Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Development ; 149(11)2022 06 01.
Article in English | MEDLINE | ID: mdl-35588250

ABSTRACT

Although lengthening of the cell cycle and G1 phase is a generic feature of tissue maturation during development, the underlying mechanism remains poorly understood. Here, we develop a time-lapse imaging strategy to measure the four cell cycle phases in single chick neural progenitor cells in their endogenous environment. We show that neural progenitors are widely heterogeneous with respect to cell cycle length. This variability in duration is distributed over all phases of the cell cycle, with the G1 phase contributing the most. Within one cell cycle, each phase duration appears stochastic and independent except for a correlation between S and M phase duration. Lineage analysis indicates that the majority of daughter cells may have a longer G1 phase than mother cells, suggesting that, at each cell cycle, a mechanism lengthens the G1 phase. We identify that the CDC25B phosphatase known to regulate the G2/M transition indirectly increases the duration of the G1 phase, partly through delaying passage through the restriction point. We propose that CDC25B increases the heterogeneity of G1 phase length, revealing a previously undescribed mechanism of G1 lengthening that is associated with tissue development.


Subject(s)
Neural Stem Cells , Cell Cycle/physiology , Cell Division , G1 Phase/physiology , cdc25 Phosphatases/genetics , cdc25 Phosphatases/metabolism
2.
J Neurosci ; 43(7): 1154-1165, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36596698

ABSTRACT

During development, cortical neurons are produced in a temporally regulated sequence from apical progenitors, directly or indirectly, through the production of intermediate basal progenitors. The balance between these major progenitor types is critical for the production of the proper number and types of neurons, and it is thus important to decipher the cellular and molecular cues controlling this equilibrium. Here we address the role of a cell cycle regulator, the CDC25B phosphatase, in this process. We show that, in the developing mouse neocortex of both sex, deleting CDC25B in apical progenitors leads to a transient increase in the production of TBR1+ neurons at the expense of TBR2+ basal progenitors. This phenotype is associated with lengthening of the G2 phase of the cell cycle, the total cell cycle length being unaffected. Using in utero electroporation and cortical slice cultures, we demonstrate that the defect in TBR2+ basal progenitor production requires interaction with CDK1 and is because of the G2 phase lengthening in CDC25B mutants. Together, this study identifies a new role for CDC25B and G2 phase length in direct versus indirect neurogenesis at early stages of cortical development.SIGNIFICANCE STATEMENT This study is the first analysis of the function of CDC25B, a G2/M regulator, in the developing neocortex. We show that removing CDC25B function leads to a transient increase in neuronal differentiation at early stages, occurring simultaneously with a decrease in basal intermediate progenitors (bIPs). Conversely, a CDC25B gain of function promotes production of bIPs, and this is directly related to CDC25B's ability to regulate CDK1 activity. This imbalance of neuron/progenitor production is linked to a G2 phase lengthening in apical progenitors; and using pharmacological treatments on cortical slice cultures, we show that shortening the G2 phase is sufficient to enhance bIP production. Our results reveal the importance of G2 phase length regulation for neural progenitor fate determination.


Subject(s)
Neocortex , Neural Stem Cells , Neurogenesis , Animals , Mice , cdc25 Phosphatases/genetics , cdc25 Phosphatases/metabolism , Neural Stem Cells/metabolism , Neurogenesis/genetics , Neurons/metabolism
3.
Development ; 139(6): 1095-104, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22318230

ABSTRACT

During embryonic development, changes in cell cycle kinetics have been associated with neurogenesis. This observation suggests that specific cell cycle regulators may be recruited to modify cell cycle dynamics and influence the decision between proliferation and differentiation. In the present study, we investigate the role of core positive cell cycle regulators, the CDC25 phosphatases, in this process. We report that, in the developing chicken spinal cord, only CDC25A is expressed in domains where neural progenitors undergo proliferative self-renewing divisions, whereas the combinatorial expression of CDC25A and CDC25B correlates remarkably well with areas where neurogenesis occurs. We also establish that neural progenitors expressing both CDC25A and CDC25B have a shorter G2 phase than those expressing CDC25A alone. We examine the functional relevance of these correlations using an RNAi-based method that allows us to knock down CDC25B efficiently and specifically. Reducing CDC25B expression results in a specific lengthening of the G2 phase, whereas the S-phase length and the total cell cycle time are not significantly modified. This modification of cell cycle kinetics is associated with a reduction in neuron production that is due to the altered conversion of proliferating neural progenitor cells to post-mitotic neurons. Thus, expression of CDC25B in neural progenitors has two functions: to change cell cycle kinetics and in particular G2-phase length and also to promote neuron production, identifying new roles for this phosphatase during neurogenesis.


Subject(s)
G2 Phase , Nervous System/embryology , Neural Stem Cells/cytology , Neurogenesis , Spinal Cord/embryology , cdc25 Phosphatases/metabolism , Animals , Cell Cycle/physiology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation , Chick Embryo , Neural Stem Cells/physiology , Neurons/metabolism , RNA Interference , RNA, Small Interfering , Spinal Cord/cytology , cdc25 Phosphatases/biosynthesis
4.
Cell Tissue Res ; 359(1): 201-13, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25260908

ABSTRACT

Deciphering the core machinery of the cell cycle and cell division has been primarily the focus of cell biologists, while developmental biologists have identified the signaling pathways and transcriptional programs controlling cell fate choices. As a result, until recently, the interplay between these two fundamental aspects of biology have remained largely unexplored. Increasing data show that the cell cycle and regulators of the core cell cycle machinery are important players in cell fate decisions during neurogenesis. Here, we summarize recent data describing how cell cycle dynamics affect the switch between proliferation and differentiation, with an emphasis on the roles played by the cell cycle regulators, the CDC25 phosphatases.


Subject(s)
Cell Cycle , Cell Lineage , Nervous System/cytology , Nervous System/enzymology , cdc25 Phosphatases/metabolism , Animals , Cell Differentiation , Humans , Neurogenesis
5.
Dev Biol ; 344(2): 611-20, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20488175

ABSTRACT

In the vertebrate central nervous system (CNS), astrocytes are the most abundant and functionally diverse glial cell population. However, the mechanisms underlying their specification and differentiation are still poorly understood. In this study, we have defined spatially and temporally the origin of astrocytes and studied the role of BMPs in astrocyte development in the embryonic chick spinal cord. Using explant cultures, we show that astrocyte precursors started migrating out of the neuroepithelium in the mantle layer from E5, and that the dorsal-most level of the neuroepithelium, from the roof plate to the dl3 level, did not generate GFAP-positive astrocytes. Using a variety of early astrocyte markers together with functional analyses, we show that dorsal-most progenitors displayed a potential for astrocyte production but that dorsally-derived BMP signalling, possibly mediated through BMP receptor 1B, promoted neuronal specification instead. BMP treatment completely prevented astrocyte development from intermediate spinal cord explants at E5, whereas it promoted it at E6. Such an abrupt change in the response of this tissue to BMP signalling could be correlated to the onset of new foci of BMP activity and enhanced expression of BMP receptor 1A, suggesting that BMP signalling could promote astrocyte development in this region.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/physiology , Spinal Cord/embryology , Spinal Cord/metabolism , Animals , Astrocytes/metabolism , Cell Differentiation , Chick Embryo , Embryo, Nonmammalian , Glial Fibrillary Acidic Protein/metabolism , Neuroglia/metabolism , Neuroglia/physiology , Neurons/metabolism , Signal Transduction/physiology , Spinal Cord/cytology
6.
Neural Dev ; 14(1): 7, 2019 03 13.
Article in English | MEDLINE | ID: mdl-30867016

ABSTRACT

In the developing neural tube in chicken and mammals, neural stem cells proliferate and differentiate according to a stereotyped spatiotemporal pattern. Several actors have been identified in the control of this process, from tissue-scale morphogens patterning to intrinsic determinants in neural progenitor cells. In a previous study (Bonnet et al. eLife 7, 2018), we have shown that the CDC25B phosphatase promotes the transition from proliferation to differentiation by stimulating neurogenic divisions, suggesting that it acts as a maturating factor for neural progenitors. In this previous study, we set up a mathematical model linking fixed progenitor modes of division to the dynamics of progenitors and differentiated populations. Here, we extend this model over time to propose a complete dynamical picture of this process. We start from the standard paradigm that progenitors are homogeneous and can perform any type of divisions (proliferative division yielding two progenitors, asymmetric neurogenic divisions yielding one progenitor and one neuron, and terminal symmetric divisions yielding two neurons). We calibrate this model using data published by Saade et al. (Cell Reports 4, 2013) about mode of divisions and population dynamics of progenitors/neurons at different developmental stages. Next, we explore the scenarios in which the progenitor population is actually split into two different pools, one of which is composed of cells that have lost the capacity to perform proliferative divisions. The scenario in which asymmetric neurogenic division would induce such a loss of proliferative capacity appears very relevant.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , Models, Biological , Neural Stem Cells/physiology , Neural Tube/cytology , Neural Tube/growth & development , Spinal Cord/cytology , Spinal Cord/growth & development , cdc25 Phosphatases/physiology , Animals
7.
Elife ; 72018 07 03.
Article in English | MEDLINE | ID: mdl-29969095

ABSTRACT

A fundamental issue in developmental biology and in organ homeostasis is understanding the molecular mechanisms governing the balance between stem cell maintenance and differentiation into a specific lineage. Accumulating data suggest that cell cycle dynamics play a major role in the regulation of this balance. Here we show that the G2/M cell cycle regulator CDC25B phosphatase is required in mammals to finely tune neuronal production in the neural tube. We show that in chick neural progenitors, CDC25B activity favors fast nuclei departure from the apical surface in early G1, stimulates neurogenic divisions and promotes neuronal differentiation. We design a mathematical model showing that within a limited period of time, cell cycle length modifications cannot account for changes in the ratio of the mode of division. Using a CDC25B point mutation that cannot interact with CDK, we show that part of CDC25B activity is independent of its action on the cell cycle.


Subject(s)
Cell Cycle/genetics , Models, Statistical , Neural Stem Cells/enzymology , Neural Tube/enzymology , Neurogenesis/genetics , cdc25 Phosphatases/genetics , Animals , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Differentiation , Chick Embryo , Chickens , Embryo, Mammalian , Gene Expression Regulation, Developmental , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Mice , Mice, Knockout , Neural Stem Cells/cytology , Neural Tube/cytology , Neural Tube/growth & development , Neurons/cytology , Neurons/enzymology , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , Point Mutation , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Spinal Cord/cytology , Spinal Cord/enzymology , Spinal Cord/growth & development , Time-Lapse Imaging , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , cdc25 Phosphatases/metabolism
8.
Neural Dev ; 13(1): 3, 2018 03 08.
Article in English | MEDLINE | ID: mdl-29519242

ABSTRACT

BACKGROUND: Most oligodendrocytes of the spinal cord originate from ventral progenitor cells of the pMN domain, characterized by expression of the transcription factor Olig2. A minority of oligodendrocytes is also recognized to emerge from dorsal progenitors during fetal development. The prevailing view is that generation of ventral oligodendrocytes depends on Sonic hedgehog (Shh) while dorsal oligodendrocytes develop under the influence of Fibroblast Growth Factors (FGFs). RESULTS: Using the well-established model of the chicken embryo, we show that ventral spinal progenitor cells activate FGF signaling at the onset of oligodendrocyte precursor cell (OPC) generation. Inhibition of FGF receptors at that time appears sufficient to prevent generation of ventral OPCs, highlighting that, in addition to Shh, FGF signaling is required also for generation of ventral OPCs. We further reveal an unsuspected interplay between Shh and FGF signaling by showing that FGFs serve dual essential functions in ventral OPC specification. FGFs are responsible for timely induction of a secondary Shh signaling center, the lateral floor plate, a crucial step to create the burst of Shh required for OPC specification. At the same time, FGFs prevent down-regulation of Olig2 in pMN progenitor cells as these cells receive higher threshold of the Shh signal. Finally, we bring arguments favoring a key role of newly differentiated neurons acting as providers of the FGF signal required to trigger OPC generation in the ventral spinal cord. CONCLUSION: Altogether our data reveal that the FGF signaling pathway is activated and required for OPC commitment in the ventral spinal cord. More generally, our data may prove important in defining strategies to produce large populations of determined oligodendrocyte precursor cells from undetermined neural progenitors, including stem cells. In the long run, these new data could be useful in attempts to stimulate the oligodendrocyte fate in residing neural stem cells.


Subject(s)
Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental/physiology , Hedgehog Proteins/metabolism , Oligodendroglia/metabolism , Signal Transduction/physiology , Spinal Cord/cytology , Animals , Chick Embryo , Electroporation , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation, Developmental/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hedgehog Proteins/genetics , In Vitro Techniques , Nerve Tissue Proteins , Oligodendrocyte Transcription Factor 2/metabolism , Organ Culture Techniques , Spinal Cord/embryology , Stem Cells/physiology
9.
J Neurosci ; 26(19): 5037-48, 2006 May 10.
Article in English | MEDLINE | ID: mdl-16687495

ABSTRACT

In the embryonic chick ventral spinal cord, the initial emergence of oligodendrocytes is a relatively late event that depends on prolonged Sonic hedgehog (Shh) signaling. In this report, we show that specification of oligodendrocyte precursors (OLPs) from ventral Nkx2.2-expressing neural progenitors occurs precisely when these progenitors stop generating neurons, indicating that the mechanism of the neuronal/oligodendroglial switch is a common feature of ventral OLP specification. We further show that an experimental early increase in the concentration of Shh is sufficient to induce premature specification of OLPs at the expense of neuronal genesis indicating that the relative doses of Shh received by ventral progenitors determine whether they become neurons or glia. Accordingly, we observe that the Shh protein accumulates at the apical surface of Nkx2.2-expressing cells just before OLP specification, providing direct evidence that these cells are subjected to a higher concentration of the morphogen when they switch to an oligodendroglial fate. Finally, we show that this abrupt change in Shh distribution is most likely attributable to the timely activity of Sulfatase 1 (Sulf1), a secreted enzym that modulates the sulfation state of heparan sulfate proteoglycans. Sulf1 is expressed in the ventral neuroepithelium just before OLP specification, and we show that its experimental overexpression leads to apical concentration of Shh on neuroepithelial cells, a decisive event for the switch of ventral neural progenitors toward an oligodendroglial fate.


Subject(s)
Oligodendroglia/cytology , Oligodendroglia/metabolism , Spinal Cord/cytology , Spinal Cord/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Sulfotransferases/metabolism , Trans-Activators/metabolism , Animals , Cell Differentiation , Cells, Cultured , Chick Embryo , Hedgehog Proteins , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Neurons/cytology , Neurons/metabolism , Nuclear Proteins , Signal Transduction/physiology , Transcription Factors
10.
Cell Rep ; 17(6): 1473-1481, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27806288

ABSTRACT

During spinal cord development, astrocyte precursors arise from neuroepithelial progenitors, delaminate from the ventricular zone, and migrate toward their final locations where they differentiate. Although the mechanisms underlying their early specification and late differentiation are being deciphered, less is known about the temporal control of their migration. Here, we show that the epithelial-mesenchymal transition regulator Zeb1 is expressed in glial precursors and report that loss of Zeb1 function specifically delays the onset of astrocyte precursor delamination from the ventricular zone, correlating with transient deregulation of the adhesion protein Cadherin-1. Consequently, astrocyte precursor invasion into the Zeb1-/- mutant white matter is delayed, and induction of their differentiation is postponed. These findings illustrate how fine regulation of adhesive properties influences the onset of neural precursor migration and further support the notion that duration of exposure of migrating astrocyte precursors to environmental cues and/or their correct positioning influence the timing of their differentiation.


Subject(s)
Astrocytes/cytology , Astrocytes/metabolism , Cell Movement , Spinal Cord/cytology , Stem Cells/cytology , Stem Cells/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism , Aging/genetics , Animals , Body Patterning , Cell Differentiation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Mice , Mutation/genetics
12.
PLoS One ; 4(3): e4900, 2009.
Article in English | MEDLINE | ID: mdl-19333380

ABSTRACT

BACKGROUND: In mammals, a temporal disconnection between mRNA transcription and protein synthesis occurs during late steps of germ cell differentiation, in contrast to most somatic tissues where transcription and translation are closely linked. Indeed, during late stages of spermatogenesis, protein synthesis relies on the appropriate storage of translationally inactive mRNAs in transcriptionally silent spermatids. The factors and cellular compartments regulating mRNA storage and the timing of their translation are still poorly understood. The chromatoid body (CB), that shares components with the P. bodies found in somatic cells, has recently been proposed to be a site of mRNA processing. Here, we describe a new component of the CB, the RNA binding protein HuR, known in somatic cells to control the stability/translation of AU-rich containing mRNAs (ARE-mRNAs). METHODOLOGY/PRINCIPAL FINDINGS: Using a combination of cell imagery and sucrose gradient fractionation, we show that HuR localization is highly dynamic during spermatid differentiation. First, in early round spermatids, HuR colocalizes with the Mouse Vasa Homolog, MVH, a marker of the CB. As spermatids differentiate, HuR exits the CB and concomitantly associates with polysomes. Using computational analyses, we identified two testis ARE-containing mRNAs, Brd2 and GCNF that are bound by HuR and MVH. We show that these target ARE-mRNAs follow HuR trafficking, accumulating successively in the CB, where they are translationally silent, and in polysomes during spermatid differentiation. CONCLUSIONS/SIGNIFICANCE: Our results reveal a temporal regulation of HuR trafficking together with its target mRNAs from the CB to polysomes as spermatids differentiate. They strongly suggest that through the transport of ARE-mRNAs from the CB to polysomes, HuR controls the appropriate timing of ARE-mRNA translation. HuR might represent a major post-transcriptional regulator, by promoting mRNA storage and then translation, during male germ cell differentiation.


Subject(s)
Antigens, Surface/metabolism , Polyribosomes/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Spermatogenesis , Animals , Antigens, Surface/genetics , Antigens, Surface/physiology , Cell Differentiation , ELAV Proteins , ELAV-Like Protein 1 , Male , Mice , Protein Transport , RNA Stability , RNA-Binding Proteins/genetics , RNA-Binding Proteins/physiology , Spermatids/cytology , Testis , Time Factors
13.
Dev Biol ; 303(2): 800-13, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17173889

ABSTRACT

During development, neural cell fate in the vertebrate optic nerve is restricted to the astroglial lineage. However, when isolated from the embryo and explanted in vitro, optic nerve progenitors generate neurons instead of astrocytes, suggesting that neuronal potentialities exist and are repressed in progenitors in vivo. Here we have investigated the mechanisms controlling cell fate in the optic nerve. The optic nerve is characterized by expression of the homeodomain transcription factor Pax2 which is maintained in differentiated astrocytes. We have observed that Pax2 is rapidly down-regulated in explanted optic nerves that generate neurons, and that its overexpression by electroporation in the optic nerve, or ectopically in the neural tube, is sufficient to block neuronal differentiation and allow glial development, showing that Pax2 plays a major role in controlling cell fate in the optic nerve. In vitro and ex vivo experiments further show that a signaling cascade that involves successively Sonic hedgehog and FGF is required to maintain Pax2 expression in optic nerve precursors whereby inhibiting the neuronal fate and promoting astroglial differentiation.


Subject(s)
Optic Nerve/embryology , Optic Nerve/metabolism , PAX2 Transcription Factor/metabolism , Animals , Apoptosis/drug effects , Bone Morphogenetic Proteins/pharmacology , Cell Differentiation , Chick Embryo , Epistasis, Genetic , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neurons/metabolism , Optic Nerve/cytology , PAX2 Transcription Factor/genetics , Pigmentation/drug effects , Signal Transduction
15.
Development ; 129(22): 5117-30, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12399304

ABSTRACT

In the vertebrate spinal cord, oligodendrocytes originate from a restricted region of the ventral neuroepithelium. This ventral localisation of oligodendrocyte precursors (OLPs) depends on the inductive influence of sonic hedgehog (Shh) secreted by ventral midline cells. We have investigated whether the ventral restriction of OLP specification might also depend on inhibiting signals mediated by bone morphogenetic proteins (BMPs). BMPs invariably and markedly inhibited oligodendrocyte development in ventral neural tissue both in vitro and in vivo. Conversely, in vivo ablation of the dorsal most part of the chick spinal cord or inactivation of BMP signalling using grafts of noggin-producing cells promoted the appearance of neuroepithelial OLPs dorsal to their normal domain of emergence, showing that endogenous BMPs contribute to the inhibition of oligodendrocyte development in the spinal cord. BMPs were able to oppose the Shh-mediated induction of OLPs in spinal cord neuroepithelial explants dissected before oligodendrocyte induction, suggesting that BMPs may repress OLP specification by interfering with Shh signalling in vivo. Strikingly, among the transcription factors involved in OLP specification, BMP treatment strongly inhibited the expression of Olig2 but not of Nkx2.2, suggesting that BMP-mediated inhibition of oligodendrogenesis is controlled through the repression of the former transcription factor. Altogether, our data show that oligodendrogenesis is not only regulated by ventral inductive signals such as Shh, but also by dorsal inhibiting signals including BMP factors. They suggest that the dorsoventral position of OLPs depends on a tightly regulated balance between Shh and BMP activities.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Oligodendroglia/metabolism , Spinal Cord/embryology , Spinal Cord/metabolism , Transforming Growth Factor beta , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/pharmacology , Cell Lineage , Chick Embryo , Culture Techniques/methods , Fetal Tissue Transplantation , Hedgehog Proteins , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/drug effects , Homeodomain Proteins/metabolism , Neural Crest/drug effects , Neural Crest/metabolism , Oligodendroglia/drug effects , Spinal Cord/cytology , Stem Cells/metabolism , Trans-Activators/drug effects , Trans-Activators/metabolism , Transcription Factors/drug effects , Transcription Factors/metabolism , Zebrafish Proteins
16.
Dev Biol ; 270(2): 308-21, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15183716

ABSTRACT

In the developing spinal cord, oligodendrocyte progenitors (OLPs) originate from the ventral neuroepithelium and the specification of this lineage depends on the inductive activity of Sonic hedgehog (Shh) produced by ventral midline cells. On the other hand, it has been shown that OLP identity is acquired by the coexpression of the transcription factors olig2 and nkx2.2. Although initially expressed in adjacent nonoverlapping domains of the ventral neuroepithelium, these transcription factors become coexpressed in the pMN domain at the time of OLP specification through dorsal extension of the Nkx2.2 domain. Here we show that Shh is sufficient to promote the coexpression of Olig2 and Nkx2.2 in neuroepithelial cells. In addition, Shh activity is necessary for this coexpression since blocking Shh signalling totally abolishes Olig2 expression and impedes dorsal extension of Nkx2.2. Although Shh at these stages affects neuroepithelial cell proliferation, the dorsal extension of the Nkx2.2 domain is not due to progenitor proliferation but to repatterning of the ventral neuroepithelium. Finally, Shh not only stimulates OLP specification but also simultaneously restricts the ventral extension of the astrocyte progenitor (AP) domain and reduces astrocyte development. We propose that specification of distinct glial lineages is the result of a choice that depends on Shh signalling.


Subject(s)
Astrocytes/physiology , Gene Expression Regulation, Developmental , Oligodendroglia/physiology , Signal Transduction , Spinal Cord/embryology , Trans-Activators/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors , Bromodeoxyuridine , Chick Embryo , DNA Primers , Hedgehog Proteins , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , In Situ Nick-End Labeling , Nerve Tissue Proteins/metabolism , Nuclear Proteins , Trans-Activators/physiology , Transcription Factors/metabolism , Zebrafish Proteins
17.
Mol Cell Neurosci ; 25(4): 612-28, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15080891

ABSTRACT

To address the question of the origin of glial cells and the mechanisms leading to their specification, we have sought to identify novel genes expressed in glial progenitors. We adopted suppression subtractive hybridization (SSH) to establish a chick cDNA library enriched for genes specifically expressed at 6 days of incubation (E6) in the ventral neuroepithelium, a tissue previously shown to contain glial progenitors. Screens were then undertaken to select differentially expressed cDNAs, and out of 82 unique SSH clones, 21 were confirmed to display a regionalized expression along the dorsoventral axis of the E6 ventral neuroepithelium. Among these, we identified a transcript coding for the chick orthologue of Sulf1, a recently identified cell surface sulfatase, as a new, early marker of oligodendrocyte (OL) precursors in the chick embryonic spinal cord. This study provides groundwork for the further identification of genes involved in glial specification.


Subject(s)
Central Nervous System/embryology , Epithelial Cells/enzymology , Oligodendroglia/enzymology , Stem Cells/enzymology , Sulfotransferases/genetics , Animals , Biomarkers/analysis , Body Patterning/genetics , Cell Differentiation/genetics , Cell Lineage/genetics , Central Nervous System/cytology , Central Nervous System/enzymology , Chick Embryo , DNA, Complementary/analysis , DNA, Complementary/genetics , Epithelial Cells/cytology , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Enzymologic/genetics , Genomic Library , Nerve Tissue Proteins/genetics , Nucleic Acid Hybridization/methods , Oligodendroglia/cytology , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL