1
|
Wang Y, Wang H, Zhang P, Zhu B, Li W, Zhao X, Yan M, Song X, Lai F, Dong J, Cui J, Guo X, Wu HJ, Li J. Single-cell atlas comparison across vertebrates reveals auditory cell evolution and mechanisms for hair cell regeneration. Commun Biol 2024; 7:1648. [PMID: 39702452 DOI: 10.1038/s42003-024-07335-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 11/29/2024] [Indexed: 12/21/2024] Open
Abstract
Mammals suffer permanent hearing impairment from the loss of auditory hair cells due to their inability to regenerate. In contrast, lower vertebrates exhibit extraordinary capacity for hair cell regeneration and hearing restoration, but the mechanisms remain unclear. Here we characterize the single-cell atlas of Xenopus laevis inner ear and perform a comprehensive comparison with mouse model. An exceptionally conserved inner ear neuronal cell type is discovered. The results reveal that the outer hair cells (OHCs) exist exclusively in mammals. Importantly, our analyses reveal an orchestrated gene expression program in Xenopus, characterized by upregulation of hair cell regeneration-related genes, coupled with downregulation of proliferation inhibitory genes. These findings unveil a natural feature of regenerative capacity in Xenopus, and provide molecular and evolutionary evidences for differential regenerative capacities across vertebrates. This work offers insights from amphibians into developing strategies to solve the challenges of hair cell regeneration in humans.
Collapse
Affiliation(s)
- Yafan Wang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610213, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Haojie Wang
- University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Penghui Zhang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610213, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Bicheng Zhu
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610213, China
| | - Wenxiu Li
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610213, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaomeng Zhao
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610213, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mengzhen Yan
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610213, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xuemei Song
- Institute of Blood Diseases, Department of Hematology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan, Chengdu, 610072, China
| | - Futing Lai
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Jieran Dong
- College of Agronomy, Sichuan Agricultural University, Chengdu, China
| | - Jianguo Cui
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610213, China
| | - Xiang Guo
- Institute of Blood Diseases, Department of Hematology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan, Chengdu, 610072, China.
| | - Hua-Jun Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China.
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
- Center for Precision Medicine Multi-Omics Research, Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China.
| | - Jun Li
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610213, China.
| |
Collapse
|
2
|
Bardhan S, Bhargava N, Dighe S, Vats N, Naganathan SR. Emergence of a left-right symmetric body plan in vertebrate embryos. Curr Top Dev Biol 2024; 159:310-342. [PMID: 38729680 DOI: 10.1016/bs.ctdb.2024.01.003] [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: 05/12/2024]
Abstract
External bilateral symmetry is a prevalent feature in vertebrates, which emerges during early embryonic development. To begin with, vertebrate embryos are largely radially symmetric before transitioning to bilaterally symmetry, after which, morphogenesis of various bilateral tissues (e.g somites, otic vesicle, limb bud), and structures (e.g palate, jaw) ensue. While a significant amount of work has probed the mechanisms behind symmetry breaking in the left-right axis leading to asymmetric positioning of internal organs, little is known about how bilateral tissues emerge at the same time with the same shape and size and at the same position on the two sides of the embryo. By discussing emergence of symmetry in many bilateral tissues and structures across vertebrate model systems, we highlight that understanding symmetry establishment is largely an open field, which will provide deep insights into fundamental problems in developmental biology for decades to come.
Collapse
Affiliation(s)
- Siddhartha Bardhan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Nandini Bhargava
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Swarali Dighe
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Neha Vats
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Sundar Ram Naganathan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India.
| |
Collapse
|
3
|
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
|
4
|
Forrester-Gauntlett B, Peters L, Oback B. Grainyhead-like 2 is required for morphological integrity of mouse embryonic stem cells and orderly formation of inner ear-like organoids. Front Cell Dev Biol 2023; 11:1112069. [PMID: 37745294 PMCID: PMC10513505 DOI: 10.3389/fcell.2023.1112069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Mutations in the transcription factor gene grainyhead-like 2 (GRHL2) are associated with progressive non-syndromic sensorineural deafness autosomal dominant type 28 (DFNA28) in humans. Since complete loss of Grhl2 is lethal in mouse embryos, we studied its role during inner ear pathology and hearing loss in vitro. To this end, we generated different homozygous deletions to knockout Grhl2 in mouse embryonic stem cells (Grhl2-KO ESCs), including some mimicking naturally occurring truncations in the dimerisation domain related to human DFNA28. Under naïve culture conditions, Grhl2-KO cells in suspension were more heterogenous in size and larger than wild-type controls. Adherent Grhl2-KO cells were also larger, with a less uniform shape, flattened, less circular morphology, forming loose monolayer colonies with poorly defined edges. These changes correlated with lower expression of epithelial cadherin Cdh1 but no changes in tight junction markers (Ocln, Tjp2) or other Grhl isoforms (Grhl1, Grhl3). Clonogenicity from single cells, proliferation rates of cell populations and proliferation markers were reduced in Grhl2-KO ESCs. We next induced stepwise directed differentiation of Grhl2-KO ESCs along an otic pathway, giving rise to three-dimensional inner ear-like organoids (IELOs). Quantitative morphometry revealed that Grhl2-KO cells initially formed larger IELOs with a less compacted structure, more eccentric shape and increased surface area. These morphological changes persisted for up to one week. They were partially rescued by forced cell aggregation and fully restored by stably overexpressing exogenous Grhl2 in Grhl2-KO ESCs, indicating that Grhl2 alters cell-cell interactions. On day 8, aggregates were transferred into minimal maturation medium to allow self-guided organogenesis for another two weeks. During this period, Grhl2-KO cells and wild-type controls developed similarly, expressing neural, neuronal and sensory hair cell markers, while maintaining their initial differences in size and shape. In summary, Grhl2 is required for morphological maintenance of ESCs and orderly formation of IELOs, consistent with an essential role in organising epithelial integrity during inner ear development. Our findings validate quantitative morphometry as a useful, non-invasive screening method for molecular phenotyping of candidate mutations during organoid development.
Collapse
Affiliation(s)
- Blaise Forrester-Gauntlett
- Animal Biotech, AgResearch, Hamilton, New Zealand
- School of Science, University of Waikato, Hamilton, New Zealand
| | - Linda Peters
- School of Science, University of Waikato, Hamilton, New Zealand
| | - Björn Oback
- Animal Biotech, AgResearch, Hamilton, New Zealand
- School of Science, University of Waikato, Hamilton, New Zealand
- School of Medical Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
5
|
Sarria-S FA, Montealegre-Z F, Gonzalez-Rodriguez J. The chemistry of an insect ear: ionic composition of a liquid-filled ear and haemolymphs of Neotropical katydids. J R Soc Interface 2023; 20:20230154. [PMID: 37464801 PMCID: PMC10354488 DOI: 10.1098/rsif.2023.0154] [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: 03/18/2023] [Accepted: 06/28/2023] [Indexed: 07/20/2023] Open
Abstract
The purpose of this study is to examine and to compare the ionic composition of the haemolymph and the ear fluid of seven species of katydids (Orthoptera: Tettigoniidae) with the aim of providing from a biochemical perspective a preliminary assessment for an insect liquid contained in the auditory organ of katydids with a hearing mechanism reminiscent of that found in vertebrates. A multi-element trace analysis by inductively coupled plasma optical-emission spectrometry was run for 16 elements for the ear liquid of seven species and the haemolymph of six of them. Based on the obtained results, it can be recognized that the ionic composition is variable among the studied insects, but sodium (Na+), potassium (K+), calcium (Ca2+) and magnesium (Mg2+) are the most prominent of the dissolved inorganic cations. However, the ion concentrations between the two fluids are considerably different and the absence or low concentration of Ca2+ is a noticeable feature in the inner ear liquid. A potential relationship between the male courtship song peak frequency and the total ion (Na+, K+, Mg2+ and Ca2+) concentration of the inner ear liquid is also reported.
Collapse
Affiliation(s)
- Fabio A. Sarria-S
- School of Life Sciences, Joseph Banks Laboratories, University of Lincoln, Lincoln LN6 7DL, UK
| | - Fernando Montealegre-Z
- School of Life Sciences, Joseph Banks Laboratories, University of Lincoln, Lincoln LN6 7DL, UK
| | | |
Collapse
|
6
|
Shen J, Xia X, Sun L, Ma X, Huang B, Hanif Q, Chen N, Qu K, Zhang J, Chen H, Jiang Y, Lei C. Genome-wide association study reveals that the IBSP locus affects ear size in cattle. Heredity (Edinb) 2023; 130:394-401. [PMID: 37016135 PMCID: PMC10238394 DOI: 10.1038/s41437-023-00614-9] [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: 02/17/2022] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 04/06/2023] Open
Abstract
Ear size is a classical model for hot climate adaptation following the evolution, but the genetic basis of the traits associated with ear size remains to be elucidated. Here, we performed a genome-wide association study on 158 cattle to explain the genetic mechanism of ear size. One region on BTA6 between 36.79 and 38.80 Mb included 50 suggestive SNPs and 4 significant SNPs that were significantly associated with ear size. The most significant locus (P = 1.30 × 10-8) was a missense mutation (T250I) on the seventh exon of integrin-binding sialoprotein (IBSP), which had an allele substitution effect of 23.46 cm2 for ear size. Furthermore, this mutation will cause changes in the three-dimensional structure of the protein. To further identify genes underlying this typical feature, we performed a genome scan among nine cattle breeds with different ear sizes by using SweeD. Results suggested that IBSP was under positive selection among four breeds with relatively large ear sizes. The expression levels of IBSP in ear tissues of large- and small-ear cattle were significantly different. A haplotype diversity survey of this missense mutation in worldwide cattle breeds strongly implied that the origin of this missense mutation event was Bos taurus. These findings have important theoretical importance for the exploration of major genes associated with ear size and provide important molecular markers for the identification of cattle germplasm resources.
Collapse
Affiliation(s)
- Jiafei Shen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, N1 Shangcheng Road, Yiwu, Zhejiang, 322000, China
| | - Xiaoting Xia
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Luyang Sun
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xiaohui Ma
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Bizhi Huang
- Yunnan Academy of Grassland and Animal Science, Kunming, Yunnan, 650212, China
| | - Quratulain Hanif
- Computational Biology Laboratory, Agricultural Biotechnology Division, National Institute for Biotechnology and Genetic Engineering, Faisalabad, 577, Pakistan
| | - Ningbo Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Kaixing Qu
- Academy of Science and Technology, Chuxiong Normal University, Chuxiong, Yunnan, 675000, China
| | - Jicai Zhang
- Yunnan Academy of Grassland and Animal Science, Kunming, Yunnan, 650212, China
| | - Hong Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yu Jiang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Chuzhao Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
7
|
Baeza-Loya S, Raible DW. Vestibular physiology and function in zebrafish. Front Cell Dev Biol 2023; 11:1172933. [PMID: 37143895 PMCID: PMC10151581 DOI: 10.3389/fcell.2023.1172933] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/06/2023] [Indexed: 05/06/2023] Open
Abstract
The vestibular system of the inner ear provides information about head motion and spatial orientation relative to gravity to ensure gaze stability, balance, and postural control. Zebrafish, like humans, have five sensory patches per ear that serve as peripheral vestibular organs, with the addition of the lagena and macula neglecta. The zebrafish inner ear can be easily studied due to its accessible location, the transparent tissue of larval fish, and the early development of vestibular behaviors. Thus, zebrafish are an excellent model for studying the development, physiology, and function of the vestibular system. Recent work has made great strides to elucidate vestibular neural circuitry in fish, tracing sensory transmission from receptors in the periphery to central computational circuits driving vestibular reflexes. Here we highlight recent work that illuminates the functional organization of vestibular sensory epithelia, innervating first-order afferent neurons, and second-order neuronal targets in the hindbrain. Using a combination of genetic, anatomical, electrophysiological, and optical techniques, these studies have probed the roles of vestibular sensory signals in fish gaze, postural, and swimming behaviors. We discuss remaining questions in vestibular development and organization that are tractable in the zebrafish model.
Collapse
Affiliation(s)
| | - David W. Raible
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology-HNS and Biological Structure, University of Washington, Seattle, WA, United States
| |
Collapse
|
8
|
Song H, Morrow BE. Tbx2 and Tbx3 regulate cell fate progression of the otic vesicle for inner ear development. Dev Biol 2023; 494:71-84. [PMID: 36521641 PMCID: PMC9870991 DOI: 10.1016/j.ydbio.2022.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/05/2022] [Accepted: 12/10/2022] [Indexed: 12/14/2022]
Abstract
The morphogenesis of the otic vesicle (OV) to form inner ear organs serves as an excellent model system to understand cell fate acquisition on a single cell level. Tbx2 and Tbx3 (Tbx2/3) encode closely related T-box transcription factors that are expressed widely in the mammalian OV. Inactivation of both genes in the OV (Tbx2/3cKO) results in failed morphogenesis into inner ear organs. To understand the basis of these defects, single cell RNA-sequencing (scRNA-seq) was performed on the OV lineage, in controls versus Tbx2/3cKO embryos. We identified a multipotent population termed otic progenitors in controls that are marked by expression of the known otic placode markers Eya1, Sox2, and Sox3 as well as new markers Fgf18, Cxcl12, and Pou3f3. The otic progenitor population was increased three-fold in Tbx2/3cKO embryos, concomitant with dysregulation of genes in these cells as well as reduced progression to more differentiated states of prosensory and nonsensory cells. An ectopic neural population of cells was detected in the posterior OV of Tbx2/3cKO embryos but had reduced maturation to delaminated neural cells. As all three cell fates were affected in Tbx2/3cKO embryos, we suggest that Tbx2/3 promotes progression of multipotent otic progenitors to more differentiated cell types in the OV.
Collapse
Affiliation(s)
- Hansoo Song
- Department of Genetics, Albert Einstein College of Medicine, 1301 Morris Park Ave., Bronx, NY, USA
| | - Bernice E Morrow
- Department of Genetics, Albert Einstein College of Medicine, 1301 Morris Park Ave., Bronx, NY, USA.
| |
Collapse
|
9
|
Nolte T, Baumgärtner W, Colbatzky F, Knippel A, Marxfeld H, Nehrbass D, Odin M, Popp A, Treumann S, Yen HY, Zellmer J, Deschl U. Proceedings of the 2020 Classic Examples in Toxicologic Pathology XXVII. Toxicol Pathol 2021; 49:1206-1228. [PMID: 34259102 DOI: 10.1177/01926233211019288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The histopathology slide seminar "Classic Examples in Toxicologic Pathology XXVII" was held on February 21 and 22, 2020, at the Department of Pathology at the University of Veterinary Medicine in Hannover, Germany, with joint organization by the European Society of Toxicologic Pathology. The goal of this annual seminar is to present and discuss classical and actual cases of toxicologic pathology. This article summarizes the presentations given during the seminar, including images of representative lesions. Ten actual and classical cases of toxicologic pathology, mostly induced by a test article, were presented. These included small intestine pathology and transcriptomics induced by a γ-secretase modulator, liver findings in nonhuman primates induced by gene therapy, drug-induced neutropenia in dogs, device-induced growth plate lesions, polycystic lesions in CAR/PXR double knockout mice, inner ear lesions in transgenic mice, findings in Beagle dogs induced by an inhibitor of the myeloid leukemia cell differentiation protein MCL1, findings induced by a monovalent fibroblast growth factor receptor 1 antagonist, kidney lesions induced by a mammalian target of rapamycin inhibitor in combination therapy, and findings in mutation-specific drugs.
Collapse
Affiliation(s)
- Thomas Nolte
- 84647Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach (Riss), Germany
| | - Wolfgang Baumgärtner
- Institut für Pathologie, Stiftung 26556Tierärztliche Hochschule Hannover, Germany
| | - Florian Colbatzky
- 84647Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach (Riss), Germany
| | | | | | - Dirk Nehrbass
- 161930AO Research Institute Davos (ARI), Davos, Switzerland
| | - Marielle Odin
- 123188Roche Innovation Center Basel, Pharma Research & Early Development, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Andreas Popp
- 385330Abbvie GmbH & Co. KG, Ludwigshafen, Germany
| | | | - Hsi-Yu Yen
- 9184Technical University, Munich, Germany
| | | | - Ulrich Deschl
- 84647Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach (Riss), Germany
| |
Collapse
|
10
|
Recommendations for Measuring the Electrically Evoked Compound Action Potential in Children With Cochlear Nerve Deficiency. Ear Hear 2021; 41:465-475. [PMID: 31567301 DOI: 10.1097/aud.0000000000000782] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES This study reports a method for measuring the electrically evoked compound action potential (eCAP) in children with cochlear nerve deficiency (CND). DESIGN This method was developed based on experience with 50 children with CND who were Cochlear Nucleus cochlear implant users. RESULTS This method includes three recommended steps conducted with recommended stimulating and recording parameters: initial screen, pulse phase duration optimization, and eCAP threshold determination (i.e., identifying the lowest stimulation level that can evoke an eCAP). Compared with the manufacturer-default parameters, the recommended parameters used in this method yielded a higher success rate for measuring the eCAP in children with CND. CONCLUSIONS The eCAP can be measured successfully in children with CND using recommended parameters. This specific method is suitable for measuring the eCAP in children with CND in clinical settings. However, it is not suitable for intraoperative eCAP recordings due to the extensive testing time required.
Collapse
|
11
|
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
|
12
|
Stojkovic M, Han D, Jeong M, Stojkovic P, Stankovic KM. Human induced pluripotent stem cells and CRISPR/Cas-mediated targeted genome editing: Platforms to tackle sensorineural hearing loss. STEM CELLS (DAYTON, OHIO) 2021; 39:673-696. [PMID: 33586253 DOI: 10.1002/stem.3353] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 12/13/2020] [Indexed: 11/09/2022]
Abstract
Hearing loss (HL) is a major global health problem of pandemic proportions. The most common type of HL is sensorineural hearing loss (SNHL) which typically occurs when cells within the inner ear are damaged. Human induced pluripotent stem cells (hiPSCs) can be generated from any individual including those who suffer from different types of HL. The development of new differentiation protocols to obtain cells of the inner ear including hair cells (HCs) and spiral ganglion neurons (SGNs) promises to expedite cell-based therapy and screening of potential pharmacologic and genetic therapies using human models. Considering age-related, acoustic, ototoxic, and genetic insults which are the most frequent causes of irreversible damage of HCs and SGNs, new methods of genome editing (GE), especially the CRISPR/Cas9 technology, could bring additional opportunities to understand the pathogenesis of human SNHL and identify novel therapies. However, important challenges associated with both hiPSCs and GE need to be overcome before scientific discoveries are correctly translated to effective and patient-safe applications. The purpose of the present review is (a) to summarize the findings from published reports utilizing hiPSCs for studies of SNHL, hence complementing recent reviews focused on animal studies, and (b) to outline promising future directions for deciphering SNHL using disruptive molecular and genomic technologies.
Collapse
Affiliation(s)
- Miodrag Stojkovic
- Eaton Peabody Laboratories, Department of Otolaryngology Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Dongjun Han
- Eaton Peabody Laboratories, Department of Otolaryngology Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Minjin Jeong
- Eaton Peabody Laboratories, Department of Otolaryngology Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Petra Stojkovic
- Eaton Peabody Laboratories, Department of Otolaryngology Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Konstantina M Stankovic
- Eaton Peabody Laboratories, Department of Otolaryngology Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, USA.,Program in Speech and Hearing Bioscience and Technology, Harvard University, Cambridge, Massachusetts, USA.,Harvard Program in Therapeutic Science, Harvard Medical School, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
13
|
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
|
14
|
Hashem S, Nisar S, Bhat AA, Yadav SK, Azeem MW, Bagga P, Fakhro K, Reddy R, Frenneaux MP, Haris M. Genetics of structural and functional brain changes in autism spectrum disorder. Transl Psychiatry 2020; 10:229. [PMID: 32661244 PMCID: PMC7359361 DOI: 10.1038/s41398-020-00921-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/05/2020] [Accepted: 06/09/2020] [Indexed: 12/21/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurological and developmental disorder characterized by social impairment and restricted interactive and communicative behaviors. It may occur as an isolated disorder or in the context of other neurological, psychiatric, developmental, and genetic disorders. Due to rapid developments in genomics and imaging technologies, imaging genetics studies of ASD have evolved in the last few years. Increased risk for ASD diagnosis is found to be related to many specific single-nucleotide polymorphisms, and the study of genetic mechanisms and noninvasive imaging has opened various approaches that can help diagnose ASD at the nascent level. Identifying risk genes related to structural and functional changes in the brain of ASD patients provide a better understanding of the disease's neuropsychiatry and can help identify targets for therapeutic intervention that could be useful for the clinical management of ASD patients.
Collapse
Affiliation(s)
- Sheema Hashem
- Functional and Molecular Imaging Laboratory, Sidra Medicine, Doha, Qatar
| | - Sabah Nisar
- Functional and Molecular Imaging Laboratory, Sidra Medicine, Doha, Qatar
| | - Ajaz A Bhat
- Functional and Molecular Imaging Laboratory, Sidra Medicine, Doha, Qatar
| | | | | | - Puneet Bagga
- Center for Magnetic Resonance and Optical Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Khalid Fakhro
- Department of Human Genetics, Sidra Medicine, Doha, Qatar
- Department of Genetic Medicine, Weill Cornell Medical College, Doha, Qatar
| | - Ravinder Reddy
- Center for Magnetic Resonance and Optical Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | - Mohammad Haris
- Functional and Molecular Imaging Laboratory, Sidra Medicine, Doha, Qatar.
- Laboratory Animal Research Center, Qatar University, Doha, Qatar.
| |
Collapse
|
15
|
Pouraghaei S, Moztarzadeh F, Chen C, Ansari S, Moshaverinia A. Microenvironment Can Induce Development of Auditory Progenitor Cells from Human Gingival Mesenchymal Stem Cells. ACS Biomater Sci Eng 2020; 6:2263-2273. [PMID: 33455314 DOI: 10.1021/acsbiomaterials.9b01795] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Sensorineural hearing loss in mammals occurs due to irreversible damage to the sensory epithelia of the inner ear and has very limited treatment options. The ability to regenerate the auditory progenitor cells is a promising approach for the treatment of sensorineural hearing loss; therefore, finding an appropriate and easily accessible stem cell source for restoring the sense of hearing would be of great interest. Here, we proposed a novel easy-to-access source of cells with the ability to recover auditory progenitor cells. In this study, gingival mesenchymal stem cells (GMSCs) were utilized, as these cells have high self-renewal and multipotent differentiation capacity and can be obtained easily from the oral cavity or discarded tissue samples at dental clinics. To manipulate the biophysical properties of the cellular microenvironment for promoting GMSC differentiation toward the target cells, we also tried to propose a candidate biomaterial. GMSCs in combination with an appropriate scaffold material can, therefore, present advantageous therapeutic options for a number of conditions. Here, we report the potential of GMSCs to differentiate into auditory progenitor cells while supporting them with an optimized three-dimensional scaffold and certain growth factors. A hybrid hydrogel scaffold based on peptide modified alginate and Matrigel was used here in addition to the presence of fibroblast growth factor-basic (bFGF), insulin-like growth factor (IGF), and epidermal growth factor (EGF). Our in vitro and in vivo studies confirmed the auditory differentiation potential of GMSCs within the engineered microenvironment.
Collapse
Affiliation(s)
- Sevda Pouraghaei
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, California, United States
| | - Fathollah Moztarzadeh
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Chider Chen
- Department of Oral and Maxillofacial Surgery and Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Sahar Ansari
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Alireza Moshaverinia
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, California, United States
- California NanoSystems Institute, University of California, Los Angeles, California, United States
| |
Collapse
|
16
|
Ghanavatinejad F, Fard Tabrizi ZP, Omidghaemi S, Sharifi E, Møller SG, Jami MS. Protein biomarkers of neural system. J Otol 2019; 14:77-88. [PMID: 31467504 PMCID: PMC6712353 DOI: 10.1016/j.joto.2019.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/14/2019] [Accepted: 03/20/2019] [Indexed: 11/30/2022] Open
Abstract
The utilization of biomarkers for in vivo and in vitro research is growing rapidly. This is mainly due to the enormous potential of biomarkers in evaluating molecular and cellular abnormalities in cell models and in tissue, and evaluating drug responses and the effectiveness of therapeutic intervention strategies. An important way to analyze the development of the human body is to assess molecular markers in embryonic specialized cells, which include the ectoderm, mesoderm, and endoderm. Neuronal development is controlled through the gene networks in the neural crest and neural tube, both components of the ectoderm. The neural crest differentiates into several different tissues including, but not limited to, the peripheral nervous system, enteric nervous system, melanocyte, and the dental pulp. The neural tube eventually converts to the central nervous system. This review provides an overview of the differentiation of the ectoderm to a fully functioning nervous system, focusing on molecular biomarkers that emerge at each stage of the cellular specialization from multipotent stem cells to completely differentiated cells. Particularly, the otic placode is the origin of most of the inner ear cell types such as neurons, sensory hair cells, and supporting cells. During the development, different auditory cell types can be distinguished by the expression of the neurogenin differentiation factor1 (Neuro D1), Brn3a, and transcription factor GATA3. However, the mature auditory neurons express other markers including βIII tubulin, the vesicular glutamate transporter (VGLUT1), the tyrosine receptor kinase B and C (Trk B, C), BDNF, neurotrophin 3 (NT3), Calretinin, etc.
Collapse
Affiliation(s)
- Fatemeh Ghanavatinejad
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Science, Shahrekord, Iran
| | - Zahra Pourteymour Fard Tabrizi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Science, Shahrekord, Iran
| | - Shadi Omidghaemi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Science, Shahrekord, Iran
| | - Esmaeel Sharifi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Simon Geir Møller
- Department of Biological Sciences, St John's University, New York, NY, USA
- The Norwegian Centre for Movement Disorders, Stavanger University Hospital, Norway
| | - Mohammad-Saeid Jami
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Science, Shahrekord, Iran
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, CA, 90095, USA
| |
Collapse
|
17
|
Cerio DG, Witmer LM. Intraspecific variation and symmetry of the inner-ear labyrinth in a population of wild turkeys: implications for paleontological reconstructions. PeerJ 2019; 7:e7355. [PMID: 31372322 PMCID: PMC6659666 DOI: 10.7717/peerj.7355] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/26/2019] [Indexed: 12/25/2022] Open
Abstract
The cochlea and semicircular canals (SCCs) of the inner ear are vital neurosensory devices. There are associations between the anatomy of these sensorineural structures, their function, and the function of related biological systems, for example, hearing ability, gaze stabilization, locomotor agility, and posture. The endosseous labyrinth is frequently used as a proxy to infer the performance of the hearing and vestibular systems, locomotor abilities, and ecology of extinct species. Such fossil inferences are often based on single specimens or even a single ear, representing an entire species. To address whether a single ear is representative of a population, we used geometric morphometrics to quantitatively assess the variation in shape and symmetry in a sample of endosseous labyrinths of wild turkeys Meleagris gallopavo of southern Ohio. We predicted that ears would be symmetrical both within individuals and across the sample; that labyrinth shape and size would covary; that labyrinth shape would vary with the size of the brain, measured as width of the endocranium at the cerebellum; and that labyrinths would be morphologically integrated. To test these predictions, we microCT-scanned the heads of 26 cadaveric turkeys, digitally segmented their endosseous labyrinths in Avizo, and assigned 15 manual landmarks and 20 sliding semilandmarks to each digital model. Following Procrustes alignment, we conducted an analysis of bilateral symmetry, a Procrustes regression analysis for allometry and other covariates including side and replicate, and analyses of global integration and modularity. Based on Procrustes distances, no individual’s left and right ears were clearly different from each other. When comparing the ears of different specimens, statistically clear differences in shape were found in only 66 of more than 1,300 contrasts. Moreover, effects of both directional and fluctuating asymmetry were very small—generally, two orders of magnitude smaller than the variance explained by individual variation. Statistical tests disagreed on whether these asymmetric effects crossed the threshold of significance, possibly due to non-isotropic variation among landmarks. Regardless, labyrinths appeared to primarily vary in shape symmetrically. Neither labyrinth size nor endocranial width was correlated with labyrinth shape, contrary to our expectations. Finally, labyrinths were found to be moderately integrated in a global sense, but four weakly separated modules—the three SCCs and cochlea—were recovered using a maximum-likelihood analysis. The results show that both fluctuating and directional asymmetry play a larger role in shape variation than expected—but nonetheless, endosseous labyrinths are symmetrical within individuals and at the level of the population, and their shape varies symmetrically. Thus, inferences about populations, and very possibly species, may be confidently made when only a single specimen, or even a single ear, is available for study.
Collapse
Affiliation(s)
- Donald G Cerio
- Department of Biological Sciences, Ohio University, Athens, OH, USA.,Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Lawrence M Witmer
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| |
Collapse
|
18
|
Varela-Nieto I, Palmero I, Magariños M. Complementary and distinct roles of autophagy, apoptosis and senescence during early inner ear development. Hear Res 2019; 376:86-96. [PMID: 30711386 DOI: 10.1016/j.heares.2019.01.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 01/13/2019] [Accepted: 01/17/2019] [Indexed: 12/25/2022]
Abstract
The development of the inner ear complex cytoarchitecture and functional geometry requires the exquisite coordination of a variety of cellular processes in a temporal manner. At early stages of inner ear development several rounds of cell proliferation in the otocyst promote the growth of the structure. The apoptotic program is initiated in exceeding cells to adjust cell type numbers. Apoptotic cells are cleared by phagocytic cells that recognize the phosphatidylserine residues exposed in the cell membrane thanks to the energy supplied by autophagy. Specific molecular programs determine hair and supporting cell fate, these populations are responsible for the functions of the adult sensory organ: detection of sound, position and acceleration. The neurons that transmit auditory and balance information to the brain are also born at the otocyst by neurogenesis facilitated by autophagy. Cellular senescence participates in tissue repair, cancer and aging, situations in which cells enter a permanent cell cycle arrest and acquire a highly secretory phenotype that modulates their microenvironment. More recently, senescence has also been proposed to take place during vertebrate development in a limited number of transitory structures and organs; among the later, the endolymphatic duct in the inner ear. Here, we review these cellular processes during the early development of the inner ear, focusing on how the most recently described cellular senescence participates and cooperates with proliferation, apoptosis and autophagy to achieve otic morphogenesis and differentiation.
Collapse
Affiliation(s)
- Isabel Varela-Nieto
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain; Centre for Biomedical Network Research on Rare Diseases (CIBERER), Madrid, Spain; Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Ignacio Palmero
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain
| | - Marta Magariños
- Institute for Biomedical Research "Alberto Sols" (IIBM), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain; Centre for Biomedical Network Research on Rare Diseases (CIBERER), Madrid, Spain; Biology Department, Faculty of Sciences, Autonomous University of Madrid (UAM), Madrid, Spain.
| |
Collapse
|
19
|
Sun P, Zhang Y, Zhao F, Wu JP, Pun SH, Peng C, Du M, Vai MI, Liu D, Chen F. An Assay for Systematically Quantifying the Vestibulo-Ocular Reflex to Assess Vestibular Function in Zebrafish Larvae. Front Cell Neurosci 2018; 12:257. [PMID: 30186115 PMCID: PMC6113563 DOI: 10.3389/fncel.2018.00257] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 07/27/2018] [Indexed: 12/25/2022] Open
Abstract
Zebrafish (Danio rerio) larvae are widely used to study otic functions because they possess all five typical vertebrate senses including hearing and balance. Powerful genetic tools and the transparent body of the embryo and larva also make zebrafish a unique vertebrate model to study otic development. Due to its small larval size and moisture requirement during experiments, accurately acquiring the vestibulo-ocular reflex (VOR) of zebrafish larva is challenging. In this report, a new VOR testing device has been developed for quantifying linear VOR (LVOR) in zebrafish larva, evoked by the head motion about the earth horizontal axis. The system has a newly designed larva-shaped chamber, by which live fish can be steadily held without anesthesia, and the system is more compact and easier to use than its predecessors. To demonstrate the efficacy of the system, the LVORs in wild-type (WT), dlx3b and dlx4b morphant zebrafish larvae were measured and the results showed that LVOR amplitudes were consistent with the morphological changes of otoliths induced by morpholino oligonucleotides (MO). Our study represents an important advance to obtain VOR and predict the vestibular conditions in zebrafish.
Collapse
Affiliation(s)
- Peng Sun
- State Key Laboratory of Analog and Mixed-Signal VLSI, University of Macau, Taipa, China.,Department of Electrical and Computer Engineering, Faculty of Science and Technology, University of Macau, Taipa, China.,Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Yingla Zhang
- School of Life Sciences, Peking University, Beijing, China
| | - Feng Zhao
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Jian-Ping Wu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China.,SUSTech Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen, China
| | - Sio Hang Pun
- State Key Laboratory of Analog and Mixed-Signal VLSI, University of Macau, Taipa, China
| | - Cheng Peng
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Meide Du
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Mang I Vai
- State Key Laboratory of Analog and Mixed-Signal VLSI, University of Macau, Taipa, China.,Department of Electrical and Computer Engineering, Faculty of Science and Technology, University of Macau, Taipa, China
| | - Dong Liu
- School of Life Sciences, Peking University, Beijing, China.,Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Fangyi Chen
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
20
|
Schrauwen I, Kari E, Mattox J, Llaci L, Smeeton J, Naymik M, Raible DW, Knowles JA, Crump JG, Huentelman MJ, Friedman RA. De novo variants in GREB1L are associated with non-syndromic inner ear malformations and deafness. Hum Genet 2018; 137:459-470. [PMID: 29955957 PMCID: PMC6082420 DOI: 10.1007/s00439-018-1898-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 06/19/2018] [Indexed: 11/24/2022]
Abstract
Congenital inner ear malformations affecting both the osseous and membranous labyrinth can have a devastating impact on hearing and language development. With the exception of an enlarged vestibular aqueduct, non-syndromic inner ear malformations are rare, and their underlying molecular biology has thus far remained understudied. To identify molecular factors that might be important in the developing inner ear, we adopted a family-based trio exome sequencing approach in young unrelated subjects with severe inner ear malformations. We identified two previously unreported de novo loss-of-function variants in GREB1L [c.4368G>T;p.(Glu1410fs) and c.982C>T;p.(Arg328*)] in two affected subjects with absent cochleae and eighth cranial nerve malformations. The cochlear aplasia in these affected subjects suggests that a developmental arrest or problem at a very early stage of inner ear development exists, e.g., during the otic pit formation. Craniofacial Greb1l RNA expression peaks in mice during this time frame (E8.5). It also peaks in the developing inner ear during E13-E16, after which it decreases in adulthood. The crucial function of Greb1l in craniofacial development is also evidenced in knockout mice, which develop severe craniofacial abnormalities. In addition, we show that Greb1l-/- zebrafish exhibit a loss of abnormal sensory epithelia innervation. An important role for Greb1l in sensory epithelia innervation development is supported by the eighth cranial nerve deficiencies seen in both affected subjects. In conclusion, we demonstrate that GREB1L is a key player in early inner ear and eighth cranial nerve development. Abnormalities in cochleovestibular anatomy can provide challenges for cochlear implantation. Combining a molecular diagnosis with imaging techniques might aid the development of individually tailored therapeutic interventions in the future.
Collapse
Affiliation(s)
- Isabelle Schrauwen
- Molecular and Human Genetics Department, Center for Statistical Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
- Neurogenomics Division and Center for Rare Childhood Disorders, Translational Genomics Research Institute, 445 N 5th str, Phoenix, AZ, 85004, USA.
| | - Elina Kari
- Division of Otolaryngology, Head and Neck Surgery, Department of Surgery, University of California, San Diego, ECOB-East Campus Office Building Room 3-013, 9444 Medical Center Drive, Mail Code 7220, La Jolla, CA, 92037, USA
| | - Jacob Mattox
- Tina and Rick Caruso Department of Otolaryngology-Head and Neck Surgery, Keck University of Southern California School of Medicine, 1975 Zonal Ave., Los Angeles, CA, 90033, USA
| | - Lorida Llaci
- Neurogenomics Division and Center for Rare Childhood Disorders, Translational Genomics Research Institute, 445 N 5th str, Phoenix, AZ, 85004, USA
| | - Joanna Smeeton
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, 1975 Zonal Ave., Los Angeles, CA, 90033, USA
| | - Marcus Naymik
- Neurogenomics Division and Center for Rare Childhood Disorders, Translational Genomics Research Institute, 445 N 5th str, Phoenix, AZ, 85004, USA
| | - David W Raible
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - James A Knowles
- Department of Cell Biology-MSC 5, SUNY Downstate Medical Center, 450 Clarkson Avenue, BSB 2-5, Brooklyn, NY, 11203, USA
| | - J Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, 1975 Zonal Ave., Los Angeles, CA, 90033, USA
| | - Matthew J Huentelman
- Neurogenomics Division and Center for Rare Childhood Disorders, Translational Genomics Research Institute, 445 N 5th str, Phoenix, AZ, 85004, USA
| | - Rick A Friedman
- Division of Otolaryngology, Head and Neck Surgery, Department of Surgery, University of California, San Diego, ECOB-East Campus Office Building Room 3-013, 9444 Medical Center Drive, Mail Code 7220, La Jolla, CA, 92037, USA
| |
Collapse
|
21
|
Nimpf S, Malkemper EP, Lauwers M, Ushakova L, Nordmann G, Wenninger-Weinzierl A, Burkard TR, Jacob S, Heuser T, Resch GP, Keays DA. Subcellular analysis of pigeon hair cells implicates vesicular trafficking in cuticulosome formation and maintenance. eLife 2017; 6:e29959. [PMID: 29140244 PMCID: PMC5699870 DOI: 10.7554/elife.29959] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 11/11/2017] [Indexed: 11/13/2022] Open
Abstract
Hair cells are specialized sensors located in the inner ear that enable the transduction of sound, motion, and gravity into neuronal impulses. In birds some hair cells contain an iron-rich organelle, the cuticulosome, that has been implicated in the magnetic sense. Here, we exploit histological, transcriptomic, and tomographic methods to investigate the development of cuticulosomes, as well as the molecular and subcellular architecture of cuticulosome positive hair cells. We show that this organelle forms rapidly after hatching in a process that involves vesicle fusion and nucleation of ferritin nanoparticles. We further report that transcripts involved in endocytosis, extracellular exosomes, and metal ion binding are differentially expressed in cuticulosome positive hair cells. These data suggest that the cuticulosome and the associated molecular machinery regulate the concentration of iron within the labyrinth of the inner ear, which might indirectly tune a magnetic sensor that relies on electromagnetic induction.
Collapse
Affiliation(s)
- Simon Nimpf
- Research Institute of Molecular PathologyVienna BiocenterViennaAustria
| | | | - Mattias Lauwers
- Research Institute of Molecular PathologyVienna BiocenterViennaAustria
| | - Lyubov Ushakova
- Research Institute of Molecular PathologyVienna BiocenterViennaAustria
| | - Gregory Nordmann
- Research Institute of Molecular PathologyVienna BiocenterViennaAustria
| | | | - Thomas R Burkard
- Research Institute of Molecular PathologyVienna BiocenterViennaAustria
| | - Sonja Jacob
- Electron Microscopy FacilityVienna BioCenter Core Facilities GmbHViennaAustria
| | - Thomas Heuser
- Electron Microscopy FacilityVienna BioCenter Core Facilities GmbHViennaAustria
| | | | - David A Keays
- Research Institute of Molecular PathologyVienna BiocenterViennaAustria
| |
Collapse
|
22
|
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
|
23
|
Olaya-Sánchez D, Sánchez-Guardado LÓ, Ohta S, Chapman SC, Schoenwolf GC, Puelles L, Hidalgo-Sánchez M. Fgf3 and Fgf16 expression patterns define spatial and temporal domains in the developing chick inner ear. Brain Struct Funct 2016; 222:131-149. [PMID: 26995070 DOI: 10.1007/s00429-016-1205-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 02/19/2016] [Indexed: 11/26/2022]
Abstract
The inner ear is a morphologically complex sensory structure with auditory and vestibular functions. The developing otic epithelium gives rise to neurosensory and non-sensory elements of the adult membranous labyrinth. Extrinsic and intrinsic signals manage the patterning and cell specification of the developing otic epithelium by establishing lineage-restricted compartments defined in turn by differential expression of regulatory genes. FGF3 and FGF16 are excellent candidates to govern these developmental events. Using the chick inner ear, we show that Fgf3 expression is present in the borders of all developing cristae. Strong Fgf16 expression was detected in a portion of the developing vertical and horizontal pouches, whereas the cristae show weaker or undetected Fgf16 expression at different developmental stages. Concerning the rest of the vestibular sensory elements, both the utricular and saccular maculae were Fgf3 positive. Interestingly, strong Fgf16 expression delimited these Fgf16-negative sensory patches. The Fgf3-negative macula neglecta and the Fgf3-positive macula lagena were included within weakly Fgf16-expressing areas. Therefore, different FGF-mediated mechanisms might regulate the specification of the anterior (utricular and saccular) and posterior (neglecta and lagena) maculae. In the developing cochlear duct, dynamic Fgf3 and Fgf16 expression suggests their cooperation in the early specification and later cell differentiation in the hearing system. The requirement of Fgf3 and Fgf16 genes in endolymphatic apparatus development and neurogenesis are discussed. Based on these observations, FGF3 and FGF16 seem to be key signaling pathways that control the inner ear plan by defining epithelial identities within the developing otic epithelium.
Collapse
Affiliation(s)
- Daniel Olaya-Sánchez
- Department of Cell Biology, School of Science, University of Extremadura, Avda. de Elvas s/n, 06071, Badajoz, Spain
| | - Luis Óscar Sánchez-Guardado
- Department of Cell Biology, School of Science, University of Extremadura, Avda. de Elvas s/n, 06071, Badajoz, Spain
| | - Sho Ohta
- Department of Neurobiology and Anatomy, University of Utah, 2R066 School of Medicine, 30 N. 1900 E., Salt Lake City, UT, 84132-3401, USA
| | - Susan C Chapman
- Department of Biological Sciences, Clemson University, 340 Long Hall, Clemson, SC, 29634, USA
| | - Gary C Schoenwolf
- Department of Neurobiology and Anatomy, University of Utah, 2R066 School of Medicine, 30 N. 1900 E., Salt Lake City, UT, 84132-3401, USA
| | - Luis Puelles
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia, 30100, Murcia, Spain
| | - Matías Hidalgo-Sánchez
- Department of Cell Biology, School of Science, University of Extremadura, Avda. de Elvas s/n, 06071, Badajoz, Spain.
| |
Collapse
|
24
|
Kanaya E, Yamahara K, Okano T, Yoshida A, Katsuno T, Takebayashi H, Ito J, Yamamoto N. Expression of the Olig gene family in the developing mouse inner ear. Gene Expr Patterns 2015; 17:79-86. [PMID: 25778822 DOI: 10.1016/j.gep.2015.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 02/15/2015] [Accepted: 03/07/2015] [Indexed: 10/23/2022]
Abstract
Transcription factors are believed to play key roles in determining cell fate in inner ear development. Olig genes, which are basic helix-loop-helix transcription factors, have been reported to play important roles in the development of the central nervous system. However, members of this family have not previously been implicated in inner ear development, despite the similarity between otocyst and neural tube development. Olig1 begins to be expressed at the ventral domain of the otocyst at embryonic day (E) 9.5, and Olig1 expression in the epithelium of the developing inner ear persists to E15.5. Olig2 expression is localized to the cochleovestibular ganglia from E12.5 through E14.5. Olig3 has a diffuse expression pattern in the developing inner ear from E12.5 through the postnatal stage. Furthermore, at early stages of inner ear development, the Olig1 expression domain overlaps a region that is positive for Sox2 and Jagged1. This observation indicates that Olig1 may play an important role in the specification of the prosensory domain in the developing inner ear. As Olig genes are expressed in the mouse developing inner ear in a temporospatially distinct fashion, they may play substantial roles in the regulation of mammalian inner ear development.
Collapse
Affiliation(s)
- Eriko Kanaya
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo, Kyoto 606-8507, Japan
| | - Kohei Yamahara
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo, Kyoto 606-8507, Japan
| | - Takayuki Okano
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo, Kyoto 606-8507, Japan
| | - Atsuhiro Yoshida
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo, Kyoto 606-8507, Japan
| | - Tatsuya Katsuno
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo, Kyoto 606-8507, Japan
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Asahimachi Chuo-ku, Niigata 951-8510, Japan
| | - Juichi Ito
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo, Kyoto 606-8507, Japan
| | - Norio Yamamoto
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo, Kyoto 606-8507, Japan.
| |
Collapse
|
25
|
Park BY. Sox9 regulates development of neural crest and otic placode in a time- and dose-dependent fashion. J Biomed Res 2015. [DOI: 10.12729/jbr.2015.16.1.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
26
|
Nakajima Y. Signaling regulating inner ear development: cell fate determination, patterning, morphogenesis, and defects. Congenit Anom (Kyoto) 2015; 55:17-25. [PMID: 25040109 DOI: 10.1111/cga.12072] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Accepted: 06/07/2014] [Indexed: 12/28/2022]
Abstract
The membranous labyrinth of the inner ear is a highly complex organ that detects sound and balance. Developmental defects in the inner ear cause congenital hearing loss and balance disorders. The membranous labyrinth consists of three semicircular ducts, the utricle, saccule, and endolymphatic ducts, and the cochlear duct. These complex structures develop from the simple otic placode, which is established in the cranial ectoderm adjacent to the neural crest at the level of the hindbrain at the early neurula stage. During development, the otic placode invaginates to form the otic vesicle, which subsequently gives rise to neurons for the vestibulocochlear ganglion, the non-sensory and sensory epithelia of the membranous labyrinth that includes three ampullary crests, two maculae, and the organ of Corti. Combined paracrine and autocrine signals including fibroblast growth factor, Wnt, retinoic acid, hedgehog, and bone morphogenetic protein regulate fate determination, axis formation, and morphogenesis in the developing inner ear. Juxtacrine signals mediated by Notch pathways play a role in establishing the sensory epithelium, which consists of mechanosensory hair cells and supporting cells. The highly differentiated organ of Corti, which consists of uniformly oriented inner/outer hair cells and specific supporting cells, develops during fetal development. Developmental alterations/arrest causes congenital malformations in the inner ear in a spatiotemporal-restricted manner. A clearer understanding of the mechanisms underlying inner ear development is important not only for the management of patients with congenital inner ear malformations, but also for the development of regenerative therapy for impaired function.
Collapse
Affiliation(s)
- Yuji Nakajima
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| |
Collapse
|
27
|
Haque KD, Pandey AK, Kelley MW, Puligilla C. Culture of embryonic mouse cochlear explants and gene transfer by electroporation. J Vis Exp 2015:52260. [PMID: 25651458 DOI: 10.3791/52260] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Auditory hair cells located within the mouse organ of Corti detect and transmit sound information to the central nervous system. The mechanosensory hair cells are aligned in one row of inner hair cells and three rows of outer hair cells that extend along the basal to apical axis of the cochlea. The explant culture technique described here provides an efficient method to isolate and maintain cochlear explants from the embryonic mouse inner ear. Also, the morphology and molecular characteristics of sensory hair cells and nonsensory supporting cells within the cochlear explant cultures resemble those observed in vivo and can be studied within its intrinsic cellular environment. The cochlear explants can serve as important experimental tools for the identification and characterization of molecular and genetic pathways that are involved in cellular specification and patterning. Although transgenic mouse models provide an effective approach for gene expression studies, a considerable number of mouse mutants die during embryonic development thereby hindering the analysis and interpretation of developmental phenotypes. The organ of Corti from mutant mice that die before birth can be cultured so that their in vitro development and responses to different factors can be analyzed. Additionally, we describe a technique for electroporating embryonic cochlear explants ex vivo which can be used to downregulate or overexpress specific gene(s) and analyze their potential endogenous function and test whether specific gene product is necessary or sufficient in a given context to influence mammalian cochlear development(1-8).
Collapse
Affiliation(s)
- Khujista D Haque
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, College of Medicine
| | - Atul K Pandey
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, College of Medicine
| | | | - Chandrakala Puligilla
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, College of Medicine;
| |
Collapse
|
28
|
Interplay of proliferation and proapoptotic and antiapoptotic factors is revealed in the early human inner ear development. Otol Neurotol 2014; 35:695-703. [PMID: 24622024 DOI: 10.1097/mao.0000000000000210] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
HYPOTHESIS Spatiotemporal interplay of factors controlling proliferation, differentiation and apoptosis within the developing human inner ear is essential for labyrinth morphogenesis and development of vestibular and cochlear functions. BACKGROUND Studies on the early human inner ear development are scarce and insufficient. METHODS The immunolocalization of Ki-67, Bcl-2, caspase-3, and IGF-1 was analyzed in 6 human inner ears, 5 to 10 gestational weeks old. Statistical data were analyzed using the Kruskal-Wallis test. RESULTS During the analyzed period, the otocyst has transformed into cochlear duct and saccule ventrally and semicircular canals and utricle dorsally. Initial differentiation of sensorineural fields characterized organ of Corti, maculae, and cristae ampullares. Intense (50%) and evenly distributed proliferation Ki-67 in the otocyst decreased to 24% to 30% and became spatially restricted within the membranous labyrinth epithelium. Simultaneously, expression of antiapoptotic Bcl-2 protein increased in sensorineural fields of organ of Corti, macula, and crista ampullaris. Throughout the investigated period, apoptotic caspase-3 positive cells were mainly distributed at the luminal and basal surfaces of labyrinth epithelium. An inhibitor of apoptosis IGF-1 co-expressed with Bcl-2 and increased in the sensorineural fields with advancing development. CONCLUSION The described expression pattern indicates roles for cell proliferation in the growth of the inner ear and Bcl-2 in differentiation of sensorineural fields and protection from apoptosis. Both IGF-1-and caspase-3-mediated apoptosis seem to contribute to proper morphogenesis, differentiation, and innervations of sensorineural fields within the cochlea, semicircular canals, saccule, and utricle. Alterations in spatiotemporal interplay of investigated factors might lead to disturbances of vestibular and cochlear function.
Collapse
|
29
|
Said-Al-Naief N, Pourian A, Cure J, Lopez R. Clinicopathologic conference case 3: a 75- year-old man with progressive right-sided hearing loss and dizziness. Oral Surg Oral Med Oral Pathol Oral Radiol 2014; 118:e89-94. [PMID: 25295344 DOI: 10.1016/j.oooo.2014.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
30
|
Willaredt MA, Ebbers L, Nothwang HG. Central auditory function of deafness genes. Hear Res 2014; 312:9-20. [DOI: 10.1016/j.heares.2014.02.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 01/31/2014] [Accepted: 02/10/2014] [Indexed: 01/11/2023]
|
31
|
Forristall CA, Stellabotte F, Castillo A, Collazo A. Embryological manipulations in the developing Xenopus inner ear reveal an intrinsic role for Wnt signaling in dorsal-ventral patterning. Dev Dyn 2014; 243:1262-74. [PMID: 24500889 DOI: 10.1002/dvdy.24116] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Revised: 01/02/2014] [Accepted: 01/17/2014] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND The inner ear develops from an ectodermal thickening known as the otic placode into a complex structure that is asymmetrical along both the anterior-posterior (A-P) and dorsal-ventral (D-V) axes. Embryological manipulations in Xenopus allow us to test regenerative potential along specific axes and timing of axis determination. We explore the role of Wnt signaling with gain and loss of function experiments. RESULTS In contrast to A or P half ablations, D or V half ablations almost never result in mirror duplications or normal ears. Instead there is a loss of structures, especially those associated with the ablated region. Rotation experiments inverting the D-V axis reveal that it is determined by stage 24-26 which is just before expression of the dorsal otic marker Wnt3a. Conditional blocking of canonical Wnt signaling results in reductions in the number of sensory organs and semicircular canals which could be placed in one of three categories, the most common phenotypes being similar to those seen after dorsal ablations. CONCLUSIONS There is less regenerative potential along the D-V axis. Wnt3a protein alone is sufficient to rescue the severe loss of inner ear structures resulting from dorsal but not ventral half ablations.
Collapse
|
32
|
Lassiter RNT, Stark MR, Zhao T, Zhou CJ. Signaling mechanisms controlling cranial placode neurogenesis and delamination. Dev Biol 2013; 389:39-49. [PMID: 24315854 DOI: 10.1016/j.ydbio.2013.11.025] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 11/22/2013] [Accepted: 11/23/2013] [Indexed: 01/17/2023]
Abstract
The neurogenic cranial placodes are a unique transient epithelial niche of neural progenitor cells that give rise to multiple derivatives of the peripheral nervous system, particularly, the sensory neurons. Placode neurogenesis occurs throughout an extended period of time with epithelial cells continually recruited as neural progenitor cells. Sensory neuron development in the trigeminal, epibranchial, otic, and olfactory placodes coincides with detachment of these neuroblasts from the encompassing epithelial sheet, leading to delamination and ingression into the mesenchyme where they continue to differentiate as neurons. Multiple signaling pathways are known to direct placodal development. This review defines the signaling pathways working at the finite spatiotemporal period when neuronal selection within the placodes occurs, and neuroblasts concomitantly delaminate from the epithelium. Examining neurogenesis and delamination after initial placodal patterning and specification has revealed a common trend throughout the neurogenic placodes, which suggests that both activated FGF and attenuated Notch signaling activities are required for neurogenesis and changes in epithelial cell adhesion leading to delamination. We also address the varying roles of other pathways such as the Wnt and BMP signaling families during sensory neurogenesis and neuroblast delamination in the differing placodes.
Collapse
Affiliation(s)
- Rhonda N T Lassiter
- Institute for Pediatric Regenerative Medicine at Shriners Hospitals for Children-Northern California, CA 95817, USA; Department of Cell Biology and Human Anatomy, University of California Davis, School of Medicine, Sacramento, CA 95817, USA.
| | - Michael R Stark
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA.
| | - Tianyu Zhao
- Institute for Pediatric Regenerative Medicine at Shriners Hospitals for Children-Northern California, CA 95817, USA; Department of Cell Biology and Human Anatomy, University of California Davis, School of Medicine, Sacramento, CA 95817, USA
| | - Chengji J Zhou
- Institute for Pediatric Regenerative Medicine at Shriners Hospitals for Children-Northern California, CA 95817, USA; Department of Cell Biology and Human Anatomy, University of California Davis, School of Medicine, Sacramento, CA 95817, USA; Department of Biochemistry and Molecular Medicine, University of California Davis, School of Medicine, Sacramento, CA 95817, USA.
| |
Collapse
|
33
|
Park JE, Son AI, Zhou R. Roles of EphA2 in Development and Disease. Genes (Basel) 2013; 4:334-57. [PMID: 24705208 PMCID: PMC3924825 DOI: 10.3390/genes4030334] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 05/22/2013] [Accepted: 05/23/2013] [Indexed: 01/12/2023] Open
Abstract
The Eph family of receptor tyrosine kinases (RTKs) has been implicated in the regulation of many aspects of mammalian development. Recent analyses have revealed that the EphA2 receptor is a key modulator for a wide variety of cellular functions. This review focuses on the roles of EphA2 in both development and disease.
Collapse
Affiliation(s)
- Jeong Eun Park
- Susan Lehman-Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA.
| | - Alexander I Son
- Susan Lehman-Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA.
| | - Renping Zhou
- Susan Lehman-Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
34
|
Tracing Sox10-expressing cells elucidates the dynamic development of the mouse inner ear. Hear Res 2013; 302:17-25. [PMID: 23684581 DOI: 10.1016/j.heares.2013.05.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 04/30/2013] [Accepted: 05/06/2013] [Indexed: 01/12/2023]
Abstract
The inner ear is constituted by complicated cochlear and vestibular compartments, which are derived from the otic vesicle, an embryonic structure of ectodermal origin. Although the inner ear development has been analyzed using various techniques, the developmental events have not been fully elucidated because of the intricate structure. We previously developed a Sox10-IRES-Venus mouse designed to express green fluorescent protein under the control of the Sox10 promoter. In the present study, we showed that the Sox10-IRES-Venus mouse enabled the non-destructive visualization and understanding of the morphogenesis during the development of the inner ear. The expression of the transcription factor Sox10 was first observed in the invaginating otic placodal epithelium, and continued to be expressed in the mature inner ear epithelium except for the hair cells and mesenchymal cells. We found that Sox10 was expressed in immature hair cells in the developing inner ear, suggesting that hair cells were generated from the Sox10-expressing prosensory cells. Furthermore, we demonstrated that scattered Sox10-expressing cells existed around the developing inner ear, some of which differentiated into pigmented melanocytes in the stria vascularis, suggesting that they were neural crest cells. Further analyzing the Sox10-IRES-Venus mice would provide important information to better understand the development of the inner ear.
Collapse
|
35
|
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
|
36
|
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]
|
37
|
Gata3 directly regulates early inner ear expression of Fgf10. Dev Biol 2013; 374:210-22. [DOI: 10.1016/j.ydbio.2012.11.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 11/23/2012] [Accepted: 11/26/2012] [Indexed: 01/19/2023]
|
38
|
Abstract
Therapies aimed at the protection and/or regeneration of inner ear hair cells are of great interest, given the significant monetary and quality of life impact of balance disorders. Different viral vectors have been shown to transfect various cell types in the inner ear. The past decade has provided tremendous advances in the use of adenoviral vectors to achieve targeted treatment delivery. Several routes of delivery have been identified to introduce vectors into the inner ear while minimizing injury to surrounding structures. Recently, the transcription factor Atoh1 was determined to play a critical role in hair cell differentiation. Adenoviral-mediated overexpression of Atoh1 in culture and in vivo has demonstrated the ability to regenerate vestibular hair cells by causing transdifferentiation of neighbouring epithelial-supporting cells. Functional recovery of the vestibular system has also been documented following adenoviral-induced Atoh1 overexpression. Experiments demonstrating gene transfer in human vestibular epithelial cells reveal that the human inner ear is a suitable target for gene therapy.
Collapse
Affiliation(s)
- Silviu Albu
- Second Department of Otolaryngology, University of Medicine and Pharmacy Cluj-Napoca, Cluj-Napoca, Romania.
| | | |
Collapse
|
39
|
Roeseler DA, Sachdev S, Buckley DM, Joshi T, Wu DK, Xu D, Hannink M, Waters ST. Elongation factor 1 alpha1 and genes associated with Usher syndromes are downstream targets of GBX2. PLoS One 2012; 7:e47366. [PMID: 23144817 PMCID: PMC3493575 DOI: 10.1371/journal.pone.0047366] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 09/12/2012] [Indexed: 11/18/2022] Open
Abstract
Gbx2 encodes a DNA-binding transcription factor that plays pivotal roles during embryogenesis. Gain-and loss-of-function studies in several vertebrate species have demonstrated a requirement for Gbx2 in development of the anterior hindbrain, spinal cord, inner ear, heart, and neural crest cells. However, the target genes through which GBX2 exerts its effects remain obscure. Using chromatin immunoprecipitation coupled with direct sequencing (ChIP-Seq) analysis in a human prostate cancer cell line, we identified cis-regulatory elements bound by GBX2 to provide insight into its direct downstream targets. The analysis revealed more than 286 highly significant candidate target genes, falling into various functional groups, of which 51% are expressed in the nervous system. Several of the top candidate genes include EEF1A1, ROBO1, PLXNA4, SLIT3, NRP1, and NOTCH2, as well as genes associated with the Usher syndrome, PCDH15 and USH2A, and are plausible candidates contributing to the developmental defects in Gbx2(-/-) mice. We show through gel shift analyses that sequences within the promoter or introns of EEF1A1, ROBO1, PCDH15, USH2A and NOTCH2, are directly bound by GBX2. Consistent with these in vitro results, analyses of Gbx2(-/-) embryos indicate that Gbx2 function is required for migration of Robo1-expressing neural crest cells out of the hindbrain. Furthermore, we show that GBX2 activates transcriptional activity through the promoter of EEF1A1, suggesting that GBX2 could also regulate gene expression indirectly via EEF1A. Taken together, our studies show that GBX2 plays a dynamic role in development and diseases.
Collapse
Affiliation(s)
- David A. Roeseler
- Division of Biological Sciences, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
| | - Shrikesh Sachdev
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
| | - Desire M. Buckley
- Division of Biological Sciences, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
| | - Trupti Joshi
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
- Department of Computer Science, University of Missouri, Columbia, Missouri, United States of America
- Informatics Institute, University of Missouri, Columbia, Missouri, United States of America
| | - Doris K. Wu
- Laboratory of Molecular Biology, NIDCD, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Dong Xu
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
- Department of Computer Science, University of Missouri, Columbia, Missouri, United States of America
- Informatics Institute, University of Missouri, Columbia, Missouri, United States of America
| | - Mark Hannink
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
| | - Samuel T. Waters
- Division of Biological Sciences, University of Missouri, Columbia, Missouri, United States of America
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
| |
Collapse
|
40
|
Lin J, Yan X, Wang C, Guo Z, Rolfs A, Luo J. Anatomical expression patterns of delta-protocadherins in developing chicken cochlea. J Anat 2012; 221:598-608. [PMID: 22998331 DOI: 10.1111/j.1469-7580.2012.01568.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2012] [Indexed: 11/29/2022] Open
Abstract
The delta-protocadherin (δ-Pcdh) family of transmembrane proteins belongs to the cadherin superfamily, which is involved in embryogenesis mediated by a homophilic binding during the embryonic development. In the present study, expression patterns of eight members of the δ-Pcdh family were investigated in the developing chicken cochlea by in situ hybridization. Our results provide a dynamical profile to show that the δ-Pcdhs are expressed spatially and temporally in the developing chicken cochleae. The earliest onset of the δ-Pcdh expression begins in the otic vesicle from embryonic incubation day (E) 3. From E11 onwards, the individual δ-Pcdh is expressed in different cell types of the cochlea. Protocadherin-1 (Pcdh1) is mainly expressed by spindle-shaped cells and acoustic ganglion cells; Pcdh7 and Pcdh17 are strongly expressed by supporting cells, cuboidal cells, hyaline cells and acoustic ganglion cells, and Pcdh9 is prominently expressed by homogene cells and acoustic ganglion cells; Pcdh8 was found to be transcribed in hair cells, spindle-shaped cells and acoustic ganglion cells; Pcdh10 mRNA is restricted to spindle-shaped cells and acoustic ganglion cells at later stages. mRNAs of Pcdh1, Pcdh18 and Pcdh19 are also expressed in blood vessels of the cochlea. The expression of the different δ-Pcdhs suggests a functional role for them during cochlear development.
Collapse
Affiliation(s)
- Juntang Lin
- Key Laboratory for medical tissue regeneration of Henan province, Xinxiang Medical University, China
| | | | | | | | | | | |
Collapse
|
41
|
Grocott T, Tambalo M, Streit A. The peripheral sensory nervous system in the vertebrate head: a gene regulatory perspective. Dev Biol 2012; 370:3-23. [PMID: 22790010 DOI: 10.1016/j.ydbio.2012.06.028] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 06/28/2012] [Accepted: 06/29/2012] [Indexed: 02/06/2023]
Abstract
In the vertebrate head, crucial parts of the sense organs and sensory ganglia develop from special regions, the cranial placodes. Despite their cellular and functional diversity, they arise from a common field of multipotent progenitors and acquire distinct identity later under the influence of local signalling. Here we present the gene regulatory network that summarises our current understanding of how sensory cells are specified, how they become different from other ectodermal derivatives and how they begin to diversify to generate placodes with different identities. This analysis reveals how sequential activation of sets of transcription factors subdivides the ectoderm over time into smaller domains of progenitors for the central nervous system, neural crest, epidermis and sensory placodes. Within this hierarchy the timing of signalling and developmental history of each cell population is of critical importance to determine the ultimate outcome. A reoccurring theme is that local signals set up broad gene expression domains, which are further refined by mutual repression between different transcription factors. The Six and Eya network lies at the heart of sensory progenitor specification. In a positive feedback loop these factors perpetuate their own expression thus stabilising pre-placodal fate, while simultaneously repressing neural and neural crest specific factors. Downstream of the Six and Eya cassette, Pax genes in combination with other factors begin to impart regional identity to placode progenitors. While our review highlights the wealth of information available, it also points to the lack information on the cis-regulatory mechanisms that control placode specification and of how the repeated use of signalling input is integrated.
Collapse
Affiliation(s)
- Timothy Grocott
- Department of Craniofacial Development and Stem Cell Biology, King's College London, Guy's Tower Wing, Floor 27, London SE1 9RT, UK
| | | | | |
Collapse
|
42
|
Harden MV, Pereiro L, Ramialison M, Wittbrodt J, Prasad MK, McCallion AS, Whitlock KE. Close association of olfactory placode precursors and cranial neural crest cells does not predestine cell mixing. Dev Dyn 2012; 241:1143-54. [PMID: 22539261 PMCID: PMC4240535 DOI: 10.1002/dvdy.23797] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2012] [Indexed: 12/22/2022] Open
Abstract
Vertebrate sensory organs originate from both cranial neural crest cells (CNCCs) and placodes. Previously, we have shown that the olfactory placode (OP) forms from a large field of cells extending caudally to the premigratory neural crest domain, and that OPs form through cell movements and not cell division. Concurrent with OP formation, CNCCs migrate rostrally to populate the frontal mass. However, little is known about the interactions between CNCCs and the placodes that form the olfactory sensory system. Previous reports suggest that the OP can generate cell types more typical of neural crest lineages such as neuroendocrine cells and glia, thus marking the OP as an unusual sensory placode. One possible explanation for this exception is that the neural crest origin of glia and neurons has been overlooked due to the intimate association of these two fields during migration. Using molecular markers and live imaging, we followed the development of OP precursors and of dorsally migrating CNCCs in zebrafish embryos. We generated a six4b:mCherry line (OP precursors) that, with a sox10:EGFP line (CNCCs), was used to follow cell migration. Our analyses showed that CNCCs associate with and eventually surround the forming OP with limited cell mixing occurring during this process.
Collapse
Affiliation(s)
- Maegan V Harden
- Department of Molecular Biology and Genetics, 445/449 Biotechnology Building, Cornell University, Ithaca, New York, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Kopecky B, Johnson S, Schmitz H, Santi P, Fritzsch B. Scanning thin-sheet laser imaging microscopy elucidates details on mouse ear development. Dev Dyn 2012; 241:465-80. [PMID: 22271591 PMCID: PMC5010664 DOI: 10.1002/dvdy.23736] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2012] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The mammalian inner ear is transformed from a flat placode into a three-dimensional (3D) structure with six sensory epithelia that allow for the perception of sound and both linear and angular acceleration. While hearing and balance problems are typically considered to be adult onset diseases, they may arise as a developmental perturbation to the developing ear. Future prevention of hearing or balance loss requires an understanding of how closely genetic mutations in model organisms reflect the human case, necessitating an objective multidimensional comparison of mouse ears with human ears that have comparable mutations in the same gene. RESULTS Here, we present improved 3D analyses of normal murine ears during embryonic development using optical sections obtained through Thin-Sheet Laser Imaging Microscopy. We chronicle the transformation of an undifferentiated otic vesicle between mouse embryonic day 11.5 to a fully differentiated inner ear at postnatal day 15. CONCLUSIONS Our analysis of ear development provides new insights into ear development, enables unique perspectives into the complex development of the ear, and allows for the first full quantification of volumetric and linear aspects of ear growth. Our data provide the framework for future analysis of mutant phenotypes that are currently under-appreciated using only two dimensional renderings.
Collapse
Affiliation(s)
- Benjamin Kopecky
- Department of Biology, University of Iowa, Iowa City, Iowa 52242, USA.
| | | | | | | | | |
Collapse
|
44
|
Noda T, Oki S, Kitajima K, Harada T, Komune S, Meno C. Restriction of Wnt signaling in the dorsal otocyst determines semicircular canal formation in the mouse embryo. Dev Biol 2011; 362:83-93. [PMID: 22166339 DOI: 10.1016/j.ydbio.2011.11.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Revised: 11/27/2011] [Accepted: 11/28/2011] [Indexed: 01/07/2023]
Abstract
The mouse inner ear develops from a simple epithelial pouch, the otocyst, with the dorsal and ventral portions giving rise to the vestibule and cochlea, respectively. The otocyst undergoes a morphological change to generate flattened saclike structures, known as outpocketings, in the dorsal and lateral regions. The semicircular canals of the vestibule form from the periphery of the outpocketings, with the central region (the fusion plate) undergoing de-epithelialization and disappearing. However, little is known of the mechanism that orchestrates formation of the semicircular canals. We now show that the area of canonical Wnt signaling changes dynamically in the dorsal otocyst during its morphogenesis. The genes for several Wnt ligands were found to be expressed in the dorsal otocyst according to specific patterns, whereas those for secreted inhibitors of Wnt ligands were expressed exclusively in the ventral otocyst. With the use of whole-embryo culture in combination with potent modulators of canonical Wnt signaling, we found that forced persistence of such signaling resulted in impaired formation both of the lateral outpocketing and of the fusion plates of the dorsal outpocketing. Canonical Wnt signaling was found to suppress Netrin1 expression and to preserve the integrity of the outpocketing epithelium. In addition, inhibition of canonical Wnt signaling reduced the size of the otocyst, likely through suppression of cell proliferation and promotion of apoptosis. Our stage-specific functional analysis suggests that strict regulation of canonical Wnt signaling in the dorsal otocyst orchestrates the process of semicircular canal formation.
Collapse
Affiliation(s)
- Teppei Noda
- Department of Developmental Biology, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, Japan
| | | | | | | | | | | |
Collapse
|
45
|
Chatterjee S, Lufkin T. The sound of silence: mouse models for hearing loss. GENETICS RESEARCH INTERNATIONAL 2011; 2011:416450. [PMID: 22567353 PMCID: PMC3335620 DOI: 10.4061/2011/416450] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 08/10/2011] [Indexed: 01/27/2023]
Abstract
Sensorineural hearing loss is one of the most common disabilities in humans. It is estimated that about 278 million people worldwide have slight to extreme hearing loss in both ears, which results in an economic loss for the country and personal loss for the individual. It is thus critical to have a deeper understanding of the causes for hearing loss to better manage and treat the affected individuals. The mouse serves as an excellent model to study and recapitulate some of these phenotypes, identify new genes which cause deafness, and to study their roles in vivo and in detail. Mutant mice have been instrumental in elucidating the function and mechanisms of the inner ear. The development and morphogenesis of the inner ear from an ectodermal layer into distinct auditory and vestibular components depends on well-coordinated gene expression and well-orchestrated signaling cascades within the otic vesicle and interactions with surrounding layers of tissues. Any disruption in these pathways can lead to hearing impairment. This review takes a look at some of the genes and their corresponding mice mutants that have shed light on the mechanism governing hearing impairment (HI) in humans.
Collapse
Affiliation(s)
- Sumantra Chatterjee
- Stem Cell and Developmental Biology, Genome Institute of Singapore, 60 Biopolis Street, Singapore 138672
| | | |
Collapse
|
46
|
Origin and segregation of cranial placodes in Xenopus laevis. Dev Biol 2011; 360:257-75. [PMID: 21989028 DOI: 10.1016/j.ydbio.2011.09.024] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2011] [Revised: 08/31/2011] [Accepted: 09/01/2011] [Indexed: 11/23/2022]
Abstract
Cranial placodes are local thickenings of the vertebrate head ectoderm that contribute to the paired sense organs (olfactory epithelium, lens, inner ear, lateral line), cranial ganglia and the adenohypophysis. Here we use tissue grafting and dye injections to generated fate maps of the dorsal cranial part of the non-neural ectoderm for Xenopus embryos between neural plate and early tailbud stages. We show that all placodes arise from a crescent-shaped area located around the anterior neural plate, the pre-placodal ectoderm. In agreement with proposed roles of Six1 and Pax genes in the specification of a panplacodal primordium and different placodal areas, respectively, we show that Six1 is expressed uniformly throughout most of the pre-placodal ectoderm, while Pax6, Pax3, Pax8 and Pax2 each are confined to specific subregions encompassing the precursors of different subsets of placodes. However, the precursors of the vagal epibranchial and posterior lateral line placodes, which arise from the posteriormost pre-placodal ectoderm, upregulate Six1 and Pax8/Pax2 only at tailbud stages. Whereas our fate map suggests that regions of origin for different placodes overlap extensively with each other and with other ectodermal fates at neural plate stages, analysis of co-labeled placodes reveals that the actual degree of overlap is much smaller. Time lapse imaging of the pre-placodal ectoderm at single cell resolution demonstrates that no directed, large-scale cell rearrangements occur, when the pre-placodal region segregates into distinct placodes at subsequent stages. Our results indicate that individuation of placodes from the pre-placodal ectoderm does not involve large-scale cell sorting in Xenopus.
Collapse
|
47
|
Yan X, Lin J, Markus A, Rolfs A, Luo J. Regional expression of ADAM19 during chicken embryonic development. Dev Growth Differ 2011; 53:333-46. [PMID: 21492148 DOI: 10.1111/j.1440-169x.2010.01238.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
ADAM19 (also named meltrin β) is a member of the ADAM (a disintegrin and metalloprotease) family of metalloproteases and is involved in morphogenesis and tissue formation during embryonic development. In the present study, chicken ADAM19 is cloned by reverse transcription-polymerase chain reaction and identified by sequencing. Its expression patterns in different parts of the developing chicken embryo are investigated by Western blot analysis and immunohistochemistry. Results show that ADAM19 protein is widely expressed in chicken embryos. It is detectable in the central nervous system, including the brain, spinal cord, cochlea, and retina. Furthermore, ADAM19 protein is also found in other tissues and organs such as digestive organs, the thymus, the lung bud, the dorsal aorta, the kidney, the gonad, muscles, and in the feather buds. All these data suggest that ADAM19 plays an important role in the embryonic development of chicken.
Collapse
Affiliation(s)
- Xin Yan
- Albrecht-Kossel-Institute for Neuroregeneration, School of Medicine University of Rostock, Gehlsheimer Strasse 20, D-18147 Rostock, Germany
| | | | | | | | | |
Collapse
|
48
|
Saeger BM, Suhm M, Neubüser A. Ephrin/ephrin receptor expression during early stages of mouse inner ear development. Dev Dyn 2011; 240:1578-85. [DOI: 10.1002/dvdy.22632] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2011] [Indexed: 12/30/2022] Open
|
49
|
Sánchez-Guardado LÓ, Ferran JL, Rodríguez-Gallardo L, Puelles L, Hidalgo-Sánchez M. Meis gene expression patterns in the developing chicken inner ear. J Comp Neurol 2011; 519:125-47. [PMID: 21120931 DOI: 10.1002/cne.22508] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We are interested in stable gene network activities operating sequentially during inner ear specification. The implementation of this patterning process is a key event in the generation of functional subdivisions of the otic vesicle during early embryonic development. The vertebrate inner ear is a complex sensory structure that is a good model system for characterization of developmental mechanisms controlling patterning and specification. Meis genes, belonging to the TALE family, encode homodomain-containing transcription factors remarkably conserved during evolution, which play a role in normal and neoplastic development. To gain understanding of the possible role of homeobox Meis genes in the developing chick inner ear, we comprehensively analyzed their spatiotemporal expression patterns from early otic specification stages onwards. In the invaginating otic placode, Meis1/2 transcripts were observed in the borders of the otic cup, being absent in the portion of otic epithelium closest to the hindbrain. As development proceeds, Meis1 and Meis2 expressions became restricted to the dorsomedial otic epithelium. Both genes were strongly expressed in the entire presumptive domain of the semicircular canals, and more weakly in all associated cristae. The endolymphatic apparatus was labeled in part by Meis1/2. Meis1 was also expressed in the lateral wall of the growing cochlear duct, while Meis2 expression was detected in a few cells of the developing acoustic-vestibular ganglion. Our results suggest a possible role of Meis assigning regional identity in the morphogenesis, patterning, and specification of the developing inner ear.
Collapse
|
50
|
Haugas M, Lilleväli K, Hakanen J, Salminen M. Gata2 is required for the development of inner ear semicircular ducts and the surrounding perilymphatic space. Dev Dyn 2011; 239:2452-69. [PMID: 20652952 DOI: 10.1002/dvdy.22373] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Gata2 has essential roles in the development of many organs. During mouse inner ear morphogenesis, it is expressed in otic vesicle and the surrounding periotic mesenchyme from early on, but no defects in the ear development of Gata2 null mice have been observed before lethality at embryonic day (E) 10.5. Here, we used conditional gene targeting to reveal the role of Gata2 at later stages of inner ear development. We show that Gata2 is critically required from E14.5-E15.5 onward for vestibular morphogenesis. Without Gata2 the semicircular ducts fail to grow to their normal size and the surrounding mesenchymal cells are not removed properly to generate the perilymphatic space. Gata2 is the first factor known to control the clearing of the vestibular perilymphatic mesenchyme, but interestingly, it is not required for the formation of the cochlear perilymphatic areas, suggesting distinct molecular control for these processes.
Collapse
Affiliation(s)
- Maarja Haugas
- Department of Veterinary Biosciences, University of Helsinki, Finland
| | | | | | | |
Collapse
|