1
|
Rumbo M, Alsina B. Cellular diversity of human inner ear organoids revealed by single-cell transcriptomics. Development 2024; 151:dev202524. [PMID: 39612289 DOI: 10.1242/dev.202524] [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] [Indexed: 12/01/2024]
Abstract
Human inner ear organoids are three-dimensional tissular structures grown in vitro that recapitulate some aspects of the fetal inner ear and allow the differentiation of inner ear cell types. These organoids offer a system in which to study human inner ear development, mutations causing hearing loss and vertigo, and new therapeutic drugs. However, the extent to which such organoids mimic in vivo human inner ear development and cellular composition remains unclear. Several recent studies have performed single-cell transcriptomics on human inner ear organoids to interrogate cellular heterogeneity, reveal the developmental trajectories of sensory lineages and compare organoid-derived vesicles to the developing human inner ear. Here, we discuss the new insights provided by these analyses that help to define new paths of investigation to understand inner ear development.
Collapse
Affiliation(s)
- Mireia Rumbo
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra - Parc de Recerca Biomèdica de Barcelona, Carrer del Doctor Aiguader 8808003 Barcelona, Spain
| | - Berta Alsina
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra - Parc de Recerca Biomèdica de Barcelona, Carrer del Doctor Aiguader 8808003 Barcelona, Spain
| |
Collapse
|
2
|
Kuroda S, Lalonde RL, Mansour TA, Mosimann C, Nakamura T. Multiple embryonic sources converge to form the pectoral girdle skeleton in zebrafish. Nat Commun 2024; 15:6313. [PMID: 39060278 PMCID: PMC11282072 DOI: 10.1038/s41467-024-50734-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
The morphological transformation of the pectoral/shoulder girdle is fundamental to the water-to-land transition in vertebrate evolution. Although previous studies have resolved the embryonic origins of tetrapod shoulder girdles, those of fish pectoral girdles remain uncharacterized, creating a gap in the understanding of girdle transformation mechanisms from fish to tetrapods. Here, we identify the embryonic origins of the zebrafish pectoral girdle, including the cleithrum as an ancestral girdle element lost in extant tetrapods. Our combinatorial approach of photoconversion and genetic lineage tracing demonstrates that cleithrum development combines four adjoining embryonic populations. A comparison of these pectoral girdle progenitors with extinct and extant vertebrates highlights that cleithrum loss, indispensable for neck evolution, is associated with the disappearance of its unique developmental environment at the head/trunk interface. Overall, our study establishes an embryological framework for pectoral/shoulder girdle formation and provides evolutionary trajectories from their origin in water to diversification on land.
Collapse
Affiliation(s)
- Shunya Kuroda
- Department of Genetics, Rutgers the State University of New Jersey, Piscataway, NJ, 08854, USA.
- Institute for Frontier Science Initiative, Kanazawa University, Kakuma-machi, Kanazawa, 920-1164, Japan.
| | - Robert L Lalonde
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Thomas A Mansour
- Department of Genetics, Rutgers the State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Christian Mosimann
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Tetsuya Nakamura
- Department of Genetics, Rutgers the State University of New Jersey, Piscataway, NJ, 08854, USA.
| |
Collapse
|
3
|
Greenberg D, Rosenblum ND, Tonelli M. The multifaceted links between hearing loss and chronic kidney disease. Nat Rev Nephrol 2024; 20:295-312. [PMID: 38287134 DOI: 10.1038/s41581-024-00808-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2024] [Indexed: 01/31/2024]
Abstract
Hearing loss affects nearly 1.6 billion people and is the third-leading cause of disability worldwide. Chronic kidney disease (CKD) is also a common condition that is associated with adverse clinical outcomes and high health-care costs. From a developmental perspective, the structures responsible for hearing have a common morphogenetic origin with the kidney, and genetic abnormalities that cause familial forms of hearing loss can also lead to kidney disease. On a cellular level, normal kidney and cochlea function both depend on cilial activities at the apical surface, and kidney tubular cells and sensory epithelial cells of the inner ear use similar transport mechanisms to modify luminal fluid. The two organs also share the same collagen IV basement membrane network. Thus, strong developmental and physiological links exist between hearing and kidney function. These theoretical considerations are supported by epidemiological data demonstrating that CKD is associated with a graded and independent excess risk of sensorineural hearing loss. In addition to developmental and physiological links between kidney and cochlear function, hearing loss in patients with CKD may be driven by specific medications or treatments, including haemodialysis. The associations between these two common conditions are not commonly appreciated, yet have important implications for research and clinical practice.
Collapse
Affiliation(s)
- Dina Greenberg
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, Toronto, Ontario, Canada
| | - Norman D Rosenblum
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, Toronto, Ontario, Canada
- Department of Paediatrics, Temerty Faculty of Medicine, Toronto, Ontario, Canada
| | - Marcello Tonelli
- Department of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
4
|
Wang SX, Streit A. Shared features in ear and kidney development - implications for oto-renal syndromes. Dis Model Mech 2024; 17:dmm050447. [PMID: 38353121 PMCID: PMC10886756 DOI: 10.1242/dmm.050447] [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] [Indexed: 02/16/2024] Open
Abstract
The association between ear and kidney anomalies has long been recognized. However, little is known about the underlying mechanisms. In the last two decades, embryonic development of the inner ear and kidney has been studied extensively. Here, we describe the developmental pathways shared between both organs with particular emphasis on the genes that regulate signalling cross talk and the specification of progenitor cells and specialised cell types. We relate this to the clinical features of oto-renal syndromes and explore links to developmental mechanisms.
Collapse
Affiliation(s)
- Scarlet Xiaoyan Wang
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Andrea Streit
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| |
Collapse
|
5
|
Koontz A, Urrutia HA, Bronner ME. Making a head: Neural crest and ectodermal placodes in cranial sensory development. Semin Cell Dev Biol 2023; 138:15-27. [PMID: 35760729 PMCID: PMC10224775 DOI: 10.1016/j.semcdb.2022.06.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 04/11/2022] [Accepted: 06/19/2022] [Indexed: 01/04/2023]
Abstract
During development of the vertebrate sensory system, many important components like the sense organs and cranial sensory ganglia arise within the head and neck. Two progenitor populations, the neural crest, and cranial ectodermal placodes, contribute to these developing vertebrate peripheral sensory structures. The interactions and contributions of these cell populations to the development of the lens, olfactory, otic, pituitary gland, and cranial ganglia are vital for appropriate peripheral nervous system development. Here, we review the origins of both neural crest and placode cells at the neural plate border of the early vertebrate embryo and investigate the molecular and environmental signals that influence specification of different sensory regions. Finally, we discuss the underlying molecular pathways contributing to the complex vertebrate sensory system from an evolutionary perspective, from basal vertebrates to amniotes.
Collapse
Affiliation(s)
- Alison Koontz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Hugo A Urrutia
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
6
|
Tan AL, Mohanty S, Guo J, Lekven AC, Riley BB. Pax2a, Sp5a and Sp5l act downstream of Fgf and Wnt to coordinate sensory-neural patterning in the inner ear. Dev Biol 2022; 492:139-153. [PMID: 36244503 DOI: 10.1016/j.ydbio.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/25/2022] [Accepted: 10/10/2022] [Indexed: 01/21/2023]
Abstract
In zebrafish, sensory epithelia and neuroblasts of the inner ear form simultaneously in abutting medial and lateral domains, respectively, in the floor of the otic vesicle. Previous studies support regulatory roles for Fgf and Wnt, but how signaling is coordinated is poorly understood. We investigated this problem using pharmacological and transgenic methods to alter Fgf or Wnt signaling from early placodal stages to evaluate later changes in growth and patterning. Blocking Fgf at any stage reduces proliferation of otic tissue and terminates both sensory and neural specification. Wnt promotes proliferation in the otic vesicle but is not required for sensory or neural development. However, sustained overactivation of Wnt laterally expands sensory epithelia and blocks neurogenesis. pax2a, sp5a and sp5l are coregulated by Fgf and Wnt and show overlapping expression in the otic placode and vesicle. Gain- and loss-of-function studies show that these genes are together required for Wnt's suppression of neurogenesis, as well as some aspects of sensory development. Thus, pax2a, sp5a and sp5l are critical for mediating Fgf and Wnt signaling to promote spatially localized sensory and neural development.
Collapse
Affiliation(s)
- Amy L Tan
- Biology Department, Texas A&M University, College Station, TX, United States
| | - Saurav Mohanty
- Department of Biology and Biochemistry, University of Houston, Houston, TX, United States
| | - Jinbai Guo
- Biology Department, Texas A&M University, College Station, TX, United States
| | - Arne C Lekven
- Department of Biology and Biochemistry, University of Houston, Houston, TX, United States
| | - Bruce B Riley
- Biology Department, Texas A&M University, College Station, TX, United States.
| |
Collapse
|
7
|
Connolly K, Gonzalez-Cordero A. Modelling inner ear development and disease using pluripotent stem cells - a pathway to new therapeutic strategies. Dis Model Mech 2022; 15:dmm049593. [PMID: 36331565 PMCID: PMC10621662 DOI: 10.1242/dmm.049593] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023] Open
Abstract
The sensory epithelia of the mammalian inner ear enable sound and movement to be perceived. Damage to these epithelia can cause irreversible sensorineural hearing loss and vestibular dysfunction because they lack regenerative capacity. The human inner ear cannot be biopsied without causing permanent damage, significantly limiting the tissue samples available for research. Investigating disease pathology and therapeutic developments have therefore traditionally relied on animal models, which often cannot completely recapitulate the human otic systems. These challenges are now being partly addressed using induced pluripotent stem cell-derived cultures, which generate the sensory epithelial-like tissues of the inner ear. Here, we review how pluripotent stem cells have been used to produce two-dimensional and three-dimensional otic cultures, the strengths and limitations of these new approaches, and how they have been employed to investigate genetic and acquired forms of audiovestibular dysfunction. This Review provides an overview of the progress in pluripotent stem cell-derived otic cultures thus far, focusing on their applications in disease modelling and therapeutic trials. We survey their current limitations and future directions, highlighting their prospective utility for high-throughput drug screening and developing personalised medicine approaches.
Collapse
Affiliation(s)
- Keeva Connolly
- Stem Cell Medicine Group, Children's Medical Research Institute, Westmead, 2145 NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, 2145 NSW, Australia
| | - Anai Gonzalez-Cordero
- Stem Cell Medicine Group, Children's Medical Research Institute, Westmead, 2145 NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, 2145 NSW, Australia
| |
Collapse
|
8
|
Abstract
Developing organs are shaped, in part, by physical interaction with their environment in the embryo. In recent years, technical advances in live-cell imaging and material science have greatly expanded our understanding of the mechanical forces driving organ formation. Here, we provide a broad overview of the types of forces generated during embryonic development and then focus on a subset of organs underlying our senses: the eyes, inner ears, nose and skin. The epithelia in these organs emerge from a common origin: the ectoderm germ layer; yet, they arrive at unique and complex forms over developmental time. We discuss exciting recent animal studies that show a crucial role for mechanical forces in, for example, the thickening of sensory placodes, the coiling of the cochlea and the lengthening of hair. Finally, we discuss how microfabricated organoid systems can now provide unprecedented insights into the physical principles of human development.
Collapse
Affiliation(s)
- Anh Phuong Le
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA 02115, USA
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Jin Kim
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA 02115, USA
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Karl R. Koehler
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA 02115, USA
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
9
|
Head B, Traber MG. Expanding role of vitamin E in protection against metabolic dysregulation: Insights gained from model systems, especially the developing nervous system of zebrafish embryos. Free Radic Biol Med 2021; 176:80-91. [PMID: 34555455 DOI: 10.1016/j.freeradbiomed.2021.09.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/27/2021] [Accepted: 09/07/2021] [Indexed: 12/20/2022]
Abstract
This review discusses why the embryo requires vitamin E (VitE) and shows that its lack causes metabolic dysregulation and impacts morphological changes at very early stages in development, which occur prior to when a woman knows she is pregnant. VitE halts the chain reactions of lipid peroxidation (LPO). Metabolomic analyses indicate that thiols become depleted in E- embryos because LPO generates products that require compensation using limited amino acids and methyl donors that are also developmentally relevant. Thus, VitE protects metabolic networks and the integrated gene expression networks that control development. VitE is critical especially for neurodevelopment, which is dependent on trafficking by the α-tocopherol transfer protein (TTPa). VitE-deficient (E-) zebrafish embryos initially appear normal, but by 12 and 24 h post-fertilization (hpf) E- embryos are developmentally abnormal with expression of pax2a and sox10 mis-localized in the midbrain-hindbrain boundary, neural crest cells and throughout the spinal neurons. These patterning defects indicate cells that are especially in need of VitE-protection. They precede obvious morphological abnormalities (cranial-facial malformation, pericardial edema, yolksac edema, skewed body-axis) and impaired behavioral responses to locomotor activity tests. The TTPA gene (ttpa) is expressed at the leading edges of the brain ventricle border. Ttpa knockdown using morpholinos is 100% lethal by 24 hpf, while E- embryo brains are often over- or under-inflated at 24 hpf. Further, E- embryos prior to 24 hpf have increased expression of genes involved in glycolysis and the pentose phosphate pathway, and decreased expression of genes involved in anabolic pathways and transcription. Combined data from both gene expression and the metabolome in E- embryos at 24 hpf suggest that the activity of the mechanistic Target of Rapamycin (mTOR) signaling pathway is decreased, which may impact both metabolism and neurodevelopment. Further evaluation of VitE deficiency in neurogenesis and its subsequent impact on learning and behavior is needed.
Collapse
Affiliation(s)
- Brian Head
- Linus Pauling Institute, Corvallis, OR, USA; Molecular and Cell Biology Program, Corvallis, OR, USA
| | - Maret G Traber
- Linus Pauling Institute, Corvallis, OR, USA; School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, USA.
| |
Collapse
|
10
|
Riley BB. Comparative assessment of Fgf's diverse roles in inner ear development: A zebrafish perspective. Dev Dyn 2021; 250:1524-1551. [PMID: 33830554 DOI: 10.1002/dvdy.343] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/26/2021] [Accepted: 03/26/2021] [Indexed: 01/21/2023] Open
Abstract
Progress in understanding mechanisms of inner ear development has been remarkably rapid in recent years. The research community has benefited from the availability of several diverse model organisms, including zebrafish, chick, and mouse. The complexity of the inner ear has proven to be a challenge, and the complexity of the mammalian cochlea in particular has been the subject of intense scrutiny. Zebrafish lack a cochlea and exhibit a number of other differences from amniote species, hence they are sometimes seen as less relevant for inner ear studies. However, accumulating evidence shows that underlying cellular and molecular mechanisms are often highly conserved. As a case in point, consideration of the diverse functions of Fgf and its downstream effectors reveals many similarities between vertebrate species, allowing meaningful comparisons the can benefit the entire research community. In this review, I will discuss mechanisms by which Fgf controls key events in early otic development in zebrafish and provide direct comparisons with chick and mouse.
Collapse
Affiliation(s)
- Bruce B Riley
- Biology Department, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
11
|
Mackowetzky K, Yoon KH, Mackowetzky EJ, Waskiewicz AJ. Development and evolution of the vestibular apparatuses of the inner ear. J Anat 2021; 239:801-828. [PMID: 34047378 PMCID: PMC8450482 DOI: 10.1111/joa.13459] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/07/2021] [Accepted: 05/06/2021] [Indexed: 12/16/2022] Open
Abstract
The vertebrate inner ear is a labyrinthine sensory organ responsible for perceiving sound and body motion. While a great deal of research has been invested in understanding the auditory system, a growing body of work has begun to delineate the complex developmental program behind the apparatuses of the inner ear involved with vestibular function. These animal studies have helped identify genes involved in inner ear development and model syndromes known to include vestibular dysfunction, paving the way for generating treatments for people suffering from these disorders. This review will provide an overview of known inner ear anatomy and function and summarize the exciting discoveries behind inner ear development and the evolution of its vestibular apparatuses.
Collapse
Affiliation(s)
- Kacey Mackowetzky
- Department of Biological SciencesUniversity of AlbertaEdmontonAlbertaCanada
| | - Kevin H. Yoon
- Department of Biological SciencesUniversity of AlbertaEdmontonAlbertaCanada
| | | | - Andrew J. Waskiewicz
- Department of Biological SciencesUniversity of AlbertaEdmontonAlbertaCanada
- Women & Children’s Health Research InstituteUniversity of AlbertaEdmontonAlbertaCanada
| |
Collapse
|
12
|
Wang J, Lu C, Zhao Y, Tang Z, Song J, Fan C. Transcriptome profiles of sturgeon lateral line electroreceptor and mechanoreceptor during regeneration. BMC Genomics 2020; 21:875. [PMID: 33287707 PMCID: PMC7720607 DOI: 10.1186/s12864-020-07293-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 11/28/2020] [Indexed: 11/10/2022] Open
Abstract
Background The electrosensory ampullary organs (AOs) and mechanosensory neuromasts (NMs) found in sturgeon and some other non-neopterygian fish or amphibians are both originated from lateral line placodes. However, these two sensory organs have characteristic morphological and physiological differences. The molecular mechanisms for the specification of AOs and NMs are not clearly understood. Results We sequenced the transcriptome for neomycin treated sturgeon AOs and NMs in the early regeneration stages, and de novo assembled a sturgeon transcriptome. By comparing the gene expression differences among untreated AOs, NMs and general epithelia (EPs), we located some specific genes for these two sensory organs. In sturgeon lateral line, the voltage-gated calcium channels and voltage-gated potassium channels were predominant calcium and potassium channel subtypes, respectively. And by correlating gene expression with the regeneration process, we predicated several candidate key transcriptional regulation related genes might be involved in AOs and NMs regeneration. Conclusions Genes with specific expression in the two lateral line sensory organs suggests their important roles in mechanoreceptor and electroreceptor formation. The candidate transcriptional regulation related genes may be important for mechano- and electro- receptor specification, in a “dosage-related” manner. These results suggested the molecular basis for specification of these two sensory organs in sturgeon.
Collapse
Affiliation(s)
- Jian Wang
- International Joint Center for Marine Biological Sciences Research, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China.,Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
| | - Chengcheng Lu
- International Joint Center for Marine Biological Sciences Research, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China.,Institute for Marine Biosystem and Neuroscience, International Center for Marine Studies, Shanghai Ocean University, Shanghai, China
| | - Yifan Zhao
- International Joint Center for Marine Biological Sciences Research, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China.,Institute for Marine Biosystem and Neuroscience, International Center for Marine Studies, Shanghai Ocean University, Shanghai, China
| | - Zhijiao Tang
- Institute for Marine Biosystem and Neuroscience, International Center for Marine Studies, Shanghai Ocean University, Shanghai, China
| | - Jiakun Song
- Institute for Marine Biosystem and Neuroscience, International Center for Marine Studies, Shanghai Ocean University, Shanghai, China
| | - Chunxin Fan
- International Joint Center for Marine Biological Sciences Research, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China. .,Institute for Marine Biosystem and Neuroscience, International Center for Marine Studies, Shanghai Ocean University, Shanghai, China.
| |
Collapse
|
13
|
Pax2a is expressed in oocytes and is responsible for early development and oogenesis in zebrafish. Biochem Biophys Res Commun 2020; 533:592-599. [PMID: 32981680 DOI: 10.1016/j.bbrc.2020.09.059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 09/15/2020] [Indexed: 11/23/2022]
Abstract
Eleven genes, including pax2a, were selected as candidate ovulation-inducing genes on the basis of microarray analysis and RNA sequencing in our previous study. The purpose of this study was to investigate the role of the pax2a gene in the ovulation-inducing process. F2 pax2a homozygous mutant zebrafish possessing a deletion of 6 nucleotides were established in this study. However, the deletion included the start codon (ATG) of the pax2a gene, and the Pax2a protein was still detected, which indicated that the deletion caused a shift in the start codon to the next ATG, resulting in a 12-amino acid deletion. F2 pax2a homozygous mutant zebrafish showed ovulation. However, the embryos showed an abnormal oval shape at the epiboly stage that resulted in yolk and tail formation abnormalities and heart edema. The surviving F3 homozygous mutants did not develop ovaries. Pax2a was detected in oocytes and eggs but not after the Prim-22 stage. It is suggested that pax2a is expressed as a maternal gene in oocytes and is necessary for oogenesis and early development.
Collapse
|
14
|
Vitamin E is necessary for zebrafish nervous system development. Sci Rep 2020; 10:15028. [PMID: 32958954 PMCID: PMC7506018 DOI: 10.1038/s41598-020-71760-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022] Open
Abstract
Vitamin E (VitE) deficiency results in embryonic lethality. Knockdown of the gene ttpa encoding for the VitE regulatory protein [α-tocopherol transfer protein (α-TTP)] in zebrafish embryos causes death within 24 h post-fertilization (hpf). To test the hypothesis that VitE, not just α-TTP, is necessary for nervous system development, adult 5D strain zebrafish, fed either VitE sufficient (E+) or deficient (E-) diets, were spawned to obtain E+ and E- embryos, which were subjected to RNA in situ hybridization and RT-qPCR. Ttpa was expressed ubiquitously in embryos up to 12 hpf. Early gastrulation (6 hpf) assessed by goosecoid expression was unaffected by VitE status. By 24 hpf, embryos expressed ttpa in brain ventricle borders, which showed abnormal closure in E- embryos. They also displayed disrupted patterns of paired box 2a (pax2a) and SRY-box transcription factor 10 (sox10) expression in the midbrain-hindbrain boundary, spinal cord and dorsal root ganglia. In E- embryos, the collagen sheath notochord markers (col2a1a and col9a2) appeared bent. Severe developmental errors in E- embryos were characterized by improper nervous system patterning of the usually carefully programmed transcriptional signals. Histological analysis also showed developmental defects in the formation of the fore-, mid- and hindbrain and somites of E- embryos at 24 hpf. Ttpa expression profile was not altered by the VitE status demonstrating that VitE itself, and not ttpa, is required for development of the brain and peripheral nervous system in this vertebrate embryo model.
Collapse
|
15
|
Heuer RA, Nella KT, Chang HT, Coots KS, Oleksijew AM, Roque CB, Silva LHA, McGuire TL, Homma K, Matsuoka AJ. Three-Dimensional Otic Neuronal Progenitor Spheroids Derived from Human Embryonic Stem Cells. Tissue Eng Part A 2020; 27:256-269. [PMID: 32580647 DOI: 10.1089/ten.tea.2020.0078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Stem cell-replacement therapies have been proposed as a potential tool to treat sensorineural hearing loss by aiding the regeneration of spiral ganglion neurons (SGNs) in the inner ear. However, transplantation procedures have yet to be explored thoroughly to ensure proper cell differentiation and optimal transplant procedures. We hypothesized that the aggregation of human embryonic stem cell (hESC)-derived otic neuronal progenitor (ONP) cells into a multicellular form would improve their function and their survival in vivo post-transplantation. We generated hESC-derived ONP spheroids-an aggregate form conducive to differentiation, transplantation, and prolonged cell survival-to optimize conditions for their transplantation. Our findings indicate that these cell spheroids maintain the molecular and functional characteristics similar to those of ONP cells, which are upstream in the SGN lineage. Moreover, our phenotypical, electrophysiological, and mechanical data suggest an optimal spheroid transplantation point after 7 days of in vitro three-dimensional (3D) culture. We have also developed a feasible transplantation protocol for these spheroids using a micropipette aided by a digital microinjection system. In summary, the present work demonstrates that the transplantation of ONP cells in spheroid form into the inner ear through micropipette 7 days after seeding for 3D spheroid culture is an expedient and viable method for stem cell replacement therapies in the inner ear.
Collapse
Affiliation(s)
- Rachel A Heuer
- Department of Otolaryngology and Head and Neck Surgery and Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Kevin T Nella
- Department of Otolaryngology and Head and Neck Surgery and Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Hsiang-Tsun Chang
- Department of Otolaryngology and Head and Neck Surgery and Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Kyle S Coots
- Department of Otolaryngology and Head and Neck Surgery and Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Andrew M Oleksijew
- Department of Otolaryngology and Head and Neck Surgery and Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Christian B Roque
- Department of Otolaryngology and Head and Neck Surgery and Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Luisa H A Silva
- Department of Otolaryngology and Head and Neck Surgery and Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Tammy L McGuire
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Kazuaki Homma
- Department of Otolaryngology and Head and Neck Surgery and Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.,Hugh Knowles Center for Hearing Research and Northwestern University, Evanston, Illinois, USA
| | - Akihiro J Matsuoka
- Department of Otolaryngology and Head and Neck Surgery and Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.,Hugh Knowles Center for Hearing Research and Northwestern University, Evanston, Illinois, USA.,Department of Communication Sciences and Disorders, Northwestern University, Evanston, Illinois, USA
| |
Collapse
|
16
|
Cell fate decisions during the development of the peripheral nervous system in the vertebrate head. Curr Top Dev Biol 2020; 139:127-167. [PMID: 32450959 DOI: 10.1016/bs.ctdb.2020.04.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Sensory placodes and neural crest cells are among the key cell populations that facilitated the emergence and diversification of vertebrates throughout evolution. Together, they generate the sensory nervous system in the head: both form the cranial sensory ganglia, while placodal cells make major contributions to the sense organs-the eye, ear and olfactory epithelium. Both are instrumental for integrating craniofacial organs and have been key to drive the concentration of sensory structures in the vertebrate head allowing the emergence of active and predatory life forms. Whereas the gene regulatory networks that control neural crest cell development have been studied extensively, the signals and downstream transcriptional events that regulate placode formation and diversity are only beginning to be uncovered. Both cell populations are derived from the embryonic ectoderm, which also generates the central nervous system and the epidermis, and recent evidence suggests that their initial specification involves a common molecular mechanism before definitive neural, neural crest and placodal lineages are established. In this review, we will first discuss the transcriptional networks that pattern the embryonic ectoderm and establish these three cell fates with emphasis on sensory placodes. Second, we will focus on how sensory placode precursors diversify using the specification of otic-epibranchial progenitors and their segregation as an example.
Collapse
|
17
|
Chang HT, Heuer RA, Oleksijew AM, Coots KS, Roque CB, Nella KT, McGuire TL, Matsuoka AJ. An engineered three-dimensional stem cell niche in the inner ear by applying a nanofibrillar cellulose hydrogel with a sustained-release neurotrophic factor delivery system. Acta Biomater 2020; 108:111-127. [PMID: 32156626 PMCID: PMC7198367 DOI: 10.1016/j.actbio.2020.03.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 02/01/2020] [Accepted: 03/03/2020] [Indexed: 11/17/2022]
Abstract
Although the application of human embryonic stem cells (hESCs) in stem cell-replacement therapy remains promising, its potential is hindered by a low cell survival rate in post-transplantation within the inner ear. Here, we aim to enhance the in vitro and in vivo survival rate and neuronal differentiation of otic neuronal progenitors (ONPs) by generating an artificial stem cell niche consisting of three-dimensional (3D) hESC-derived ONP spheroids with a nanofibrillar cellulose hydrogel and a sustained-release brain-derivative neurotrophic factor delivery system. Our results demonstrated that the transplanted hESC-derived ONP spheroids survived and neuronally differentiated into otic neuronal lineages in vitro and in vivo and also extended neurites toward the bony wall of the cochlea 90 days after the transplantation without the use of immunosuppressant medication. Our data in vitro and in vivo presented here provide sufficient evidence that we have established a robust, reproducible protocol for in vivo transplantation of hESC-derived ONPs to the inner ear. Using our protocol to create an artificial stem cell niche in the inner ear, it is now possible to work on integrating transplanted hESC-derived ONPs further and also to work toward achieving functional auditory neurons generated from hESCs. Our findings suggest that the provision of an artificial stem cell niche can be a future approach to stem cell-replacement therapy for inner-ear regeneration. STATEMENT OF SIGNIFICANCE: Inner ear regeneration utilizing human embryonic stem cell-derived otic neuronal progenitors (hESC-derived ONPs) has remarkable potential for treating sensorineural hearing loss. However, the local environment of the inner ear requires a suitable stem cell niche to allow hESC-derived ONP engraftment as well as neuronal differentiation. To overcome this obstacle, we utilized three-dimensional spheroid formation (direct contact), nanofibrillar cellulose hydrogel (extracellular matrix), and a neurotrophic factor delivery system to artificially create a stem cell niche in vitro and in vivo. Our in vitro and in vivo data presented here provide sufficient evidence that we have established a robust, reproducible protocol for in vivo transplantation of hESC-derived ONPs to the inner ear.
Collapse
Affiliation(s)
- Hsiang-Tsun Chang
- Department of Otolaryngology and Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Rachel A Heuer
- Department of Otolaryngology and Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Andrew M Oleksijew
- Department of Otolaryngology and Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Kyle S Coots
- Department of Otolaryngology and Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Christian B Roque
- Department of Otolaryngology and Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Kevin T Nella
- Department of Otolaryngology and Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Tammy L McGuire
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago IL 60611, USA
| | - Akihiro J Matsuoka
- Department of Otolaryngology and Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL 60201, USA; Hugh Knowles Center for Hearing Research, Northwestern University, Evanston, IL 60201, USA.
| |
Collapse
|
18
|
Wang L, Xie J, Zhang H, Tsang LH, Tsang SL, Braune EB, Lendahl U, Sham MH. Notch signalling regulates epibranchial placode patterning and segregation. Development 2020; 147:dev.183665. [PMID: 31988190 PMCID: PMC7044445 DOI: 10.1242/dev.183665] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 01/14/2020] [Indexed: 11/20/2022]
Abstract
Epibranchial placodes are the geniculate, petrosal and nodose placodes that generate parts of cranial nerves VII, IX and X, respectively. How the three spatially separated placodes are derived from the common posterior placodal area is poorly understood. Here, we reveal that the broad posterior placode area is first patterned into a Vgll2+/Irx5+ rostral domain and a Sox2+/Fgf3+/Etv5+ caudal domain relative to the first pharyngeal cleft. This initial rostral and caudal patterning is then sequentially repeated along each pharyngeal cleft for each epibranchial placode. The caudal domains give rise to the neuronal and non-neuronal cells in the placode, whereas the rostral domains are previously unrecognized structures, serving as spacers between the final placodes. Notch signalling regulates the balance between the rostral and caudal domains: high levels of Notch signalling expand the caudal domain at the expense of the rostral domain, whereas loss of Notch signalling produces the converse phenotype. Collectively, these data unravel a new patterning principle for the early phases of epibranchial placode development and a role for Notch signalling in orchestrating epibranchial placode segregation and differentiation.
Collapse
Affiliation(s)
- Li Wang
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Junjie Xie
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Haoran Zhang
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Long Hin Tsang
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Sze Lan Tsang
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Eike-Benjamin Braune
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Urban Lendahl
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Mai Har Sham
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| |
Collapse
|
19
|
Cao M, Ouyang J, Liang H, Guo J, Lin S, Yang S, Xie T, Chen S. Regional Gene Expression Profile Comparison Reveals the Unique Transcriptome of the Optic Fissure. Invest Ophthalmol Vis Sci 2019; 59:5773-5784. [PMID: 30521666 DOI: 10.1167/iovs.18-23962] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The optic fissure (OF) is a transient opening in the ventral optic cup (OC) that acts as a passage for blood vessels and retinal ganglion cell axons during early eye development. Failure to close the OF is the developmental basis for uveal coloboma, a congenital blinding eye disease that significantly contributes to childhood blindness. Genes specifically expressed in the OF region may play important roles in OF development and function. The aim of this study was to characterize the transcriptome of OC cells in the OF region and investigate the function of OF-specific genes during OF closure. Methods Laser-assisted microdissection was used to collect different regions of OC tissues. Microarray analysis was used to obtain and compare gene expression profiles of different OC regions. RNA in situ hybridization (ISH) was used to further characterize OF-specific gene expression patterns. Morpholino knockdown in zebrafish was used to study the function of a newly discovered OF-specific gene during OF closure. Results Microarray comparison revealed that the OC at the OF region exhibited a unique gene expression profile. OC expression patterns of a number of newly discovered OF-specific genes were confirmed by ISH. Morpholino knockdown and downstream target expression and function analysis demonstrated that afap1l2, a newly discovered OF-specific gene, controls OF closure by regulating pax2a expression. Conclusions Our study characterized the unique transcriptome of the OF region of the OC and demonstrated the essential role of a newly discovered OF-specific gene in OF closure. This study provides a valuable foundation for future mechanism dissection in OF development and physiology, and for human coloboma etiology exploration.
Collapse
Affiliation(s)
- Mingzhe Cao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jiamin Ouyang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Huilin Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jingyi Guo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Siyuan Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Shulan Yang
- Translational Medicine Centre, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ting Xie
- Stowers Institute for Medical Research, Kansas City, Missouri, United States
| | - Shuyi Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
20
|
Lu M, Guo S, Hong F, Zhang Y, Yuan L, Ma C, Ma J. Pax2 is essential for proliferation and osteogenic differentiation of mouse mesenchymal stem cells via Runx2. Exp Cell Res 2018; 371:342-352. [DOI: 10.1016/j.yexcr.2018.08.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 08/19/2018] [Accepted: 08/21/2018] [Indexed: 01/09/2023]
|
21
|
Olson HM, Nechiporuk AV. Using Zebrafish to Study Collective Cell Migration in Development and Disease. Front Cell Dev Biol 2018; 6:83. [PMID: 30175096 PMCID: PMC6107837 DOI: 10.3389/fcell.2018.00083] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 07/16/2018] [Indexed: 12/24/2022] Open
Abstract
Cellular migration is necessary for proper embryonic development as well as maintenance of adult health. Cells can migrate individually or in groups in a process known as collective cell migration. Collectively migrating cohorts maintain cell-cell contacts, group polarization, and exhibit coordinated behavior. This mode of migration is important during numerous developmental processes including tracheal branching, blood vessel sprouting, neural crest cell migration and others. In the adult, collective cell migration is important for proper wound healing and is often misappropriated during cancer cell invasion. A variety of genetic model systems are used to examine and define the cellular and molecular mechanisms behind collective cell migration including border cell migration and tracheal branching in Drosophila melanogaster, neural crest cell migration in chick and Xenopus embryos, and posterior lateral line primordium (pLLP) migration in zebrafish. The pLLP is a group of about 100 cells that begins migrating around 22 hours post-fertilization along the lateral aspect of the trunk of the developing embryo. During migration, clusters of cells are deposited from the trailing end of the pLLP; these ultimately differentiate into mechanosensory organs of the lateral line system. As zebrafish embryos are transparent during early development and the pLLP migrates close to the surface of the skin, this system can be easily visualized and manipulated in vivo. These advantages together with the amenity to advance genetic methods make the zebrafish pLLP one of the premier model systems for studying collective cell migration. This review will describe the cellular behaviors and signaling mechanisms of the pLLP and compare the pLLP to collective cell migration in other popular model systems. In addition, we will examine how this type of migration is hijacked by collectively invading cancer cells.
Collapse
Affiliation(s)
- Hannah M Olson
- Department Cell, Developmental & Cancer Biology, The Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States.,Neuroscience Graduate Program, Oregon Health & Science University, Portland, OR, United States
| | - Alex V Nechiporuk
- Department Cell, Developmental & Cancer Biology, The Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
22
|
Lahlou H, Lopez-Juarez A, Fontbonne A, Nivet E, Zine A. Modeling human early otic sensory cell development with induced pluripotent stem cells. PLoS One 2018; 13:e0198954. [PMID: 29902227 PMCID: PMC6002076 DOI: 10.1371/journal.pone.0198954] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 05/24/2018] [Indexed: 11/18/2022] Open
Abstract
The inner ear represents a promising system to develop cell-based therapies from human induced pluripotent stem cells (hiPSCs). In the developing ear, Notch signaling plays multiple roles in otic region specification and for cell fate determination. Optimizing hiPSC induction for the generation of appropriate numbers of otic progenitors and derivatives, such as hair cells, may provide an unlimited supply of cells for research and cell-based therapy. In this study, we used monolayer cultures, otic-inducing agents, Notch modulation, and marker expression to track early and otic sensory lineages during hiPSC differentiation. Otic/placodal progenitors were derived from hiPSC cultures in medium supplemented with FGF3/FGF10 for 13 days. These progenitor cells were then treated for 7 days with retinoic acid (RA) and epidermal growth factor (EGF) or a Notch inhibitor. The differentiated cultures were analyzed in parallel by qPCR and immunocytochemistry. After the 13 day induction, hiPSC-derived cells displayed an upregulated expression of a panel of otic/placodal markers. Strikingly, a subset of these induced progenitor cells displayed key-otic sensory markers, the percentage of which was increased in cultures under Notch inhibition as compared to RA/EGF-treated cultures. Our results show that modulating Notch pathway during in vitro differentiation of hiPSC-derived otic/placodal progenitors is a valuable strategy to promote the expression of human otic sensory lineage genes.
Collapse
Affiliation(s)
- Hanae Lahlou
- Aix Marseille Université, CNRS, LNIA UMR 7260, Marseille, France
| | | | - Arnaud Fontbonne
- Aix Marseille Université, CNRS, LNIA UMR 7260, Marseille, France
| | - Emmanuel Nivet
- Aix Marseille Université, CNRS, NICN UMR 7259, Marseille, France
| | - Azel Zine
- Aix Marseille Université, CNRS, LNIA UMR 7260, Marseille, France
- Université de Montpellier, Faculté de Pharmacie, Montpellier, France
- * E-mail: ,
| |
Collapse
|
23
|
Sato S, Furuta Y, Kawakami K. Regulation of continuous but complex expression pattern of Six1 during early sensory development. Dev Dyn 2017; 247:250-261. [PMID: 29106072 DOI: 10.1002/dvdy.24603] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 10/17/2017] [Accepted: 10/31/2017] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND In vertebrates, cranial sensory placodes give rise to neurosensory and endocrine structures, such as the olfactory epithelium, inner ear, and anterior pituitary. We report here the establishment of a transgenic mouse line that expresses Cre recombinase under the control of Six1-21, a major placodal enhancer of the homeobox gene Six1. RESULTS In the new Cre-expressing line, mSix1-21-NLSCre, the earliest Cre-mediated recombination was induced at embryonic day 8.5 in the region overlapping with the otic-epibranchial progenitor domain (OEPD), a transient, common precursor domain for the otic and epibranchial placodes. Recombination was later observed in the OEPD-derived structures (the entire inner ear and the VIIth-Xth cranial sensory ganglia), olfactory epithelium, anterior pituitary, pharyngeal ectoderm and pouches. Other Six1-positive structures, such as salivary/lacrimal glands and limb buds, were also positive for recombination. Moreover, comparison with another mouse line expressing Cre under the control of the sensory neuron enhancer, Six1-8, indicated that the continuous and complex expression pattern of Six1 during sensory organ formation is pieced together by separate enhancers. CONCLUSIONS mSix1-21-NLSCre has several unique characteristics to make it suitable for analysis of cell lineage and gene function in sensory placodes as well as nonplacodal Six1-positive structures. Developmental Dynamics 247:250-261, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Shigeru Sato
- Division of Biology, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Yasuhide Furuta
- Animal Resource Development Unit and Genetic Engineering Team, Division of Bio-function Dynamics Imaging, RIKEN Center for Life Science Technologies (CLST), Kobe, Hyogo, Japan
| | - Kiyoshi Kawakami
- Division of Biology, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| |
Collapse
|
24
|
Schwarzer S, Spieß S, Brand M, Hans S. Dlx3b/4b is required for early-born but not later-forming sensory hair cells during zebrafish inner ear development. Biol Open 2017; 6:1270-1278. [PMID: 28751305 PMCID: PMC5612237 DOI: 10.1242/bio.026211] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Morpholino-mediated knockdown has shown that the homeodomain transcription factors Dlx3b and Dlx4b are essential for proper induction of the otic-epibranchial progenitor domain (OEPD), as well as subsequent formation of sensory hair cells in the developing zebrafish inner ear. However, increasing use of reverse genetic approaches has revealed poor correlation between morpholino-induced and mutant phenotypes. Using CRISPR/Cas9-mediated mutagenesis, we generated a defined deletion eliminating the entire open reading frames of dlx3b and dlx4b (dlx3b/4b) and investigated a potential phenotypic difference between mutants and morpholino-mediated knockdown. Consistent with previous findings obtained by morpholino-mediated knockdown of Dlx3b and Dlx4b, dlx3b/4b mutants display compromised otic induction, the development of smaller otic vesicles and an elimination of all indications of otic specification when combined with loss of foxi1, a second known OEPD competence factor in zebrafish. Furthermore, sensorigenesis is also affected in dlx3b/4b mutants. However, we find that only early-born sensory hair cells (tether cells), that seed and anchor the formation of otoliths, are affected. Later-forming sensory hair cells are present, indicating that two genetically distinct pathways control the development of early-born and later-forming sensory hair cells. Finally, impairment of early-born sensory hair cell formation in dlx3b/4b mutant embryos reverses the common temporal sequence of neuronal and sensory hair cell specification in zebrafish, resembling the order of cell specification in amniotes; Neurog1 expression before Atoh1 expression. We conclude that the Dlx3b/4b-dependent pathway has been either acquired newly in the fish lineage or lost in other vertebrate species during evolution, and that the events during early inner ear development are remarkably similar in fish and amniotes in the absence of this pathway. Summary: The transcription factors Dlx3b and Dlx4b control the formation of early-born sensory hair cells or tether cells in the developing zebrafish inner ear.
Collapse
Affiliation(s)
- Simone Schwarzer
- Technische Universität Dresden, Biotechnology Center and DFG-Center for Regenerative Therapies Dresden Cluster of Excellence, Tatzberg 47-49, 01307 Dresden, Germany
| | - Sandra Spieß
- Technische Universität Dresden, Biotechnology Center and DFG-Center for Regenerative Therapies Dresden Cluster of Excellence, Tatzberg 47-49, 01307 Dresden, Germany
| | - Michael Brand
- Technische Universität Dresden, Biotechnology Center and DFG-Center for Regenerative Therapies Dresden Cluster of Excellence, Tatzberg 47-49, 01307 Dresden, Germany
| | - Stefan Hans
- Technische Universität Dresden, Biotechnology Center and DFG-Center for Regenerative Therapies Dresden Cluster of Excellence, Tatzberg 47-49, 01307 Dresden, Germany
| |
Collapse
|
25
|
A gene network regulated by FGF signalling during ear development. Sci Rep 2017; 7:6162. [PMID: 28733657 DOI: 10.1038/s41598-017-05472-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 05/31/2017] [Indexed: 02/08/2023] Open
Abstract
During development cell commitment is regulated by inductive signals that are tightly controlled in time and space. In response, cells activate specific programmes, but the transcriptional circuits that maintain cell identity in a changing signalling environment are often poorly understood. Specification of inner ear progenitors is initiated by FGF signalling. Here, we establish the genetic hierarchy downstream of FGF by systematic analysis of many ear factors combined with a network inference approach. We show that FGF rapidly activates a small circuit of transcription factors forming positive feedback loops to stabilise otic progenitor identity. Our predictive network suggests that subsequently, transcriptional repressors ensure the transition of progenitors to mature otic cells, while simultaneously repressing alternative fates. Thus, we reveal the regulatory logic that initiates ear formation and highlight the hierarchical organisation of the otic gene network.
Collapse
|
26
|
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
|
27
|
Ladher RK. Changing shape and shaping change: Inducing the inner ear. Semin Cell Dev Biol 2017; 65:39-46. [DOI: 10.1016/j.semcdb.2016.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 10/20/2016] [Accepted: 10/25/2016] [Indexed: 12/21/2022]
|
28
|
Sculpting the labyrinth: Morphogenesis of the developing inner ear. Semin Cell Dev Biol 2017; 65:47-59. [DOI: 10.1016/j.semcdb.2016.09.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 07/26/2016] [Accepted: 09/25/2016] [Indexed: 01/23/2023]
|
29
|
Matsuoka AJ, Morrissey ZD, Zhang C, Homma K, Belmadani A, Miller CA, Chadly DM, Kobayashi S, Edelbrock AN, Tanaka‐Matakatsu M, Whitlon DS, Lyass L, McGuire TL, Stupp SI, Kessler JA. Directed Differentiation of Human Embryonic Stem Cells Toward Placode-Derived Spiral Ganglion-Like Sensory Neurons. Stem Cells Transl Med 2017; 6:923-936. [PMID: 28186679 PMCID: PMC5442760 DOI: 10.1002/sctm.16-0032] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 08/31/2016] [Accepted: 10/19/2016] [Indexed: 12/15/2022] Open
Abstract
The ability to generate spiral ganglion neurons (SGNs) from stem cells is a necessary prerequisite for development of cell-replacement therapies for sensorineural hearing loss. We present a protocol that directs human embryonic stem cells (hESCs) toward a purified population of otic neuronal progenitors (ONPs) and SGN-like cells. Between 82% and 95% of these cells express SGN molecular markers, they preferentially extend neurites to the cochlear nucleus rather than nonauditory nuclei, and they generate action potentials. The protocol follows an in vitro stepwise recapitulation of developmental events inherent to normal differentiation of hESCs into SGNs, resulting in efficient sequential generation of nonneuronal ectoderm, preplacodal ectoderm, early prosensory ONPs, late ONPs, and cells with cellular and molecular characteristics of human SGNs. We thus describe the sequential signaling pathways that generate the early and later lineage species in the human SGN lineage, thereby better describing key developmental processes. The results indicate that our protocol generates cells that closely replicate the phenotypic characteristics of human SGNs, advancing the process of guiding hESCs to states serving inner-ear cell-replacement therapies and possible next-generation hybrid auditory prostheses. © Stem Cells Translational Medicine 2017;6:923-936.
Collapse
Affiliation(s)
- Akihiro J. Matsuoka
- Department of Otolaryngology and Head and Neck SurgeryChicagoILUSA
- Department of Communication Sciences and DisordersChicagoILUSA
- Knowles Hearing CenterChicagoILUSA
| | | | - Chaoying Zhang
- Department of Otolaryngology and Head and Neck SurgeryChicagoILUSA
| | - Kazuaki Homma
- Department of Otolaryngology and Head and Neck SurgeryChicagoILUSA
- Knowles Hearing CenterChicagoILUSA
| | - Abdelhak Belmadani
- Department of Molecular Pharmacology and Biological ChemistryChicagoILUSA
| | | | - Duncan M. Chadly
- Department of Otolaryngology and Head and Neck SurgeryChicagoILUSA
| | - Shun Kobayashi
- Department of Otolaryngology and Head and Neck SurgeryChicagoILUSA
| | | | | | - Donna S. Whitlon
- Department of Otolaryngology and Head and Neck SurgeryChicagoILUSA
- Knowles Hearing CenterChicagoILUSA
| | - Ljuba Lyass
- Department of Biomedical EngineeringChicagoILUSA
| | | | - Samuel I. Stupp
- Department of MedicineChicagoILUSA
- Department of Biomedical EngineeringChicagoILUSA
- Simpson Querrey Institute for BioNanotechnologyChicagoILUSA
- Department of ChemistryNorthwestern University
- Department of Materials Science & EngineeringNorthwestern University
| | - John A. Kessler
- Department of NeurologyFeinberg School of Medicine, Northwestern UniversityChicagoILUSA
| |
Collapse
|
30
|
Washausen S, Knabe W. Pax2/Pax8-defined subdomains and the occurrence of apoptosis in the posterior placodal area of mice. Brain Struct Funct 2017; 222:2671-2695. [PMID: 28160066 DOI: 10.1007/s00429-016-1364-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 12/28/2016] [Indexed: 11/28/2022]
Abstract
The present work aims to improve our understanding of the causes and functions of apoptosis during the morphogenesis of epibranchial placodes in mice. Schematic maps helped to compare the spatiotemporal sequence of apoptotic events with the protein expression patterns of general (Six1) and specific placodal markers (Pax2, Pax8). Our findings challenge the view that, in mammals, all three epibranchial placodes spring from the original posterior placodal area (PPA) of presomite or early somite embryos. Instead, close-meshed analysis of the Pax2/Pax8 expression patterns demonstrates the stepwise emergence of two subdomains which both belong to the gradually expanding PPA, and which largely give rise to the otic placode and epibranchial placode 1 (anterior subdomain), or to the caudal epibranchial placodes (posterior subdomain). Our observations reinforce previous doubts raised on the PPA progeny of early somite Xenopus embryos (Schlosser and Ahrens, Dev Biol 271:439-466, 2004). They also demonstrate that partly different Pax2/Pax8 codes accompany epibranchial placode development in Xenopus laevis and mice. In mice, interplacodal apoptosis assists in the establishment of the two PPA subdomains and, subsequently, of individualized placodes by predominantly eliminating Six1+ placodal precursor cells. Onset of interplacodal and intraplacodal large-scale apoptosis is almost always preceded and/or paralleled by Pax2/Pax8 expression minima in the very same region. Future work will demand the use of knock-out mice and whole embryo culture to experimentally test, whether the combined action of differentially expressed Pax2 and Pax8 genes exerts antiapoptotic effects in the mammalian PPA.
Collapse
Affiliation(s)
- Stefan Washausen
- Department Prosektur Anatomie, Westfälische Wilhelms-University, Vesaliusweg 2-4, 48149, Münster, Germany
| | - Wolfgang Knabe
- Department Prosektur Anatomie, Westfälische Wilhelms-University, Vesaliusweg 2-4, 48149, Münster, Germany.
| |
Collapse
|
31
|
Nie J, Koehler KR, Hashino E. Directed Differentiation of Mouse Embryonic Stem Cells Into Inner Ear Sensory Epithelia in 3D Culture. Methods Mol Biol 2017; 1597:67-83. [PMID: 28361311 DOI: 10.1007/978-1-4939-6949-4_6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The inner ear sensory epithelium harbors mechanosensory hair cells responsible for detecting sound and maintaining balance. This protocol describes a three-dimensional (3D) culture system that efficiently generates inner ear sensory epithelia from aggregates of mouse embryonic stem (mES) cells. By mimicking the activations and repressions of key signaling pathways during in vivo inner ear development, mES cell aggregates are sequentially treated with recombinant proteins and small molecule inhibitors for activating or inhibiting the Bmp, TGFβ, Fgf, and Wnt signaling pathways. These stepwise treatments promote mES cells to sequentially differentiate into epithelia representing the non-neural ectoderm, preplacodal ectoderm, otic placodal ectoderm, and ultimately, the hair cell-containing sensory epithelia. The derived hair cells are surrounded by a layer of supporting cells and are innervated by sensory neurons. This in vitro inner ear organoid culture system may serve as a valuable tool in developmental and physiological research, disease modeling, drug testing, and potential cell-based therapies.
Collapse
Affiliation(s)
- Jing Nie
- Department of Otolaryngology-Head and Neck Surgery, and Stark Neurosciences Research Institute, Indiana University School of Medicine, 980 W. Walnut St., Indianapolis, IN, 46202, USA
| | - Karl R Koehler
- Department of Otolaryngology-Head and Neck Surgery, and Stark Neurosciences Research Institute, Indiana University School of Medicine, 980 W. Walnut St., Indianapolis, IN, 46202, USA
| | - Eri Hashino
- Department of Otolaryngology-Head and Neck Surgery, and Stark Neurosciences Research Institute, Indiana University School of Medicine, 980 W. Walnut St., Indianapolis, IN, 46202, USA.
| |
Collapse
|
32
|
Steventon B, Mayor R, Streit A. Directional cell movements downstream of Gbx2 and Otx2 control the assembly of sensory placodes. Biol Open 2016; 5:1620-1624. [PMID: 27659690 PMCID: PMC5155537 DOI: 10.1242/bio.020966] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Cranial placodes contribute to sensory structures including the inner ear, the lens and olfactory epithelium and the neurons of the cranial sensory ganglia. At neurula stages, placode precursors are interspersed in the ectoderm surrounding the anterior neural plate before segregating into distinct placodes by as yet unknown mechanisms. Here, we perform live imaging to follow placode progenitors as they aggregate to form the lens and otic placodes. We find that while placode progenitors move with the same speed as their non-placodal neighbours, they exhibit increased persistence and directionality and these properties are required to assemble morphological placodes. Furthermore, we demonstrate that these factors are components of the transcriptional networks that coordinate placode cell behaviour including their directional movements. Together with previous work, our results support a dual role for Otx and Gbx transcription factors in both the early patterning of the neural plate border and the later segregation of its derivatives into distinct placodes. Summary: Using spatial and temporally controlled perturbations followed by live cell tracking in vivo, this paper demonstrates that directional movements downstream of Gbx2 and Otx2 are important for otic and lens placode formation.
Collapse
Affiliation(s)
- Ben Steventon
- Department of Craniofacial Development, King's College London, Guy's Campus, Tower Wing Floor 27, London SE1 9RT, UK .,Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Andrea Streit
- Department of Craniofacial Development, King's College London, Guy's Campus, Tower Wing Floor 27, London SE1 9RT, UK
| |
Collapse
|
33
|
Johnson Chacko L, Pechriggl EJ, Fritsch H, Rask-Andersen H, Blumer MJF, Schrott-Fischer A, Glueckert R. Neurosensory Differentiation and Innervation Patterning in the Human Fetal Vestibular End Organs between the Gestational Weeks 8-12. Front Neuroanat 2016; 10:111. [PMID: 27895556 PMCID: PMC5108762 DOI: 10.3389/fnana.2016.00111] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 10/28/2016] [Indexed: 11/13/2022] Open
Abstract
Balance orientation depends on the precise operation of the vestibular end organs and the vestibular ganglion neurons. Previous research on the assemblage of the neuronal network in the developing fetal vestibular organ has been limited to data from animal models. Insights into the molecular expression profiles and signaling moieties involved in embryological development of the human fetal inner ear have been limited. We present an investigation of the cells of the vestibular end organs with specific focus on the hair cell differentiation and innervation pattern using an uninterrupted series of unique specimens from gestational weeks 8-12. Nerve fibers positive for peripherin innervate the entire fetal crista and utricle. While in rodents only the peripheral regions of the cristae and the extra-striolar region of the statolithic organs are stained. At week 9, transcription factors PAX2 and PAX8 were observed in the hair cells whereas PAX6 was observed for the first time among the supporting cells of the cristae and the satellite glial cells of the vestibular ganglia. Glutamine synthetase, a regulator of the neurotransmitter glutamate, is strongly expressed among satellite glia cells, transitional zones of the utricle and supporting cells in the sensory epithelium. At gestational week 11, electron microscopic examination reveals bouton contacts at hair cells and first signs of the formation of a protocalyx at type I hair cells. Our study provides first-hand insight into the fetal development of the vestibular end organs as well as their pattern of innervation by means of immunohistochemical and EM techniques, with the aim of contributing toward our understanding of balance development.
Collapse
Affiliation(s)
- Lejo Johnson Chacko
- Department of Otolaryngology, Medical University of Innsbruck Innsbruck, Austria
| | - Elisabeth J Pechriggl
- Department of Anatomy, Histology, and Embryology, Division of Clinical and Functional Anatomy, Medical University of Innsbruck Innsbruck, Austria
| | - Helga Fritsch
- Department of Anatomy, Histology, and Embryology, Division of Clinical and Functional Anatomy, Medical University of Innsbruck Innsbruck, Austria
| | | | - Michael J F Blumer
- Department of Anatomy, Histology, and Embryology, Division of Clinical and Functional Anatomy, Medical University of Innsbruck Innsbruck, Austria
| | | | - Rudolf Glueckert
- Department of Otolaryngology, Medical University of InnsbruckInnsbruck, Austria; University Clinics Innsbruck, Tirol KlinikenInnsbruck, Austria
| |
Collapse
|
34
|
DeJonge RE, Liu XP, Deig CR, Heller S, Koehler KR, Hashino E. Modulation of Wnt Signaling Enhances Inner Ear Organoid Development in 3D Culture. PLoS One 2016; 11:e0162508. [PMID: 27607106 PMCID: PMC5015985 DOI: 10.1371/journal.pone.0162508] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 08/01/2016] [Indexed: 01/17/2023] Open
Abstract
Stem cell-derived inner ear sensory epithelia are a promising source of tissues for treating patients with hearing loss and dizziness. We recently demonstrated how to generate inner ear sensory epithelia, designated as inner ear organoids, from mouse embryonic stem cells (ESCs) in a self-organizing 3D culture. Here we improve the efficiency of this culture system by elucidating how Wnt signaling activity can drive the induction of otic tissue. We found that a carefully timed treatment with the potent Wnt agonist CHIR99021 promotes induction of otic vesicles—a process that was previously self-organized by unknown mechanisms. The resulting otic-like vesicles have a larger lumen size and contain a greater number of Pax8/Pax2-positive otic progenitor cells than organoids derived without the Wnt agonist. Additionally, these otic-like vesicles give rise to large inner ear organoids with hair cells whose morphological, biochemical and functional properties are indistinguishable from those of vestibular hair cells in the postnatal mouse inner ear. We conclude that Wnt signaling plays a similar role during inner ear organoid formation as it does during inner ear development in the embryo.
Collapse
Affiliation(s)
- Rachel E. DeJonge
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States of America
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, United States of America
| | - Xiao-Ping Liu
- Department of Otolaryngology, F.M. Kirby Neurobiology Center Boston Children’s Hospital, and Harvard Medical School, Boston, MA, 02115, United States of America
| | - Christopher R. Deig
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States of America
| | - Stefan Heller
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, Palo Alto, CA, 94305, United States of America
| | - Karl R. Koehler
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States of America
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, United States of America
- * E-mail: (EH); (KRK)
| | - Eri Hashino
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, United States of America
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, United States of America
- * E-mail: (EH); (KRK)
| |
Collapse
|
35
|
Adams DS, Uzel SGM, Akagi J, Wlodkowic D, Andreeva V, Yelick PC, Devitt-Lee A, Pare JF, Levin M. Bioelectric signalling via potassium channels: a mechanism for craniofacial dysmorphogenesis in KCNJ2-associated Andersen-Tawil Syndrome. J Physiol 2016; 594:3245-70. [PMID: 26864374 PMCID: PMC4908029 DOI: 10.1113/jp271930] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 02/01/2016] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS Xenopus laevis craniofacial development is a good system for the study of Andersen-Tawil Syndrome (ATS)-associated craniofacial anomalies (CFAs) because (1) Kcnj2 is expressed in the nascent face; (2) molecular-genetic and biophysical techniques are available for the study of ion-dependent signalling during craniofacial morphogenesis; (3) as in humans, expression of variant Kcnj2 forms in embryos causes a muscle phenotype; and (4) variant forms of Kcnj2 found in human patients, when injected into frog embryos, cause CFAs in the same cell lineages. Forced expression of WT or variant Kcnj2 changes the normal pattern of Vmem (resting potential) regionalization found in the ectoderm of neurulating embryos, and changes the normal pattern of expression of ten different genetic regulators of craniofacial development, including markers of cranial neural crest and of placodes. Expression of other potassium channels and two different light-activated channels, all of which have an effect on Vmem , causes CFAs like those induced by injection of Kcnj2 variants. In contrast, expression of Slc9A (NHE3), an electroneutral ion channel, and of GlyR, an inactive Cl(-) channel, do not cause CFAs, demonstrating that correct craniofacial development depends on a pattern of bioelectric states, not on ion- or channel-specific signalling. Using optogenetics to control both the location and the timing of ion flux in developing embryos, we show that affecting Vmem of the ectoderm and no other cell layers is sufficient to cause CFAs, but only during early neurula stages. Changes in Vmem induced late in neurulation do not affect craniofacial development. We interpret these data as strong evidence, consistent with our hypothesis, that ATS-associated CFAs are caused by the effect of variant Kcnj2 on the Vmem of ectodermal cells of the developing face. We predict that the critical time is early during neurulation, and the critical cells are the ectodermal cranial neural crest and placode lineages. This points to the potential utility of extant, ion flux-modifying drugs as treatments to prevent CFAs associated with channelopathies such as ATS. ABSTRACT Variants in potassium channel KCNJ2 cause Andersen-Tawil Syndrome (ATS); the induced craniofacial anomalies (CFAs) are entirely unexplained. We show that KCNJ2 is expressed in Xenopus and mouse during the earliest stages of craniofacial development. Misexpression in Xenopus of KCNJ2 carrying ATS-associated mutations causes CFAs in the same structures affected in humans, changes the normal pattern of membrane voltage potential regionalization in the developing face and disrupts expression of important craniofacial patterning genes, revealing the endogenous control of craniofacial patterning by bioelectric cell states. By altering cells' resting potentials using other ion translocators, we show that a change in ectodermal voltage, not tied to a specific protein or ion, is sufficient to cause CFAs. By adapting optogenetics for use in non-neural cells in embryos, we show that developmentally patterned K(+) flux is required for correct regionalization of the resting potentials and for establishment of endogenous early gene expression domains in the anterior ectoderm, and that variants in KCNJ2 disrupt this regionalization, leading to the CFAs seen in ATS patients.
Collapse
Affiliation(s)
- Dany Spencer Adams
- Department of Biology and Tufts Centre for Regenerative and Developmental Biology, Tufts University, 200 Boston Avenue, Medford, MA, 02155, USA
| | - Sebastien G M Uzel
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Jin Akagi
- School of Applied Sciences, RMIT University, Melbourne, Australia
| | - Donald Wlodkowic
- School of Applied Sciences, RMIT University, Melbourne, Australia
| | - Viktoria Andreeva
- Department of Orthodontics, Division of Craniofacial and Molecular Genetics, Tufts University School of Dental Medicine, Boston, MA 02111, USA
| | - Pamela Crotty Yelick
- Department of Orthodontics, Division of Craniofacial and Molecular Genetics, Tufts University School of Dental Medicine, Boston, MA 02111, USA
| | - Adrian Devitt-Lee
- Department of Biology and Tufts Centre for Regenerative and Developmental Biology, Tufts University, 200 Boston Avenue, Medford, MA, 02155, USA
| | - Jean-Francois Pare
- Department of Biology and Tufts Centre for Regenerative and Developmental Biology, Tufts University, 200 Boston Avenue, Medford, MA, 02155, USA
| | - Michael Levin
- Department of Biology and Tufts Centre for Regenerative and Developmental Biology, Tufts University, 200 Boston Avenue, Medford, MA, 02155, USA
| |
Collapse
|
36
|
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
|
37
|
Aguillon R, Blader P, Batut J. Patterning, morphogenesis, and neurogenesis of zebrafish cranial sensory placodes. Methods Cell Biol 2016; 134:33-67. [PMID: 27312490 DOI: 10.1016/bs.mcb.2016.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Peripheral sensory organs and ganglia found in the vertebrate head arise during embryonic development from distinct ectodermal thickenings, called cranial sensory placodes (adenohypophyseal, olfactory, lens, trigeminal, epibranchial, and otic). A series of patterning events leads to the establishment of these placodes. Subsequently, these placodes undergo specific morphogenetic movements and cell-type specification in order to shape the final placodal derivatives and to produce differentiated cell types necessary for their function. In this chapter, we will focus on recent studies in the zebrafish that have advanced our understanding of cranial sensory placode development. We will summarize the signaling events and their molecular effectors guiding the formation of the so-called preplacodal region, and the subsequent subdivision of this region along the anteroposterior axis that gives rise to specific placode identities as well as those controlling morphogenesis and neurogenesis. Finally, we will highlight the approaches used in zebrafish that have been established to precisely label cell populations, to follow their development, and/or to characterize cell fates within a specific placode.
Collapse
Affiliation(s)
- R Aguillon
- Centre de Biologie du Développement (CBD, UMR5547), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - P Blader
- Centre de Biologie du Développement (CBD, UMR5547), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - J Batut
- Centre de Biologie du Développement (CBD, UMR5547), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| |
Collapse
|
38
|
Żak M, Klis SFL, Grolman W. The Wnt and Notch signalling pathways in the developing cochlea: Formation of hair cells and induction of regenerative potential. Int J Dev Neurosci 2015; 47:247-58. [PMID: 26471908 DOI: 10.1016/j.ijdevneu.2015.09.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 09/25/2015] [Accepted: 09/26/2015] [Indexed: 12/21/2022] Open
Abstract
The Wnt and Notch signalling pathways control proliferation, specification, and cell fate choices during embryonic development and in adult life. Hence, there is much interest in both signalling pathways in the context of stem cell biology and tissue regeneration. In the developing ear, the Wnt and Notch signalling pathways specify otic cells and refine the ventral boundary of the otic placode. Since both signalling pathways control events essential for the formation of sensory cells, such as proliferation and hair cell differentiation, these pathways could hold promise for the regeneration of hair cells in adult mammalian cochlea. Indeed, modulating either the Wnt or Notch pathways can trigger the regenerative potential of supporting cells. In the neonatal mouse cochlea, Notch-mediated regeneration of hair cells partially depends on Wnt signalling, which implies an interaction between the pathways. This review presents how the Wnt and Notch signalling pathways regulate the formation of sensory hair cells and how modulating their activity induces regenerative potential in the mammalian cochlea.
Collapse
Affiliation(s)
- Magdalena Żak
- Department of Otorhinolaryngology and Head & Neck Surgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Room G.02.531, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands.
| | - Sjaak F L Klis
- Department of Otorhinolaryngology and Head & Neck Surgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Room G.02.531, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands
| | - Wilko Grolman
- Department of Otorhinolaryngology and Head & Neck Surgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Room G.02.531, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands
| |
Collapse
|
39
|
Wright KD, Mahoney Rogers AA, Zhang J, Shim K. Cooperative and independent functions of FGF and Wnt signaling during early inner ear development. BMC DEVELOPMENTAL BIOLOGY 2015; 15:33. [PMID: 26443994 PMCID: PMC4594887 DOI: 10.1186/s12861-015-0083-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 09/18/2015] [Indexed: 12/28/2022]
Abstract
Background In multiple vertebrate organisms, including chick, Xenopus, and zebrafish, Fibroblast Growth Factor (FGF) and Wnt signaling cooperate during formation of the otic placode. However, in the mouse, although FGF signaling induces Wnt8a expression during induction of the otic placode, it is unclear whether these two signaling pathways functionally cooperate. Sprouty (Spry) genes encode intracellular antagonists of receptor tyrosine kinase signaling, including FGF signaling. We previously demonstrated that the Sprouty1 (Spry1) and Sprouty2 (Spry2) genes antagonize FGF signaling during induction of the otic placode. Here, we investigate cross talk between FGF/SPRY and Wnt signaling during otic placode induction and assess whether these two signaling pathways functionally cooperate during early inner ear development in the mouse. Methods Embryos were generated carrying combinations of a Spry1 null allele, Spry2 null allele, β-catenin null allele, or a Wnt reporter transgene. Otic phenotypes were assessed by in situ hybridization, semi-quantitative reverse transcriptase PCR, immunohistochemistry, and morphometric analysis of sectioned tissue. Results Comparison of Spry1, Spry2, and Wnt reporter expression in pre-otic and otic placode cells indicates that FGF signaling precedes and is active in more cells than Wnt signaling. We provide in vivo evidence that FGF signaling activates the Wnt signaling pathway upstream of TCF/Lef transcriptional activation. FGF regulation of Wnt signaling is functional, since early inner ear defects in Spry1 and Spry2 compound mutant embryos can be genetically rescued by reducing the activity of the Wnt signaling pathway. Interestingly, we find that although the entire otic placode increases in size in Spry1 and Spry2 compound mutant embryos, the size of the Wnt-reporter-positive domain does not increase to the same extent as the Wnt-reporter-negative domain. Conclusions This study provides genetic evidence that FGF and Wnt signaling cooperate during early inner ear development in the mouse. Furthermore, our data suggest that although specification of the otic placode may be globally regulated by FGF signaling, otic specification of cells in which both FGF and Wnt signaling are active may be more tightly regulated. Electronic supplementary material The online version of this article (doi:10.1186/s12861-015-0083-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kevin D Wright
- Department of Pediatrics, Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| | - Amanda A Mahoney Rogers
- Department of Pediatrics, Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| | - Jian Zhang
- Department of Pediatrics, Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| | - Katherine Shim
- Department of Pediatrics, Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| |
Collapse
|
40
|
D’Aniello E, Ravisankar P, Waxman JS. Rdh10a Provides a Conserved Critical Step in the Synthesis of Retinoic Acid during Zebrafish Embryogenesis. PLoS One 2015; 10:e0138588. [PMID: 26394147 PMCID: PMC4578954 DOI: 10.1371/journal.pone.0138588] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 09/01/2015] [Indexed: 01/15/2023] Open
Abstract
The first step in the conversion of vitamin A into retinoic acid (RA) in embryos requires retinol dehydrogenases (RDHs). Recent studies have demonstrated that RDH10 is a critical core component of the machinery that produces RA in mouse and Xenopus embryos. If the conservation of Rdh10 function in the production of RA extends to teleost embryos has not been investigated. Here, we report that zebrafish Rdh10a deficient embryos have defects consistent with loss of RA signaling, including anteriorization of the nervous system and enlarged hearts with increased cardiomyocyte number. While knockdown of Rdh10a alone produces relatively mild RA deficient phenotypes, Rdh10a can sensitize embryos to RA deficiency and enhance phenotypes observed when Aldh1a2 function is perturbed. Moreover, excess Rdh10a enhances embryonic sensitivity to retinol, which has relatively mild teratogenic effects compared to retinal and RA treatment. Performing Rdh10a regulatory expression analysis, we also demonstrate that a conserved teleost rdh10a enhancer requires Pax2 sites to drive expression in the eyes of transgenic embryos. Altogether, our results demonstrate that Rdh10a has a conserved requirement in the first step of RA production within vertebrate embryos.
Collapse
Affiliation(s)
- Enrico D’Aniello
- The Heart Institute, Molecular Cardiovascular Biology and Developmental Biology Divisions, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | - Padmapriyadarshini Ravisankar
- The Heart Institute, Molecular Cardiovascular Biology and Developmental Biology Divisions, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | - Joshua S. Waxman
- The Heart Institute, Molecular Cardiovascular Biology and Developmental Biology Divisions, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
- * E-mail:
| |
Collapse
|
41
|
Whitfield TT. Development of the inner ear. Curr Opin Genet Dev 2015; 32:112-8. [DOI: 10.1016/j.gde.2015.02.006] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 02/11/2015] [Accepted: 02/13/2015] [Indexed: 02/04/2023]
|
42
|
Qian G, Zhu L, Li G, Liu X, Wang Y. Distinct expression patterns and functions of the pygopus genes in amphioxus and zebrafish early embryogenesis. Genes Genomics 2015. [DOI: 10.1007/s13258-015-0266-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
43
|
Sai X, Ladher RK. Early steps in inner ear development: induction and morphogenesis of the otic placode. Front Pharmacol 2015; 6:19. [PMID: 25713536 PMCID: PMC4322616 DOI: 10.3389/fphar.2015.00019] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 01/21/2015] [Indexed: 01/09/2023] Open
Abstract
Various cellular replacement therapies using in vitro generated cells to replace damaged tissue have been proposed as strategies to alleviate hearing loss. All such therapies must involve a complete understanding of the earliest steps in inner ear development; its induction as a thickened plate of cells in the non-neural, surface ectoderm of the embryo, to its internalization as an otocyst embedded in the head mesenchyme of the embryo. Such knowledge informs researchers addressing the feasibility of the proposed strategy and present alternatives if needed. In this review we describe the mechanisms of inner ear induction, concentrating on the factors that steer the fate of ectoderm into precursors of the inner ear. Induction then leads to inner ear morphogenesis and we describe the cellular changes that occur as the inner ear is converted from a superficial placode to an internalized otocyst, and how they are coordinated with a particular emphasis on how the signaling environment surrounding the inner ear influences these processes.
Collapse
Affiliation(s)
- Xiaorei Sai
- Laboratory for Sensory Development, RIKEN Center for Developmental Biology Kobe, Japan
| | - Raj K Ladher
- Laboratory for Sensory Development, RIKEN Center for Developmental Biology Kobe, Japan
| |
Collapse
|
44
|
Pechriggl EJ, Bitsche M, Glueckert R, Rask‐Andersen H, Blumer MJF, Schrott‐Fischer A, Fritsch H. Development of the innervation of the human inner ear. Dev Neurobiol 2014; 75:683-702. [DOI: 10.1002/dneu.22242] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 10/28/2014] [Accepted: 10/28/2014] [Indexed: 01/04/2023]
Affiliation(s)
- Elisabeth J. Pechriggl
- Department of Anatomy, Histology, and Embryology, Division of Clinical and Functional AnatomyMedical University of InnsbruckMüllerstrasse 596020Innsbruck Austria
| | - Mario Bitsche
- Department of Anatomy, Histology, and Embryology, Division of Clinical and Functional AnatomyMedical University of InnsbruckMüllerstrasse 596020Innsbruck Austria
| | - Rudolf Glueckert
- Department of OtolaryngologyMedical University of InnsbruckAnichstrasse 356020Innsbruck Austria
- University Clinics InnsbruckTiroler LandeskrankenanstaltenInnsbruck Austria
| | - Helge Rask‐Andersen
- Departments of OtolaryngologyUppsala University Hospital751 85Uppsala Sweden
| | - Michael J. F. Blumer
- Department of Anatomy, Histology, and Embryology, Division of Clinical and Functional AnatomyMedical University of InnsbruckMüllerstrasse 596020Innsbruck Austria
| | | | - Helga Fritsch
- Department of Anatomy, Histology, and Embryology, Division of Clinical and Functional AnatomyMedical University of InnsbruckMüllerstrasse 596020Innsbruck Austria
| |
Collapse
|
45
|
Gerlach GF, Wingert RA. Zebrafish pronephros tubulogenesis and epithelial identity maintenance are reliant on the polarity proteins Prkc iota and zeta. Dev Biol 2014; 396:183-200. [PMID: 25446529 DOI: 10.1016/j.ydbio.2014.08.038] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 08/21/2014] [Accepted: 08/26/2014] [Indexed: 02/06/2023]
Abstract
The zebrafish pronephros provides an excellent in vivo system to study the mechanisms of vertebrate nephron development. When and how renal progenitors in the zebrafish embryo undergo tubulogenesis to form nephrons is poorly understood, but is known to involve a mesenchymal to epithelial transition (MET) and the acquisition of polarity. Here, we determined the precise timing of these events in pronephros tubulogenesis. As the ternary polarity complex is an essential regulator of epithelial cell polarity across tissues, we performed gene knockdown studies to assess the roles of the related factors atypical protein kinase C iota and zeta (prkcι, prkcζ). We found that prkcι and prkcζ serve partially redundant functions to establish pronephros tubule epithelium polarity. Further, the loss of prkcι or the combined knockdown of prkcι/ζ disrupted proximal tubule morphogenesis and podocyte migration due to cardiac defects that prevented normal fluid flow to the kidney. Surprisingly, tubule cells in prkcι/ζ morphants displayed ectopic expression of the transcription factor pax2a and the podocyte-associated genes wt1a, wt1b, and podxl, suggesting that prkcι/ζ are needed to maintain renal epithelial identity. Knockdown of genes essential for cardiac contractility and vascular flow to the kidney, such as tnnt2a, or elimination of pronephros fluid output through knockdown of the intraflagellar transport gene ift88, was not associated with ectopic pronephros gene expression, thus suggesting a unique role for prkcι/ζ in maintaining tubule epithelial identity separate from the consequence of disruptions to renal fluid flow. Interestingly, knockdown of pax2a, but not wt1a, was sufficient to rescue ectopic tubule gene expression in prkcι/ζ morphants. These data suggest a model in which the redundant activities of prkcι and prkcζ are essential to establish tubule epithelial polarity and also serve to maintain proper epithelial cell type identity in the tubule by inhibiting pax2a expression. These studies provide a valuable foundation for further analysis of MET during nephrogenesis, and have implications for understanding the pathways that affect nephron epithelial cells during kidney disease and regeneration.
Collapse
Affiliation(s)
- Gary F Gerlach
- Department of Biological Sciences and Center for Zebrafish Research, University of Notre Dame, 100 Galvin Life Sciences, Notre Dame, IN 46556, USA
| | - Rebecca A Wingert
- Department of Biological Sciences and Center for Zebrafish Research, University of Notre Dame, 100 Galvin Life Sciences, Notre Dame, IN 46556, USA.
| |
Collapse
|
46
|
Maulding K, Padanad MS, Dong J, Riley BB. Mesodermal Fgf10b cooperates with other fibroblast growth factors during induction of otic and epibranchial placodes in zebrafish. Dev Dyn 2014; 243:1275-85. [PMID: 24677486 PMCID: PMC4313390 DOI: 10.1002/dvdy.24119] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 01/27/2014] [Accepted: 02/16/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Vertebrate otic and epibranchial placodes develop in close proximity in response to localized fibroblast growth factor (Fgf) signaling. Although less is known about epibranchial induction, the process of otic induction in highly conserved, with important roles for Fgf3 and Fgf8 reported in all species examined. Fgf10 is also critical for otic induction in mouse, but the only zebrafish ortholog examined to date, fgf10a, is not expressed early enough to play such a role. A second zebrafish ortholog, fgf10b, has not been previously examined. RESULTS We find that zebrafish fgf10b is expressed at tailbud stage in paraxial cephalic mesoderm beneath prospective epibranchial tissue, lateral to the developing otic placode. Knockdown of fgf10b does not affect initial otic induction but impairs subsequent accumulation of otic cells. Formation of epibranchial placodes and ganglia are also moderately impaired. Combinatorial disruption of fgf10b and fgf3 exacerbates the deficiency of otic cells and eliminates epibranchial induction entirely. Disruption of fgf10b and fgf24 also strongly reduces, but does not eliminate, epibranchial induction. CONCLUSIONS fgf10b participates in a late phase of otic induction and, in combination with fgf3, is especially critical for epibranchial induction.
Collapse
Affiliation(s)
- Kirstin Maulding
- Biology Department, Texas A&M University, College Station, TX 77843-3258
| | - Mahesh S. Padanad
- Biology Department, Texas A&M University, College Station, TX 77843-3258
| | - Jennifer Dong
- Biology Department, Texas A&M University, College Station, TX 77843-3258
| | - Bruce B. Riley
- Biology Department, Texas A&M University, College Station, TX 77843-3258
| |
Collapse
|
47
|
Sánchez-Guardado LÓ, Puelles L, Hidalgo-Sánchez M. Fate map of the chicken otic placode. Development 2014; 141:2302-12. [PMID: 24821982 DOI: 10.1242/dev.101667] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The inner ear is an intricate three-dimensional sensory organ that arises from a flat, thickened portion of the ectoderm termed the otic placode. There is evidence that the ontogenetic steps involved in the progressive specification of the highly specialized inner ear of vertebrates involve the concerted actions of diverse patterning signals that originate from nearby tissues, providing positional identity and instructive context. The topology of the prospective inner ear portions at placode stages when such patterning begins has remained largely unknown. The chick-quail model was used to perform a comprehensive fate mapping study of the chick otic placode, shedding light on the precise topological position of each presumptive inner ear component relative to the dorsoventral and anteroposterior axes of the otic placode and, implicitly, to the possible sources of inducing signals. The findings reveal the existence of three dorsoventrally arranged anteroposterior domains from which the endolymphatic system, the maculae and basilar papilla, and the cristae develop. This study provides new bases for the interpretation of earlier and future descriptive and experimental studies that aim to understand the molecular genetic mechanisms involved in otic placode patterning.
Collapse
Affiliation(s)
| | - Luis Puelles
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia, 30003 Murcia, Spain
| | - Matías Hidalgo-Sánchez
- Department of Cell Biology, Faculty of Science, University of Extremadura, 06071 Badajoz, Spain
| |
Collapse
|
48
|
Steventon B, Mayor R, Streit A. Neural crest and placode interaction during the development of the cranial sensory system. Dev Biol 2014; 389:28-38. [PMID: 24491819 PMCID: PMC4439187 DOI: 10.1016/j.ydbio.2014.01.021] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 01/19/2014] [Accepted: 01/25/2014] [Indexed: 01/13/2023]
Abstract
In the vertebrate head, the peripheral components of the sensory nervous system are derived from two embryonic cell populations, the neural crest and cranial sensory placodes. Both arise in close proximity to each other at the border of the neural plate: neural crest precursors abut the future central nervous system, while placodes originate in a common preplacodal region slightly more lateral. During head morphogenesis, complex events organise these precursors into functional sensory structures, raising the question of how their development is coordinated. Here we review the evidence that neural crest and placode cells remain in close proximity throughout their development and interact repeatedly in a reciprocal manner. We also review recent controversies about the relative contribution of the neural crest and placodes to the otic and olfactory systems. We propose that a sequence of mutual interactions between the neural crest and placodes drives the coordinated morphogenesis that generates functional sensory systems within the head.
Collapse
Affiliation(s)
- Ben Steventon
- Department of Developmental and Stem Cell Biology, Insitut Pasteur, France
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Andrea Streit
- Department of Craniofacial Development and Stem Cell Biology, King׳s College London, London, UK.
| |
Collapse
|
49
|
Blake JA, Ziman MR. Pax genes: regulators of lineage specification and progenitor cell maintenance. Development 2014; 141:737-51. [PMID: 24496612 DOI: 10.1242/dev.091785] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Pax genes encode a family of transcription factors that orchestrate complex processes of lineage determination in the developing embryo. Their key role is to specify and maintain progenitor cells through use of complex molecular mechanisms such as alternate RNA splice forms and gene activation or inhibition in conjunction with protein co-factors. The significance of Pax genes in development is highlighted by abnormalities that arise from the expression of mutant Pax genes. Here, we review the molecular functions of Pax genes during development and detail the regulatory mechanisms by which they specify and maintain progenitor cells across various tissue lineages. We also discuss mechanistic insights into the roles of Pax genes in regeneration and in adult diseases, including cancer.
Collapse
Affiliation(s)
- Judith A Blake
- School of Medical Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
| | | |
Collapse
|
50
|
Becherucci F, Lazzeri E, Lasagni L, Romagnani P. Renal progenitors and childhood: from development to disorders. Pediatr Nephrol 2014; 29:711-9. [PMID: 24389601 DOI: 10.1007/s00467-013-2686-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 10/17/2013] [Accepted: 10/28/2013] [Indexed: 12/11/2022]
Abstract
Nephropathies arise from conditions that alter nephron development or trigger nephron damage during neonatal, juvenile, and adult stages of life. Much evidence suggests that a key role in maintaining kidney integrity, homeostasis, and regenerative capacity is played by a population of progenitor cells resident in the organ. Although the primary goals in the field of renal progenitor cells are understanding their ability to regenerate nephrons and to restore damaged kidney function, the discovery of these cells could also be used to elucidate the molecular and pathophysiological basis of kidney diseases. As a result, once the identification of a subset of progenitor cells capable of kidney regeneration has been obtained, the increasing knowledge about their characteristics and about the mechanisms of renal development had pointed out the possibility of understanding the molecular basis of kidney diseases, so that, nowadays, some renal disorders could also be related to renal progenitor dysfunction. In this review, we summarize the evidence on the existence of renal progenitors in fetal and adult kidneys and discuss their role in physiology as well as in the pathogenesis of renal disorders with a particular focus on childhood age.
Collapse
Affiliation(s)
- Francesca Becherucci
- Pediatric Nephrology and Dialysis Unit, Meyer Children's University Hospital, Florence, Italy
| | | | | | | |
Collapse
|