1
|
Kamenev D, Kameneva P, Adameyko I. The role of microheterogeneity in cell fate decisions in neural progenitors and neural crest. Curr Opin Neurobiol 2025; 92:103031. [PMID: 40288017 DOI: 10.1016/j.conb.2025.103031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 01/23/2025] [Accepted: 04/05/2025] [Indexed: 04/29/2025]
Abstract
Neuroprogenitors must integrate a multitude of signals, including gradients of morphogens, transcriptional programs, and temporal cues to generate an astonishing diversity of cell types inhabiting the nervous system. How do these different layers of information come together to influence cell fate in progenitor cells in a coordinated way? Here we provide a nuanced perspective on cell fate selection in the nervous system and neural crest lineage, suggesting that it is not a straightforward, deterministic process governed by rigid on-off switches. Instead, the process involves probabilistic transitions influenced by small variations - termed "microheterogeneity" - within a progenitor cell population. These minuscule differences between individual neural progenitor cells can result in significantly different outcomes, making certain fates more probable for some cells than others. Here we discuss the diversity of such examples and the theory behind, also providing future perspectives.
Collapse
Affiliation(s)
- Dmitrii Kamenev
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, 1090 Vienna, Austria
| | - Polina Kameneva
- St. Anna Children's Cancer Research Institute (CCRI), 1090 Vienna, Austria.
| | - Igor Adameyko
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, 1090 Vienna, Austria; Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden.
| |
Collapse
|
2
|
Edens BM, Bronner ME. Making sense of vertebrate senses from a neural crest and cranial placode evo-devo perspective. Trends Neurosci 2025; 48:213-226. [PMID: 39848838 PMCID: PMC11903184 DOI: 10.1016/j.tins.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 11/27/2024] [Accepted: 12/20/2024] [Indexed: 01/25/2025]
Abstract
The evolution of vertebrates from protochordate ancestors marked the beginning of the gradual transition to predatory lifestyles. Enabled by the acquisition of multipotent neural crest and cranial placode cell populations, vertebrates developed an elaborate peripheral nervous system, equipped with paired sense organs, which aided in adaptive behaviors and ultimately, successful colonization of diverse environmental niches. Underpinning the enduring success of vertebrates is the highly adaptable nature of the peripheral nervous system, which is enabled by the exceptional malleability of the neural crest and placode developmental programs. Here, we explore the embryonic origins of the vertebrate senses from the neural crest and cranial placodes and discuss the evolutionary trajectory of the senses in the context of adaptation to novel environments.
Collapse
Affiliation(s)
- Brittany M Edens
- 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
|
3
|
Drake PM, Franz‐Odendaal TA. Hydrocortisone treatment as a tool to study conjunctival placode induction. Dev Dyn 2025; 254:74-93. [PMID: 39096180 PMCID: PMC11737293 DOI: 10.1002/dvdy.729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/27/2024] [Accepted: 07/10/2024] [Indexed: 08/05/2024] Open
Abstract
BACKGROUND Conjunctival placodes are a series of placodes that develop into the conjunctival (scleral) papillae and ultimately induce a series of scleral ossicles in the eyes of many vertebrates. This study establishes a hydrocortisone injection procedure (incl. dosage) that consistently inhibits all conjunctival papillae in the embryonic chicken eye. The effects of this hydrocortisone treatment on apoptosis, vasculature, and placode-related gene expression were assessed. RESULTS Hydrocortisone treatment does not increase apoptotic cell death or have a major effect on the ciliary artery or vascular plexus in the eye. β-catenin and Eda expression levels were not significantly altered following hydrocortisone treatment, despite the absence of conjunctival papillae. Notably, Fgf20 expression was significantly reduced following hydrocortisone treatment, and the distribution of β-catenin was altered. CONCLUSIONS Our study showed that conjunctival papillae induction begins as early as HH27.5 (E5.5). Hydrocortisone treatment reduces Fgf20 expression independently of β-catenin and Eda and may instead affect other members of the Wnt/β-catenin or Eda/Edar pathways, or it may affect the ability of morphogens to diffuse through the extracellular matrix. This study contributes to a growing profile of gene expression data during placode development and enhances our understanding of how some vertebrate eyes develop these fascinating bones.
Collapse
Affiliation(s)
- Paige M. Drake
- Department of Medical NeuroscienceDalhousie UniversityHalifaxNova ScotiaCanada
| | | |
Collapse
|
4
|
Ge H, Di G, Song P, Han W, Chen P, Wang Y. Role of vitamin A on the ocular surface. Exp Eye Res 2025; 250:110179. [PMID: 39581361 DOI: 10.1016/j.exer.2024.110179] [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: 07/24/2024] [Revised: 11/14/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
Vitamin A is an essential fat-soluble vitamin that cannot be endogenously synthesized by the human body. Retinoic acid (RA) is the biologically active form of vitamin A. Utilizing both nuclear and non-nuclear receptor-mediated pathways, RA plays a crucial role in regulating various biological processes, including apoptosis, differentiation, and anti-inflammatory properties within the cornea and conjunctiva. In addition, RA has been demonstrated to exert a significant influence on anti-tumor mechanisms. Disruption of RA signaling can result in corneal defects, anophthalmia, and microphthalmia. However, the beneficial effects of RA are only observed when it is administered at appropriate dosages, and higher doses have an adverse impact. Ocular abnormalities are often early indicators of a vitamin A deficiency. The lacrimal gland secretes vitamin A onto the ocular surface, where it is metabolized into RA via two sequential steps. This article provides a comprehensive overview of how vitamin A is transformed and transported from the intestine to the ocular surface, ultimately contributing to the maintenance of the normal physiological function of the ocular surface.
Collapse
Affiliation(s)
- Huanhuan Ge
- School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong Province, China
| | - Guohu Di
- School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong Province, China; Institute of Stem Cell Regeneration Medicine, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Peirong Song
- School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong Province, China
| | - Wenshuo Han
- School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong Province, China
| | - Peng Chen
- School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong Province, China; Department of Ophthalmology, Qingdao Eighth People's Hospital, Qingdao, Shandong, 266121, China; Institute of Stem Cell Regeneration Medicine, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| | - Ye Wang
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, Shandong, 266042, China.
| |
Collapse
|
5
|
Neal SJ, Rajasekaran A, Jusić N, Taylor L, Read M, Alfandari D, Pignoni F, Moody SA. Using Xenopus to discover new candidate genes involved in BOR and other congenital hearing loss syndromes. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART B, MOLECULAR AND DEVELOPMENTAL EVOLUTION 2024; 342:212-240. [PMID: 37830236 PMCID: PMC11014897 DOI: 10.1002/jez.b.23222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/15/2023] [Accepted: 09/14/2023] [Indexed: 10/14/2023]
Abstract
Hearing in infants is essential for brain development, acquisition of verbal language skills, and development of social interactions. Therefore, it is important to diagnose hearing loss soon after birth so that interventions can be provided as early as possible. Most newborns in the United States are screened for hearing deficits and commercially available next-generation sequencing hearing loss panels often can identify the causative gene, which may also identify congenital defects in other organs. One of the most prevalent autosomal dominant congenital hearing loss syndromes is branchio-oto-renal syndrome (BOR), which also presents with defects in craniofacial structures and the kidney. Currently, mutations in three genes, SIX1, SIX5, and EYA1, are known to be causative in about half of the BOR patients that have been tested. To uncover new candidate genes that could be added to congenital hearing loss genetic screens, we have combined the power of Drosophila mutants and protein biochemical assays with the embryological advantages of Xenopus, a key aquatic animal model with a high level of genomic similarity to human, to identify potential Six1 transcriptional targets and interacting proteins that play a role during otic development. We review our transcriptomic, yeast 2-hybrid, and proteomic approaches that have revealed a large number of new candidates. We also discuss how we have begun to identify how Six1 and co-factors interact to direct developmental events necessary for normal otic development.
Collapse
Affiliation(s)
- Scott J. Neal
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY, USA
| | - Anindita Rajasekaran
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY, USA
| | - Nisveta Jusić
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY, USA
| | - Louis Taylor
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, USA
| | - Mai Read
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, USA
| | - Dominique Alfandari
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, USA
| | - Francesca Pignoni
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY, USA
| | - Sally A. Moody
- Department of Anatomy and Cell Biology, George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
6
|
Anselmi C, Fuller GK, Stolfi A, Groves AK, Manni L. Sensory cells in tunicates: insights into mechanoreceptor evolution. Front Cell Dev Biol 2024; 12:1359207. [PMID: 38550380 PMCID: PMC10973136 DOI: 10.3389/fcell.2024.1359207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Tunicates, the sister group of vertebrates, offer a unique perspective for evolutionary developmental studies (Evo-Devo) due to their simple anatomical organization. Moreover, the separation of tunicates from vertebrates predated the vertebrate-specific genome duplications. As adults, they include both sessile and pelagic species, with very limited mobility requirements related mainly to water filtration. In sessile species, larvae exhibit simple swimming behaviors that are required for the selection of a suitable substrate on which to metamorphose. Despite their apparent simplicity, tunicates display a variety of mechanoreceptor structures involving both primary and secondary sensory cells (i.e., coronal sensory cells). This review encapsulates two decades of research on tunicate mechanoreception focusing on the coronal organ's sensory cells as prime candidates for understanding the evolution of vertebrate hair cells of the inner ear and the lateral line organ. The review spans anatomical, cellular and molecular levels emphasizing both similarity and differences between tunicate and vertebrate mechanoreception strategies. The evolutionary significance of mechanoreception is discussed within the broader context of Evo-Devo studies, shedding light on the intricate pathways that have shaped the sensory system in chordates.
Collapse
Affiliation(s)
- Chiara Anselmi
- Hopkins Marine Station, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Pacific Grove, CA, United States
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, United States
| | - Gwynna K. Fuller
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Alberto Stolfi
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States
| | - Andrew K. Groves
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Lucia Manni
- Dipartimento di Biologia, Università degli Studi di Padova, Padova, Italy
| |
Collapse
|
7
|
Edens BM, Bronner ME. Making developmental sense of the senses, their origin and function. Curr Top Dev Biol 2024; 159:132-167. [PMID: 38729675 DOI: 10.1016/bs.ctdb.2024.01.015] [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: 05/12/2024]
Abstract
The primary senses-touch, taste, sight, smell, and hearing-connect animals with their environments and with one another. Aside from the eyes, the primary sense organs of vertebrates and the peripheral sensory pathways that relay their inputs arise from two transient stem cell populations: the neural crest and the cranial placodes. In this chapter we consider the senses from historical and cultural perspectives, and discuss the senses as biological faculties. We begin with the embryonic origin of the neural crest and cranial placodes from within the neural plate border of the ectodermal germ layer. Then, we describe the major chemical (i.e. olfactory and gustatory) and mechanical (i.e. vestibulo-auditory and somatosensory) senses, with an emphasis on the developmental interactions between neural crest and cranial placodes that shape their structures and functions.
Collapse
Affiliation(s)
- Brittany M Edens
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States.
| |
Collapse
|
8
|
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: 0] [Impact Index Per Article: 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
|
9
|
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
|
10
|
Roccio M. Directed differentiation and direct reprogramming: Applying stem cell technologies to hearing research. Stem Cells 2020; 39:375-388. [PMID: 33378797 DOI: 10.1002/stem.3315] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/24/2020] [Accepted: 12/01/2020] [Indexed: 12/26/2022]
Abstract
Hearing loss is the most widely spread sensory disorder in our society. In the majority of cases, it is caused by the loss or malfunctioning of cells in the cochlea: the mechanosensory hair cells, which act as primary sound receptors, and the connecting auditory neurons of the spiral ganglion, which relay the signal to upper brain centers. In contrast to other vertebrates, where damage to the hearing organ can be repaired through the activity of resident cells, acting as tissue progenitors, in mammals, sensory cell damage or loss is irreversible. The understanding of gene and cellular functions, through analysis of different animal models, has helped to identify causes of disease and possible targets for hearing restoration. Translation of these findings to novel therapeutics is, however, hindered by the lack of cellular assays, based on human sensory cells, to evaluate the conservation of molecular pathways across species and the efficacy of novel therapeutic strategies. In the last decade, stem cell technologies enabled to generate human sensory cell types in vitro, providing novel tools to study human inner ear biology, model disease, and validate therapeutics. This review focuses specifically on two technologies: directed differentiation of pluripotent stem cells and direct reprogramming of somatic cell types to sensory hair cells and neurons. Recent development in the field are discussed as well as how these tools could be implemented to become routinely adopted experimental models for hearing research.
Collapse
Affiliation(s)
- Marta Roccio
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich (USZ), and University of Zurich (UZH), Zurich, Switzerland
| |
Collapse
|
11
|
Taste buds are not derived from neural crest in mouse, chicken, and zebrafish. Dev Biol 2020; 471:76-88. [PMID: 33326797 DOI: 10.1016/j.ydbio.2020.12.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
Our lineage tracing studies using multiple Cre mouse lines showed a concurrent labeling of abundant taste bud cells and the underlying connective tissue with a neural crest (NC) origin, warranting a further examination on the issue of whether there is an NC derivation of taste bud cells. In this study, we mapped NC cell lineages in three different models, Sox10-iCreERT2/tdT mouse, GFP+ neural fold transplantation to GFP- chickens, and Sox10-Cre/GFP-RFP zebrafish model. We found that in mice, Sox10-iCreERT2 specifically labels NC cell lineages with a single dose of tamoxifen at E7.5 and that the labeled cells were widely distributed in the connective tissue of the tongue. No labeled cells were found in taste buds or the surrounding epithelium in the postnatal mice. In the GFP+/GFP- chicken chimera model, GFP+ cells migrated extensively to the cranial region of chicken embryos ipsilateral to the surgery side but were absent in taste buds in the base of oral cavity and palate. In zebrafish, Sox10-Cre/GFP-RFP faithfully labeled known NC-derived tissues but did not label taste buds in lower jaw or the barbel. Our data, together with previous findings in axolotl, indicate that taste buds are not derived from NC cells in rodents, birds, amphibians or teleost fish.
Collapse
|
12
|
Wang L, Xie J, Zhang H, Tsang LH, Tsang SL, Braune EB, Lendahl U, Sham MH. Notch signalling regulates epibranchial placode patterning and segregation. Development 2020; 147:dev.183665. [PMID: 31988190 PMCID: PMC7044445 DOI: 10.1242/dev.183665] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 01/14/2020] [Indexed: 11/20/2022]
Abstract
Epibranchial placodes are the geniculate, petrosal and nodose placodes that generate parts of cranial nerves VII, IX and X, respectively. How the three spatially separated placodes are derived from the common posterior placodal area is poorly understood. Here, we reveal that the broad posterior placode area is first patterned into a Vgll2+/Irx5+ rostral domain and a Sox2+/Fgf3+/Etv5+ caudal domain relative to the first pharyngeal cleft. This initial rostral and caudal patterning is then sequentially repeated along each pharyngeal cleft for each epibranchial placode. The caudal domains give rise to the neuronal and non-neuronal cells in the placode, whereas the rostral domains are previously unrecognized structures, serving as spacers between the final placodes. Notch signalling regulates the balance between the rostral and caudal domains: high levels of Notch signalling expand the caudal domain at the expense of the rostral domain, whereas loss of Notch signalling produces the converse phenotype. Collectively, these data unravel a new patterning principle for the early phases of epibranchial placode development and a role for Notch signalling in orchestrating epibranchial placode segregation and differentiation.
Collapse
Affiliation(s)
- Li Wang
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Junjie Xie
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Haoran Zhang
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Long Hin Tsang
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Sze Lan Tsang
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Eike-Benjamin Braune
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Urban Lendahl
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Mai Har Sham
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| |
Collapse
|
13
|
Yuan T, York JR, McCauley DW. Neural crest and placode roles in formation and patterning of cranial sensory ganglia in lamprey. Genesis 2020; 58:e23356. [PMID: 32049434 DOI: 10.1002/dvg.23356] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/30/2020] [Accepted: 01/31/2020] [Indexed: 11/07/2022]
Abstract
Vertebrates possess paired cranial sensory ganglia derived from two embryonic cell populations, neural crest and placodes. Cranial sensory ganglia arose prior to the divergence of jawed and jawless vertebrates, but the developmental mechanisms that facilitated their evolution are unknown. Using gene expression and cell lineage tracing experiments in embryos of the sea lamprey, Petromyzon marinus, we find that in the cranial ganglia we targeted, development consists of placode-derived neuron clusters in the core of ganglia, with neural crest cells mostly surrounding these neuronal clusters. To dissect functional roles of neural crest and placode cell associations in these developing cranial ganglia, we used CRISPR/Cas9 gene editing experiments to target genes critical for the development of each population. Genetic ablation of SoxE2 and FoxD-A in neural crest cells resulted in differentiated cranial sensory neurons with abnormal morphologies, whereas deletion of DlxB in cranial placodes resulted in near-total loss of cranial sensory neurons. Taken together, our cell-lineage, gene expression, and gene editing results suggest that cranial neural crest cells may not be required for cranial ganglia specification but are essential for shaping the morphology of these sensory structures. We propose that the association of neural crest and placodes in the head of early vertebrates was a key step in the organization of neurons and glia into paired sensory ganglia.
Collapse
Affiliation(s)
- Tian Yuan
- Department of Biology, University of Oklahoma, Norman, Oklahoma
| | - Joshua R York
- Department of Biology, University of Oklahoma, Norman, Oklahoma
| | | |
Collapse
|
14
|
Novel insights into inner ear development and regeneration for targeted hearing loss therapies. Hear Res 2019; 397:107859. [PMID: 31810596 DOI: 10.1016/j.heares.2019.107859] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/06/2019] [Accepted: 11/25/2019] [Indexed: 02/06/2023]
Abstract
Sensorineural hearing loss is the most common sensory deficit in humans. Despite the global scale of the problem, only limited treatment options are available today. The mammalian inner ear is a highly specialized postmitotic organ, which lacks proliferative or regenerative capacity. Since the discovery of hair cell regeneration in non-mammalian species however, much attention has been placed on identifying possible strategies to reactivate similar responses in humans. The development of successful regenerative approaches for hearing loss strongly depends on a detailed understanding of the mechanisms that control human inner ear cellular specification, differentiation and function, as well as on the development of robust in vitro cellular assays, based on human inner ear cells, to study these processes and optimize therapeutic interventions. We summarize here some aspects of inner ear development and strategies to induce regeneration that have been investigated in rodents. Moreover, we discuss recent findings in human inner ear development and compare the results with findings from animal models. Finally, we provide an overview of strategies for in vitro generation of human sensory cells from pluripotent and somatic progenitors that may provide a platform for drug development and validation of therapeutic strategies in vitro.
Collapse
|
15
|
Roccio M, Edge ASB. Inner ear organoids: new tools to understand neurosensory cell development, degeneration and regeneration. Development 2019; 146:146/17/dev177188. [PMID: 31477580 DOI: 10.1242/dev.177188] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The development of therapeutic interventions for hearing loss requires fundamental knowledge about the signaling pathways controlling tissue development as well as the establishment of human cell-based assays to validate therapeutic strategies ex vivo Recent advances in the field of stem cell biology and organoid culture systems allow the expansion and differentiation of tissue-specific progenitors and pluripotent stem cells in vitro into functional hair cells and otic-like neurons. We discuss how inner ear organoids have been developed and how they offer for the first time the opportunity to validate drug-based therapies, gene-targeting approaches and cell replacement strategies.
Collapse
Affiliation(s)
- Marta Roccio
- Inner Ear Research Laboratory, Department of Biomedical Research (DBMR), University of Bern, Bern 3008, Switzerland .,Department of Otorhinolaryngology, Head & Neck Surgery, Inselspital, Bern University Hospital, University of Bern, Bern 3010, Switzerland
| | - Albert S B Edge
- Department of Otolaryngology, Harvard Medical School, Boston, MA 02115, USA.,Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, MA 02114, USA.,Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
16
|
Marzban H, Rahimi-Balaei M, Hawkes R. Early trigeminal ganglion afferents enter the cerebellum before the Purkinje cells are born and target the nuclear transitory zone. Brain Struct Funct 2019; 224:2421-2436. [PMID: 31256239 DOI: 10.1007/s00429-019-01916-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 06/25/2019] [Indexed: 12/20/2022]
Abstract
In the standard model for the development of climbing and mossy fiber afferent pathways to the cerebellum, the ingrowing axons target the embryonic Purkinje cell somata (around embryonic ages (E13-E16 in mice). In this report, we describe a novel earlier stage in afferent development. Immunostaining for a neurofilament-associated antigen (NAA) reveals the early axon distributions with remarkable clarity. Using a combination of DiI axon tract tracing, analysis of neurogenin1 null mice, which do not develop trigeminal ganglia, and mouse embryos maintained in vitro, we show that the first axons to innervate the cerebellar primordium as early as E9 arise from the trigeminal ganglion. Therefore, early trigeminal axons are in situ before the Purkinje cells are born. Double immunostaining for NAA and markers of the different domains in the cerebellar primordium reveal that afferents first target the nuclear transitory zone (E9-E10), and only later (E10-E11) are the axons, either collaterals from the trigeminal ganglion or a new afferent source (e.g., vestibular ganglia), seen in the Purkinje cell plate. The finding that the earliest axons to the cerebellum derive from the trigeminal ganglion and enter the cerebellar primordium before the Purkinje cells are born, where they seem to target the cerebellar nuclei, reveals a novel stage in the development of the cerebellar afferents.
Collapse
Affiliation(s)
- Hassan Marzban
- Department of Human Anatomy and Cell Science, The Children's Hospital Research Institute of Manitoba (CHRIM), Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada. .,Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Rm 129 BMSB, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada.
| | - Maryam Rahimi-Balaei
- Department of Human Anatomy and Cell Science, The Children's Hospital Research Institute of Manitoba (CHRIM), Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Richard Hawkes
- Department of Cell Biology and Anatomy and Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| |
Collapse
|
17
|
Szabó A, Theveneau E, Turan M, Mayor R. Neural crest streaming as an emergent property of tissue interactions during morphogenesis. PLoS Comput Biol 2019; 15:e1007002. [PMID: 31009457 PMCID: PMC6497294 DOI: 10.1371/journal.pcbi.1007002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 05/02/2019] [Accepted: 04/03/2019] [Indexed: 12/05/2022] Open
Abstract
A fundamental question in embryo morphogenesis is how a complex pattern is established in seemingly uniform tissues. During vertebrate development, neural crest cells differentiate as a continuous mass of tissue along the neural tube and subsequently split into spatially distinct migratory streams to invade the rest of the embryo. How these streams are established is not well understood. Inhibitory signals surrounding the migratory streams led to the idea that position and size of streams are determined by a pre-pattern of such signals. While clear evidence for a pre-pattern in the cranial region is still lacking, all computational models of neural crest migration published so far have assumed a pre-pattern of negative signals that channel the neural crest into streams. Here we test the hypothesis that instead of following a pre-existing pattern, the cranial neural crest creates their own migratory pathway by interacting with the surrounding tissue. By combining theoretical modeling with experimentation, we show that streams emerge from the interaction of the hindbrain neural crest and the neighboring epibranchial placodal tissues, without the need for a pre-existing guidance cue. Our model suggests that the initial collective neural crest invasion is based on short-range repulsion and asymmetric attraction between neighboring tissues. The model provides a coherent explanation for the formation of cranial neural crest streams in concert with previously reported findings and our new in vivo observations. Our results point to a general mechanism of inducing collective invasion patterns.
Collapse
Affiliation(s)
- András Szabó
- Research Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Eric Theveneau
- Research Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Melissa Turan
- Research Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Roberto Mayor
- Research Department of Cell and Developmental Biology, University College London, London, United Kingdom
| |
Collapse
|
18
|
Sudiwala S, Knox SM. The emerging role of cranial nerves in shaping craniofacial development. Genesis 2019; 57:e23282. [PMID: 30628162 DOI: 10.1002/dvg.23282] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/05/2019] [Accepted: 01/07/2019] [Indexed: 12/17/2022]
Abstract
Organs and structures of the vertebrate head perform a plethora of tasks including visualization, digestion, vocalization/communication, auditory functions, and respiration in response to neuronal input. This input is primarily derived from afferent and efferent fibers of the cranial nerves (sensory and motor respectively) and efferent fibers of the cervical sympathetic trunk. Despite their essential contribution to the function and integration of processes necessary for survival, how organ innervation is established remains poorly understood. Furthermore, while it has been appreciated for some time that innervation of organs by cranial nerves is regulated in part by secreted factors and cell surface ligands expressed by those organs, whether nerves also regulate the development of facial organs is only beginning to be elucidated. This review will provide an overview of cranial nerve development in relation to the organs they innervate, and outline their known contributions to craniofacial development, thereby providing insight into how nerves may shape the organs they innervate during development. Throughout, the interaction between different cell and tissue types will be highlighted.
Collapse
Affiliation(s)
- Sonia Sudiwala
- Program in Craniofacial Biology, Department of Cell and Tissue Biology, University of California, San Francisco, California
| | - Sarah M Knox
- Program in Craniofacial Biology, Department of Cell and Tissue Biology, University of California, San Francisco, California
| |
Collapse
|
19
|
Jenkins BA, Fontecilla NM, Lu CP, Fuchs E, Lumpkin EA. The cellular basis of mechanosensory Merkel-cell innervation during development. eLife 2019; 8:42633. [PMID: 30794158 PMCID: PMC6386521 DOI: 10.7554/elife.42633] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 02/06/2019] [Indexed: 02/06/2023] Open
Abstract
Touch sensation is initiated by mechanosensory neurons that innervate distinct skin structures; however, little is known about how these neurons are patterned during mammalian skin development. We explored the cellular basis of touch-receptor patterning in mouse touch domes, which contain mechanosensory Merkel cell-neurite complexes and abut primary hair follicles. At embryonic stage 16.5 (E16.5), touch domes emerge as patches of Merkel cells and keratinocytes clustered with a previously unsuspected population of Bmp4-expressing dermal cells. Epidermal Noggin overexpression at E14.5 disrupted touch-dome formation but not hair-follicle specification, demonstrating a temporally distinct requirement for BMP signaling in placode-derived structures. Surprisingly, two neuronal populations preferentially targeted touch domes during development but only one persisted in mature touch domes. Finally, Keratin-17-expressing keratinocytes but not Merkel cells were necessary to establish innervation patterns during development. These findings identify key cell types and signaling pathways required for targeting Merkel-cell afferents to discrete mechanosensory compartments.
Collapse
Affiliation(s)
- Blair A Jenkins
- Department of Physiology and Cellular BiophysicsColumbia UniversityNew YorkUnited States
- Department of DermatologyColumbia UniversityNew YorkUnited States
| | - Natalia M Fontecilla
- Department of Physiology and Cellular BiophysicsColumbia UniversityNew YorkUnited States
| | - Catherine P Lu
- Robin Neustein Laboratory of Mammalian Development and Cell BiologyHoward Hughes Medical Institute, The Rockefeller UniversityNew YorkUnited States
| | - Elaine Fuchs
- Robin Neustein Laboratory of Mammalian Development and Cell BiologyHoward Hughes Medical Institute, The Rockefeller UniversityNew YorkUnited States
| | - Ellen A Lumpkin
- Department of Physiology and Cellular BiophysicsColumbia UniversityNew YorkUnited States
| |
Collapse
|
20
|
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
|
21
|
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
|
22
|
Drake PM, Franz-Odendaal TA. A Potential Role for MMPs during the Formation of Non-Neurogenic Placodes. J Dev Biol 2018; 6:jdb6030020. [PMID: 30049947 PMCID: PMC6162748 DOI: 10.3390/jdb6030020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/16/2018] [Accepted: 07/24/2018] [Indexed: 12/16/2022] Open
Abstract
The formation of non-neurogenic placodes is critical prior to the development of several epithelial derivatives (e.g., feathers, teeth, etc.) and their development frequently involves morphogenetic proteins (or morphogens). Matrix metalloproteinases (MMPs) are important enzymes involved in extracellular matrix remodeling, and recent research has shown that the extracellular matrix (ECM) can modulate morphogen diffusion and cell behaviors. This review summarizes the known roles of MMPs during the development of non-neurogenic structures that involve a placodal stage. Specifically, we discuss feather, hair, tooth, mammary gland and lens development. This review highlights the potential critical role MMPs may play during placode formation in these systems.
Collapse
Affiliation(s)
- Paige M Drake
- Department of Medical Neuroscience, Dalhousie University, 5850 College Street, Halifax, NS B3H 4R2, Canada.
- Department of Biology, Mount Saint Vincent University, 166 Bedford Highway, Halifax, NS B3M 2J6, Canada.
| | - Tamara A Franz-Odendaal
- Department of Medical Neuroscience, Dalhousie University, 5850 College Street, Halifax, NS B3H 4R2, Canada.
- Department of Biology, Mount Saint Vincent University, 166 Bedford Highway, Halifax, NS B3M 2J6, Canada.
| |
Collapse
|
23
|
Shannon ML, Fame RM, Chau KF, Dani N, Calicchio ML, Géléoc GS, Lidov HGW, Alexandrescu S, Lehtinen MK. Mice Expressing Myc in Neural Precursors Develop Choroid Plexus and Ciliary Body Tumors. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1334-1344. [PMID: 29545198 PMCID: PMC5971223 DOI: 10.1016/j.ajpath.2018.02.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/25/2018] [Accepted: 02/20/2018] [Indexed: 12/18/2022]
Abstract
Choroid plexus tumors and ciliary body medulloepithelioma are predominantly pediatric neoplasms. Progress in understanding the pathogenesis of these tumors has been hindered by their rarity and lack of models that faithfully recapitulate the disease. Here, we find that endogenous Myc proto-oncogene protein is down-regulated in the forebrain neuroepithelium, whose neural plate border domains give rise to the anterior choroid plexus and ciliary body. To uncover the consequences of persistent Myc expression, MYC expression was forced in multipotent neural precursors (nestin-Cre:Myc), which produced fully penetrant models of choroid plexus carcinoma and ciliary body medulloepithelioma. Nestin-mediated MYC expression in the epithelial cells of choroid plexus leads to the regionalized formation of choroid plexus carcinoma in the posterior domain of the lateral ventricle choroid plexus and the fourth ventricle choroid plexus that is accompanied by loss of multiple cilia, up-regulation of protein biosynthetic machinery, and hydrocephalus. Parallel MYC expression in the ciliary body leads also to up-regulation of protein biosynthetic machinery. Additionally, Myc expression in human choroid plexus tumors increases with aggressiveness of disease. Collectively, our findings expose a select vulnerability of the neuroepithelial lineage to postnatal tumorigenesis and provide a new mouse model for investigating the pathogenesis of these rare pediatric neoplasms.
Collapse
Affiliation(s)
- Morgan L Shannon
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts
| | - Ryann M Fame
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts
| | - Kevin F Chau
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts; Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts
| | - Neil Dani
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts
| | - Monica L Calicchio
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts
| | - Gwenaelle S Géléoc
- Department of Otolaryngology, Boston Children's Hospital, Boston, Massachusetts; F.M. Kirby Center for Neurobiology, Boston Children's Hospital, Boston, Massachusetts
| | - Hart G W Lidov
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts
| | - Sanda Alexandrescu
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts; Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
24
|
Okamoto Y, Nishimura N, Matsuda K, Ranawakage DC, Kamachi Y, Kondoh H, Uchikawa M. Cooperation of Sall4 and Sox8 transcription factors in the regulation of the chicken Sox3
gene during otic placode development. Dev Growth Differ 2018. [DOI: 10.1111/dgd.12427] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Yu Okamoto
- Graduate School of Frontier Biosciences; Osaka University; Osaka Japan
| | - Naoko Nishimura
- Graduate School of Frontier Biosciences; Osaka University; Osaka Japan
| | - Kazunari Matsuda
- Graduate School of Frontier Biosciences; Osaka University; Osaka Japan
| | - Deshani C. Ranawakage
- School of Environmental Science and Engineering; Kochi University of Technology; Kochi Japan
| | - Yusuke Kamachi
- School of Environmental Science and Engineering; Kochi University of Technology; Kochi Japan
| | - Hisato Kondoh
- Graduate School of Frontier Biosciences; Osaka University; Osaka Japan
- Faculty of Life Sciences; Kyoto Sangyo University; Kyoto Japan
| | - Masanori Uchikawa
- Graduate School of Frontier Biosciences; Osaka University; Osaka Japan
| |
Collapse
|
25
|
Nasal and otic placode specific regulation of Sox2 involves both activation by Sox-Sall4 synergism and multiple repression mechanisms. Dev Biol 2018; 433:61-74. [DOI: 10.1016/j.ydbio.2017.11.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 11/02/2017] [Accepted: 11/10/2017] [Indexed: 01/21/2023]
|
26
|
Neural induction by the node and placode induction by head mesoderm share an initial state resembling neural plate border and ES cells. Proc Natl Acad Sci U S A 2017; 115:355-360. [PMID: 29259119 DOI: 10.1073/pnas.1719674115] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Around the time of gastrulation in higher vertebrate embryos, inductive interactions direct cells to form central nervous system (neural plate) or sensory placodes. Grafts of different tissues into the periphery of a chicken embryo elicit different responses: Hensen's node induces a neural plate whereas the head mesoderm induces placodes. How different are these processes? Transcriptome analysis in time course reveals that both processes start by induction of a common set of genes, which later diverge. These genes are remarkably similar to those induced by an extraembryonic tissue, the hypoblast, and are normally expressed in the pregastrulation stage epiblast. Explants of this epiblast grown in the absence of further signals develop as neural plate border derivatives and eventually express lens markers. We designate this state as "preborder"; its transcriptome resembles embryonic stem cells. Finally, using sequential transplantation experiments, we show that the node, head mesoderm, and hypoblast are interchangeable to begin any of these inductions while the final outcome depends on the tissue emitting the later signals.
Collapse
|
27
|
Perny M, Ting CC, Kleinlogel S, Senn P, Roccio M. Generation of Otic Sensory Neurons from Mouse Embryonic Stem Cells in 3D Culture. Front Cell Neurosci 2017; 11:409. [PMID: 29311837 PMCID: PMC5742223 DOI: 10.3389/fncel.2017.00409] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 12/05/2017] [Indexed: 12/29/2022] Open
Abstract
The peripheral hearing process taking place in the cochlea mainly depends on two distinct sensory cell types: the mechanosensitive hair cells and the spiral ganglion neurons (SGNs). The first respond to the mechanical stimulation exerted by sound pressure waves on their hair bundles by releasing neurotransmitters and thereby activating the latter. Loss of these sensorineural cells is associated with permanent hearing loss. Stem cell-based approaches aiming at cell replacement or in vitro drug testing to identify potential ototoxic, otoprotective, or regenerative compounds have lately gained attention as putative therapeutic strategies for hearing loss. Nevertheless, they rely on efficient and reliable protocols for the in vitro generation of cochlear sensory cells for their implementation. To this end, we have developed a differentiation protocol based on organoid culture systems, which mimics the most important steps of in vivo otic development, robustly guiding mouse embryonic stem cells (mESCs) toward otic sensory neurons (OSNs). The stepwise differentiation of mESCs toward ectoderm was initiated using a quick aggregation method in presence of Matrigel in serum-free conditions. Non-neural ectoderm was induced via activation of bone morphogenetic protein (BMP) signaling and concomitant inhibition of transforming growth factor beta (TGFβ) signaling to prevent mesendoderm induction. Preplacodal and otic placode ectoderm was further induced by inhibition of BMP signaling and addition of fibroblast growth factor 2 (FGF2). Delamination and differentiation of SGNs was initiated by plating of the organoids on a 2D Matrigel-coated substrate. Supplementation with brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3) was used for further maturation until 15 days of in vitro differentiation. A large population of neurons with a clear bipolar morphology and functional excitability was derived from these cultures. Immunostaining and gene expression analysis performed at different time points confirmed the transition trough the otic lineage and final expression of the key OSN markers. Moreover, the stem cell-derived OSNs exhibited functional electrophysiological properties of native SGNs. Our established in vitro model of OSNs development can be used for basic developmental studies, for drug screening or for the exploration of their regenerative potential.
Collapse
Affiliation(s)
- Michael Perny
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland.,Laboratory of Inner Ear Research, Department for BioMedical Research, University of Bern, Bern, Switzerland.,Department of Otorhinolaryngology, Head and Neck Surgery, Inselspital, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Ching-Chia Ting
- Laboratory of Inner Ear Research, Department for BioMedical Research, University of Bern, Bern, Switzerland.,Department of Otorhinolaryngology, Head and Neck Surgery, Inselspital, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, Department of Biomedical Research, University of Bern, Bern, Switzerland
| | | | - Pascal Senn
- Laboratory of Inner Ear Research, Department for BioMedical Research, University of Bern, Bern, Switzerland.,Department of Otorhinolaryngology, Head and Neck Surgery, Inselspital, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, Department of Biomedical Research, University of Bern, Bern, Switzerland.,Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Geneva (HUG), Geneva, Switzerland
| | - Marta Roccio
- Laboratory of Inner Ear Research, Department for BioMedical Research, University of Bern, Bern, Switzerland.,Department of Otorhinolaryngology, Head and Neck Surgery, Inselspital, University of Bern, Bern, Switzerland.,Cluster for Regenerative Neuroscience, Department of Biomedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
28
|
Sato S, Furuta Y, Kawakami K. Regulation of continuous but complex expression pattern of Six1 during early sensory development. Dev Dyn 2017; 247:250-261. [PMID: 29106072 DOI: 10.1002/dvdy.24603] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 10/17/2017] [Accepted: 10/31/2017] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND In vertebrates, cranial sensory placodes give rise to neurosensory and endocrine structures, such as the olfactory epithelium, inner ear, and anterior pituitary. We report here the establishment of a transgenic mouse line that expresses Cre recombinase under the control of Six1-21, a major placodal enhancer of the homeobox gene Six1. RESULTS In the new Cre-expressing line, mSix1-21-NLSCre, the earliest Cre-mediated recombination was induced at embryonic day 8.5 in the region overlapping with the otic-epibranchial progenitor domain (OEPD), a transient, common precursor domain for the otic and epibranchial placodes. Recombination was later observed in the OEPD-derived structures (the entire inner ear and the VIIth-Xth cranial sensory ganglia), olfactory epithelium, anterior pituitary, pharyngeal ectoderm and pouches. Other Six1-positive structures, such as salivary/lacrimal glands and limb buds, were also positive for recombination. Moreover, comparison with another mouse line expressing Cre under the control of the sensory neuron enhancer, Six1-8, indicated that the continuous and complex expression pattern of Six1 during sensory organ formation is pieced together by separate enhancers. CONCLUSIONS mSix1-21-NLSCre has several unique characteristics to make it suitable for analysis of cell lineage and gene function in sensory placodes as well as nonplacodal Six1-positive structures. Developmental Dynamics 247:250-261, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Shigeru Sato
- Division of Biology, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Yasuhide Furuta
- Animal Resource Development Unit and Genetic Engineering Team, Division of Bio-function Dynamics Imaging, RIKEN Center for Life Science Technologies (CLST), Kobe, Hyogo, Japan
| | - Kiyoshi Kawakami
- Division of Biology, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| |
Collapse
|
29
|
Abstract
The sensation of touch is mediated by mechanosensory neurons that are embedded in skin and relay signals from the periphery to the central nervous system. During embryogenesis, axons elongate from these neurons to make contact with the developing skin. Concurrently, the epithelium of skin transforms from a homogeneous tissue into a heterogeneous organ that is made up of distinct layers and microdomains. Throughout this process, each neuronal terminal must form connections with an appropriate skin region to serve its function. This Review presents current knowledge of the development of the sensory microdomains in mammalian skin and the mechanosensory neurons that innervate them.
Collapse
Affiliation(s)
- Blair A Jenkins
- Department of Physiology & Cellular Biophysics and Department of Dermatology, Columbia University in the City of New York, New York, NY 10032, USA
| | - Ellen A Lumpkin
- Department of Physiology & Cellular Biophysics and Department of Dermatology, Columbia University in the City of New York, New York, NY 10032, USA
| |
Collapse
|
30
|
Sanchez-Arrones L, Sandonís Á, Cardozo MJ, Bovolenta P. Adenohypophysis placodal precursors exhibit distinctive features within the rostral preplacodal ectoderm. Development 2017; 144:3521-3532. [PMID: 28974641 DOI: 10.1242/dev.149724] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 08/14/2017] [Indexed: 11/20/2022]
Abstract
Placodes are discrete thickenings of the vertebrate cranial ectoderm that generate morpho-functionally distinct structures, such as the adenohypophysis, olfactory epithelium and lens. All placodes arise from a horseshoe-shaped preplacodal ectoderm in which the precursors of individual placodes are intermingled. However, fate-map studies indicated that cells positioned at the preplacodal midline give rise to only the adenohypophyseal placode, suggesting a unique organization of these precursors within the preplacode. To test this possibility, we combined embryological and molecular approaches in chick embryos to show that, at gastrula stage, adenohypophyseal precursors are clustered in the median preplacodal ectoderm, largely segregated from those of the adjacent olfactory placode. Median precursors are elongated, densely packed and, at neurula stage, express a molecular signature that distinguishes them from the remaining preplacodal cells. Olfactory placode precursors and midline neural cells can replace ablated adenohypophyseal precursors up to head-fold stage, although with a more plastic organization. We thus propose that adenohypophyseal placode precursors are unique within the preplacodal ectoderm possibly because they originate the only single placode and the only one with an endocrine character.
Collapse
Affiliation(s)
- Luisa Sanchez-Arrones
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, c/ Nicolás Cabrera 1, Madrid 28049, Spain.,CIBER de Enfermedades Raras (CIBERER), c/ Nicolás Cabrera 1, Madrid 28049, Spain
| | - África Sandonís
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, c/ Nicolás Cabrera 1, Madrid 28049, Spain.,CIBER de Enfermedades Raras (CIBERER), c/ Nicolás Cabrera 1, Madrid 28049, Spain
| | - Marcos Julián Cardozo
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, c/ Nicolás Cabrera 1, Madrid 28049, Spain.,CIBER de Enfermedades Raras (CIBERER), c/ Nicolás Cabrera 1, Madrid 28049, Spain
| | - Paola Bovolenta
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, c/ Nicolás Cabrera 1, Madrid 28049, Spain .,CIBER de Enfermedades Raras (CIBERER), c/ Nicolás Cabrera 1, Madrid 28049, Spain
| |
Collapse
|
31
|
Nikaido M, Navajas Acedo J, Hatta K, Piotrowski T. Retinoic acid is required and Fgf, Wnt, and Bmp signaling inhibit posterior lateral line placode induction in zebrafish. Dev Biol 2017; 431:215-225. [PMID: 28923486 DOI: 10.1016/j.ydbio.2017.09.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 09/12/2017] [Accepted: 09/14/2017] [Indexed: 10/18/2022]
Abstract
The lateral line system is a mechanosensory systems present in aquatic animals. The anterior and posterior lateral lines develop from anterior and posterior lateral line placodes (aLLp and pLLp), respectively. Although signaling molecules required for the induction of other cranial placodes have been well studied, the molecular mechanisms underlying formation of the lateral line placodes are unknown. In this study we tested the requirement of multiple signaling pathways, such as Wnt, Bmp Fgf, and Retinoic Acid for aLLp and pLLp induction. We determined that aLLp specification requires Fgf signaling, whilst pLLp specification requires retinoic acid which inhibits Fgf signaling. pLLp induction is also independent of Wnt and Bmp activities, even though these pathways limit the boundaries of the pLLp. This is the first report that the aLLp and pLLp depend on different inductive mechanisms and that pLLp induction requires the inhibition of Fgf, Wnt and Bmp signaling.
Collapse
Affiliation(s)
- Masataka Nikaido
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA; Graduate School of Life Sciences, University of Hyogo, Hyogo Pref. 678-1297, Japan
| | | | - Kohei Hatta
- Graduate School of Life Sciences, University of Hyogo, Hyogo Pref. 678-1297, Japan
| | | |
Collapse
|
32
|
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]
|
33
|
Shigetani Y, Wakamatsu Y, Tachibana T, Okabe M. Conversion of neural plate explants to pre-placodal ectoderm-like tissue in vitro. Biochem Biophys Res Commun 2016; 477:807-813. [PMID: 27369078 DOI: 10.1016/j.bbrc.2016.06.139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 06/27/2016] [Indexed: 10/21/2022]
Abstract
Neural crest and cranial sensory placodes arise from ectodermal epithelium lying between the neural plate and non-neural ectoderm (neural border). BMP signaling is important for both an induction of the neural border and a subsequent induction of the neural crest within the neural border. In contrast, FGF signaling is important for the neural border induction and the following induction of the pre-placodal ectoderm (PPE), which later gives rise to the cranial sensory placodes. While previous studies have demonstrated that the neural plate explants could be converted to the neural crest cells by adding BMP4 in a culture medium, there is no report showing a similar conversion of the neural plate to the PPE. We therefore examined the effect of FGF2 along with BMP4 on the rostral neural plate explants and found that the explants became the simple squamous epithelia, which were characterized by the desmosomes/tonofilaments in membranes of adjacent cells. Such epithelia expressed sets of neural border markers and the PPE genes, suggesting that the neural plate explants were converted to a PPE-like tissue. This method will be useful for further studying mechanisms of PPE induction and subsequent specifications of the cranial placodes.
Collapse
Affiliation(s)
- Yasuyo Shigetani
- Department of Anatomy, The Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Minato-ku, Tokyo 105-0003, Japan.
| | - Yoshio Wakamatsu
- Department of Developmental Neuroscience, United Centers for Advance Research and Translational Medicine, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Toshiaki Tachibana
- Division of Molecular Cell Biology, Core Research Facilities for Basic Science, Research Center for Medical Science, The Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Minato-ku, Tokyo 105-0003, Japan
| | - Masataka Okabe
- Department of Anatomy, The Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Minato-ku, Tokyo 105-0003, Japan
| |
Collapse
|
34
|
Differentiation of Induced Pluripotent Stem Cells to Lentoid Bodies Expressing a Lens Cell-Specific Fluorescent Reporter. PLoS One 2016; 11:e0157570. [PMID: 27322380 PMCID: PMC4913943 DOI: 10.1371/journal.pone.0157570] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 06/01/2016] [Indexed: 12/18/2022] Open
Abstract
Curative approaches for eye cataracts and other eye abnormalities, such as myopia and hyperopia currently suffer from a lack of appropriate models. Here, we present a new approach for in vitro growth of lentoid bodies from induced pluripotent stem (iPS) cells as a tool for ophthalmological research. We generated a transgenic mouse line with lens-specific expression of a fluorescent reporter driven by the alphaA crystallin promoter. Fetal fibroblasts were isolated from transgenic fetuses, reprogrammed to iPS cells, and differentiated to lentoid bodies exploiting the specific fluorescence of the lens cell-specific reporter. The employment of cell type-specific reporters for establishing and optimizing differentiation in vitro seems to be an efficient and generally applicable approach for developing differentiation protocols for desired cell populations.
Collapse
|
35
|
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
|
36
|
Audette DS, Anand D, So T, Rubenstein TB, Lachke SA, Lovicu FJ, Duncan MK. Prox1 and fibroblast growth factor receptors form a novel regulatory loop controlling lens fiber differentiation and gene expression. Development 2015; 143:318-28. [PMID: 26657765 DOI: 10.1242/dev.127860] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 11/26/2015] [Indexed: 01/19/2023]
Abstract
Lens epithelial cells differentiate into lens fibers (LFs) in response to a fibroblast growth factor (FGF) gradient. This cell fate decision requires the transcription factor Prox1, which has been hypothesized to promote cell cycle exit in differentiating LF cells. However, we find that conditional deletion of Prox1 from mouse lenses results in a failure in LF differentiation despite maintenance of normal cell cycle exit. Instead, RNA-seq demonstrated that Prox1 functions as a global regulator of LF cell gene expression. Intriguingly, Prox1 also controls the expression of fibroblast growth factor receptors (FGFRs) and can bind to their promoters, correlating with decreased downstream signaling through MAPK and AKT in Prox1 mutant lenses. Further, culturing rat lens explants in FGF increased their expression of Prox1, and this was attenuated by the addition of inhibitors of MAPK. Together, these results describe a novel feedback loop required for lens differentiation and morphogenesis, whereby Prox1 and FGFR signaling interact to mediate LF differentiation in response to FGF.
Collapse
Affiliation(s)
- Dylan S Audette
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Deepti Anand
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Tammy So
- Discipline of Anatomy & Histology, Bosch Institute & Save Sight Institute, University of Sydney, Sydney, New South Wales 2000, Australia
| | - Troy B Rubenstein
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Salil A Lachke
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA
| | - Frank J Lovicu
- Discipline of Anatomy & Histology, Bosch Institute & Save Sight Institute, University of Sydney, Sydney, New South Wales 2000, Australia
| | - Melinda K Duncan
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
37
|
Moody SA, Neilson KM, Kenyon KL, Alfandari D, Pignoni F. Using Xenopus to discover new genes involved in branchiootorenal spectrum disorders. Comp Biochem Physiol C Toxicol Pharmacol 2015; 178:16-24. [PMID: 26117063 PMCID: PMC4662879 DOI: 10.1016/j.cbpc.2015.06.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 06/16/2015] [Accepted: 06/17/2015] [Indexed: 12/14/2022]
Abstract
Congenital hearing loss is an important clinical problem because, without early intervention, affected children do not properly acquire language and consequently have difficulties developing social skills. Although most newborns in the US are screened for hearing deficits, even earlier diagnosis can be made with prenatal genetic screening. Genetic screening that identifies the relevant mutated gene can also warn about potential congenital defects in organs not related to hearing. We will discuss efforts to identify new candidate genes that underlie the Branchiootorenal spectrum disorders in which affected children have hearing deficits and are also at risk for kidney defects. Mutations in two genes, SIX1 and EYA1, have been identified in about half of the patients tested. To uncover new candidate genes, we have used the aquatic animal model, Xenopus laevis, to identify genes that are part of the developmental genetic pathway of Six1 during otic and kidney development. We have already identified a large number of potential Six1 transcriptional targets and candidate co-factor proteins that are expressed at the right time and in the correct tissues to interact with Six1 during development. We discuss the advantages of using this system for gene discovery in a human congenital hearing loss syndrome.
Collapse
Affiliation(s)
- Sally A Moody
- Department of Anatomy and Regenerative Biology, George Washington University, School of Medicine and Health Sciences, Washington, DC, USA.
| | - Karen M Neilson
- Department of Anatomy and Regenerative Biology, George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
| | - Kristy L Kenyon
- Department of Biology, Hobart and William Smith Colleges, Geneva, NY, USA
| | - Dominique Alfandari
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, USA
| | - Francesca Pignoni
- Department of Ophthalmology, Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
38
|
Solute Carrier Family 26 Member a2 (slc26a2) Regulates Otic Development and Hair Cell Survival in Zebrafish. PLoS One 2015; 10:e0136832. [PMID: 26375458 PMCID: PMC4573323 DOI: 10.1371/journal.pone.0136832] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 08/10/2015] [Indexed: 12/16/2022] Open
Abstract
Hearing loss is one of the most prevalent human birth defects. Genetic factors contribute to the pathogenesis of deafness. It is estimated that one-third of deafness genes have already been identified. The current work is an attempt to find novel genes relevant to hearing loss using guilt-by-profiling and guilt-by-association bioinformatics analyses of approximately 80 known non-syndromic hereditary hearing loss (NSHL) genes. Among the 300 newly identified candidate deafness genes, slc26a2 were selected for functional studies in zebrafish. The slc26a2 gene was knocked down using an antisense morpholino (MO), and significant defects were observed in otolith patterns, semicircular canal morphology, and lateral neuromast distributions in morphants. Loss-of-function defects are caused primarily by apoptosis, and morphants are insensitive to sound stimulation and imbalanced swimming behaviours. Morphant defects were found to be partially rescued by co-injection of human SLC26A2 mRNA. All the results suggest that bioinformatics is capable of predicting new deafness genes and this showed slc26a2 is to be a critical otic gene whose dysfunction may induce hearing impairment.
Collapse
|
39
|
Sato S, Yajima H, Furuta Y, Ikeda K, Kawakami K. Activation of Six1 Expression in Vertebrate Sensory Neurons. PLoS One 2015; 10:e0136666. [PMID: 26313368 PMCID: PMC4551851 DOI: 10.1371/journal.pone.0136666] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/05/2015] [Indexed: 12/31/2022] Open
Abstract
SIX1 homeodomain protein is one of the essential key regulators of sensory organ development. Six1-deficient mice lack the olfactory epithelium, vomeronasal organs, cochlea, vestibule and vestibuloacoustic ganglion, and also show poor neural differentiation in the distal part of the cranial ganglia. Simultaneous loss of both Six1 and Six4 leads to additional abnormalities such as small trigeminal ganglion and abnormal dorsal root ganglia (DRG). The aim of this study was to understand the molecular mechanism that controls Six1 expression in sensory organs, particularly in the trigeminal ganglion and DRG. To this end, we focused on the sensory ganglia-specific Six1 enhancer (Six1-8) conserved between chick and mouse. In vivo reporter assays using both animals identified an important core region comprising binding consensus sequences for several transcription factors including nuclear hormone receptors, TCF/LEF, SMAD, POU homeodomain and basic-helix-loop-helix proteins. The results provided information on upstream factors and signals potentially relevant to Six1 regulation in sensory neurons. We also report the establishment of a new transgenic mouse line (mSix1-8-NLSCre) that expresses Cre recombinase under the control of mouse Six1-8. Cre-mediated recombination was detected specifically in ISL1/2-positive sensory neurons of Six1-positive cranial sensory ganglia and DRG. The unique features of the mSix1-8-NLSCre line are the absence of Cre-mediated recombination in SOX10-positive glial cells and central nervous system and ability to induce recombination in a subset of neurons derived from the olfactory placode/epithelium. This mouse model can be potentially used to advance research on sensory development.
Collapse
Affiliation(s)
- Shigeru Sato
- Division of Biology, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
- * E-mail:
| | - Hiroshi Yajima
- Division of Biology, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Yasuhide Furuta
- Animal Resource Development Unit and Genetic Engineering Team, Division of Bio-function Dynamics Imaging, RIKEN Center for Life Science Technologies (CLST), Kobe, Hyogo, Japan
| | - Keiko Ikeda
- Division of Biology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Kiyoshi Kawakami
- Division of Biology, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| |
Collapse
|
40
|
Szabó A, Mayor R. Cell traction in collective cell migration and morphogenesis: the chase and run mechanism. Cell Adh Migr 2015; 9:380-3. [PMID: 26267782 DOI: 10.1080/19336918.2015.1019997] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Directional collective cell migration plays an important role in development, physiology, and disease. An increasing number of studies revealed key aspects of how cells coordinate their movement through distances surpassing several cell diameters. While physical modeling and measurements of forces during collective cell movements helped to reveal key mechanisms, most of these studies focus on tightly connected epithelial cultures. Less is known about collective migration of mesenchymal cells. A typical example of such behavior is the migration of the neural crest cells, which migrate large distances as a group. A recent study revealed that this persistent migration is aided by the interaction between the neural crest and the neighboring placode cells, whereby neural crest chase the placodes via chemotaxis, but upon contact both populations undergo contact inhibition of locomotion and a rapid reorganization of cellular traction. The resulting asymmetric traction field of the placodes forces them to run away from the chasers. We argue that this chase and run interaction may not be specific only to the neural crest system, but could serve as the underlying mechanism for several morphogenetic processes involving collective cell migration.
Collapse
Affiliation(s)
- András Szabó
- a Department of Cell and Developmental Biology ; University College London ; London UK
| | - Roberto Mayor
- a Department of Cell and Developmental Biology ; University College London ; London UK
| |
Collapse
|
41
|
Vega‐López GA, Bonano M, Tríbulo C, Fernández JP, Agüero TH, Aybar MJ. Functional analysis of
Hairy
genes in
Xenopus
neural crest initial specification and cell migration. Dev Dyn 2015; 244:988-1013. [DOI: 10.1002/dvdy.24295] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 04/25/2015] [Accepted: 05/14/2015] [Indexed: 01/28/2023] Open
Affiliation(s)
| | - Marcela Bonano
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET‐UNT
| | - Celeste Tríbulo
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET‐UNT
- Instituto de Biología “Dr. Francisco D. Barbieri”, Facultad de Bioquímica, Química y FarmaciaUniversidad Nacional de TucumánChacabuco San Miguel de Tucumán Argentina
| | - Juan P. Fernández
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET‐UNT
| | - Tristán H. Agüero
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET‐UNT
| | - Manuel J. Aybar
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET‐UNT
- Instituto de Biología “Dr. Francisco D. Barbieri”, Facultad de Bioquímica, Química y FarmaciaUniversidad Nacional de TucumánChacabuco San Miguel de Tucumán Argentina
| |
Collapse
|
42
|
Shida H, Mende M, Takano-Yamamoto T, Osumi N, Streit A, Wakamatsu Y. Otic placode cell specification and proliferation are regulated by Notch signaling in avian development. Dev Dyn 2015; 244:839-51. [PMID: 25970828 DOI: 10.1002/dvdy.24291] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 04/26/2015] [Accepted: 05/01/2015] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND The entire inner ear including the cochlear-vestibular ganglion arises from a simple epithelium, the otic placode. Precursors for the placode originate from a pool of progenitors located in ectoderm next to the future hindbrain, the pre-otic field, where they are intermingled with future epibranchial and epidermal cells. While the importance of secreted proteins, such as FGFs and Wnts, in imparting otic identity has been well studied, how precursors for these different fates segregate locally is less well understood. RESULTS (1) The Notch ligand Delta1 and the Notch target Hes5-2 are expressed in a part of pre-otic field before otic commitment, indicative of active Notch signaling, and this is confirmed using a Notch reporter. (2) Loss and gain-of-function approaches reveal that Notch signaling regulates both proliferation and specification of pre-otic progenitors. CONCLUSIONS Our results identify a novel function of Notch signaling in cell fate determination in the pre-otic field of avian embryos.
Collapse
Affiliation(s)
- Hiroko Shida
- Department of Developmental Neuroscience, Center for Translational and Advanced Animal Research, Tohoku University Graduate School of Medicine, Miyagi, Japan.,Division of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Miyagi, Japan
| | - Michael Mende
- Department of Craniofacial Development and Stem Cell Biology, King's College London
| | - Teruko Takano-Yamamoto
- Division of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Miyagi, Japan
| | - Noriko Osumi
- Department of Developmental Neuroscience, Center for Translational and Advanced Animal Research, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Andrea Streit
- Department of Craniofacial Development and Stem Cell Biology, King's College London
| | - Yoshio Wakamatsu
- Department of Developmental Neuroscience, Center for Translational and Advanced Animal Research, Tohoku University Graduate School of Medicine, Miyagi, Japan
| |
Collapse
|
43
|
Cvekl A, McGreal R, Liu W. Lens Development and Crystallin Gene Expression. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 134:129-67. [PMID: 26310154 DOI: 10.1016/bs.pmbts.2015.05.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The eye and lens represent excellent models to understand embryonic development at cellular and molecular levels. Initial 3D formation of the eye depends on a reciprocal invagination of the lens placode/optic vesicle to form the eye primordium, i.e., the optic cup partially surrounding the lens vesicle. Subsequently, the anterior part of the lens vesicle gives rise to the lens epithelium, while the posterior cells of the lens vesicle differentiate into highly elongated lens fibers. Lens fiber differentiation involves cytoskeletal rearrangements, cellular elongation, accumulation of crystallin proteins, production of extracellular matrix for the lens capsule, and degradation of organelles. This chapter summarizes recent advances in lens development and provides insights into the regulatory mechanisms and differentiation at the level of chromatin structure and dynamics, the emerging field of noncoding RNAs, and novel strategies to fill the gaps in our understanding of lens development.
Collapse
Affiliation(s)
- Ales Cvekl
- Departments of Genetics and Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, New York, USA.
| | - Rebecca McGreal
- Departments of Genetics and Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Wei Liu
- Departments of Genetics and Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
44
|
Park BY. Sox9 regulates development of neural crest and otic placode in a time- and dose-dependent fashion. J Biomed Res 2015. [DOI: 10.12729/jbr.2015.16.1.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
45
|
Sai X, Ladher RK. Early steps in inner ear development: induction and morphogenesis of the otic placode. Front Pharmacol 2015; 6:19. [PMID: 25713536 PMCID: PMC4322616 DOI: 10.3389/fphar.2015.00019] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 01/21/2015] [Indexed: 01/09/2023] Open
Abstract
Various cellular replacement therapies using in vitro generated cells to replace damaged tissue have been proposed as strategies to alleviate hearing loss. All such therapies must involve a complete understanding of the earliest steps in inner ear development; its induction as a thickened plate of cells in the non-neural, surface ectoderm of the embryo, to its internalization as an otocyst embedded in the head mesenchyme of the embryo. Such knowledge informs researchers addressing the feasibility of the proposed strategy and present alternatives if needed. In this review we describe the mechanisms of inner ear induction, concentrating on the factors that steer the fate of ectoderm into precursors of the inner ear. Induction then leads to inner ear morphogenesis and we describe the cellular changes that occur as the inner ear is converted from a superficial placode to an internalized otocyst, and how they are coordinated with a particular emphasis on how the signaling environment surrounding the inner ear influences these processes.
Collapse
Affiliation(s)
- Xiaorei Sai
- Laboratory for Sensory Development, RIKEN Center for Developmental Biology Kobe, Japan
| | - Raj K Ladher
- Laboratory for Sensory Development, RIKEN Center for Developmental Biology Kobe, Japan
| |
Collapse
|
46
|
Abstract
Cranial sensory placodes derive from discrete patches of the head ectoderm and give rise to numerous sensory structures. During gastrulation, a specialized "neural border zone" forms around the neural plate in response to interactions between the neural and nonneural ectoderm and signals from adjacent mesodermal and/or endodermal tissues. This zone subsequently gives rise to two distinct precursor populations of the peripheral nervous system: the neural crest and the preplacodal ectoderm (PPE). The PPE is a common field from which all cranial sensory placodes arise (adenohypophyseal, olfactory, lens, trigeminal, epibranchial, otic). Members of the Six family of transcription factors are major regulators of PPE specification, in partnership with cofactor proteins such as Eya. Six gene activity also maintains tissue boundaries between the PPE, neural crest, and epidermis by repressing genes that specify the fates of those adjacent ectodermally derived domains. As the embryo acquires anterior-posterior identity, the PPE becomes transcriptionally regionalized, and it subsequently becomes subdivided into specific placodes with distinct developmental fates in response to signaling from adjacent tissues. Each placode is characterized by a unique transcriptional program that leads to the differentiation of highly specialized cells, such as neurosecretory cells, sensory receptor cells, chemosensory neurons, peripheral glia, and supporting cells. In this review, we summarize the transcriptional and signaling factors that regulate key steps of placode development, influence subsequent sensory neuron specification, and discuss what is known about mutations in some of the essential PPE genes that underlie human congenital syndromes.
Collapse
Affiliation(s)
- Sally A Moody
- Department of Anatomy and Regenerative Biology, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA; George Washington University Institute for Neuroscience, Washington, DC, USA.
| | - Anthony-Samuel LaMantia
- George Washington University Institute for Neuroscience, Washington, DC, USA; Department of Pharmacology and Physiology, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
47
|
Edlund RK, Birol O, Groves AK. The role of foxi family transcription factors in the development of the ear and jaw. Curr Top Dev Biol 2015; 111:461-95. [PMID: 25662269 DOI: 10.1016/bs.ctdb.2014.11.014] [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] [Indexed: 12/24/2022]
Abstract
The mammalian outer, middle, and inner ears have different embryonic origins and evolved at different times in the vertebrate lineage. The outer ear is derived from first and second branchial arch ectoderm and mesoderm, the middle ear ossicles are derived from neural crest mesenchymal cells that invade the first and second branchial arches, whereas the inner ear and its associated vestibule-acoustic (VIIIth) ganglion are derived from the otic placode. In this chapter, we discuss recent findings in the development of these structures and describe the contributions of members of a Forkhead transcription factor family, the Foxi family to their formation. Foxi transcription factors are critical for formation of the otic placode, survival of the branchial arch neural crest, and developmental remodeling of the branchial arch ectoderm.
Collapse
Affiliation(s)
- Renée K Edlund
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Onur Birol
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Andrew K Groves
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA; Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA.
| |
Collapse
|
48
|
Schlosser G. Vertebrate cranial placodes as evolutionary innovations--the ancestor's tale. Curr Top Dev Biol 2015; 111:235-300. [PMID: 25662263 DOI: 10.1016/bs.ctdb.2014.11.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Evolutionary innovations often arise by tinkering with preexisting components building new regulatory networks by the rewiring of old parts. The cranial placodes of vertebrates, ectodermal thickenings that give rise to many of the cranial sense organs (ear, nose, lateral line) and ganglia, originated as such novel structures, when vertebrate ancestors elaborated their head in support of a more active and exploratory life style. This review addresses the question of how cranial placodes evolved by tinkering with ectodermal patterning mechanisms and sensory and neurosecretory cell types that have their own evolutionary history. With phylogenetic relationships among the major branches of metazoans now relatively well established, a comparative approach is used to infer, which structures evolved in which lineages and allows us to trace the origin of placodes and their components back from ancestor to ancestor. Some of the core networks of ectodermal patterning and sensory and neurosecretory differentiation were already established in the common ancestor of cnidarians and bilaterians and were greatly elaborated in the bilaterian ancestor (with BMP- and Wnt-dependent patterning of dorsoventral and anteroposterior ectoderm and multiple neurosecretory and sensory cell types). Rostral and caudal protoplacodal domains, giving rise to some neurosecretory and sensory cells, were then established in the ectoderm of the chordate and tunicate-vertebrate ancestor, respectively. However, proper cranial placodes as clusters of proliferating progenitors producing high-density arrays of neurosecretory and sensory cells only evolved and diversified in the ancestors of vertebrates.
Collapse
Affiliation(s)
- Gerhard Schlosser
- School of Natural Sciences & Regenerative Medicine Institute (REMEDI), National University of Ireland, Galway, Ireland.
| |
Collapse
|
49
|
Yan B, Neilson KM, Ranganathan R, Maynard T, Streit A, Moody SA. Microarray identification of novel genes downstream of Six1, a critical factor in cranial placode, somite, and kidney development. Dev Dyn 2014; 244:181-210. [PMID: 25403746 DOI: 10.1002/dvdy.24229] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 11/03/2014] [Accepted: 11/12/2014] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Six1 plays an important role in the development of several vertebrate organs, including cranial sensory placodes, somites, and kidney. Although Six1 mutations cause one form of branchio-otic syndrome (BOS), the responsible gene in many patients has not been identified; genes that act downstream of Six1 are potential BOS candidates. RESULTS We sought to identify novel genes expressed during placode, somite and kidney development by comparing gene expression between control and Six1-expressing ectodermal explants. The expression patterns of 19 of the significantly up-regulated and 11 of the significantly down-regulated genes were assayed from cleavage to larval stages. A total of 28/30 genes are expressed in the otocyst, a structure that is functionally disrupted in BOS, and 26/30 genes are expressed in the nephric mesoderm, a structure that is functionally disrupted in the related branchio-otic-renal (BOR) syndrome. We also identified the chick homologues of five genes and show that they have conserved expression patterns. CONCLUSIONS Of the 30 genes selected for expression analyses, all are expressed at many of the developmental times and appropriate tissues to be regulated by Six1. Many have the potential to play a role in the disruption of hearing and kidney function seen in BOS/BOR patients.
Collapse
Affiliation(s)
- Bo Yan
- Department of Anatomy and Regenerative Biology, The George Washington University, School of Medicine and Health Sciences, Washington, DC
| | | | | | | | | | | |
Collapse
|
50
|
Cajal M, Creuzet SE, Papanayotou C, Sabéran-Djoneidi D, Chuva de Sousa Lopes SM, Zwijsen A, Collignon J, Camus A. A conserved role for non-neural ectoderm cells in early neural development. Development 2014; 141:4127-38. [DOI: 10.1242/dev.107425] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
During the early steps of head development, ectodermal patterning leads to the emergence of distinct non-neural and neural progenitor cells. The induction of the preplacodal ectoderm and the neural crest depends on well-studied signalling interactions between the non-neural ectoderm fated to become epidermis and the prospective neural plate. By contrast, the involvement of the non-neural ectoderm in the morphogenetic events leading to the development and patterning of the central nervous system has been studied less extensively. Here, we show that the removal of the rostral non-neural ectoderm abutting the prospective neural plate at late gastrulation stage leads, in mouse and chick embryos, to morphological defects in forebrain and craniofacial tissues. In particular, this ablation compromises the development of the telencephalon without affecting that of the diencephalon. Further investigations of ablated mouse embryos established that signalling centres crucial for forebrain regionalization, namely the axial mesendoderm and the anterior neural ridge, form normally. Moreover, changes in cell death or cell proliferation could not explain the specific loss of telencephalic tissue. Finally, we provide evidence that the removal of rostral tissues triggers misregulation of the BMP, WNT and FGF signalling pathways that may affect telencephalon development. This study opens new perspectives on the role of the neural/non-neural interface and reveals its functional relevance across higher vertebrates.
Collapse
Affiliation(s)
- Marieke Cajal
- Université Paris Diderot, Sorbonne Paris Cité, Institut Jacques Monod, UMR7592 CNRS, Paris F-75013, France
| | - Sophie E. Creuzet
- Institut de Neurobiologie, Laboratoire Neurobiologie et Développement, CNRS-UPR3294, avenue de la Terrasse, Gif-sur-Yvette 91198, France
| | - Costis Papanayotou
- Université Paris Diderot, Sorbonne Paris Cité, Institut Jacques Monod, UMR7592 CNRS, Paris F-75013, France
| | - Délara Sabéran-Djoneidi
- Université Paris Diderot, Sorbonne Paris Cité, Institut Jacques Monod, UMR7592 CNRS, Paris F-75013, France
| | | | - An Zwijsen
- Laboratory of Developmental Signaling, VIB Center for the Biology of Disease, and KU Leuven, Department for Human Genetics, Leuven 3000, Belgium
| | - Jérôme Collignon
- Université Paris Diderot, Sorbonne Paris Cité, Institut Jacques Monod, UMR7592 CNRS, Paris F-75013, France
| | - Anne Camus
- Université Paris Diderot, Sorbonne Paris Cité, Institut Jacques Monod, UMR7592 CNRS, Paris F-75013, France
| |
Collapse
|