component relative to the dorsoventral and anteroposterior axes of the otic placode and, implicitly, to the possible sources of inducing signals. The findings reveal the existence of three dorsoventrally arranged anteroposterior domains from which the endolymphatic system, the maculae and basilar papilla, and the cristae develop. This study provides new bases for the interpretation of earlier and future descriptive and experimental studies that aim to understand the molecular genetic mechanisms involved in otic placode patterning.
Collapse
Affiliation(s)
| | - Luis Puelles
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia, 30003 Murcia, Spain
| | - Matías Hidalgo-Sánchez
- Department of Cell Biology, Faculty of Science, University of Extremadura, 06071 Badajoz, Spain
| |
Collapse
|
33
|
Battisti AC, Fantetti KN, Moyers BA, Fekete DM. A subset of chicken statoacoustic ganglion neurites are repelled by Slit1 and Slit2. Hear Res 2014; 310:1-12. [PMID: 24456709 PMCID: PMC3979322 DOI: 10.1016/j.heares.2014.01.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 12/20/2013] [Accepted: 01/09/2014] [Indexed: 01/23/2023]
Abstract
Mechanosensory hair cells in the chicken inner ear are innervated by bipolar afferent neurons of the statoacoustic ganglion (SAG). During development, individual SAG neurons project their peripheral process to only one of eight distinct sensory organs. These neuronal subtypes may respond differently to guidance cues as they explore the periphery in search of their target. Previous gene expression data suggested that Slit repellants might channel SAG neurites into the sensory primordia, based on the presence of robo transcripts in the neurons and the confinement of slit transcripts to the flanks of the prosensory domains. This led to the prediction that excess Slit proteins would impede the outgrowth of SAG neurites. As predicted, axonal projections to the primordium of the anterior crista were reduced 2-3 days after electroporation of either slit1 or slit2 expression plasmids into the anterior pole of the otocyst on embryonic day 3 (E3). The posterior crista afferents, which normally grow through and adjacent to slit expression domains as they are navigating towards the posterior pole of the otocyst, did not show Slit responsiveness when similarly challenged by ectopic delivery of slit to their targets. The sensitivity to ectopic Slits shown by the anterior crista afferents was more the exception than the rule: responsiveness to Slits was not observed when the entire E4 SAG was challenged with Slits for 40 h in vitro. The corona of neurites emanating from SAG explants was unaffected by the presence of purified human Slit1 and Slit2 in the culture medium. Reduced axon outgrowth from E8 olfactory bulbs cultured under similar conditions for 24 h confirmed bioactivity of purified human Slits on chicken neurons. In summary, differential sensitivity to Slit repellents may influence the directional outgrowth of otic axons toward either the anterior or posterior otocyst.
Collapse
Affiliation(s)
- Andrea C Battisti
- Department of Biological Sciences and Purdue University Center for Cancer Research, Purdue University, 915 W State St., West Lafayette, IN 47907-1392, USA.
| | - Kristen N Fantetti
- Department of Biological Sciences and Purdue University Center for Cancer Research, Purdue University, 915 W State St., West Lafayette, IN 47907-1392, USA.
| | - Belle A Moyers
- Department of Biological Sciences and Purdue University Center for Cancer Research, Purdue University, 915 W State St., West Lafayette, IN 47907-1392, USA.
| | - Donna M Fekete
- Department of Biological Sciences and Purdue University Center for Cancer Research, Purdue University, 915 W State St., West Lafayette, IN 47907-1392, USA.
| |
Collapse
|
34
|
Wittmann W, Iulianella A, Gunhaga L. Cux2 acts as a critical regulator for neurogenesis in the olfactory epithelium of vertebrates. Dev Biol 2014; 388:35-47. [PMID: 24512687 DOI: 10.1016/j.ydbio.2014.01.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 01/30/2014] [Accepted: 01/31/2014] [Indexed: 01/17/2023]
Abstract
Signaling pathways and transcription factors are crucial regulators of vertebrate neurogenesis, exerting their function in a spatial and temporal manner. Despite recent advances in our understanding of the molecular regulation of embryonic neurogenesis, little is known regarding how different signaling pathways interact to tightly regulate this process during the development of neuroepithelia. To address this, we have investigated the events lying upstream and downstream of a key neurogenic factor, the Cut-like homeodomain transcription factor-2 (Cux2), during embryonic neurogenesis in chick and mouse. By using the olfactory epithelium as a model for neurogenesis we have analyzed mouse embryos deficient in Cux2, as well as chick embryos exposed to Cux2 silencing (si) RNA or a Cux2 over-expression construct. We provide evidence that enhanced BMP activity increases Cux2 expression and suppresses olfactory neurogenesis in the chick olfactory epithelium. In addition, our results show that up-regulation of Cux2, either BMP-induced or ectopically over-expressed, reduce Delta1 expression and suppress proliferation. Interestingly, the loss of Cux2 activity, using mutant mice or siRNA in chick, also diminishes neurogenesis, Notch activity and cell proliferation in the olfactory epithelium. Our results suggest that controlled low levels of Cux2 activity are necessary for proper Notch signaling, maintenance of the proliferative pool and ongoing neurogenesis in the olfactory epithelium. Thus, we demonstrate a novel conserved mechanism in vertebrates in which levels of Cux2 activity play an important role for ongoing neurogenesis in the olfactory epithelium.
Collapse
Affiliation(s)
- Walter Wittmann
- Umeå Centre for Molecular Medicine, Umeå University, Building 6M 4th floor, 901 87 Umeå, Sweden.
| | - Angelo Iulianella
- Department of Medical Neuroscience, Dalhousie University, Halifax, Canada.
| | - Lena Gunhaga
- Umeå Centre for Molecular Medicine, Umeå University, Building 6M 4th floor, 901 87 Umeå, Sweden.
| |
Collapse
|
35
|
Sensational placodes: neurogenesis in the otic and olfactory systems. Dev Biol 2014; 389:50-67. [PMID: 24508480 PMCID: PMC3988839 DOI: 10.1016/j.ydbio.2014.01.023] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 01/27/2014] [Accepted: 01/28/2014] [Indexed: 11/22/2022]
Abstract
For both the intricate morphogenetic layout of the sensory cells in the ear and the elegantly radial arrangement of the sensory neurons in the nose, numerous signaling molecules and genetic determinants are required in concert to generate these specialized neuronal populations that help connect us to our environment. In this review, we outline many of the proteins and pathways that play essential roles in the differentiation of otic and olfactory neurons and their integration into their non-neuronal support structures. In both cases, well-known signaling pathways together with region-specific factors transform thickened ectodermal placodes into complex sense organs containing numerous, diverse neuronal subtypes. Olfactory and otic placodes, in combination with migratory neural crest stem cells, generate highly specialized subtypes of neuronal cells that sense sound, position and movement in space, odors and pheromones throughout our lives.
Collapse
|
36
|
Patthey C, Schlosser G, Shimeld SM. The evolutionary history of vertebrate cranial placodes--I: cell type evolution. Dev Biol 2014; 389:82-97. [PMID: 24495912 DOI: 10.1016/j.ydbio.2014.01.017] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 01/21/2014] [Accepted: 01/24/2014] [Indexed: 10/25/2022]
Abstract
Vertebrate cranial placodes are crucial contributors to the vertebrate cranial sensory apparatus. Their evolutionary origin has attracted much attention from evolutionary and developmental biologists, yielding speculation and hypotheses concerning their putative homologues in other lineages and the developmental and genetic innovations that might have underlain their origin and diversification. In this article we first briefly review our current understanding of placode development and the cell types and structures they form. We next summarise previous hypotheses of placode evolution, discussing their strengths and caveats, before considering the evolutionary history of the various cell types that develop from placodes. In an accompanying review, we also further consider the evolution of ectodermal patterning. Drawing on data from vertebrates, tunicates, amphioxus, other bilaterians and cnidarians, we build these strands into a scenario of placode evolutionary history and of the genes, cells and developmental processes that underlie placode evolution and development.
Collapse
Affiliation(s)
- Cedric Patthey
- Department of Zoology, University of Oxford, South Parks Road, Oxford OX1 3PS, UK.
| | - Gerhard Schlosser
- Zoology, School of Natural Sciences & Regenerative Medicine Institute (REMEDI), National University of Ireland, Galway, University Road, Galway, Ireland
| | - Sebastian M Shimeld
- Department of Zoology, University of Oxford, South Parks Road, Oxford OX1 3PS, UK
| |
Collapse
|
37
|
Allen-Sharpley MR, Tjia M, Cramer KS. Differential roles for EphA and EphB signaling in segregation and patterning of central vestibulocochlear nerve projections. PLoS One 2013; 8:e78658. [PMID: 24130906 PMCID: PMC3795076 DOI: 10.1371/journal.pone.0078658] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 09/13/2013] [Indexed: 11/18/2022] Open
Abstract
Auditory and vestibular afferents enter the brainstem through the VIIIth cranial nerve and find targets in distinct brain regions. We previously reported that the axon guidance molecules EphA4 and EphB2 have largely complementary expression patterns in the developing avian VIIIth nerve. Here, we tested whether inhibition of Eph signaling alters central targeting of VIIIth nerve axons. We first identified the central compartments through which auditory and vestibular axons travel. We then manipulated Eph-ephrin signaling using pharmacological inhibition of Eph receptors and in ovo electroporation to misexpress EphA4 and EphB2. Anterograde labeling of auditory afferents showed that inhibition of Eph signaling did not misroute axons to non-auditory target regions. Similarly, we did not find vestibular axons within auditory projection regions. However, we found that pharmacologic inhibition of Eph receptors reduced the volume of the vestibular projection compartment. Inhibition of EphB signaling alone did not affect auditory or vestibular central projection volumes, but it significantly increased the area of the auditory sensory epithelium. Misexpression of EphA4 and EphB2 in VIIIth nerve axons resulted in a significant shift of dorsoventral spacing between the axon tracts, suggesting a cell-autonomous role for the partitioning of projection areas along this axis. Cochlear ganglion volumes did not differ among treatment groups, indicating the changes seen were not due to a gain or loss of cochlear ganglion cells. These results suggest that Eph-ephrin signaling does not specify auditory versus vestibular targets but rather contributes to formation of boundaries for patterning of inner ear projections in the hindbrain.
Collapse
Affiliation(s)
- Michelle R. Allen-Sharpley
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, California, United States of America
| | - Michelle Tjia
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, California, United States of America
| | - Karina S. Cramer
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, California, United States of America
- * E-mail:
| |
Collapse
|
38
|
Lin J, Yan X, Wang C, Talabattula VAN, Guo Z, Rolfs A, Luo J. Expression patterns of the ADAMs in early developing chicken cochlea. Dev Growth Differ 2013; 55:368-76. [PMID: 23496030 DOI: 10.1111/dgd.12051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 01/29/2013] [Accepted: 02/04/2013] [Indexed: 12/30/2022]
Abstract
Members of the ADAM (a disintegrin and metalloprotease) family are type I transmembrane proteins involved in biological processes of proteolysis, cell adhesion, cell-matrix interaction, as well as in the intracellular signaling transduction. In the present study, expression patterns of seven members of the ADAM family were investigated at the early stages of the developing cochlea by in situ hybridization. The results show that each individual ADAM is expressed and regulated in the early developing cochlea. ADAM9, ADAM10, ADAM17, and ADAM23 are initially and widely expressed in the otic vesicle at embryonic day 2.5 (E2.5) and in the differential elements of the cochlear duct at E9, while ADAM12 is expressed in acoustic ganglion cells at E7. ADAM22 is detectable in cochlear ganglion cells as early as from E4 and in the basilar papilla from E7. Therefore, the present study extends our previous results and suggests that ADAMs also play a role in the early cochlear development.
Collapse
Affiliation(s)
- Juntang Lin
- Key Laboratory for Medical Tissue Regeneration of Henan Province, Xinxiang Medical University, Xinxiang City, 453003, Henan, China
| | | | | | | | | | | | | |
Collapse
|
39
|
Neves J, Vachkov I, Giraldez F. Sox2 regulation of hair cell development: incoherence makes sense. Hear Res 2013; 297:20-9. [DOI: 10.1016/j.heares.2012.11.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 10/17/2012] [Accepted: 11/05/2012] [Indexed: 01/09/2023]
|
40
|
Kim HJ, Woo HM, Ryu J, Bok J, Kim JW, Choi SB, Park MH, Park HY, Koo SK. Conditional deletion of pten leads to defects in nerve innervation and neuronal survival in inner ear development. PLoS One 2013; 8:e55609. [PMID: 23393595 PMCID: PMC3564925 DOI: 10.1371/journal.pone.0055609] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 01/02/2013] [Indexed: 12/25/2022] Open
Abstract
All cellular phenomena and developmental events, including inner ear development, are modulated through harmonized signaling networks. Phosphatase and tensin homolog deleted on chromosome 10 (PTEN), a tumor suppressor, is a major signaling component involved in cross talk with key regulators of development; i.e., Wnt, Notch, and bone morphogenetic proteins. Although Pten function has been studied in various systems, its role in inner ear development is poorly understood. Here, we used inner ear-specific Pten conditional knockout mice and examined the characteristics of the inner ear. In a detailed analysis of the phenotype, reduced cochlear turning and widened epithelia were observed. Phalloidin staining of sensory epithelium revealed that hair cell patterns were disturbed; i.e., additional rows of hair cells were discovered. The neural abnormality revealed a reduction in and disorganization of nerve fibers, including apoptosis at the neural precursor stage. Pten deficiency induced increased phosphorylation of Akt at Ser473. The elevation of inhibitory glycogen synthase kinase 3β Ser9 phosphorylation (pGSK3β) was sustained until the neuronal differentiation stage at embryonic day 14.5, instead of pGSK3β downregulation. This is the first report on the influence of Pten/Akt/GSK3β signaling on the development of spiral ganglia. These results suggest that Pten is required for the maintenance of neuroblast number, neural precursors, and differentiation in the inner ear.
Collapse
Affiliation(s)
- Hyung Jin Kim
- Center for Biomedical Sciences, National Institute of Health, Osong Health Technology Administration Complex 643, Yeonje-ri, Osong-eup, Cheongwon-gun, Chungcheongbuk-do, South Korea
| | - Hae-Mi Woo
- Center for Biomedical Sciences, National Institute of Health, Osong Health Technology Administration Complex 643, Yeonje-ri, Osong-eup, Cheongwon-gun, Chungcheongbuk-do, South Korea
| | - Jihee Ryu
- Center for Biomedical Sciences, National Institute of Health, Osong Health Technology Administration Complex 643, Yeonje-ri, Osong-eup, Cheongwon-gun, Chungcheongbuk-do, South Korea
| | - Jinwoong Bok
- Department of Anatomy, BK21 Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Jin Woo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, South Korea
| | - Sang Back Choi
- Center for Biomedical Sciences, National Institute of Health, Osong Health Technology Administration Complex 643, Yeonje-ri, Osong-eup, Cheongwon-gun, Chungcheongbuk-do, South Korea
| | - Mi-Hyun Park
- Center for Biomedical Sciences, National Institute of Health, Osong Health Technology Administration Complex 643, Yeonje-ri, Osong-eup, Cheongwon-gun, Chungcheongbuk-do, South Korea
| | - Hyun-Young Park
- Center for Biomedical Sciences, National Institute of Health, Osong Health Technology Administration Complex 643, Yeonje-ri, Osong-eup, Cheongwon-gun, Chungcheongbuk-do, South Korea
| | - Soo Kyung Koo
- Center for Biomedical Sciences, National Institute of Health, Osong Health Technology Administration Complex 643, Yeonje-ri, Osong-eup, Cheongwon-gun, Chungcheongbuk-do, South Korea
- * E-mail:
| |
Collapse
|
41
|
Neves J, Abelló G, Petrovic J, Giraldez F. Patterning and cell fate in the inner ear: a case for Notch in the chicken embryo. Dev Growth Differ 2012; 55:96-112. [PMID: 23252974 DOI: 10.1111/dgd.12016] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 10/09/2012] [Accepted: 10/09/2012] [Indexed: 01/08/2023]
Abstract
The development of the inner ear provides a beautiful example of one basic problem in development, that is, to understand how different cell types are generated at specific times and domains throughout embryonic life. The functional unit of the inner ear consists of hair cells, supporting cells and neurons, all deriving from progenitor cells located in the neurosensory competent domain of the otic placode. Throughout development, the otic placode resolves into the complex inner ear labyrinth, which holds the auditory and vestibular sensory organs that are innervated in a highly specific manner. How does the early competent domain of the otic placode give rise to the diverse specialized cell types of the different sensory organs of the inner ear? We review here our current understanding on the role of Notch signaling in coupling patterning and cell fate determination during inner ear development, with a particular emphasis on contributions from the chicken embryo as a model organism. We discuss further the question of how these two processes rely on two modes of operation of the Notch signaling pathway named lateral induction and lateral inhibition.
Collapse
Affiliation(s)
- Joana Neves
- CEXS, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona, Barcelona, Spain
| | | | | | | |
Collapse
|
42
|
Vemaraju S, Kantarci H, Padanad MS, Riley BB. A spatial and temporal gradient of Fgf differentially regulates distinct stages of neural development in the zebrafish inner ear. PLoS Genet 2012; 8:e1003068. [PMID: 23166517 PMCID: PMC3499369 DOI: 10.1371/journal.pgen.1003068] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 09/21/2012] [Indexed: 01/13/2023] Open
Abstract
Neuroblasts of the statoacoustic ganglion (SAG) initially form in the floor of the otic vesicle during a relatively brief developmental window. They soon delaminate and undergo a protracted phase of proliferation and migration (transit-amplification). Neuroblasts eventually differentiate and extend processes bi-directionally to synapse with hair cells in the inner ear and various targets in the hindbrain. Our studies in zebrafish have shown that Fgf signaling controls multiple phases of this complex developmental process. Moderate levels of Fgf in a gradient emanating from the nascent utricular macula specify SAG neuroblasts in laterally adjacent otic epithelium. At a later stage, differentiating SAG neurons express Fgf5, which serves two functions: First, as SAG neurons accumulate, increasing levels of Fgf exceed an upper threshold that terminates the initial phase of neuroblast specification. Second, elevated Fgf delays differentiation of transit-amplifying cells, balancing the rate of progenitor renewal with neuronal differentiation. Laser-ablation of mature SAG neurons abolishes feedback-inhibition and causes precocious neuronal differentiation. Similar effects are obtained by Fgf5-knockdown or global impairment of Fgf signaling, whereas Fgf misexpression has the opposite effect. Thus Fgf signaling renders SAG development self-regulating, ensuring steady production of an appropriate number of neurons as the larva grows. Neurons of the statoacoustic ganglion (SAG), which innervate the inner ear, are derived from neuroblasts originating from the floor of the otic vesicle. Neuroblasts quickly delaminate from the otic vesicle to form dividing progenitors, which eventually differentiate into mature neurons of the SAG. Fgf has been implicated in initial neuroblast specification in multiple vertebrate species. However, the role of Fgf at later stages remains uncertain, because previous studies have not been able to evaluate the effects of changing levels of Fgf, nor have they been able to clearly distinguish the effects of Fgf at different stages of SAG development. We have combined conditional loss of function, misexpression, and laser-ablation studies in zebrafish to elucidate how graded Fgf coordinates distinct steps in SAG development. Initially moderate Fgf in a spatial gradient specifies neuroblasts within the otic vesicle. Later, mature SAG neurons express Fgf5 and, as additional neurons accumulate outside the otic vesicle, rising levels of Fgf terminate further specification. Elevated Fgf also slows maturation of progenitors, maintaining a stable progenitor pool in which growth and differentiation are evenly balanced. This feedback facilitates steady production of new neurons as the animal grows through larval and adults stages.
Collapse
Affiliation(s)
- Shruti Vemaraju
- Biology Department, Texas A&M University, College Station, Texas, United States of America
| | | | | | | |
Collapse
|
43
|
Duncan JS, Fritzsch B. Evolution of Sound and Balance Perception: Innovations that Aggregate Single Hair Cells into the Ear and Transform a Gravistatic Sensor into the Organ of Corti. Anat Rec (Hoboken) 2012; 295:1760-74. [DOI: 10.1002/ar.22573] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 07/24/2012] [Indexed: 01/20/2023]
|
44
|
Magariños M, Contreras J, Aburto MR, Varela-Nieto I. Early development of the vertebrate inner ear. Anat Rec (Hoboken) 2012; 295:1775-90. [PMID: 23044927 DOI: 10.1002/ar.22575] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 07/24/2012] [Indexed: 12/12/2022]
Abstract
This is a review of the biological processes and the main signaling pathways required to generate the different otic cell types, with particular emphasis on the actions of insulin-like growth factor I. The sensory organs responsible of hearing and balance have a common embryonic origin in the otic placode. Lineages of neural, sensory, and support cells are generated from common otic neuroepithelial progenitors. The sequential generation of the cell types that will form the adult inner ear requires the coordination of cell proliferation with cell differentiation programs, the strict regulation of cell survival, and the metabolic homeostasis of otic precursors. A network of intracellular signals operates to coordinate the transcriptional response to the extracellular input. Understanding the molecular clues that direct otic development is fundamental for the design of novel treatments for the protection and repair of hearing loss and balance disorders.
Collapse
Affiliation(s)
- Marta Magariños
- Instituto de Investigaciones Biomédicas, Alberto Sols, CSIC-UAM, Madrid, Spain
| | | | | | | |
Collapse
|
45
|
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
|
46
|
Duncan JS, Fritzsch B. Transforming the vestibular system one molecule at a time: the molecular and developmental basis of vertebrate auditory evolution. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 739:173-86. [PMID: 22399402 DOI: 10.1007/978-1-4614-1704-0_11] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
Abstract
We review the molecular basis of auditory development and evolution. We propose that the auditory periphery (basilar papilla, organ of Corti) evolved by transforming a newly created and redundant vestibular (gravistatic) endorgan into a sensory epithelium that could respond to sound instead of gravity. Evolution altered this new epithelia's mechanoreceptive properties through changes of hair cells, positioned the epithelium in a unique position near perilymphatic space to extract sound moving between the round and the oval window, and transformed its otolith covering into a tympanic membrane. Another important step in the evolution of an auditory system was the evolution of a unique set of "auditory neurons" that apparently evolved from vestibular neurons. Evolution of mammalian auditory (spiral ganglion) neurons coincides with GATA3 being a transcription factor found selectively in the auditory afferents. For the auditory information to be processed, the CNS required a dedicated center for auditory processing, the auditory nuclei. It is not known whether the auditory nucleus is ontogenetically related to the vestibular or electroreceptive nuclei, two sensory systems found in aquatic but not in amniotic vertebrates, or a de-novo formation of the rhombic lip in line with other novel hindbrain structures such as pontine nuclei. Like other novel hindbrain structures, the auditory nuclei express exclusively the bHLH gene Atoh1, and loss of Atoh1 results in loss of most of this nucleus in mice. Only after the basilar papilla, organ of Corti evolved could efferent neurons begin to modulate their activity. These auditory efferents most likely evolved from vestibular efferent neurons already present. The most simplistic interpretation of available data suggest that the ear, sensory neurons, auditory nucleus, and efferent neurons have been transformed by altering the developmental genetic modules necessary for their development into a novel direction conducive for sound extraction, conduction, and processing.
Collapse
Affiliation(s)
- Jeremy S Duncan
- Department of Biology, University of Iowa, Iowa City, Iowa, USA.
| | | |
Collapse
|
47
|
Sabado V, Barraud P, Baker CVH, Streit A. Specification of GnRH-1 neurons by antagonistic FGF and retinoic acid signaling. Dev Biol 2012; 362:254-62. [PMID: 22200593 PMCID: PMC4561506 DOI: 10.1016/j.ydbio.2011.12.016] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Revised: 12/08/2011] [Accepted: 12/09/2011] [Indexed: 11/25/2022]
Abstract
A small population of neuroendocrine cells in the rostral hypothalamus and basal forebrain is the key regulator of vertebrate reproduction. They secrete gonadotropin-releasing hormone (GnRH-1), communicate with many areas of the brain and integrate multiple inputs to control gonad maturation, puberty and sexual behavior. In humans, disruption of the GnRH-1 system leads to hypogonadotropic gonadism and Kallmann syndrome. Unlike other neurons in the central nervous system, GnRH-1 neurons arise in the periphery, however their embryonic origin is controversial, and the molecular mechanisms that control their initial specification are not clear. Here, we provide evidence that in chick GnRH-1 neurons originate in the olfactory placode, where they are specified shortly after olfactory sensory neurons. FGF signaling is required and sufficient to induce GnRH-1 neurons, while retinoic acid represses their formation. Both pathways regulate and antagonize each other and our results suggest that the timing of signaling is critical for normal GnRH-1 neuron formation. While Kallmann's syndrome has generally been attributed to a failure of GnRH-1 neuron migration due to impaired FGF signaling, our findings suggest that in at least some Kallmann patients these neurons may never be specified. In addition, this study highlights the intimate embryonic relationship between GnRH-1 neurons and their targets and modulators in the adult.
Collapse
Affiliation(s)
- Virginie Sabado
- Department of Craniofacial Development, King’s College London, Guy’s Campus, London, SE1 9RT, UK
| | - Perrine Barraud
- Department of Physiology, Development & Neuroscience, Anatomy Building, Downing Street, Cambridge, CB2 3DY, UK
| | - Clare V. H. Baker
- Department of Physiology, Development & Neuroscience, Anatomy Building, Downing Street, Cambridge, CB2 3DY, UK
| | - Andrea Streit
- Department of Craniofacial Development, King’s College London, Guy’s Campus, London, SE1 9RT, UK
| |
Collapse
|
48
|
Neves J, Uchikawa M, Bigas A, Giraldez F. The prosensory function of Sox2 in the chicken inner ear relies on the direct regulation of Atoh1. PLoS One 2012; 7:e30871. [PMID: 22292066 PMCID: PMC3264626 DOI: 10.1371/journal.pone.0030871] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Accepted: 12/22/2011] [Indexed: 11/18/2022] Open
Abstract
The proneural gene Atoh1 is crucial for the development of inner ear hair cells and it requires the function of the transcription factor Sox2 through yet unknown mechanisms. In the present work, we used the chicken embryo and HEK293T cells to explore the regulation of Atoh1 by Sox2. The results show that hair cells derive from Sox2-positive otic progenitors and that Sox2 directly activates Atoh1 through a transcriptional activator function that requires the integrity of Sox2 DNA binding domain. Atoh1 activation depends on Sox transcription factor binding sites (SoxTFBS) present in the Atoh1 3′ enhancer where Sox2 directly binds, as shown by site directed mutagenesis and chromatin immunoprecipitation (ChIP). In the inner ear, Atoh1 enhancer activity is detected in the neurosensory domain and it depends on Sox2. Dominant negative competition (Sox2HMG-Engrailed) and mutation of the SoxTFBS abolish the reporter activity in vivo. Moreover, ChIP assay in isolated otic vesicles shows that Sox2 is bound to the Atoh1 enhancer in vivo. However, besides activating Atoh1, Sox2 also promotes the expression of Atoh1 negative regulators and the temporal profile of Atoh1 activation by Sox2 is transient suggesting that Sox2 triggers an incoherent feed-forward loop. These results provide a mechanism for the prosensory function of Sox2 in the inner ear. We suggest that sensory competence is established early in otic development through the activation of Atoh1 by Sox2, however, hair cell differentiation is prevented until later stages by the parallel activation of negative regulators of Atoh1 function.
Collapse
Affiliation(s)
- Joana Neves
- CEXS, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona (PRBB), Barcelona, Spain
| | - Masanori Uchikawa
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Anna Bigas
- Program in Cancer Research, IMIM-Hospital del Mar, Parc de Recerca Biomèdica de Barcelona (PRBB), Barcelona, Spain
| | - Fernando Giraldez
- CEXS, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona (PRBB), Barcelona, Spain
- * E-mail:
| |
Collapse
|
49
|
Appler JM, Goodrich LV. Connecting the ear to the brain: Molecular mechanisms of auditory circuit assembly. Prog Neurobiol 2011; 93:488-508. [PMID: 21232575 PMCID: PMC3078955 DOI: 10.1016/j.pneurobio.2011.01.004] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 12/09/2010] [Accepted: 01/03/2011] [Indexed: 12/21/2022]
Abstract
Our sense of hearing depends on precisely organized circuits that allow us to sense, perceive, and respond to complex sounds in our environment, from music and language to simple warning signals. Auditory processing begins in the cochlea of the inner ear, where sounds are detected by sensory hair cells and then transmitted to the central nervous system by spiral ganglion neurons, which faithfully preserve the frequency, intensity, and timing of each stimulus. During the assembly of auditory circuits, spiral ganglion neurons establish precise connections that link hair cells in the cochlea to target neurons in the auditory brainstem, develop specific firing properties, and elaborate unusual synapses both in the periphery and in the CNS. Understanding how spiral ganglion neurons acquire these unique properties is a key goal in auditory neuroscience, as these neurons represent the sole input of auditory information to the brain. In addition, the best currently available treatment for many forms of deafness is the cochlear implant, which compensates for lost hair cell function by directly stimulating the auditory nerve. Historically, studies of the auditory system have lagged behind other sensory systems due to the small size and inaccessibility of the inner ear. With the advent of new molecular genetic tools, this gap is narrowing. Here, we summarize recent insights into the cellular and molecular cues that guide the development of spiral ganglion neurons, from their origin in the proneurosensory domain of the otic vesicle to the formation of specialized synapses that ensure rapid and reliable transmission of sound information from the ear to the brain.
Collapse
Affiliation(s)
- Jessica M Appler
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
50
|
Neves J, Parada C, Chamizo M, Giráldez F. Jagged 1 regulates the restriction of Sox2 expression in the developing chicken inner ear: a mechanism for sensory organ specification. Development 2011; 138:735-44. [PMID: 21266409 DOI: 10.1242/dev.060657] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Hair cells of the inner ear sensory organs originate from progenitor cells located at specific domains of the otic vesicle: the prosensory patches. Notch signalling is necessary for sensory development and loss of function of the Notch ligand jagged 1 (Jag1, also known as serrate 1) results in impaired sensory organs. However, the underlying mechanism of Notch function is unknown. Our results show that in the chicken otic vesicle, the Sox2 expression domain initially contains the nascent patches of Jag1 expression but, later on, Sox2 is only maintained in the Jag1-positive domains. Ectopic human JAG1 (hJag1) is able to induce Sox2 expression and enlarged sensory organs. The competence to respond to hJag1, however, is confined to the regions that expressed Sox2 early in development, suggesting that hJag1 maintains Sox2 expression rather than inducing it de novo. The effect is non-cell-autonomous and requires Notch signalling. hJag1 activates Notch, induces Hes/Hey genes and endogenous Jag1 in a non-cell-autonomous manner, which is consistent with lateral induction. The effects of hJag1 are mimicked by Jag2 but not by Dl1. Sox2 is sufficient to activate the Atoh1 enhancer and to ectopically induce sensory cell fate outside neurosensory-competent domains. We suggest that the prosensory function of Jag1 resides in its ability to generate discrete domains of Notch activity that maintain Sox2 expression within restricted areas of an extended neurosensory-competent domain. This provides a mechanism to couple patterning and cell fate specification during the development of sensory organs.
Collapse
Affiliation(s)
- Joana Neves
- CEXS, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona, C/Dr Aiguader 88, 08003-Barcelona, Spain
| | | | | | | |
Collapse
|