1
|
El Amri M, Pandit A, Schlosser G. Marcks and Marcks-like 1 proteins promote spinal cord development and regeneration in Xenopus. eLife 2024; 13:e98277. [PMID: 39665418 PMCID: PMC11637466 DOI: 10.7554/elife.98277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 11/15/2024] [Indexed: 12/13/2024] Open
Abstract
Marcks and Marcksl1 are abundant proteins that shuttle between the cytoplasm and membrane to modulate multiple cellular processes, including cytoskeletal dynamics, proliferation, and secretion. Here, we performed loss- and gain-of-function experiments in Xenopus laevis to reveal the novel roles of these proteins in spinal cord development and regeneration. We show that Marcks and Marcksl1 have partly redundant functions and are required for normal neurite formation and proliferation of neuro-glial progenitors during embryonic spinal cord development and for its regeneration during tadpole stages. Rescue experiments in Marcks and Marcksl1 loss-of-function animals further suggested that some of the functions of Marcks and Marcksl1 in the spinal cord are mediated by phospholipid signaling. Taken together, these findings identify Marcks and Marcksl1 as critical new players in spinal cord development and regeneration and suggest new pathways to be targeted for therapeutic stimulation of spinal cord regeneration in human patients.
Collapse
Affiliation(s)
- Mohamed El Amri
- School of Biological and Chemical Sciences, University of GalwayGalwayIreland
- Research Ireland Center for Medical Devices (CÚRAM), University of GalwayGalwayIreland
| | - Abhay Pandit
- Research Ireland Center for Medical Devices (CÚRAM), University of GalwayGalwayIreland
| | - Gerhard Schlosser
- School of Biological and Chemical Sciences, University of GalwayGalwayIreland
| |
Collapse
|
2
|
Chung K, Millet M, Rouillon L, Zine A. Timing and Graded BMP Signalling Determines Fate of Neural Crest and Ectodermal Placode Derivatives from Pluripotent Stem Cells. Biomedicines 2024; 12:2262. [PMID: 39457575 PMCID: PMC11504183 DOI: 10.3390/biomedicines12102262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 09/29/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Pluripotent stem cells (PSCs) offer many potential research and clinical benefits due to their ability to differentiate into nearly every cell type in the body. They are often used as model systems to study early stages of ontogenesis to better understand key developmental pathways, as well as for drug screening. However, in order to fully realise the potential of PSCs and their translational applications, a deeper understanding of developmental pathways, especially in humans, is required. Several signalling molecules play important roles during development and are required for proper differentiation of PSCs. The concentration and timing of signal activation are important, with perturbations resulting in improper development and/or pathology. Bone morphogenetic proteins (BMPs) are one such key group of signalling molecules involved in the specification and differentiation of various cell types and tissues in the human body, including those related to tooth and otic development. In this review, we describe the role of BMP signalling and its regulation, the consequences of BMP dysregulation in disease and differentiation, and how PSCs can be used to investigate the effects of BMP modulation during development, mainly focusing on otic development. Finally, we emphasise the unique role of BMP4 in otic specification and how refined understanding of controlling its regulation could lead to the generation of more robust and reproducible human PSC-derived otic organoids for research and translational applications.
Collapse
Affiliation(s)
- Keshi Chung
- LBN, Laboratory of Bioengineering and Nanoscience, University of Montpellier, 34193 Montpellier, France
| | - Malvina Millet
- LBN, Laboratory of Bioengineering and Nanoscience, University of Montpellier, 34193 Montpellier, France
- Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA
| | - Ludivine Rouillon
- LBN, Laboratory of Bioengineering and Nanoscience, University of Montpellier, 34193 Montpellier, France
| | - Azel Zine
- LBN, Laboratory of Bioengineering and Nanoscience, University of Montpellier, 34193 Montpellier, France
| |
Collapse
|
3
|
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
|
4
|
Griffin C, Saint-Jeannet JP. In vitro modeling of cranial placode differentiation: Recent advances, challenges, and perspectives. Dev Biol 2024; 506:20-30. [PMID: 38052294 PMCID: PMC10843546 DOI: 10.1016/j.ydbio.2023.11.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 12/07/2023]
Abstract
Cranial placodes are transient ectodermal thickenings that contribute to a diverse array of organs in the vertebrate head. They develop from a common territory, the pre-placodal region that over time segregates along the antero-posterior axis into individual placodal domains: the adenohypophyseal, olfactory, lens, trigeminal, otic, and epibranchial placodes. These placodes terminally differentiate into the anterior pituitary, the lens, and contribute to sensory organs including the olfactory epithelium, and inner ear, as well as several cranial ganglia. To study cranial placodes and their derivatives and generate cells for therapeutic purposes, several groups have turned to in vitro derivation of placodal cells from human embryonic stem cells (hESCs) or induced pluripotent stem cells (hiPSCs). In this review, we summarize the signaling cues and mechanisms involved in cranial placode induction, specification, and differentiation in vivo, and discuss how this knowledge has informed protocols to derive cranial placodes in vitro. We also discuss the benefits and limitations of these protocols, and the potential of in vitro cranial placode modeling in regenerative medicine to treat cranial placode-related pathologies.
Collapse
Affiliation(s)
- Casey Griffin
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Jean-Pierre Saint-Jeannet
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA.
| |
Collapse
|
5
|
Heikes KL, Goldstein B. Expression patterns of FGF and BMP pathway genes in the tardigrade Hypsibius exemplaris. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.29.577774. [PMID: 38352320 PMCID: PMC10862696 DOI: 10.1101/2024.01.29.577774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
A small number of conserved signaling pathways regulate development of most animals, yet we do not know where these pathways are deployed in most embryos. This includes tardigrades, a phylum with a unique body shape. We examined expression patterns of components of the BMP and FGF signaling pathways during embryonic segmentation and mesoderm development of the tardigrade Hypsibius exemplaris. Among the patterns examined, we found that an FGF ligand gene is expressed in ectodermal segment posteriors and an FGF receptor gene is expressed in underlying endomesodermal pouches, suggesting possible FGF signaling between these developing germ layers. We found that a BMP ligand gene is expressed in lateral ectoderm and dorsolateral bands along segment posteriors, while the BMP antagonist Sog gene is expressed in lateral ectoderm and also in a subset of endomesodermal cells, suggesting a possible role of BMP signaling in dorsal-ventral patterning of lateral ectoderm. In combination with known roles of these pathways during development of common model systems, we developed hypotheses for how the BMP and FGF pathways might regulate embryo segmentation and mesoderm formation of the tardigrade H. exemplaris. These results identify the expression patterns of genes from two conserved signaling pathways for the first time in the tardigrade phylum.
Collapse
Affiliation(s)
- Kira L. Heikes
- Biology Department, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bob Goldstein
- Biology Department, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
6
|
Thiery AP, Buzzi AL, Hamrud E, Cheshire C, Luscombe NM, Briscoe J, Streit A. scRNA-sequencing in chick suggests a probabilistic model for cell fate allocation at the neural plate border. eLife 2023; 12:e82717. [PMID: 37530410 PMCID: PMC10425176 DOI: 10.7554/elife.82717] [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: 08/15/2022] [Accepted: 08/01/2023] [Indexed: 08/03/2023] Open
Abstract
The vertebrate 'neural plate border' is a transient territory located at the edge of the neural plate containing precursors for all ectodermal derivatives: the neural plate, neural crest, placodes and epidermis. Elegant functional experiments in a range of vertebrate models have provided an in-depth understanding of gene regulatory interactions within the ectoderm. However, these experiments conducted at tissue level raise seemingly contradictory models for fate allocation of individual cells. Here, we carry out single cell RNA sequencing of chick ectoderm from primitive streak to neurulation stage, to explore cell state diversity and heterogeneity. We characterise the dynamics of gene modules, allowing us to model the order of molecular events which take place as ectodermal fates segregate. Furthermore, we find that genes previously classified as neural plate border 'specifiers' typically exhibit dynamic expression patterns and are enriched in either neural, neural crest or placodal fates, revealing that the neural plate border should be seen as a heterogeneous ectodermal territory and not a discrete transitional transcriptional state. Analysis of neural, neural crest and placodal markers reveals that individual NPB cells co-express competing transcriptional programmes suggesting that their ultimate identify is not yet fixed. This population of 'border located undecided progenitors' (BLUPs) gradually diminishes as cell fate decisions take place. Considering our findings, we propose a probabilistic model for cell fate choice at the neural plate border. Our data suggest that the probability of a progenitor's daughters to contribute to a given ectodermal derivative is related to the balance of competing transcriptional programmes, which in turn are regulated by the spatiotemporal position of a progenitor.
Collapse
Affiliation(s)
- Alexandre P Thiery
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College LondonLondonUnited Kingdom
| | - Ailin Leticia Buzzi
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College LondonLondonUnited Kingdom
| | - Eva Hamrud
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College LondonLondonUnited Kingdom
| | - Chris Cheshire
- Bioinformatics and Computational Biology Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Nicholas M Luscombe
- Bioinformatics and Computational Biology Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - James Briscoe
- Bioinformatics and Computational Biology Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Andrea Streit
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College LondonLondonUnited Kingdom
| |
Collapse
|
7
|
Liu B, Ren X, Satou Y. BMP signaling is required to form the anterior neural plate border in ascidian embryos. Dev Genes Evol 2023:10.1007/s00427-023-00702-0. [PMID: 37079132 DOI: 10.1007/s00427-023-00702-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 03/29/2023] [Indexed: 04/21/2023]
Abstract
Cranial neurogenic placodes have been considered vertebrate innovations. However, anterior neural plate border (ANB) cells of ascidian embryos share many properties with vertebrate neurogenic placodes; therefore, it is now believed that the last common ancestor of vertebrates and ascidians had embryonic structures similar to neurogenic placodes of vertebrate embryos. Because BMP signaling is important for specifying the placode region in vertebrate embryos, we examined whether BMP signaling is also involved in gene expression in the ANB region of ascidian embryos. Our data indicated that Admp, a divergent BMP family member, is mainly responsible for BMP signaling in the ANB region, and that two BMP-antagonists, Noggin and Chordin, restrict the domain, in which BMP signaling is activated, to the ANB region, and prevent it from expanding to the neural plate. BMP signaling is required for expression of Foxg and Six1/2 at the late gastrula stage, and also for expression of Zf220, which encodes a zinc finger transcription factor in late neurula embryos. Because Zf220 negatively regulates Foxg, when we downregulated Zf220 by inhibiting BMP signaling, Foxg was upregulated, resulting in one large palp instead of three palps (adhesive organs derived from ANB cells). Functions of BMP signaling in specification of the ANB region give further support to the hypothesis that ascidian ANB cells share an evolutionary origin with vertebrate cranial placodes.
Collapse
Affiliation(s)
- Boqi Liu
- Department of Zoology, Graduate School of Science, Kyoto University, Sakyo, Kyoto, 606-8502, Japan
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Life Sciences, Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, China
| | - Ximan Ren
- Department of Zoology, Graduate School of Science, Kyoto University, Sakyo, Kyoto, 606-8502, Japan
| | - Yutaka Satou
- Department of Zoology, Graduate School of Science, Kyoto University, Sakyo, Kyoto, 606-8502, Japan.
| |
Collapse
|
8
|
Yamamoto T, Kambayashi Y, Tsukano K, Michiue T. Ndst1, a heparan sulfate modification enzyme, regulates neuroectodermal patterning by enhancing Wnt signaling in Xenopus. Dev Growth Differ 2023; 65:153-160. [PMID: 36726238 PMCID: PMC11520968 DOI: 10.1111/dgd.12843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 01/05/2023] [Accepted: 01/10/2023] [Indexed: 02/03/2023]
Abstract
Neural tissue is derived from three precursor regions: neural plate, neural crest, and preplacodal ectoderm. These regions are determined by morphogen-mediated signaling. Morphogen distribution is generally regulated by binding to an extracellular matrix component, heparan sulfate (HS) proteoglycan. HS is modified by many enzymes, such as N-deacetyl sulfotransferase 1 (Ndst1), which is highly expressed in early development. However, functions of HS modifications in ectodermal patterning are largely unknown. In this study, we analyzed the role of Ndst1 using Xenopus embryos. We found that ndst1 was expressed in anterior neural plate and the trigeminal region at the neurula stage. ndst1 overexpression expanded the neural crest (NC) region, whereas translational inhibition reduced not only the trigeminal region, but also the adjacent NC region, especially the anterior part. At a later stage, ndst1 knocked-down embryos showed defects in cranial ganglion formation. We also found that Ndst1 activates Wnt signaling pathway at the neurula stage. Taken together, our results suggest that N-sulfonated HS accumulates Wnt ligand and activates Wnt signaling in ndst1-expressing cells, but that it inhibits signaling in non-ndst1-expressing cells, leading to proper neuroectodermal patterning.
Collapse
Grants
- JPMJSP2108 Japan Science and Technology Agency
- 18K06244 Ministry of Education, Culture, Sports, Science and Technology
- 19K16138 Ministry of Education, Culture, Sports, Science and Technology
- 21K06183 Ministry of Education, Culture, Sports, Science and Technology
- Narishige Zoological Science Award
- Japan Science and Technology Agency
- Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Takayoshi Yamamoto
- Department of Life Sciences, Graduate School of Arts and SciencesThe University of TokyoTokyoJapan
| | - Yuta Kambayashi
- Department of Life Sciences, Graduate School of Arts and SciencesThe University of TokyoTokyoJapan
| | - Kohei Tsukano
- Department of Life Sciences, Graduate School of Arts and SciencesThe University of TokyoTokyoJapan
| | - Tatsuo Michiue
- Department of Life Sciences, Graduate School of Arts and SciencesThe University of TokyoTokyoJapan
- Department of Biological Sciences, Graduate School of ScienceThe University of TokyoTokyoJapan
| |
Collapse
|
9
|
Yamamoto T, Kaneshima T, Tsukano K, Michiue T. The heparan sulfate modification enzyme, Hs6st1, governs Xenopus neuroectodermal patterning by regulating distributions of Fgf and Noggin. Dev Biol 2023; 496:87-94. [PMID: 36739958 DOI: 10.1016/j.ydbio.2023.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 01/20/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
The nervous system has various types of cells derived from three neuroectodermal regions: neural plate (NP), neural crest (NC), and preplacodal ectoderm (PPE). Differentiation of these regions is regulated by various morphogens. However, regulatory mechanisms of morphogen distribution in neural patterning are still debated. In general, an extracellular component, heparan sulfate (HS), is essential to regulate morphogen gradients by modulating morphogen binding. The present study focused on an HS modification enzyme, heparan sulfate 6-O-sulfotransferase 1 (Hs6st1), which is highly expressed during the neurula stage in Xenopus. Our present in situ hybridization analysis revealed that Hs6st1 is expressed in the lateral sensorial layer of neuroectoderm. Overexpression of Hs6st1 expands Sox3 (NP marker gene) expression, and slightly dampens FoxD3 (NC marker) expression. Hs6st1 knockout using the CRISPR/Cas9 system also expands the neural plate region, followed by retinal malformation. These results imply that 6-O sulfation, mediated by Hs6st1, selectively regulates morphogen distribution required for neuroectodermal patterning. Among morphogens required for patterning, Fgf8a accumulates on Hs6st1-expressing cells, whereas a secreted BMP antagonist, Noggin, diffuses away from those cells. Thus, cell-autonomous 6-O sulfation of HS at the sensorial layer of neuroectoderm also affects neuroectodermal patterning in neighboring regions, including neural plate and neural crest, not only through accumulation, but also through dispersal of specific morphogens.
Collapse
Affiliation(s)
- Takayoshi Yamamoto
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan.
| | - Toki Kaneshima
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan
| | - Kohei Tsukano
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan
| | - Tatsuo Michiue
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo, 153-8902, Japan; Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
10
|
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
|
11
|
Conti E, Harschnitz O. Human stem cell models to study placode development, function and pathology. Development 2022; 149:276462. [DOI: 10.1242/dev.200831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
ABSTRACT
Placodes are embryonic structures originating from the rostral ectoderm that give rise to highly diverse organs and tissues, comprising the anterior pituitary gland, paired sense organs and cranial sensory ganglia. Their development, including the underlying gene regulatory networks and signalling pathways, have been for the most part characterised in animal models. In this Review, we describe how placode development can be recapitulated by the differentiation of human pluripotent stem cells towards placode progenitors and their derivatives, highlighting the value of this highly scalable platform as an optimal in vitro tool to study the development of human placodes, and identify human-specific mechanisms in their development, function and pathology.
Collapse
Affiliation(s)
- Eleonora Conti
- Neurogenomics Research Centre, Human Technopole , Viale Rita Levi-Montalcini, 1, 20157 Milan , Italy
| | - Oliver Harschnitz
- Neurogenomics Research Centre, Human Technopole , Viale Rita Levi-Montalcini, 1, 20157 Milan , Italy
| |
Collapse
|
12
|
Gao L, Jin N, Ye Z, Ma T, Huang Y, Li H, Du J, Li Z. A possible connection between reactive oxygen species and the unfolded protein response in lens development: From insight to foresight. Front Cell Dev Biol 2022; 10:820949. [PMID: 36211466 PMCID: PMC9535091 DOI: 10.3389/fcell.2022.820949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 08/31/2022] [Indexed: 11/28/2022] Open
Abstract
The lens is a relatively special and simple organ. It has become an ideal model to study the common developmental characteristics among different organic systems. Lens development is a complex process influenced by numerous factors, including signals from the intracellular and extracellular environment. Reactive oxygen species (ROS) are a group of highly reactive and oxygen-containing molecules that can cause endoplasmic reticulum stress in lens cells. As an adaptive response to ER stress, lens cells initiate the unfolded protein response (UPR) to maintain normal protein synthesis by selectively increasing/decreasing protein synthesis and increasing the degradation of misfolded proteins. Generally, the UPR signaling pathways have been well characterized in the context of many pathological conditions. However, recent studies have also confirmed that all three UPR signaling pathways participate in a variety of developmental processes, including those of the lens. In this review, we first briefly summarize the three stages of lens development and present the basic profiles of ROS and the UPR. We then discuss the interconnections between lens development and these two mechanisms. Additionally, the potential adoption of human pluripotent stem-cell-based lentoids in lens development research is proposed to provide a novel perspective on future developmental studies.
Collapse
Affiliation(s)
- Lixiong Gao
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ni Jin
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Endocrinology, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, The Chinese PLA General Hospital, Beijing, China
| | - Zi Ye
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Tianju Ma
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yang Huang
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hongyu Li
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jinlin Du
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhaohui Li
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
- *Correspondence: Zhaohui Li,
| |
Collapse
|
13
|
Michiue T, Tsukano K. Feedback Regulation of Signaling Pathways for Precise Pre-Placodal Ectoderm Formation in Vertebrate Embryos. J Dev Biol 2022; 10:35. [PMID: 36135368 PMCID: PMC9504399 DOI: 10.3390/jdb10030035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/19/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Intracellular signaling pathways are essential to establish embryonic patterning, including embryonic axis formation. Ectodermal patterning is also governed by a series of morphogens. Four ectodermal regions are thought to be controlled by morphogen gradients, but some perturbations are expected to occur during dynamic morphogenetic movement. Therefore, a mechanism to define areas precisely and reproducibly in embryos, including feedback regulation of signaling pathways, is necessary. In this review, we outline ectoderm pattern formation and signaling pathways involved in the establishment of the pre-placodal ectoderm (PPE). We also provide an example of feedback regulation of signaling pathways for robust formation of the PPE, showing the importance of this regulation.
Collapse
Affiliation(s)
- Tatsuo Michiue
- Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | | |
Collapse
|
14
|
Tsukano K, Yamamoto T, Watanabe T, Michiue T. Xenopus Dusp6 modulates FGF signaling precisely to pattern pre-placodal ectoderm. Dev Biol 2022; 488:81-90. [DOI: 10.1016/j.ydbio.2022.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 05/07/2022] [Accepted: 05/16/2022] [Indexed: 12/23/2022]
|
15
|
Klein SL, Tavares ALP, Peterson M, Sullivan CH, Moody SA. Repressive Interactions Between Transcription Factors Separate Different Embryonic Ectodermal Domains. Front Cell Dev Biol 2022; 10:786052. [PMID: 35198557 PMCID: PMC8859430 DOI: 10.3389/fcell.2022.786052] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/10/2022] [Indexed: 11/13/2022] Open
Abstract
The embryonic ectoderm is composed of four domains: neural plate, neural crest, pre-placodal region (PPR) and epidermis. Their formation is initiated during early gastrulation by dorsal-ventral and anterior-posterior gradients of signaling factors that first divide the embryonic ectoderm into neural and non-neural domains. Next, the neural crest and PPR domains arise, either via differential competence of the neural and non-neural ectoderm (binary competence model) or via interactions between the neural and non-neural ectoderm tissues to produce an intermediate neural border zone (NB) (border state model) that subsequently separates into neural crest and PPR. Many previous gain- and loss-of-function experiments demonstrate that numerous TFs are expressed in initially overlapping zones that gradually resolve into patterns that by late neurula stages are characteristic of each of the four domains. Several of these studies suggested that this is accomplished by a combination of repressive TF interactions and competence to respond to local signals. In this study, we ectopically expressed TFs that at neural plate stages are characteristic of one domain in a different domain to test whether they act cell autonomously as repressors. We found that almost all tested TFs caused reduced expression of the other TFs. At gastrulation these effects were strictly within the lineage-labeled cells, indicating that the effects were cell autonomous, i.e., due to TF interactions within individual cells. Analysis of previously published single cell RNAseq datasets showed that at the end of gastrulation, and continuing to neural tube closure stages, many ectodermal cells express TFs characteristic of more than one neural plate stage domain, indicating that different TFs have the opportunity to interact within the same cell. At neurula stages repression was observed both in the lineage-labeled cells and in adjacent cells not bearing detectable lineage label, suggesting that cell-to-cell signaling has begun to contribute to the separation of the domains. Together, these observations directly demonstrate previous suggestions in the literature that the segregation of embryonic ectodermal domains initially involves cell autonomous, repressive TF interactions within an individual cell followed by the subsequent advent of non-cell autonomous signaling to neighbors.
Collapse
Affiliation(s)
- Steven L Klein
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, D.C., DC, United States
| | - Andre L P Tavares
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, D.C., DC, United States
| | - Meredith Peterson
- Department of Biology, State College, Penn State University, University Park, PA, United States
| | | | - Sally A Moody
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, D.C., DC, United States
| |
Collapse
|
16
|
Ueda Y, Moore ST, Hashino E. Directed Differentiation of Human Pluripotent Stem Cells into Inner Ear Organoids. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2021; 2520:135-150. [PMID: 34724191 DOI: 10.1007/7651_2021_448] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The sensory epithelia of the inner ear contain mechanosensitive hair cells that detect sound and head acceleration. This protocol details a 3D differentiation method to generate inner ear organoids containing sensory epithelia with hair cells. Human pluripotent stem cells are aggregated in low-binding 96-well plates and treated in chemically defined media with extracellular matrix to promote epithelialization. Small molecules and recombinant proteins are applied in a stepwise manner to recapitulate the morphogenic cues (BMP, TGF-β, FGF, and WNT) present during inner ear development in vivo. These treatments induce the sequential formation of nonneural ectoderm, otic-epibranchial progenitor domain, and otic placodes. The derived otic placodes then undergo self-guided morphogenesis to form otic vesicles, which eventually give rise to sensory epithelia containing hair cells and supporting cells, as well as neurons with synaptic formations to hair cells. This human stem cell-derived inner ear organoid system provides an ideal platform to study human inner ear development and disease in vitro.
Collapse
Affiliation(s)
- Yoshitomo Ueda
- Department of Otolaryngology - Head and Neck Surgery, and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Stephen T Moore
- Department of Otolaryngology - Head and Neck Surgery, and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Eri Hashino
- Department of Otolaryngology - Head and Neck Surgery, and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
17
|
Almasoudi SH, Schlosser G. Otic Neurogenesis in Xenopus laevis: Proliferation, Differentiation, and the Role of Eya1. Front Neuroanat 2021; 15:722374. [PMID: 34616280 PMCID: PMC8488300 DOI: 10.3389/fnana.2021.722374] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/27/2021] [Indexed: 11/15/2022] Open
Abstract
Using immunostaining and confocal microscopy, we here provide the first detailed description of otic neurogenesis in Xenopus laevis. We show that the otic vesicle comprises a pseudostratified epithelium with apicobasal polarity (apical enrichment of Par3, aPKC, phosphorylated Myosin light chain, N-cadherin) and interkinetic nuclear migration (apical localization of mitotic, pH3-positive cells). A Sox3-immunopositive neurosensory area in the ventromedial otic vesicle gives rise to neuroblasts, which delaminate through breaches in the basal lamina between stages 26/27 and 39. Delaminated cells congregate to form the vestibulocochlear ganglion, whose peripheral cells continue to proliferate (as judged by EdU incorporation), while central cells differentiate into Islet1/2-immunopositive neurons from stage 29 on and send out neurites at stage 31. The central part of the neurosensory area retains Sox3 but stops proliferating from stage 33, forming the first sensory areas (utricular/saccular maculae). The phosphatase and transcriptional coactivator Eya1 has previously been shown to play a central role for otic neurogenesis but the underlying mechanism is poorly understood. Using an antibody specifically raised against Xenopus Eya1, we characterize the subcellular localization of Eya1 proteins, their levels of expression as well as their distribution in relation to progenitor and neuronal differentiation markers during otic neurogenesis. We show that Eya1 protein localizes to both nuclei and cytoplasm in the otic epithelium, with levels of nuclear Eya1 declining in differentiating (Islet1/2+) vestibulocochlear ganglion neurons and in the developing sensory areas. Morpholino-based knockdown of Eya1 leads to reduction of proliferating, Sox3- and Islet1/2-immunopositive cells, redistribution of cell polarity proteins and loss of N-cadherin suggesting that Eya1 is required for maintenance of epithelial cells with apicobasal polarity, progenitor proliferation and neuronal differentiation during otic neurogenesis.
Collapse
Affiliation(s)
| | - Gerhard Schlosser
- School of Natural Sciences, National University of Galway, Galway, Ireland
| |
Collapse
|
18
|
Washausen S, Knabe W. Responses of Epibranchial Placodes to Disruptions of the FGF and BMP Signaling Pathways in Embryonic Mice. Front Cell Dev Biol 2021; 9:712522. [PMID: 34589483 PMCID: PMC8473811 DOI: 10.3389/fcell.2021.712522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/17/2021] [Indexed: 11/21/2022] Open
Abstract
Placodes are ectodermal thickenings of the embryonic vertebrate head. Their descendants contribute to sensory organ development, but also give rise to sensory neurons of the cranial nerves. In mammals, the signaling pathways which regulate the morphogenesis and neurogenesis of epibranchial placodes, localized dorsocaudally to the pharyngeal clefts, are poorly understood. Therefore, we performed mouse whole embryo culture experiments to assess the impact of pan-fibroblast growth factor receptor (FGFR) inhibitors, anti-FGFR3 neutralizing antibodies or the pan-bone morphogenetic protein receptor (BMPR) inhibitor LDN193189 on epibranchial development. We demonstrate that each of the three paired epibranchial placodes is regulated by a unique combination of FGF and/or bone morphogenetic protein (BMP) signaling. Thus, neurogenesis depends on fibroblast growth factor (FGF) signals, albeit to different degrees, in all epibranchial placodes (EP), whereas only EP1 and EP3 significantly rely on neurogenic BMP signals. Furthermore, individual epibranchial placodes vary in the extent to which FGF and/or BMP signals (1) have access to certain receptor subtypes, (2) affect the production of Neurogenin (Ngn)2+ and/or Ngn1+ neuroblasts, and (3) regulate either neurogenesis alone or together with structural maintenance. In EP2 and EP3, all FGF-dependent production of Ngn2+ neuroblasts is mediated via FGFR3 whereas, in EP1, it depends on FGFR1 and FGFR3. Differently, production of FGF-dependent Ngn1+ neuroblasts almost completely depends on FGFR3 in EP1 and EP2, but not in EP3. Finally, FGF signals turned out to be responsible for the maintenance of both placodal thickening and neurogenesis in all epibranchial placodes, whereas administration of the pan-BMPR inhibitor, apart from its negative neurogenic effects in EP1 and EP3, causes only decreases in the thickness of EP3. Experimentally applied inhibitors most probably not only blocked receptors in the epibranchial placodes, but also endodermal receptors in the pharyngeal pouches, which act as epibranchial signaling centers. While high doses of pan-FGFR inhibitors impaired the development of all pharyngeal pouches, high doses of the pan-BMPR inhibitor negatively affected only the pharyngeal pouches 3 and 4. In combination with partly concordant, partly divergent findings in other vertebrate classes our observations open up new approaches for research into the complex regulation of neurogenic placode development.
Collapse
Affiliation(s)
- Stefan Washausen
- Prosektur Anatomie, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Wolfgang Knabe
- Prosektur Anatomie, Westfälische Wilhelms-Universität Münster, Münster, Germany
| |
Collapse
|
19
|
Abstract
Neural crest stem/progenitor cells arise early during vertebrate embryogenesis at the border of the forming central nervous system. They subsequently migrate throughout the body, eventually differentiating into diverse cell types ranging from neurons and glia of the peripheral nervous system to bones of the face, portions of the heart, and pigmentation of the skin. Along the body axis, the neural crest is heterogeneous, with different subpopulations arising in the head, neck, trunk, and tail regions, each characterized by distinct migratory patterns and developmental potential. Modern genomic approaches like single-cell RNA- and ATAC-sequencing (seq) have greatly enhanced our understanding of cell lineage trajectories and gene regulatory circuitry underlying the developmental progression of neural crest cells. Here, we discuss how genomic approaches have provided new insights into old questions in neural crest biology by elucidating transcriptional and posttranscriptional mechanisms that govern neural crest formation and the establishment of axial level identity. Expected final online publication date for the Annual Review of Genetics, Volume 55 is November 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Shashank Gandhi
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA; ,
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA; ,
| |
Collapse
|
20
|
Almasoudi SH, Schlosser G. Eya1 protein distribution during embryonic development of Xenopus laevis. Gene Expr Patterns 2021; 42:119213. [PMID: 34536585 DOI: 10.1016/j.gep.2021.119213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/10/2021] [Accepted: 09/10/2021] [Indexed: 11/24/2022]
Abstract
Eya1 and other Eya proteins are important regulators of progenitor proliferation, cell differentiation and morphogenesis in all three germ layers. At present, most of our knowledge of Eya1 distribution is based on in situ hybridization for Eya1 mRNA. However, to begin to dissect the mechanisms underlying Eya1 functions, we need a better understanding of the spatiotemporal distribution of Eya1 proteins during embryonic development, their subcellular localization and their levels of expression in various tissues. Here we report the localization of Eya1 protein throughout embryonic development from neural plate stages to tadpole stages of Xenopus laevis using a specific antibody for Xenopus Eya1. Our study confirms the expression of Eya1 protein in cranial placodes, placodally derived sensory primordia (olfactory epithelium, otic vesicle, lateral line primordia) and cranial ganglia, as well as in somites, secondary heart field and pharyngeal endoderm. In addition, we report here a novel expression of Eya1 proteins in scattered epidermal cells in Xenopus. Our findings also reveal that, while being predominantly expressed in nuclei in most expression domains, Eya1 protein is also localized to the cytoplasm, in particular in the early preplacodal ectoderm, some placode-derived ganglia and a subset of epidermal cells. While some cytoplasmic roles of Eya1 have been previously described in other contexts, the functions of cytoplasmic Eya1 in the preplacodal ectoderm, cranial ganglia and epidermal cells remain to be investigated.
Collapse
Affiliation(s)
| | - Gerhard Schlosser
- School of Natural Sciences, National University of Galway, Galway, Ireland.
| |
Collapse
|
21
|
Dubey A, Yu J, Liu T, Kane MA, Saint-Jeannet JP. Retinoic acid production, regulation and containment through Zic1, Pitx2c and Cyp26c1 control cranial placode specification. Development 2021; 148:dev193227. [PMID: 33531433 PMCID: PMC7903997 DOI: 10.1242/dev.193227] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 01/12/2021] [Indexed: 12/28/2022]
Abstract
All paired sensory organs arise from a common precursor domain called the pre-placodal region (PPR). In Xenopus, Zic1 non-cell autonomously regulates PPR formation by activating retinoic acid (RA) production. Here, we have identified two Zic1 targets, the RA catabolizing enzyme Cyp26c1 and the transcription factor Pitx2c, expressed in the vicinity of the PPR as being crucially required for maintaining low RA levels in a spatially restricted, PPR-adjacent domain. Morpholino- or CRISPR/Cas9-mediated Cyp26c1 knockdown abrogated PPR gene expression, yielding defective cranial placodes. Direct measurement of RA levels revealed that this is mediated by a mechanism involving excess RA accumulation. Furthermore, we show that pitx2c is activated by RA and required for Cyp26c1 expression in a domain-specific manner through induction of FGF8. We propose that Zic1 anteriorly establishes a program of RA containment and regulation through activation of Cyp26c1 and Pitx2c that cooperates to promote PPR specification in a spatially restricted domain.
Collapse
Affiliation(s)
- Aditi Dubey
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Jianshi Yu
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Tian Liu
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Jean-Pierre Saint-Jeannet
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| |
Collapse
|
22
|
Stundl J, Bertucci PY, Lauri A, Arendt D, Bronner ME. Evolution of new cell types at the lateral neural border. Curr Top Dev Biol 2021; 141:173-205. [PMID: 33602488 DOI: 10.1016/bs.ctdb.2020.11.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
During the course of evolution, animals have become increasingly complex by the addition of novel cell types and regulatory mechanisms. A prime example is represented by the lateral neural border, known as the neural plate border in vertebrates, a region of the developing ectoderm where presumptive neural and non-neural tissue meet. This region has been intensively studied as the source of two important embryonic cell types unique to vertebrates-the neural crest and the ectodermal placodes-which contribute to diverse differentiated cell types including the peripheral nervous system, pigment cells, bone, and cartilage. How did these multipotent progenitors originate in animal evolution? What triggered the elaboration of the border during the course of chordate evolution? How is the lateral neural border patterned in various bilaterians and what is its fate? Here, we review and compare the development and fate of the lateral neural border in vertebrates and invertebrates and we speculate about its evolutionary origin. Taken together, the data suggest that the lateral neural border existed in bilaterian ancestors prior to the origin of vertebrates and became a developmental source of exquisite evolutionary change that frequently enabled the acquisition of new cell types.
Collapse
Affiliation(s)
- Jan Stundl
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| | | | | | - Detlev Arendt
- European Molecular Biology Laboratory, Heidelberg, Germany.
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States.
| |
Collapse
|
23
|
Thawani A, Groves AK. Building the Border: Development of the Chordate Neural Plate Border Region and Its Derivatives. Front Physiol 2020; 11:608880. [PMID: 33364980 PMCID: PMC7750469 DOI: 10.3389/fphys.2020.608880] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/19/2020] [Indexed: 01/04/2023] Open
Abstract
The paired cranial sensory organs and peripheral nervous system of vertebrates arise from a thin strip of cells immediately adjacent to the developing neural plate. The neural plate border region comprises progenitors for four key populations of cells: neural plate cells, neural crest cells, the cranial placodes, and epidermis. Putative homologues of these neural plate border derivatives can be found in protochordates such as amphioxus and tunicates. In this review, we summarize key signaling pathways and transcription factors that regulate the inductive and patterning events at the neural plate border region that give rise to the neural crest and placodal lineages. Gene regulatory networks driven by signals from WNT, fibroblast growth factor (FGF), and bone morphogenetic protein (BMP) signaling primarily dictate the formation of the crest and placodal lineages. We review these studies and discuss the potential of recent advances in spatio-temporal transcriptomic and epigenomic analyses that would allow a mechanistic understanding of how these signaling pathways and their downstream transcriptional cascades regulate the formation of the neural plate border region.
Collapse
Affiliation(s)
- Ankita Thawani
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Andrew K Groves
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
24
|
Seal S, Monsoro-Burq AH. Insights Into the Early Gene Regulatory Network Controlling Neural Crest and Placode Fate Choices at the Neural Border. Front Physiol 2020; 11:608812. [PMID: 33324244 PMCID: PMC7726110 DOI: 10.3389/fphys.2020.608812] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/02/2020] [Indexed: 12/30/2022] Open
Abstract
The neural crest (NC) cells and cranial placodes are two ectoderm-derived innovations in vertebrates that led to the acquisition of a complex head structure required for a predatory lifestyle. They both originate from the neural border (NB), a portion of the ectoderm located between the neural plate (NP), and the lateral non-neural ectoderm. The NC gives rise to a vast array of tissues and cell types such as peripheral neurons and glial cells, melanocytes, secretory cells, and cranial skeletal and connective cells. Together with cells derived from the cranial placodes, which contribute to sensory organs in the head, the NC also forms the cranial sensory ganglia. Multiple in vivo studies in different model systems have uncovered the signaling pathways and genetic factors that govern the positioning, development, and differentiation of these tissues. In this literature review, we give an overview of NC and placode development, focusing on the early gene regulatory network that controls the formation of the NB during early embryonic stages, and later dictates the choice between the NC and placode progenitor fates.
Collapse
Affiliation(s)
- Subham Seal
- Université Paris-Saclay, CNRS UMR 3347, INSERM U1021, Orsay, France.,Institut Curie Research Division, PSL Research University, Orsay Cedex, France
| | - Anne H Monsoro-Burq
- Université Paris-Saclay, CNRS UMR 3347, INSERM U1021, Orsay, France.,Institut Curie Research Division, PSL Research University, Orsay Cedex, France.,Institut Universitaire de France, Paris, France
| |
Collapse
|
25
|
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
|
26
|
Abstract
The sensory epithelia of the inner ear contain mechanosensitive hair cells that transmit sound, gravity and head motion signals. This protocol describes an in vitro 3D differentiation method, by which the inner ear sensory epithelium harboring hair cells are derived from human pluripotent stem cells (hPSCs). To begin the differentiation, hPSCs are aggregated in low-binding 96-well plates and treated with extracellular matrix proteins to promote epithelialization. By recapitulating signaling pathway activation and attenuation during in vivo inner ear development, the aggregates are treated with small molecules and recombinant proteins that modulate signaling pathways such as BMP, FGF and WNT in a stepwise manner. These treatments induce sequential formation of non-neural ectoderm (NNE), otic-epibranchial progenitor domain (OEPD), and otic placodes. The otic placodes subsequently undergo self-guided morphogenesis to form otic vesicles, which eventually give rise to sensory epithelia containing inner ear hair cells and supporting cells, as well as neurons forming synapses with the hair cells. These hPSC-derived inner ear sensory structures are designated human inner ear organoids. As human inner ear biopsies are nearly impossible to obtain without causing severe injuries to the auditory system of the patients, the human inner ear organoid system provides a powerful in vitro platform for studying human inner ear disease and development.
Collapse
Affiliation(s)
- Jing Nie
- Department of Otolaryngology-Head and Neck Surgery, and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Eri Hashino
- Department of Otolaryngology-Head and Neck Surgery, and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
27
|
Tambalo M, Anwar M, Ahmed M, Streit A. Enhancer activation by FGF signalling during otic induction. Dev Biol 2020; 457:69-82. [PMID: 31539539 PMCID: PMC6902270 DOI: 10.1016/j.ydbio.2019.09.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/16/2019] [Accepted: 09/16/2019] [Indexed: 02/07/2023]
Abstract
Vertebrate ear progenitors are induced by fibroblast growth factor signalling, however the molecular mechanisms leading to the coordinate activation of downstream targets are yet to be discovered. The ear, like other sensory placodes, arises from the pre-placodal region at the border of the neural plate. Using a multiplex NanoString approach, we determined the response of these progenitors to FGF signalling by examining the changes of more than 200 transcripts that define the otic and other placodes, neural crest and neural plate territories. This analysis identifies new direct and indirect FGF targets during otic induction. Investigating changes in histone marks by ChIP-seq reveals that FGF exposure of pre-placodal cells leads to rapid deposition of active chromatin marks H3K27ac near FGF-response genes, while H3K27ac is depleted in the vicinity of non-otic genes. Genomic regions that gain H3K27ac act as cis-regulatory elements controlling otic gene expression in time and space and define a unique transcription factor signature likely to control their activity. Finally, we show that in response to FGF signalling the transcription factor dimer AP1 recruits the histone acetyl transferase p300 to selected otic enhancers. Thus, during ear induction FGF signalling modifies the chromatin landscape to promote enhancer activation and chromatin accessibility.
Collapse
Affiliation(s)
- Monica Tambalo
- Centre for Craniofacial and Regenerative Biology, Faculty of Dental, Oral and Craniofacial Sciences, King's College London, London, SE1 9RT, UK
| | - Maryam Anwar
- Centre for Craniofacial and Regenerative Biology, Faculty of Dental, Oral and Craniofacial Sciences, King's College London, London, SE1 9RT, UK
| | - Mohi Ahmed
- Centre for Craniofacial and Regenerative Biology, Faculty of Dental, Oral and Craniofacial Sciences, King's College London, London, SE1 9RT, UK
| | - Andrea Streit
- Centre for Craniofacial and Regenerative Biology, Faculty of Dental, Oral and Craniofacial Sciences, King's College London, London, SE1 9RT, UK.
| |
Collapse
|
28
|
Britton G, Heemskerk I, Hodge R, Qutub AA, Warmflash A. A novel self-organizing embryonic stem cell system reveals signaling logic underlying the patterning of human ectoderm. Development 2019; 146:dev.179093. [PMID: 31519692 DOI: 10.1242/dev.179093] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 09/06/2019] [Indexed: 12/20/2022]
Abstract
During development, the ectoderm is patterned by a combination of BMP and WNT signaling. Research in model organisms has provided substantial insight into this process; however, there are currently no systems in which to study ectodermal patterning in humans. Further, the complexity of neural plate border specification has made it difficult to transition from discovering the genes involved to deeper mechanistic understanding. Here, we develop an in vitro model of human ectodermal patterning, in which human embryonic stem cells self-organize to form robust and quantitatively reproducible patterns corresponding to the complete medial-lateral axis of the embryonic ectoderm. Using this platform, we show that the duration of endogenous WNT signaling is a crucial control parameter, and that cells sense relative levels of BMP and WNT signaling in making fate decisions. These insights allowed us to develop an improved protocol for placodal differentiation. Thus, our platform is a powerful tool for studying human ectoderm patterning and for improving directed differentiation protocols.This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- George Britton
- Systems Synthetic and Physical Biology Program, Rice University Houston, Houston, TX 77005, USA
| | - Idse Heemskerk
- Department of Biosciences, Rice University Houston, Houston, TX 77005, USA
| | - Rachel Hodge
- Department of Biosciences, Rice University Houston, Houston, TX 77005, USA
| | - Amina A Qutub
- Department of Bioengineering, Rice University Houston, Houston, TX 77005, USA
| | - Aryeh Warmflash
- Department of Biosciences, Rice University Houston, Houston, TX 77005, USA .,Department of Bioengineering, Rice University Houston, Houston, TX 77005, USA
| |
Collapse
|
29
|
Sullivan CH, Majumdar HD, Neilson KM, Moody SA. Six1 and Irx1 have reciprocal interactions during cranial placode and otic vesicle formation. Dev Biol 2019; 446:68-79. [PMID: 30529252 PMCID: PMC6349505 DOI: 10.1016/j.ydbio.2018.12.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 12/02/2018] [Accepted: 12/03/2018] [Indexed: 01/04/2023]
Abstract
The specialized sensory organs of the vertebrate head are derived from thickened patches of cells in the ectoderm called cranial sensory placodes. The developmental program that generates these placodes and the genes that are expressed during the process have been studied extensively in a number of animals, yet very little is known about how these genes regulate one another. We previously found via a microarray screen that Six1, a known transcriptional regulator of cranial placode fate, up-regulates Irx1 in ectodermal explants. In this study, we investigated the transcriptional relationship between Six1 and Irx1 and found that they reciprocally regulate each other throughout cranial placode and otic vesicle formation. Although Irx1 expression precedes that of Six1 in the neural border zone, its continued and appropriately patterned expression in the pre-placodal region (PPR) and otic vesicle requires Six1. At early PPR stages, Six1 expands the Irx1 domain, but this activity subsides over time and changes to a predominantly repressive effect. Likewise, Irx1 initially expands Six1 expression in the PPR, but later represses it. We also found that Irx1 and Sox11, a known direct target of Six1, reciprocally affect each other. This work demonstrates that the interactions between Six1 and Irx1 are continuous during PPR and placode development and their transcriptional effects on one another change over developmental time.
Collapse
Affiliation(s)
- Charles H Sullivan
- Department of Biology, Grinnell College, Grinnell, IA, 50112, USA; bDepartment of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, 2300 I (eye) Street, N.W., Washington DC 20037, USA
| | - Himani D Majumdar
- bDepartment of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, 2300 I (eye) Street, N.W., Washington DC 20037, USA
| | - Karen M Neilson
- bDepartment of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, 2300 I (eye) Street, N.W., Washington DC 20037, USA
| | - Sally A Moody
- bDepartment of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, 2300 I (eye) Street, N.W., Washington DC 20037, USA.
| |
Collapse
|
30
|
Watanabe T, Yamamoto T, Tsukano K, Hirano S, Horikawa A, Michiue T. Fam46a regulates BMP-dependent pre-placodal ectoderm differentiation in Xenopus. Development 2018; 145:dev.166710. [PMID: 30291163 DOI: 10.1242/dev.166710] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 09/06/2018] [Indexed: 12/30/2022]
Abstract
The pre-placodal ectoderm (PPE) is a specialized ectodermal region which gives rise to the sensory organs and other systems. The PPE is induced from the neural plate border during neurulation, but the molecular mechanism of PPE formation is not fully understood. In this study, we examined the role of a newly identified PPE gene, Fam46a, during embryogenesis. Fam46a contains a nucleoside triphosphate transferase domain, but its function in early development was previously unclear. We show that Fam46a is expressed in the PPE in Xenopus embryos, and Fam46a knockdown induces abnormalities in the eye formation and the body color. At the neurula stage, Fam46a upregulates the expression of PPE genes and inhibits neural crest formation. We also show that Fam46a physically interacts with Smad1/Smad4 and positively regulates BMP signaling. From these results, we conclude that Fam46a is required for PPE formation via the positive regulation of BMP signaling. Our study provides a new mechanism of ectodermal patterning via cell-autonomous regulation of BMP signaling in the PPE.
Collapse
Affiliation(s)
- Tomoko Watanabe
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Takayoshi Yamamoto
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Kohei Tsukano
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Sayuki Hirano
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Ayumi Horikawa
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Tatsuo Michiue
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8902, Japan
| |
Collapse
|
31
|
Shared evolutionary origin of vertebrate neural crest and cranial placodes. Nature 2018; 560:228-232. [PMID: 30069052 PMCID: PMC6390964 DOI: 10.1038/s41586-018-0385-7] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 06/13/2018] [Indexed: 12/21/2022]
|
32
|
A gene regulatory network underlying the formation of pre-placodal ectoderm in Xenopus laevis. BMC Biol 2018; 16:79. [PMID: 30012125 PMCID: PMC6048776 DOI: 10.1186/s12915-018-0540-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/14/2018] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND The neural plate border ectoderm gives rise to key developmental structures during embryogenesis, including the neural crest and the preplacodal ectoderm. Many sensory organs and ganglia of vertebrates develop from cranial placodes, which themselves arise from preplacodal ectoderm, defined by expression of transcription factor Six1 and its coactivator Eya1. Here we elucidate the gene regulatory network underlying the specification of the preplacodal ectoderm in Xenopus, and the functional interactions among transcription factors that give rise to this structure. RESULTS To elucidate the gene regulatory network upstream of preplacodal ectoderm formation, we use gain- and loss-of-function studies to explore the role of early ectodermal transcription factors for establishing the preplacodal ectoderm and adjacent ectodermal territories, and the role of Six1 and Eya1 in feedback regulation of these transcription factors. Our findings suggest that transcription factors with expression restricted to ventral (non-neural) ectoderm (AP2, Msx1, FoxI1, Vent2, Dlx3, GATA2) and those restricted to dorsal (neural) ectoderm (Pax3, Hairy2b, Zic1) are required for specification of both preplacodal ectoderm and neural crest in a context-dependent fashion and are cross-regulated by Eya1 and Six1. CONCLUSION These findings allow us to elucidate a detailed gene regulatory network at the neural plate border upstream of preplacodal ectoderm formation based on functional interactions between ectodermal transcription factors. We propose a new model to explain the formation of immediately juxtaposed preplacodal ectoderm and neural crest territories at the neural plate border, uniting previous models.
Collapse
|
33
|
Pla P, Monsoro-Burq AH. The neural border: Induction, specification and maturation of the territory that generates neural crest cells. Dev Biol 2018; 444 Suppl 1:S36-S46. [PMID: 29852131 DOI: 10.1016/j.ydbio.2018.05.018] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 05/23/2018] [Accepted: 05/23/2018] [Indexed: 11/17/2022]
Abstract
The neural crest is induced at the edge between the neural plate and the nonneural ectoderm, in an area called the neural (plate) border, during gastrulation and neurulation. In recent years, many studies have explored how this domain is patterned, and how the neural crest is induced within this territory, that also participates to the prospective dorsal neural tube, the dorsalmost nonneural ectoderm, as well as placode derivatives in the anterior area. This review highlights the tissue interactions, the cell-cell signaling and the molecular mechanisms involved in this dynamic spatiotemporal patterning, resulting in the induction of the premigratory neural crest. Collectively, these studies allow building a complex neural border and early neural crest gene regulatory network, mostly composed by transcriptional regulations but also, more recently, including novel signaling interactions.
Collapse
Affiliation(s)
- Patrick Pla
- Univ. Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Centre Universitaire, 15, rue Georges Clémenceau, F-91405 Orsay, France; Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, F-91405 Orsay, France
| | - Anne H Monsoro-Burq
- Univ. Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Centre Universitaire, 15, rue Georges Clémenceau, F-91405 Orsay, France; Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, F-91405 Orsay, France; Institut Universitaire de France, F-75005, Paris.
| |
Collapse
|
34
|
Dubey A, Rose RE, Jones DR, Saint-Jeannet JP. Generating retinoic acid gradients by local degradation during craniofacial development: One cell's cue is another cell's poison. Genesis 2018; 56:10.1002/dvg.23091. [PMID: 29330906 PMCID: PMC5818312 DOI: 10.1002/dvg.23091] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 01/02/2023]
Abstract
Retinoic acid (RA) is a vital morphogen for early patterning and organogenesis in the developing embryo. RA is a diffusible, lipophilic molecule that signals via nuclear RA receptor heterodimeric units that regulate gene expression by interacting with RA response elements in promoters of a significant number of genes. For precise RA signaling, a robust gradient of the morphogen is required. The developing embryo contains regions that produce RA, and specific intracellular concentrations of RA are created through local degradation mediated by Cyp26 enzymes. In order to elucidate the mechanisms by which RA executes precise developmental programs, the kinetics of RA metabolism must be clearly understood. Recent advances in techniques for endogenous RA detection and quantification have paved the way for mechanistic studies to shed light on downstream gene expression regulation coordinated by RA. It is increasingly coming to light that RA signaling operates not only at precise concentrations but also employs mechanisms of degradation and feedback inhibition to self-regulate its levels. A global gradient of RA throughout the embryo is often found concurrently with several local gradients, created by juxtaposed domains of RA synthesis and degradation. The existence of such local gradients has been found especially critical for the proper development of craniofacial structures that arise from the neural crest and the cranial placode populations. In this review, we summarize the current understanding of how local gradients of RA are established in the embryo and their impact on craniofacial development.
Collapse
Affiliation(s)
- Aditi Dubey
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry
| | - Rebecca E. Rose
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Health, New York, NY, USA
| | - Drew R. Jones
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Health, New York, NY, USA
| | | |
Collapse
|
35
|
Identification of novel cis-regulatory elements of Eya1 in Xenopus laevis using BAC recombineering. Sci Rep 2017; 7:15033. [PMID: 29101371 PMCID: PMC5670250 DOI: 10.1038/s41598-017-15153-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 10/23/2017] [Indexed: 12/13/2022] Open
Abstract
The multifunctional Eya1 protein plays important roles during the development of cranial sensory organs and ganglia, kidneys, hypaxial muscles and several other organs in vertebrates. Eya1 is encoded by a complex locus with candidate cis-regulatory elements distributed over a 329 kbp wide genomic region in Xenopus. Consequently, very little is currently known about how expression of Eya1 is controlled by upstream regulators. Here we use a library of Xenopus tropicalis genomic sequences in bacterial artificial chromosomes (BAC) to analyze the genomic region surrounding the Eya1 locus for enhancer activity. We used BAC recombineering to first create GFP reporter constructs, which were analysed for enhancer activity by injection into Xenopus laevis embryos. We then used a second round of BAC recombineering to create deletion constructs of these BAC reporters to localize enhancer activity more precisely. This double recombineering approach allowed us to probe a large genomic region for enhancer activity without assumptions on sequence conservation. Using this approach we were able to identify two novel cis-regulatory regions, which direct Eya1 expression to the somites, pharyngeal pouches, the preplacodal ectoderm (the common precursor region of many cranial sensory organs and ganglia), and other ectodermal domains.
Collapse
|
36
|
Six1 and Eya1 both promote and arrest neuronal differentiation by activating multiple Notch pathway genes. Dev Biol 2017; 431:152-167. [PMID: 28947179 DOI: 10.1016/j.ydbio.2017.09.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/19/2017] [Accepted: 09/19/2017] [Indexed: 12/12/2022]
Abstract
The transcription factor Six1 and its cofactor Eya1 are important regulators of neurogenesis in cranial placodes, activating genes promoting both a progenitor state, such as hes8, and neuronal differentiation, such as neurog1. Here, we use gain and loss of function studies in Xenopus laevis to elucidate how these genes function during placodal neurogenesis. We first establish that hes8 is activated by Notch signaling and represses neurog1 and neuronal differentiation, indicating that it mediates lateral inhibition. Using hes8 knockdown we demonstrate that hes8 is essential for limiting neuronal differentiation during normal placode development. We next show that Six1 and Eya1 cell autonomously activate both hes8 and neurog1 in a dose-dependent fashion, with increasing upregulation at higher doses, while neuronal differentiation is increasingly repressed. However, high doses of Six1 and Eya1 upregulate neurog1 only transiently, whereas low doses of Six1 and Eya1 ultimately promote both neurog1 expression and neuronal differentiation. Finally, we show that Six1 and Eya1 can activate hes8 and arrest neuronal differentiation even when Notch signaling is blocked. Our findings indicate that Six1 and Eya1 can both promote and arrest neuronal differentiation by activating the Notch pathway genes neurog1 and hes8, respectively, revealing a novel mechanism of Six1/Eya1 action during placodal neurogenesis.
Collapse
|
37
|
Hinaux H, Devos L, Blin M, Elipot Y, Bibliowicz J, Alié A, Rétaux S. Sensory evolution in blind cavefish is driven by early embryonic events during gastrulation and neurulation. Development 2017; 143:4521-4532. [PMID: 27899509 DOI: 10.1242/dev.141291] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 10/20/2016] [Indexed: 11/20/2022]
Abstract
Natural variations in sensory systems constitute adaptive responses to the environment. Here, we compared sensory placode development in the blind cave-adapted morph and the eyed river-dwelling morph of Astyanax mexicanus Focusing on the lens and olfactory placodes, we found a trade-off between these two sensory components in the two morphs: from neural plate stage onwards, cavefish have larger olfactory placodes and smaller lens placodes. In a search for developmental mechanisms underlying cavefish sensory evolution, we analyzed the roles of Shh, Fgf8 and Bmp4 signaling, which are known to be fundamental in patterning the vertebrate head and are subtly modulated in space and time during cavefish embryogenesis. Modulating these signaling systems at the end of gastrulation shifted the balance toward a larger olfactory derivative. Olfactory tests to assess potential behavioral outcomes of such developmental evolution revealed that Astyanax cavefish are able to respond to a 105-fold lower concentration of amino acids than their surface-dwelling counterparts. We suggest that similar evolutionary developmental mechanisms may be used throughout vertebrates to drive adaptive sensory specializations according to lifestyle and habitat.
Collapse
Affiliation(s)
- Hélène Hinaux
- DECA group, Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Université Paris-Saclay, Avenue de la terrasse, Gif-sur-Yvette 91198, France
| | - Lucie Devos
- DECA group, Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Université Paris-Saclay, Avenue de la terrasse, Gif-sur-Yvette 91198, France
| | - Maryline Blin
- DECA group, Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Université Paris-Saclay, Avenue de la terrasse, Gif-sur-Yvette 91198, France
| | - Yannick Elipot
- DECA group, Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Université Paris-Saclay, Avenue de la terrasse, Gif-sur-Yvette 91198, France
| | - Jonathan Bibliowicz
- DECA group, Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Université Paris-Saclay, Avenue de la terrasse, Gif-sur-Yvette 91198, France
| | - Alexandre Alié
- DECA group, Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Université Paris-Saclay, Avenue de la terrasse, Gif-sur-Yvette 91198, France
| | - Sylvie Rétaux
- DECA group, Paris-Saclay Institute of Neuroscience, CNRS, Université Paris Sud, Université Paris-Saclay, Avenue de la terrasse, Gif-sur-Yvette 91198, France
| |
Collapse
|
38
|
Hintze M, Prajapati RS, Tambalo M, Christophorou NAD, Anwar M, Grocott T, Streit A. Cell interactions, signals and transcriptional hierarchy governing placode progenitor induction. Development 2017; 144:2810-2823. [PMID: 28684624 PMCID: PMC5560042 DOI: 10.1242/dev.147942] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 06/21/2017] [Indexed: 12/18/2022]
Abstract
In vertebrates, cranial placodes contribute to all sense organs and sensory ganglia and arise from a common pool of Six1/Eya2+ progenitors. Here we dissect the events that specify ectodermal cells as placode progenitors using newly identified genes upstream of the Six/Eya complex. We show in chick that two different tissues, namely the lateral head mesoderm and the prechordal mesendoderm, gradually induce placode progenitors: cells pass through successive transcriptional states, each identified by distinct factors and controlled by different signals. Both tissues initiate a common transcriptional state but over time impart regional character, with the acquisition of anterior identity dependent on Shh signalling. Using a network inference approach we predict the regulatory relationships among newly identified transcription factors and verify predicted links in knockdown experiments. Based on this analysis we propose a new model for placode progenitor induction, in which the initial induction of a generic transcriptional state precedes regional divergence.
Collapse
Affiliation(s)
- Mark Hintze
- Department of Craniofacial Development & Stem Cell Biology, King's College London, Dental Institute, London SE1 9RT, UK
| | - Ravindra Singh Prajapati
- Department of Craniofacial Development & Stem Cell Biology, King's College London, Dental Institute, London SE1 9RT, UK
| | - Monica Tambalo
- Department of Craniofacial Development & Stem Cell Biology, King's College London, Dental Institute, London SE1 9RT, UK
| | - Nicolas A D Christophorou
- Department of Craniofacial Development & Stem Cell Biology, King's College London, Dental Institute, London SE1 9RT, UK
| | - Maryam Anwar
- Department of Craniofacial Development & Stem Cell Biology, King's College London, Dental Institute, London SE1 9RT, UK
| | - Timothy Grocott
- Department of Craniofacial Development & Stem Cell Biology, King's College London, Dental Institute, London SE1 9RT, UK
| | - Andrea Streit
- Department of Craniofacial Development & Stem Cell Biology, King's College London, Dental Institute, London SE1 9RT, UK
| |
Collapse
|
39
|
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]
|
40
|
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]
|
41
|
Chen J, Tambalo M, Barembaum M, Ranganathan R, Simões-Costa M, Bronner ME, Streit A. A systems-level approach reveals new gene regulatory modules in the developing ear. Development 2017; 144:1531-1543. [PMID: 28264836 PMCID: PMC5399671 DOI: 10.1242/dev.148494] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 02/24/2017] [Indexed: 01/23/2023]
Abstract
The inner ear is a complex vertebrate sense organ, yet it arises from a simple epithelium, the otic placode. Specification towards otic fate requires diverse signals and transcriptional inputs that act sequentially and/or in parallel. Using the chick embryo, we uncover novel genes in the gene regulatory network underlying otic commitment and reveal dynamic changes in gene expression. Functional analysis of selected transcription factors reveals the genetic hierarchy underlying the transition from progenitor to committed precursor, integrating known and novel molecular players. Our results not only characterize the otic transcriptome in unprecedented detail, but also identify new gene interactions responsible for inner ear development and for the segregation of the otic lineage from epibranchial progenitors. By recapitulating the embryonic programme, the genes and genetic sub-circuits discovered here might be useful for reprogramming naïve cells towards otic identity to restore hearing loss. Summary: Transcriptome analysis and knock down of select transcription factors reveals a genetic hierarchy as cells become committed to inner ear fate.
Collapse
Affiliation(s)
- Jingchen Chen
- Department of Craniofacial Development and Stem Cell Biology, King's College London, London SE1 9RT, UK
| | - Monica Tambalo
- Department of Craniofacial Development and Stem Cell Biology, King's College London, London SE1 9RT, UK
| | - Meyer Barembaum
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Ramya Ranganathan
- Department of Craniofacial Development and Stem Cell Biology, King's College London, London SE1 9RT, UK
| | - Marcos Simões-Costa
- 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
| | - Andrea Streit
- Department of Craniofacial Development and Stem Cell Biology, King's College London, London SE1 9RT, UK
| |
Collapse
|
42
|
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
|
43
|
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
|
44
|
Riddiford N, Schlosser G. Dissecting the pre-placodal transcriptome to reveal presumptive direct targets of Six1 and Eya1 in cranial placodes. eLife 2016; 5. [PMID: 27576864 PMCID: PMC5035141 DOI: 10.7554/elife.17666] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 08/29/2016] [Indexed: 11/13/2022] Open
Abstract
The pre-placodal ectoderm, marked by the expression of the transcription factor Six1 and its co-activator Eya1, develops into placodes and ultimately into many cranial sensory organs and ganglia. Using RNA-Seq in Xenopus laevis we screened for presumptive direct placodal target genes of Six1 and Eya1 by overexpressing hormone-inducible constructs of Six1 and Eya1 in pre-placodal explants, and blocking protein synthesis before hormone-inducing nuclear translocation of Six1 or Eya1. Comparing the transcriptome of explants with non-induced controls, we identified hundreds of novel Six1/Eya1 target genes with potentially important roles for placode development. Loss-of-function studies confirmed that target genes encoding known transcriptional regulators of progenitor fates (e.g. Sox2, Hes8) and neuronal/sensory differentiation (e.g. Ngn1, Atoh1, Pou4f1, Gfi1) require Six1 and Eya1 for their placodal expression. Our findings provide insights into the gene regulatory network regulating placodal neurogenesis downstream of Six1 and Eya1 suggesting new avenues of research into placode development and disease.
Collapse
Affiliation(s)
- Nick Riddiford
- School of Natural Sciences, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute (REMEDI), National University of Ireland, Galway, Ireland
| | - Gerhard Schlosser
- School of Natural Sciences, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute (REMEDI), National University of Ireland, Galway, Ireland
| |
Collapse
|
45
|
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
|
46
|
Kawasaki T, Takahashi M, Yajima H, Mori Y, Kawakami K. Six1 is required for mouse dental follicle cell and human periodontal ligament-derived cell proliferation. Dev Growth Differ 2016; 58:530-45. [DOI: 10.1111/dgd.12291] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/28/2016] [Accepted: 04/12/2016] [Indexed: 12/23/2022]
Affiliation(s)
- Tatsuki Kawasaki
- Department of Oral and Maxillofacial Surgery; Jichi Medical University; 3311-1, Yakushiji Shimotsuke Tochigi 329-0498 Japan
- Division of Biology; Center for Molecular Medicine; Jichi Medical University; 3311-1, Yakushiji Shimotsuke Tochigi 329-0498 Japan
| | - Masanori Takahashi
- Division of Biology; Center for Molecular Medicine; Jichi Medical University; 3311-1, Yakushiji Shimotsuke Tochigi 329-0498 Japan
| | - Hiroshi Yajima
- Division of Biology; Center for Molecular Medicine; Jichi Medical University; 3311-1, Yakushiji Shimotsuke Tochigi 329-0498 Japan
| | - Yoshiyuki Mori
- Department of Oral and Maxillofacial Surgery; Jichi Medical University; 3311-1, Yakushiji Shimotsuke Tochigi 329-0498 Japan
| | - Kiyoshi Kawakami
- Division of Biology; Center for Molecular Medicine; Jichi Medical University; 3311-1, Yakushiji Shimotsuke Tochigi 329-0498 Japan
| |
Collapse
|
47
|
Singh S, Groves AK. The molecular basis of craniofacial placode development. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2016; 5:363-76. [PMID: 26952139 DOI: 10.1002/wdev.226] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/22/2015] [Accepted: 12/27/2015] [Indexed: 12/20/2022]
Abstract
The sensory organs of the vertebrate head originate from simple ectodermal structures known as cranial placodes. All cranial placodes derive from a common domain adjacent to the neural plate, the preplacodal region, which is induced at the border of neural and non-neural ectoderm during gastrulation. Induction and specification of the preplacodal region is regulated by the fibroblast growth factor, bone morphogenetic protein, WNT, and retinoic acid signaling pathways, and characterized by expression of the EYA and SIX family of transcriptional regulators. Once the preplacodal region is specified, different combinations of local signaling molecules and placode-specific transcription factors, including competence factors, promote the induction of individual cranial placodes along the neural axis of the head region. In this review, we summarize the steps of cranial placode development and discuss the roles of the main signaling molecules and transcription factors that regulate these steps during placode induction, specification, and development. For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Sunita Singh
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Andrew K Groves
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
48
|
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
|
49
|
Hirayama M, Kawakita T, Tsubota K, Shimmura S. Challenges and Strategies for Regenerating the Lacrimal Gland. Ocul Surf 2015; 14:135-43. [PMID: 26738799 DOI: 10.1016/j.jtos.2015.11.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 11/02/2015] [Accepted: 11/04/2015] [Indexed: 02/04/2023]
Abstract
The lacrimal gland produces the aqueous component of tears, including electrolytes, peptides, and glycoproteins necessary to maintain homeostasis and optical properties of the ocular surface. Stem cells that contribute to the homeostasis of the lacrimal gland are under extensive study. It is still unclear whether such stem cells are of mesenchymal or epithelial origin. It is also possible that a unique epithelial stem cell undergoes epithelial-mesenchymal transition and contributes to the mesenchyme. Developmental studies in mice have shown that a network of growth factors contributes to epithelial-mesenchymal interaction during morphogenesis of the lacrimal gland. Recently, the developmental process was successfully recapitulated in vitro, providing a valuable tool for study of lacrimal gland development and possibly opening doors to regenerative therapy. While further studies are required to identify and appreciate the potential of lacrimal gland stem cells, advances in stem cell biology in general should become a catalyst towards developing regenerative therapy of the lacrimal gland.
Collapse
Affiliation(s)
- Masatoshi Hirayama
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Tetsuya Kawakita
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Shigeto Shimmura
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
50
|
Birol O, Ohyama T, Edlund RK, Drakou K, Georgiades P, Groves AK. The mouse Foxi3 transcription factor is necessary for the development of posterior placodes. Dev Biol 2015; 409:139-151. [PMID: 26550799 DOI: 10.1016/j.ydbio.2015.09.022] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 09/22/2015] [Indexed: 02/01/2023]
Abstract
The inner ear develops from the otic placode, one of the cranial placodes that arise from a region of ectoderm adjacent to the anterior neural plate called the pre-placodal domain. We have identified a Forkhead family transcription factor, Foxi3, that is expressed in the pre-placodal domain and down-regulated when the otic placode is induced. We now show that Foxi3 mutant mice do not form otic placodes as evidenced by expression changes in early molecular markers and the lack of thickened placodal ectoderm, an otic cup or otocyst. Some preplacodal genes downstream of Foxi3-Gata3, Six1 and Eya1-are not expressed in the ectoderm of Foxi3 mutant mice, and the ectoderm exhibits signs of increased apoptosis. We also show that Fgf signals from the hindbrain and cranial mesoderm, which are necessary for otic placode induction, are received by pre-placodal ectoderm in Foxi3 mutants, but do not initiate otic induction. Finally, we show that the epibranchial placodes that develop in close proximity to the otic placode and the mandibular division of the trigeminal ganglion are missing in Foxi3 mutants. Our data suggest that Foxi3 is necessary to prime pre-placodal ectoderm for the correct interpretation of inductive signals for the otic and epibranchial placodes.
Collapse
Affiliation(s)
- Onur Birol
- Program in Developmental Biology, Baylor College of Medicine, BCM295, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Takahiro Ohyama
- USC Caruso Department of Otolaryngology - Head & Neck Surgery, Keck Medicine of USC, University of Southern California, 1501 San Pablo Street, Los Angeles, CA 90033-4503, USA; Zilkha Neurogenetic Institute, Keck Medicine of USC, University of Southern California, 1501 San Pablo Street, Los Angeles, CA 90033-4503, USA
| | - Renée K Edlund
- Program in Developmental Biology, Baylor College of Medicine, BCM295, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Katerina Drakou
- Department of Biological Sciences, University of Cyprus, 1 University Avenue, P.O. Box 20537, 1678 Nicosia, Cyprus
| | - Pantelis Georgiades
- Department of Biological Sciences, University of Cyprus, 1 University Avenue, P.O. Box 20537, 1678 Nicosia, Cyprus
| | - Andrew K Groves
- Program in Developmental Biology, Baylor College of Medicine, BCM295, 1 Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, BCM295, 1 Baylor Plaza, Houston, TX 77030, USA; Department of Neurosc ience, Baylor College of Medicine, BCM295, 1 Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|