Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 49
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Dev Dyn ; 250(7): 955-973, 2021 07.
Article de Anglais | MEDLINE | ID: mdl-33501723

RÉSUMÉ

BACKGROUND: Neural tube (NT) closure is a complex developmental process that takes place in the early stages of embryogenesis and that is a key step in neurulation. In mammals, the process by which the neural plate generates the NT requires organized cell movements and tissue folding, and it terminates with the fusion of the apposed ends of the neural folds. RESULTS: Here we describe how almost identical cellular and molecular machinery is used to fuse the spinal neural folds as that involved in the repair of epithelial injury in the same area of the embryo. For both natural and wound activated closure of caudal neural tissue, hyaluronic acid and platelet-derived growth factor signaling appear to be crucial for the final fusion step. CONCLUSIONS: There seems to be no general wound healing machinery for all tissues but rather, a tissue-specific epithelial fusion machinery that embryos activate when necessary after abnormal epithelial opening.


Sujet(s)
Cellules épithéliales/physiologie , Tube neural/embryologie , Neurulation/physiologie , Cicatrisation de plaie/physiologie , Animaux , Fusion cellulaire , Cellules cultivées , Embryon de mammifère , Développement embryonnaire/physiologie , Cellules épithéliales/cytologie , Femelle , Foetus/embryologie , Acide hyaluronique/métabolisme , Mâle , Souris , Crête neurale/embryologie , Crête neurale/physiologie , Plaque neurale/embryologie , Plaque neurale/physiologie , Anomalies du tube neural/embryologie , Facteur de croissance dérivé des plaquettes/physiologie , Grossesse
2.
Biosystems ; 198: 104286, 2020 Dec.
Article de Anglais | MEDLINE | ID: mdl-33181236

RÉSUMÉ

This essay represents a critical analysis of the literary data on various types of waves occurring in the amphibian embryos during gastrulation. A surface contraction wave travels through the presumptive neurectoderm during Mexican axolotl gastrulation. This wave coincides temporally and spatially with involution of the inducing chordomesoderm and with the prospective neural plate. By contrast, there is no similar surface contraction wave during African clawed frog gastrulation. However, the clawed frog displays the waves of DNA synthesis and mitosis in the presumptive neurectoderm during gastrulation, whereas no such waves were discovered in axolotl gastrulae. These sets of experimental data are in accordance with the contemporary concept of considerable ontogenetic diversity of the class Amphibia.


Sujet(s)
Différenciation cellulaire/physiologie , Prolifération cellulaire/physiologie , Gastrula/physiologie , Gastrulation/physiologie , Plaque neurale/physiologie , Ambystoma mexicanum , Animaux , Différenciation cellulaire/génétique , Prolifération cellulaire/génétique , Réplication de l'ADN/génétique , Réplication de l'ADN/physiologie , Gastrula/cytologie , Gastrulation/génétique , Mitose/génétique , Mitose/physiologie , Plaque neurale/cytologie , Spécificité d'espèce , Xenopus laevis
3.
Dev Biol ; 462(2): 152-164, 2020 06 15.
Article de Anglais | MEDLINE | ID: mdl-32243887

RÉSUMÉ

The process that partitions the nascent vertebrate central nervous system into forebrain, midbrain, hindbrain, and spinal cord after neural induction is of fundamental interest in developmental biology, and is known to be dependent on Wnt/ß-catenin signaling at multiple steps. Neural induction specifies neural ectoderm with forebrain character that is subsequently posteriorized by graded Wnt signaling: embryological and mutant analyses have shown that progressively higher levels of Wnt signaling induce progressively more posterior fates. However, the mechanistic link between Wnt signaling and the molecular subdivision of the neural ectoderm into distinct domains in the anteroposterior (AP) axis is still not clear. To better understand how Wnt mediates neural AP patterning, we performed a temporal dissection of neural patterning in response to manipulations of Wnt signaling in zebrafish. We show that Wnt-mediated neural patterning in zebrafish can be divided into three phases: (I) a primary AP patterning phase, which occurs during gastrulation, (II) a mes/r1 (mesencephalon-rhombomere 1) specification and refinement phase, which occurs immediately after gastrulation, and (III) a midbrain-hindbrain boundary (MHB) morphogenesis phase, which occurs during segmentation stages. A major outcome of these Wnt signaling phases is the specification of the major compartment divisions of the developing brain: first the MHB, then the diencephalic-mesencephalic boundary (DMB). The specification of these lineage divisions depends upon the dynamic changes of gene transcription in response to Wnt signaling, which we show primarily involves transcriptional repression or indirect activation. We show that otx2b is directly repressed by Wnt signaling during primary AP patterning, but becomes resistant to Wnt-mediated repression during late gastrulation. Also during late gastrulation, Wnt signaling becomes both necessary and sufficient for expression of wnt8b, en2a, and her5 in mes/r1. We suggest that the change in otx2b response to Wnt regulation enables a transition to the mes/r1 phase of Wnt-mediated patterning, as it ensures that Wnts expressed in the midbrain and MHB do not suppress midbrain identity, and consequently reinforce formation of the DMB. These findings integrate important temporal elements into our spatial understanding of Wnt-mediated neural patterning and may serve as an important basis for a better understanding of neural patterning defects that have implications in human health.


Sujet(s)
Plan d'organisation du corps/physiologie , Plaque neurale/physiologie , Voie de signalisation Wnt/physiologie , Animaux , Diencéphale/métabolisme , Ectoderme/métabolisme , Embryon non mammalien/métabolisme , Facteurs de croissance fibroblastique/métabolisme , Gastrula/métabolisme , Gastrulation/physiologie , Régulation de l'expression des gènes au cours du développement/génétique , Protéines à homéodomaine/métabolisme , Mésencéphale/métabolisme , Système nerveux/métabolisme , Plaque neurale/métabolisme , Rhombencéphale/métabolisme , Danio zébré/métabolisme , Protéines de poisson-zèbre/métabolisme
4.
Cell Rep ; 30(2): 465-480.e6, 2020 01 14.
Article de Anglais | MEDLINE | ID: mdl-31940490

RÉSUMÉ

How ubiquitous transcription factors (TFs) coordinate temporal inputs from broadly expressed epigenetic factors to control cell fate remains poorly understood. Here, we uncover a molecular relationship between p53, an abundant embryonic TF, and WDR5, an essential member of the MLL chromatin modifying complex, that regulates mouse embryonic stem cell fate. Wild-type Wdr5 or transient Wdr5 knockout promotes a distinct pattern of global chromatin accessibility and spurs neuroectodermal differentiation through an RbBP5-dependent process in which WDR5 binds to, and activates transcription of, neural genes. Wdr5 rescue after its prolonged inhibition targets WDR5 to mesoderm lineage-specifying genes, stimulating differentiation toward mesoderm fates in a p53-dependent fashion. Finally, we identify a direct interaction between WDR5 and p53 that enables their co-recruitment to, and regulation of, genes known to control cell proliferation and fate. Our results unmask p53-dependent mechanisms that temporally integrate epigenetic WDR5 inputs to drive neuroectoderm and mesoderm differentiation from pluripotent cells.


Sujet(s)
Protéines et peptides de signalisation intracellulaire/métabolisme , Mésoderme/physiologie , Cellules souches embryonnaires de souris/métabolisme , Plaque neurale/physiologie , Protéine p53 suppresseur de tumeur/métabolisme , Animaux , Différenciation cellulaire , Humains , Souris
5.
Nat Cell Biol ; 21(12): 1518-1531, 2019 12.
Article de Anglais | MEDLINE | ID: mdl-31792383

RÉSUMÉ

The first lineage specification of pluripotent mouse epiblast segregates neuroectoderm (NE) from mesoderm and definitive endoderm (ME) by mechanisms that are not well understood. Here we demonstrate that the induction of ME gene programs critically relies on the T-box transcription factors Eomesodermin (also known as Eomes) and Brachyury, which concomitantly repress pluripotency and NE gene programs. Cells deficient in these T-box transcription factors retain pluripotency and differentiate to NE lineages despite the presence of ME-inducing signals transforming growth factor ß (TGF-ß)/Nodal and Wnt. Pluripotency and NE gene networks are additionally repressed by ME factors downstream of T-box factor induction, demonstrating a redundancy in program regulation to safeguard mutually exclusive lineage specification. Analyses of chromatin revealed that accessibility of ME enhancers depends on T-box factor binding, whereas NE enhancers are accessible and already activation primed at pluripotency. This asymmetry of the chromatin landscape thus explains the default differentiation of pluripotent cells to NE in the absence of ME induction that depends on activating and repressive functions of Eomes and Brachyury.


Sujet(s)
Chromatine/génétique , Protéines foetales/génétique , Feuillets embryonnaires/physiologie , Cellules souches pluripotentes/physiologie , Protéines à domaine boîte-T/génétique , Animaux , Différenciation cellulaire/génétique , Lignée cellulaire , Séparation cellulaire/méthodes , Endoderme/physiologie , Femelle , Régulation de l'expression des gènes au cours du développement/génétique , Mâle , Souris , Plaque neurale/physiologie , Facteur de croissance transformant bêta/génétique
6.
J Biol Chem ; 294(8): 2924-2934, 2019 02 22.
Article de Anglais | MEDLINE | ID: mdl-30573686

RÉSUMÉ

Neural tube closure requires apical constriction during which contraction of the apical F-actin network forces the cell into a wedged shape, facilitating the folding of the neural plate into a tube. However, how F-actin assembly at the apical surface is regulated in mammalian neurulation remains largely unknown. We report here that formin homology 2 domain-containing 3 (Fhod3), a formin protein that mediates F-actin assembly, is essential for cranial neural tube closure in mouse embryos. We found that Fhod3 is expressed in the lateral neural plate but not in the floor region of the closing neural plate at the hindbrain. Consistently, in Fhod3-null embryos, neural plate bending at the midline occurred normally, but lateral plates seemed floppy and failed to flex dorsomedially. Because the apical accumulation of F-actin and constriction were impaired specifically at the lateral plates in Fhod3-null embryos, we concluded that Fhod3-mediated actin assembly contributes to lateral plate-specific apical constriction to advance closure. Intriguingly, Fhod3 expression at the hindbrain was restricted to neuromeric segments called rhombomeres. The rhombomere-specific accumulation of apical F-actin induced by the rhombomere-restricted expression of Fhod3 was responsible for the outward bulging of rhombomeres involving apical constriction along the anteroposterior axis, as rhombomeric bulging was less prominent in Fhod3-null embryos than in the wild type. Fhod3 thus plays a crucial role in the morphological changes associated with neural tube closure at the hindbrain by mediating apical constriction not only in the mediolateral but also in the anteroposterior direction, thereby contributing to tube closure and rhombomere segmentation, respectively.


Sujet(s)
Protéines des microfilaments/physiologie , Morphogenèse , Plaque neurale/cytologie , Tube neural/cytologie , Neurulation , Cytosquelette d'actine/métabolisme , Animaux , Cellules cultivées , Femelle , Formines , Souris , Souris knockout , Plaque neurale/physiologie , Tube neural/physiologie
7.
Biosystems ; 173: 214-220, 2018 Nov.
Article de Anglais | MEDLINE | ID: mdl-30554603

RÉSUMÉ

We have designed and constructed a Flipping Stage for a light microscope that can view the whole exterior surface of a 2 mm diameter developing axolotl salamander embryo. It works by rapidly inverting the bottom-heavy embryo, imaging it as it rights itself. The images are then montaged to reconstruct the whole 3D surface versus time, for a full 4D record of the surface. Imaging early stage axolotl development will help discover how cell differentiation and movement takes place in the early embryo. For example, the switch from ectodermal to neural plate cells takes place on the top, animal surface portion the egg/embryo and can be observed using the flipping stage microscope. Detailed pictures of the whole surface need to be obtained so that cell tracking and event histories, such as cell divisions and participation in differentiation waves, of individual cells can be recorded. Imaging the whole exterior of the eggs/embryos will allow for the analysis of cell behavior and the forces the cells experience in their natural setting in the intact or manipulated embryo. This will give insights into embryogenesis, development, developmental disruptions, birth defects, cell differentiation and tissue engineering.


Sujet(s)
Ambystoma mexicanum/physiologie , Développement embryonnaire , Microscopie confocale/instrumentation , Robotique/instrumentation , Animaux , Différenciation cellulaire , Mouvement cellulaire , Embryon non mammalien , Traitement d'image par ordinateur , Microscopie , Microscopie confocale/méthodes , Plaque neurale/physiologie , Robotique/méthodes , Ingénierie tissulaire
8.
Nat Commun ; 9(1): 2583, 2018 07 03.
Article de Anglais | MEDLINE | ID: mdl-29968706

RÉSUMÉ

Embryonic stem cells (ESCs) maintain pluripotency through unique epigenetic states. When ESCs commit to a specific lineage, epigenetic changes in histones and DNA accompany the transition to specialized cell types. Investigating how epigenetic regulation controls lineage specification is critical in order to generate the required cell types for clinical applications. Uhrf1 is a widely known hemi-methylated DNA-binding protein, playing a role in DNA methylation through the recruitment of Dnmt1 and in heterochromatin formation alongside G9a, Trim28, and HDACs. Although Uhrf1 is not essential in ESC self-renewal, it remains elusive how Uhrf1 regulates cell specification. Here we report that Uhrf1 forms a complex with the active trithorax group, the Setd1a/COMPASS complex, to maintain bivalent histone marks, particularly those associated with neuroectoderm and mesoderm specification. Overall, our data demonstrate that Uhrf1 safeguards proper differentiation via bivalent histone modifications.


Sujet(s)
Reprogrammation cellulaire/génétique , Code histone/génétique , Histone-lysine N-methyltransferase/métabolisme , Protéines nucléaires/métabolisme , Animaux , Protéines liant les séquences stimulatrices de type CCAAT , Techniques de reprogrammation cellulaire , Chimère , Méthylation de l'ADN/physiologie , Épigenèse génétique , Femelle , Fibroblastes , Techniques de knock-out de gènes , Cellules HEK293 , Histone-lysine N-methyltransferase/génétique , Histone-lysine N-methyltransferase/isolement et purification , Histone/métabolisme , Humains , Mâle , Mésoderme/cytologie , Mésoderme/physiologie , Souris , Cellules souches embryonnaires de souris , Plaque neurale/cytologie , Plaque neurale/physiologie , Protéines nucléaires/génétique , Culture de cellules primaires , Protéines recombinantes/génétique , Protéines recombinantes/isolement et purification , Protéines recombinantes/métabolisme , Ubiquitin-protein ligases
9.
Dev Biol ; 444 Suppl 1: S3-S13, 2018 12 01.
Article de Anglais | MEDLINE | ID: mdl-30048640

RÉSUMÉ

FOREWORD: The neural crest has been the main object of my investigations during my career in science, up to now. It is a fascinating topic for an embryologist because of its two unique characteristics: its large degree of multipotency and the fact that its development involves a phase during which its component cells migrate all over the embryo and settle in elected sites where they differentiate into a large variety of cell types. Thus, neural crest development raises several specific questions that are at the same time, of general interest: what are the mechanisms controlling the migratory behavior of the cells that detach from the neural plate borders? What are the migration routes taken by the neural crest cells and the environmental factors that make these cells stop in elected sites where they differentiate into a definite series of cell types? When I started to be interested in the neural crest, in the late 1960s, this embryonic structure was the subject of investigations of only a small number of developmental biologists. Fifty years later, it has become the center of interest of many laboratories over the world. The 150th anniversary of its discovery is a relevant opportunity to consider the progress that has been accomplished in our knowledge on the development of this ubiquitous structure, the roles it plays in the physiology of the organism through its numerous and widespread derivatives and its relationships with its environment, as well as the evolutionary advantages it has conferred to the vertebrate phylum. I wish to thank Pr Marianne Bronner, Chief Editor of Developmental Biology and Special Issue Guest Editor, for dedicating a special issue of this journal to this particular structure of the vertebrate embryo. In the following pages, Elisabeth Dupin and I will report some of the highlights of our own acquaintance with the neural crest of the avian embryo, after retracing the main trends of the discoveries of the historical pioneers.


Sujet(s)
Crête neurale/cytologie , Crête neurale/métabolisme , Crête neurale/physiologie , Animaux , Évolution biologique , Plan d'organisation du corps , Différenciation cellulaire/physiologie , Mouvement cellulaire , Embryon de poulet , Mélanocytes/cytologie , Plaque neurale/physiologie , Neurogenèse/physiologie , Caille , Vertébrés
10.
Dev Biol ; 444 Suppl 1: S36-S46, 2018 12 01.
Article de Anglais | MEDLINE | ID: mdl-29852131

RÉSUMÉ

The neural crest is induced at the edge between the neural plate and the nonneural ectoderm, in an area called the neural (plate) border, during gastrulation and neurulation. In recent years, many studies have explored how this domain is patterned, and how the neural crest is induced within this territory, that also participates to the prospective dorsal neural tube, the dorsalmost nonneural ectoderm, as well as placode derivatives in the anterior area. This review highlights the tissue interactions, the cell-cell signaling and the molecular mechanisms involved in this dynamic spatiotemporal patterning, resulting in the induction of the premigratory neural crest. Collectively, these studies allow building a complex neural border and early neural crest gene regulatory network, mostly composed by transcriptional regulations but also, more recently, including novel signaling interactions.


Sujet(s)
Crête neurale/cytologie , Crête neurale/métabolisme , Crête neurale/physiologie , Animaux , Évolution biologique , Plan d'organisation du corps/génétique , Protéines morphogénétiques osseuses/métabolisme , Différenciation cellulaire/physiologie , Mouvement cellulaire , Embryon de poulet , Ectoderme/métabolisme , Facteurs de croissance fibroblastique/métabolisme , Gastrulation/génétique , Régulation de l'expression des gènes au cours du développement/génétique , Régulation de l'expression des gènes au cours du développement/physiologie , Humains , Mélanocytes/cytologie , Système nerveux/métabolisme , Plaque neurale/métabolisme , Plaque neurale/physiologie , Neurogenèse/physiologie , Neurulation/physiologie , Transduction du signal , Voie de signalisation Wnt/physiologie , Protéines de Xénope/génétique , Xenopus laevis/génétique , Danio zébré/génétique , Protéines de poisson-zèbre/génétique
11.
Dev Biol ; 444 Suppl 1: S47-S59, 2018 12 01.
Article de Anglais | MEDLINE | ID: mdl-29614271

RÉSUMÉ

In the neural primordium of vertebrate embryos, the neural crest (NC) displays a unique character: the capacity of its component cells to leave the neural primordium, migrate along definite (and, for long, not identified) routes in the developing embryo and invade virtually all tissues and organs, while producing a large array of differentiated cell types. The most striking diversity of the NC derivatives is found in its cephalic domain that produces, not only melanocytes and peripheral nerves and ganglia, but also various mesenchymal derivatives (connective tissues, bones, cartilages…) which, in other parts of the body, are mesoderm-derived. The aim of this article was to review the large amount of work that has been devoted to solving the problem of the differentiation capacities of individual NC cells (NCC) arising from both the cephalic and trunk levels of the neural axis. A variety of experimental designs applied to NCC either in vivo or in vitro are evaluated, including the possibility to culture them in crestospheres, a technique previously designed for cells of the CNS, and which reinforces the notion, previously put forward, of the existence of NC stem cells. At the trunk level, the developmental potentialities of the NCC are more restricted than in their cephalic counterparts, but, in addition to the neural-melanocytic fate that they exclusively express in vivo, it was clearly shown that they harbor mesenchymal capacities that can be revealed in vitro. Finally, a large amount of evidence has been obtained that, during the migration process, most of the NCC are multipotent with a variable array of potentialities among the cells considered. Investigations carried out in adults have shown that multipotent NC stem cells persist in the various sites of the body occupied by NCC. Enlightening new developments concerning the invasive capacity of NCC, the growing peripheral nerves were revealed as migration routes for NCC travelling to distant ventrolateral regions of the body. Designated "Schwann cell precursors" in the mouse embryo, these NCC can leave the nerves and are able to convert to a novel fate. The convertibility of the NC-derived cells, particularly evident in the Schwann cell-melanocyte lineage transition, has also been demonstrated for neuroendocrine cells of the adult carotid body and for the differentiation of parasympathetic neurons of ganglia distant from their origin, the NC. All these new developments attest the vitality of the research on the NC, a field that characterizes vertebrate development and for which the interest has constantly increased during the last decades.


Sujet(s)
Cellules souches multipotentes/cytologie , Crête neurale/cytologie , Crête neurale/physiologie , Animaux , Évolution biologique , Plan d'organisation du corps , Différenciation cellulaire/physiologie , Mouvement cellulaire/physiologie , Système nerveux central/physiologie , Développement embryonnaire , Transition épithélio-mésenchymateuse/physiologie , Humains , Mélanocytes/cytologie , Mésoderme , Crête neurale/métabolisme , Plaque neurale/physiologie , Cellules souches neurales/cytologie , Neurogenèse/physiologie , Plasticité neuronale/physiologie , Neurones/cytologie , Cellules de Schwann , Vertébrés
12.
Nat Commun ; 8(1): 1339, 2017 11 07.
Article de Anglais | MEDLINE | ID: mdl-29109536

RÉSUMÉ

The neuroectoderm is patterned along a rostral-caudal axis in response to localized factors in the embryo, but exactly how these factors act as positional information for this patterning is not yet fully understood. Here, using the self-organizing properties of mouse embryonic stem cell (ESC), we report that ESC-derived neuroectoderm self-generates a Six3+ rostral and a Irx3+ caudal bipolarized patterning. In this instance, localized Fgf signaling performs dual roles, as it regulates Six3+ rostral polarization at an earlier stage and promotes Wnt signaling at a later stage. The Wnt signaling components are differentially expressed in the polarized tissues, leading to genome-wide Irx3+ caudal-polarization signals. Surprisingly, differentially expressed Wnt agonists and antagonists have essential roles in orchestrating the formation of a balanced rostral-caudal neuroectoderm pattern. Together, our findings provide key processes for dynamic self-patterning and evidence that a temporally and locally regulated interaction between Fgf and Wnt signaling controls self-patterning in ESC-derived neuroectoderm.


Sujet(s)
Facteurs de croissance fibroblastique/métabolisme , Plaque neurale/métabolisme , Protéines de type Wingless/métabolisme , Animaux , Plan d'organisation du corps , Cellules souches embryonnaires , Protéines de l'oeil/génétique , Protéines de l'oeil/métabolisme , Facteur de croissance fibroblastique de type 5/génétique , Facteur de croissance fibroblastique de type 5/métabolisme , Facteurs de croissance fibroblastique/génétique , Régulation de l'expression des gènes au cours du développement , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Protéines et peptides de signalisation intercellulaire/génétique , Protéines et peptides de signalisation intercellulaire/métabolisme , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Souris de lignée ICR , Souris transgéniques , Protéines de tissu nerveux/génétique , Protéines de tissu nerveux/métabolisme , Plaque neurale/physiologie , Transduction du signal , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Protéines de type Wingless/génétique ,
13.
Nat Commun ; 8(1): 1806, 2017 11 27.
Article de Anglais | MEDLINE | ID: mdl-29180618

RÉSUMÉ

Enhancers and long noncoding RNAs (lncRNAs) are key determinants of lineage specification during development. Here, we evaluate remodeling of the enhancer landscape and modulation of the lncRNA transcriptome during mesendoderm specification. We sort mesendodermal progenitors from differentiating embryonic stem cells (ESCs) according to Eomes expression, and find that enhancer usage is coordinated with mesendoderm-specific expression of key lineage-determining transcription factors. Many of these enhancers are associated with the expression of lncRNAs. Examination of ESC-specific enhancers interacting in three-dimensional space with mesendoderm-specifying transcription factor loci identifies MesEndoderm Transcriptional Enhancer Organizing Region (Meteor). Genetic and epigenetic manipulation of the Meteor enhancer reveal its indispensable role during mesendoderm specification and subsequent cardiogenic differentiation via transcription-independent and -dependent mechanisms. Interestingly, Meteor-deleted ESCs are epigenetically redirected towards neuroectodermal lineages. Loci, topologically associating a transcribed enhancer and its cognate protein coding gene, appear to represent therefore a class of genomic elements controlling developmental competence in pluripotency.


Sujet(s)
Ectoderme/physiologie , Cellules souches embryonnaires/physiologie , Éléments activateurs (génétique)/physiologie , Mésoderme/physiologie , ARN long non codant/physiologie , Animaux , Différenciation cellulaire/génétique , Lignée cellulaire , Lignage cellulaire/génétique , Ectoderme/cytologie , Analyse de profil d'expression de gènes/méthodes , Régulation de l'expression des gènes au cours du développement/physiologie , Humains , Cellules souches pluripotentes induites , Mésoderme/cytologie , Souris , Plaque neurale/cytologie , Plaque neurale/physiologie
14.
Int J Dev Neurosci ; 59: 21-30, 2017 Jun.
Article de Anglais | MEDLINE | ID: mdl-28285945

RÉSUMÉ

Inspired by in vivo developmental process, several studies were conducted to design a protocol for differentiating of mesenchymal stem cells into neural cells in vitro. Human adipose-derived stem cells (hADSCs) as mesenchymal stem cells are a promising source for this purpose. At current study, we applied a defined neural induction medium by using small molecules for direct differentiation of hADSCs into anterior neuroectodermal cells. Anterior neuroectodermal differentiation of hADSCs was performed by hanging drop and monolayer protocols. At these methods, three small molecules were used to suppress the BMP, Nodal, and Wnt signaling pathways in order to obtain anterior neuroectodermal (eye field) cells from hADSCs. After two and three weeks of induction, the differentiated cells with neural morphology expressed anterior neuroectodermal markers such as OTX2, SIX3, ß-TUB III and PAX6. The protein expression of such markers was confirmed by real time, RT-PCR and immunocytochemistry methods According to our data, it seems that the hanging drop method is a proper approach for neuroectodermal induction of hADSCs. Considering wide availability and immunosuppressive properties of hADSCs, these cells may open a way for autologous cell therapy of neurodegenerative disorders.


Sujet(s)
Tissu adipeux/cytologie , Techniques de culture cellulaire/méthodes , Différenciation cellulaire/physiologie , Cellules souches mésenchymateuses/physiologie , Plaque neurale/physiologie , Adulte , Antigènes CD/métabolisme , Cellules cultivées , Protéines de l'oeil/génétique , Protéines de l'oeil/métabolisme , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Humains , Protéines de tissu nerveux/génétique , Protéines de tissu nerveux/métabolisme , Facteurs de transcription Otx/génétique , Facteurs de transcription Otx/métabolisme , Facteur de transcription PAX6/génétique , Facteur de transcription PAX6/métabolisme , Tubuline/génétique , Tubuline/métabolisme ,
15.
J Cell Biochem ; 118(7): 1630-1633, 2017 07.
Article de Anglais | MEDLINE | ID: mdl-28059469

RÉSUMÉ

Formation of organs that consist primarily or exclusively of tubes is essential for metazoan development. Increasing evidence suggests that the morphogenesis and homeostasis of these tubular organs depend on proper orientation of the mitotic spindle during cell division. Consequently, improper spindle orientation can perturb spatial arrangement of daughter cells, resulting in congenital malformations or dysfunctions of tubular organs. Over the past decade, the association of spindle misorientation with brain diseases and cancer has been extensively studied. However, few studies have explored the effects of spindle misorientation on developmental disorders impacting tubular organs. Here, we examine and interpret recent literature that shows how tubular organ development relies on proper spindle orientation and discuss how defects in this cellular process are associated with the pathogenesis of tubular organ diseases. J. Cell. Biochem. 118: 1630-1633, 2017. © 2017 Wiley Periodicals, Inc.


Sujet(s)
Appareil du fuseau/physiologie , Animaux , Division cellulaire/génétique , Division cellulaire/physiologie , Polarité de la cellule/génétique , Polarité de la cellule/physiologie , Humains , Plaque neurale/cytologie , Plaque neurale/physiologie , Appareil du fuseau/génétique
16.
Genes Dev ; 30(22): 2538-2550, 2016 11 15.
Article de Anglais | MEDLINE | ID: mdl-27920086

RÉSUMÉ

Mitotic bookmarking transcription factors remain bound to chromosomes during mitosis and were proposed to regulate phenotypic maintenance of stem and progenitor cells at the mitosis-to-G1 (M-G1) transition. However, mitotic bookmarking remains largely unexplored in most stem cell types, and its functional relevance for cell fate decisions remains unclear. Here we screened for mitotic chromosome binding within the pluripotency network of embryonic stem (ES) cells and show that SOX2 and OCT4 remain bound to mitotic chromatin through their respective DNA-binding domains. Dynamic characterization using photobleaching-based methods and single-molecule imaging revealed quantitatively similar specific DNA interactions, but different nonspecific DNA interactions, of SOX2 and OCT4 with mitotic chromatin. Using ChIP-seq (chromatin immunoprecipitation [ChIP] combined with high-throughput sequencing) to assess the genome-wide distribution of SOX2 on mitotic chromatin, we demonstrate the bookmarking activity of SOX2 on a small set of genes. Finally, we investigated the function of SOX2 mitotic bookmarking in cell fate decisions and show that its absence at the M-G1 transition impairs pluripotency maintenance and abrogates its ability to induce neuroectodermal differentiation but does not affect reprogramming efficiency toward induced pluripotent stem cells. Our study demonstrates the mitotic bookmarking property of SOX2 and reveals its functional importance in pluripotency maintenance and ES cell differentiation.


Sujet(s)
Différenciation cellulaire/génétique , Mitose/génétique , Facteurs de transcription SOX-B1/génétique , Facteurs de transcription SOX-B1/métabolisme , Animaux , Reprogrammation cellulaire/génétique , Chromatine/métabolisme , Cellules souches embryonnaires , Phase G1 , Cellules HEK293 , Humains , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Souris , Cellules NIH 3T3 , Plaque neurale/cytologie , Plaque neurale/physiologie , Facteur de transcription Oct-3/génétique , Facteur de transcription Oct-3/métabolisme , Liaison aux protéines
17.
Biochem Biophys Res Commun ; 477(4): 807-813, 2016 09 02.
Article de Anglais | MEDLINE | ID: mdl-27369078

RÉSUMÉ

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.


Sujet(s)
Protéine morphogénétique osseuse de type 4/métabolisme , Ectoderme/embryologie , Ectoderme/métabolisme , Facteur de croissance fibroblastique de type 2/métabolisme , Plaque neurale/embryologie , Plaque neurale/physiologie , Animaux , Poulets , Ectoderme/physiologie , Développement embryonnaire/physiologie , Plaque neurale/cytologie , Techniques de culture d'organes/méthodes , Organogenèse/physiologie , Caille , Ingénierie tissulaire/méthodes
18.
Dev Biol ; 411(2): 294-300, 2016 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-26826496

RÉSUMÉ

Cranial placodes are thickenings in the ectoderm that give rise to sensory organs and peripheral ganglia of the vertebrate head. At gastrula and neurula stages, placodal precursors are intermingled in the neural plate border with future neural and neural crest cells. Here, we show that the epigenetic modifier, DNA methyl transferase (DNMT) 3A, expressed in the neural plate border region, influences development of the otic placode which will contribute to the ear. DNMT3A is expressed in the presumptive otic region at gastrula through neurula stages and later in the otic placode itself. Whereas neural plate border and non-neural ectoderm markers Erni, Dlx5, Msx1 and Six1 are unaltered, DNMT3A loss of function leads to early reduction in the expression of the key otic placode specifier genes Pax2 and Gbx2 and later otic markers Sox10 and Soho1. Reduction of Gbx2 was first observed at HH7, well before loss of other otic markers. Later, this translates to significant reduction in the size of the otic vesicle. Based on these results, we propose that DNMT3A is important for enabling the activation of Gbx2 expression, necessary for normal development of the inner ear.


Sujet(s)
DNA (cytosine-5-)-methyltransferase/génétique , DNA (cytosine-5-)-methyltransferase/physiologie , Oreille interne/embryologie , Oreille interne/physiologie , Épigenèse génétique , Régulation de l'expression des gènes au cours du développement , Animaux , Embryon de poulet , Poulets , Méthylation de l'ADN , DNA methyltransferase 3A , Ectoderme/métabolisme , Développement embryonnaire , Gastrula/métabolisme , Protéines à homéodomaine/physiologie , Hybridation in situ , Crête neurale/embryologie , Plaque neurale/métabolisme , Plaque neurale/physiologie , Facteur de transcription PAX2/métabolisme , Facteurs de transcription PAX/métabolisme
19.
PLoS One ; 10(9): e0137894, 2015.
Article de Anglais | MEDLINE | ID: mdl-26368825

RÉSUMÉ

Bipotent axial stem cells residing in the caudal epiblast during late gastrulation generate neuroectodermal and presomitic mesodermal progeny that coordinate somitogenesis with neural tube formation, but the mechanism that controls these two fates is not fully understood. Retinoic acid (RA) restricts the anterior extent of caudal fibroblast growth factor 8 (Fgf8) expression in both mesoderm and neural plate to control somitogenesis and neurogenesis, however it remains unclear where RA acts to control the spatial expression of caudal Fgf8. Here, we found that mouse Raldh2-/- embryos, lacking RA synthesis and displaying a consistent small somite defect, exhibited abnormal expression of key markers of axial stem cell progeny, with decreased Sox2+ and Sox1+ neuroectodermal progeny and increased Tbx6+ presomitic mesodermal progeny. The Raldh2-/- small somite defect was rescued by treatment with an FGF receptor antagonist. Rdh10 mutants, with a less severe RA synthesis defect, were found to exhibit a small somite defect and anterior expansion of caudal Fgf8 expression only for somites 1-6, with normal somite size and Fgf8 expression thereafter. Rdh10 mutants were found to lack RA activity during the early phase when somites are small, but at the 6-somite stage RA activity was detected in neural plate although not in presomitic mesoderm. Expression of a dominant-negative RA receptor in mesoderm eliminated RA activity in presomitic mesoderm but did not affect somitogenesis. Thus, RA activity in the neural plate is sufficient to prevent anterior expansion of caudal Fgf8 expression associated with a small somite defect. Our studies provide evidence that RA restriction of Fgf8 expression in undifferentiated neural progenitors stimulates neurogenesis while also restricting the anterior extent of the mesodermal Fgf8 mRNA gradient that controls somite size, providing new insight into the mechanism that coordinates somitogenesis with neurogenesis.


Sujet(s)
Aldehyde oxidoreductases/génétique , Facteur de croissance fibroblastique de type 8/métabolisme , Plaque neurale/physiologie , Somites/croissance et développement , Aldehyde oxidoreductases/métabolisme , Animaux , Techniques de culture d'embryons , Régulation de l'expression des gènes au cours du développement , Souris , Plaque neurale/métabolisme , Neurogenèse , Somites/malformations , Trétinoïne
20.
Dev Biol ; 399(2): 218-25, 2015 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-25576927

RÉSUMÉ

The ascidian Ciona intestinalis is a marine invertebrate belonging to the sister group of the vertebrates, the tunicates. Its compact genome and simple, experimentally tractable embryos make Ciona well-suited for the study of cell-fate specification in chordates. Tunicate larvae possess a characteristic chordate body plan, and many developmental pathways are conserved between tunicates and vertebrates. Previous studies have shown that FGF signals are essential for neural induction and patterning at sequential steps of Ciona embryogenesis. Here we show that two different ETS family transcription factors, Ets1/2 and Elk1/3/4, have partially redundant activities in the anterior neural plate of gastrulating embryos. Whereas Ets1/2 promotes pigment cell formation in lateral lineages, both Ets1/2 and Elk1/3/4 are involved in the activation of Myt1L in medial lineages and the restriction of Six3/6 expression to the anterior-most regions of the neural tube. We also provide evidence that photoreceptor cells arise from posterior regions of the presumptive sensory vesicle, and do not depend on FGF signaling. Cells previously identified as photoreceptor progenitors instead form ependymal cells and neurons of the larval brain. Our results extend recent findings on FGF-dependent patterning of anterior-posterior compartments in the Ciona central nervous system.


Sujet(s)
Lignage cellulaire/physiologie , Ciona intestinalis/embryologie , Facteurs de croissance fibroblastique/métabolisme , Plaque neurale/physiologie , Protéine proto-oncogène c-ets-1/métabolisme , Transduction du signal/physiologie , Protéine Elk-1 à domaine ets/métabolisme , Animaux , Clonage moléculaire , Amorces ADN/génétique , Embryon non mammalien/physiologie , Régulation de l'expression des gènes au cours du développement/physiologie , Immunohistochimie , Hybridation in situ , Microscopie confocale
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...