Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
iScience ; 15: 257-273, 2019 May 31.
Article in English | MEDLINE | ID: mdl-31082736

ABSTRACT

Visual system development involves the formation of neuronal projections connecting the retina to the thalamic dorso-lateral geniculate nucleus (dLGN) and the thalamus to the visual cerebral cortex. Patients carrying mutations in the SOX2 transcription factor gene present severe visual defects, thought to be linked to SOX2 functions in the retina. We show that Sox2 is strongly expressed in mouse postmitotic thalamic projection neurons. Cre-mediated deletion of Sox2 in these neurons causes reduction of the dLGN, abnormal distribution of retino-thalamic and thalamo-cortical projections, and secondary defects in cortical patterning. Reduced expression, in mutants, of Sox2 target genes encoding ephrin-A5 and the serotonin transport molecules SERT and vMAT2 (important for establishment of thalamic connectivity) likely provides a molecular contribution to these defects. These findings unveil thalamic SOX2 function as a novel regulator of visual system development and a plausible additional cause of brain-linked genetic blindness in humans.

2.
Cell Stem Cell ; 24(3): 462-476.e6, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30849367

ABSTRACT

The SOX2 transcription factor is critical for neural stem cell (NSC) maintenance and brain development. Through chromatin immunoprecipitation (ChIP) and chromatin interaction analysis (ChIA-PET), we determined genome-wide SOX2-bound regions and Pol II-mediated long-range chromatin interactions in brain-derived NSCs. SOX2-bound DNA was highly enriched in distal chromatin regions interacting with promoters and carrying epigenetic enhancer marks. Sox2 deletion caused widespread reduction of Pol II-mediated long-range interactions and decreased gene expression. Genes showing reduced expression in Sox2-deleted cells were significantly enriched in interactions between promoters and SOX2-bound distal enhancers. Expression of one such gene, Suppressor of Cytokine Signaling 3 (Socs3), rescued the self-renewal defect of Sox2-ablated NSCs. Our work identifies SOX2 as a major regulator of gene expression through connections to the enhancer network in NSCs. Through the definition of such a connectivity network, our study shows the way to the identification of genes and enhancers involved in NSC maintenance and neurodevelopmental disorders.


Subject(s)
Chromatin/metabolism , Neural Stem Cells/metabolism , SOXB1 Transcription Factors/metabolism , Animals , Cells, Cultured , Gene Regulatory Networks/genetics , Mice , Mice, Knockout , Mice, Transgenic , Mutation , SOXB1 Transcription Factors/deficiency , SOXB1 Transcription Factors/genetics , Zebrafish
4.
Neuron ; 93(2): 348-361, 2017 Jan 18.
Article in English | MEDLINE | ID: mdl-28041881

ABSTRACT

Mutations in NIPBL are the most frequent cause of Cornelia de Lange syndrome (CdLS), a developmental disorder encompassing several neurological defects, including intellectual disability and seizures. How NIPBL mutations affect brain development is not understood. Here we identify Nipbl as a functional interaction partner of the neural transcription factor Zfp609 in brain development. Depletion of Zfp609 or Nipbl from cortical neural progenitors in vivo is detrimental to neuronal migration. Zfp609 and Nipbl overlap at genomic binding sites independently of cohesin and regulate genes that control cortical neuron migration. We find that Zfp609 and Nipbl interact with the Integrator complex, which functions in RNA polymerase 2 pause release. Indeed, Zfp609 and Nipbl co-localize at gene promoters containing paused RNA polymerase 2, and Integrator similarly regulates neuronal migration. Our data provide a rationale and mechanistic insights for the role of Nipbl in the neurological defects associated with CdLS.


Subject(s)
Cell Movement/genetics , Cerebral Cortex/growth & development , De Lange Syndrome/genetics , Gene Expression Regulation, Developmental , Neural Stem Cells/cytology , Neurons/cytology , Trans-Activators/genetics , Transcription Factors/genetics , Animals , Cell Cycle Proteins/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Mice , Neural Stem Cells/metabolism , Neurons/metabolism , Promoter Regions, Genetic , RNA Polymerase II/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Cohesins
7.
EMBO Rep ; 16(9): 1177-91, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26265007

ABSTRACT

Embryonic stem cell (ESC) identity is orchestrated by co-operativity between the transcription factors (TFs) Sox2 and the class V POU-TF Oct4 at composite Sox/Oct motifs. Neural stem cells (NSCs) lack Oct4 but express Sox2 and class III POU-TFs Oct6, Brn1 and Brn2. This raises the question of how Sox2 interacts with POU-TFs to transcriptionally specify ESCs versus NSCs. Here, we show that Oct4 alone binds the Sox/Oct motif and the octamer-containing palindromic MORE equally well. Sox2 binding selectively increases the affinity of Oct4 for the Sox/Oct motif. In contrast, Oct6 binds preferentially to MORE and is unaffected by Sox2. ChIP-Seq in NSCs shows the MORE to be the most enriched motif for class III POU-TFs, including MORE subtypes, and that the Sox/Oct motif is not enriched. These results suggest that in NSCs, co-operativity between Sox2 and class III POU-TFs may not occur and that POU-TF-driven transcription uses predominantly the MORE cis architecture. Thus, distinct interactions between Sox2 and POU-TF subclasses distinguish pluripotent ESCs from multipotent NSCs, providing molecular insight into how Oct4 alone can convert NSCs to pluripotency.


Subject(s)
Embryonic Stem Cells/metabolism , Neural Stem Cells/metabolism , POU Domain Factors/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Animals , Embryo, Mammalian , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , POU Domain Factors/genetics , Promoter Regions, Genetic , Transcription Factors/metabolism , Transcription, Genetic
8.
Stem Cells Dev ; 24(18): 2114-26, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26083238

ABSTRACT

Murine postnatal neural stem cells (NSCs) give rise to neurons, astrocytes, or oligodendrocytes (OLs); however, our knowledge of the genes that control this lineage specification is incomplete. In this study, we show that nuclear factor I X (NFIX), a transcription factor known to regulate NSC quiescence, also suppresses oligodendrogenesis (ODG) from NSCs. Immunostaining reveals little or no expression of NFIX in OL lineage cells both in vivo and in vitro. Loss of NFIX from subventricular zone (SVZ) NSCs results in enhanced ODG both in vivo and in vitro, while forced expression of NFIX blocks NSC differentiation into OLs in vitro. RNA-seq analysis shows that genes previously shown to be differentially expressed in OL progenitors are significantly enriched in RNA from Nfix(-/-) versus wild-type NSCs. These data indicate that NFIX influences the lineage specification of postnatal SVZ NSCs, specifically suppressing ODG.


Subject(s)
Lateral Ventricles/embryology , NFI Transcription Factors/genetics , Neural Stem Cells/cytology , Neurogenesis/physiology , Oligodendroglia/cytology , Animals , Astrocytes/cytology , Cell Lineage , Cells, Cultured , Lateral Ventricles/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , SOXE Transcription Factors/metabolism
9.
Genome Res ; 25(1): 41-56, 2015 01.
Article in English | MEDLINE | ID: mdl-25294244

ABSTRACT

The gene regulatory network (GRN) that supports neural stem cell (NS cell) self-renewal has so far been poorly characterized. Knowledge of the central transcription factors (TFs), the noncoding gene regulatory regions that they bind to, and the genes whose expression they modulate will be crucial in unlocking the full therapeutic potential of these cells. Here, we use DNase-seq in combination with analysis of histone modifications to identify multiple classes of epigenetically and functionally distinct cis-regulatory elements (CREs). Through motif analysis and ChIP-seq, we identify several of the crucial TF regulators of NS cells. At the core of the network are TFs of the basic helix-loop-helix (bHLH), nuclear factor I (NFI), SOX, and FOX families, with CREs often densely bound by several of these different TFs. We use machine learning to highlight several crucial regulatory features of the network that underpin NS cell self-renewal and multipotency. We validate our predictions by functional analysis of the bHLH TF OLIG2. This TF makes an important contribution to NS cell self-renewal by concurrently activating pro-proliferation genes and preventing the untimely activation of genes promoting neuronal differentiation and stem cell quiescence.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Cells, Cultured , Cluster Analysis , Epigenomics , Logistic Models , Mice , Microarray Analysis , Models, Theoretical , NFI Transcription Factors/genetics , NFI Transcription Factors/metabolism , Nerve Tissue Proteins/genetics , Oligodendrocyte Transcription Factor 2 , Regulatory Sequences, Nucleic Acid , SOX Transcription Factors/genetics , SOX Transcription Factors/metabolism , Sequence Analysis, DNA
10.
Cereb Cortex ; 25(10): 3758-78, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25331604

ABSTRACT

Transcription factors of the nuclear factor one (NFI) family play a pivotal role in the development of the nervous system. One member, NFIX, regulates the development of the neocortex, hippocampus, and cerebellum. Postnatal Nfix(-/-) mice also display abnormalities within the subventricular zone (SVZ) lining the lateral ventricles, a region of the brain comprising a neurogenic niche that provides ongoing neurogenesis throughout life. Specifically, Nfix(-/-) mice exhibit more PAX6-expressing progenitor cells within the SVZ. However, the mechanism underlying the development of this phenotype remains undefined. Here, we reveal that NFIX contributes to multiple facets of SVZ development. Postnatal Nfix(-/-) mice exhibit increased levels of proliferation within the SVZ, both in vivo and in vitro as assessed by a neurosphere assay. Furthermore, we show that the migration of SVZ-derived neuroblasts to the olfactory bulb is impaired, and that the olfactory bulbs of postnatal Nfix(-/-) mice are smaller. We also demonstrate that gliogenesis within the rostral migratory stream is delayed in the absence of Nfix, and reveal that Gdnf (glial-derived neurotrophic factor), a known attractant for SVZ-derived neuroblasts, is a target for transcriptional activation by NFIX. Collectively, these findings suggest that NFIX regulates both proliferation and migration during the development of the SVZ neurogenic niche.


Subject(s)
Cell Movement , Cell Proliferation , Lateral Ventricles/embryology , NFI Transcription Factors/physiology , Neural Stem Cells/physiology , Neurogenesis , Animals , Female , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Interneurons/physiology , Lateral Ventricles/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NFI Transcription Factors/genetics , NFI Transcription Factors/metabolism , Neuroglia/physiology , Olfactory Bulb/embryology , Olfactory Bulb/metabolism , Stem Cell Niche
11.
Neuron ; 83(5): 1085-97, 2014 Sep 03.
Article in English | MEDLINE | ID: mdl-25189209

ABSTRACT

The activity of adult stem cells is regulated by signals emanating from the surrounding tissue. Many niche signals have been identified, but it is unclear how they influence the choice of stem cells to remain quiescent or divide. Here we show that when stem cells of the adult hippocampus receive activating signals, they first induce the expression of the transcription factor Ascl1 and only subsequently exit quiescence. Moreover, lowering Ascl1 expression reduces the proliferation rate of hippocampal stem cells, and inactivating Ascl1 blocks quiescence exit completely, rendering them unresponsive to activating stimuli. Ascl1 promotes the proliferation of hippocampal stem cells by directly regulating the expression of cell-cycle regulatory genes. Ascl1 is similarly required for stem cell activation in the adult subventricular zone. Our results support a model whereby Ascl1 integrates inputs from both stimulatory and inhibitory signals and converts them into a transcriptional program activating adult neural stem cells.


Subject(s)
Adult Stem Cells/cytology , Gene Expression Regulation, Developmental/genetics , Hippocampus/cytology , Neurogenesis/genetics , Adult Stem Cells/metabolism , Age Factors , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Proliferation , Cerebral Ventricles/cytology , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 1/metabolism , Gene Expression Regulation, Developmental/drug effects , Glial Fibrillary Acidic Protein/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Kainic Acid/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Box Domain Proteins/metabolism , p300-CBP Transcription Factors/metabolism
12.
Cereb Cortex ; 24(11): 2822-34, 2014 Nov.
Article in English | MEDLINE | ID: mdl-23720416

ABSTRACT

We aimed to identify cis-regulatory elements that control gene expression in progenitors of the cerebral cortex. A list of 975 putative enhancers were retrieved from a ChIP-Seq experiment performed in NS5 mouse stem cells with antibodies to Sox2, Brn2/Pou3f2, or Brn1/Pou3f3. Through a selection pipeline including gene ontology and expression pattern, we reduced the number of candidate enhancer sequences to 20. Ex vivo electroporation of green fluorescent pProtein (GFP) reporter constructs in the telencephalon of mouse embryos showed that 35% of the 20 selected candidate sequences displayed enhancer activity in the developing cortex at E13.5. In silico transcription factor binding site (TFBS) searches and mutagenesis experiments showed that enhancer activity is related to the presence of Sox/Pou TFBS pairs in the sequence. Comparative genomic analyses showed that enhancer activity is not related to the evolutionary conservation of the sequence. Finally, the combination of in utero electroporation of GFP reporter constructs with immunostaining for Tbr2 (basal progenitor marker) and phospho-histoneH3 (mitotic activity marker) demonstrated that each enhancer is specifically active in precise subpopulations of progenitors in the cortical germinal zone, highlighting the heterogeneity of these progenitors in terms of cis-regulation.


Subject(s)
Cerebral Cortex/cytology , Cerebral Cortex/embryology , Gene Expression Regulation, Developmental/physiology , POU Domain Factors/metabolism , SOXB1 Transcription Factors/metabolism , Stem Cells/physiology , Animals , Binding Sites/genetics , Biological Evolution , Cadherins/genetics , Cadherins/metabolism , Cell Line , Embryo, Mammalian , Female , Histones/genetics , Histones/metabolism , In Vitro Techniques , Mice , Mice, Transgenic , Mutagenesis/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Organ Culture Techniques , POU Domain Factors/genetics , Pregnancy , SOXB1 Transcription Factors/genetics , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
13.
Cell ; 155(3): 621-35, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24243019

ABSTRACT

Direct lineage reprogramming is a promising approach for human disease modeling and regenerative medicine, with poorly understood mechanisms. Here, we reveal a hierarchical mechanism in the direct conversion of fibroblasts into induced neuronal (iN) cells mediated by the transcription factors Ascl1, Brn2, and Myt1l. Ascl1 acts as an "on-target" pioneer factor by immediately occupying most cognate genomic sites in fibroblasts. In contrast, Brn2 and Myt1l do not access fibroblast chromatin productively on their own; instead, Ascl1 recruits Brn2 to Ascl1 sites genome wide. A unique trivalent chromatin signature in the host cells predicts the permissiveness for Ascl1 pioneering activity among different cell types. Finally, we identified Zfp238 as a key Ascl1 target gene that can partially substitute for Ascl1 during iN cell reprogramming. Thus, a precise match between pioneer factors and the chromatin context at key target genes is determinative for transdifferentiation to neurons and likely other cell types.


Subject(s)
Cellular Reprogramming , Embryo, Mammalian/cytology , Fibroblasts/cytology , Gene Regulatory Networks , Neurons/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Chromatin/metabolism , Fibroblasts/metabolism , Genome-Wide Association Study , Humans , Mice , Nerve Tissue Proteins/metabolism , Neurons/metabolism , POU Domain Factors/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism
14.
Genes Dev ; 27(16): 1769-86, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23964093

ABSTRACT

The majority of neural stem cells (NSCs) in the adult brain are quiescent, and this fraction increases with aging. Although signaling pathways that promote NSC quiescence have been identified, the transcriptional mechanisms involved are mostly unknown, largely due to lack of a cell culture model. In this study, we first demonstrate that NSC cultures (NS cells) exposed to BMP4 acquire cellular and transcriptional characteristics of quiescent cells. We then use epigenomic profiling to identify enhancers associated with the quiescent NS cell state. Motif enrichment analysis of these enhancers predicts a major role for the nuclear factor one (NFI) family in the gene regulatory network controlling NS cell quiescence. Interestingly, we found that the family member NFIX is robustly induced when NS cells enter quiescence. Using genome-wide location analysis and overexpression and silencing experiments, we demonstrate that NFIX has a major role in the induction of quiescence in cultured NSCs. Transcript profiling of NS cells overexpressing or silenced for Nfix and the phenotypic analysis of the hippocampus of Nfix mutant mice suggest that NFIX controls the quiescent state by regulating the interactions of NSCs with their microenvironment.


Subject(s)
Epigenesis, Genetic , NFI Transcription Factors/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Animals , Bone Morphogenetic Protein 4/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Enhancer Elements, Genetic , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , HEK293 Cells , Humans , Mice , NFI Transcription Factors/genetics , Neural Stem Cells/drug effects , Protein Binding
15.
Cell Rep ; 4(3): 477-91, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23891001

ABSTRACT

FOXO transcription factors are central regulators of longevity from worms to humans. FOXO3, the FOXO isoform associated with exceptional human longevity, preserves adult neural stem cell pools. Here, we identify FOXO3 direct targets genome-wide in primary cultures of adult neural progenitor cells (NPCs). Interestingly, FOXO3-bound sites are enriched for motifs for bHLH transcription factors, and FOXO3 shares common targets with the proneuronal bHLH transcription factor ASCL1/MASH1 in NPCs. Analysis of the chromatin landscape reveals that FOXO3 and ASCL1 are particularly enriched at the enhancers of genes involved in neurogenic pathways. Intriguingly, FOXO3 inhibits ASCL1-dependent neurogenesis in NPCs and direct neuronal conversion in fibroblasts. FOXO3 also restrains neurogenesis in vivo. Our study identifies a genome-wide interaction between the prolongevity transcription factor FOXO3 and the cell-fate determinant ASCL1 and raises the possibility that FOXO3's ability to restrain ASCL1-dependent neurogenesis may help preserve the neural stem cell pool.


Subject(s)
Forkhead Transcription Factors/metabolism , Neural Stem Cells/physiology , Neurogenesis/physiology , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Binding Sites , Cell Differentiation/physiology , Cell Growth Processes/physiology , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Genome , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism
16.
Article in English | MEDLINE | ID: mdl-23028117

ABSTRACT

The mammalian nervous system is the most complex organ of any living organism. How this complexity is generated during neural development is just beginning to be elucidated. This article discusses the signaling, transcriptional, and epigenetic mechanisms that are involved in neural development. The first part focuses on molecules that control neuronal numbers through regulation of the timing of onset of neurogenesis, the timing of the neuronal-to-glial switch, and the rate of progenitor proliferation. The second part focuses on molecules that control neuronal diversity by generating spatially or temporally distinct populations of neuronal progenitors. Most of the studies discussed in this article are focused on the developing mammalian cerebral cortex, because this is one of the main model systems for neural developmental studies and many of the mechanisms identified in this tissue also operate elsewhere in the developing brain and spinal cord.


Subject(s)
Cerebral Cortex/growth & development , Epigenesis, Genetic , Mammals/growth & development , Neurogenesis/physiology , Signal Transduction , Animals , Cell Differentiation/genetics , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Gene Expression Regulation, Developmental , Mammals/genetics , Mammals/metabolism , Models, Biological , Neural Plate/embryology , Neural Plate/metabolism , Neural Plate/physiology , Neurogenesis/genetics
17.
Neuron ; 74(2): 209-11, 2012 Apr 26.
Article in English | MEDLINE | ID: mdl-22542173

ABSTRACT

A crucial event in the birth of a neuron is the detachment of its apical process from the neuroepithelium. In this issue of Neuron, Rousso et al. (2012) show that repression of N-cadherin by Foxp transcription factors disrupts apical adherens junctions and triggers neurogenesis.

18.
Genes Dev ; 25(9): 930-45, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21536733

ABSTRACT

Proneural genes such as Ascl1 are known to promote cell cycle exit and neuronal differentiation when expressed in neural progenitor cells. The mechanisms by which proneural genes activate neurogenesis--and, in particular, the genes that they regulate--however, are mostly unknown. We performed a genome-wide characterization of the transcriptional targets of Ascl1 in the embryonic brain and in neural stem cell cultures by location analysis and expression profiling of embryos overexpressing or mutant for Ascl1. The wide range of molecular and cellular functions represented among these targets suggests that Ascl1 directly controls the specification of neural progenitors as well as the later steps of neuronal differentiation and neurite outgrowth. Surprisingly, Ascl1 also regulates the expression of a large number of genes involved in cell cycle progression, including canonical cell cycle regulators and oncogenic transcription factors. Mutational analysis in the embryonic brain and manipulation of Ascl1 activity in neural stem cell cultures revealed that Ascl1 is indeed required for normal proliferation of neural progenitors. This study identified a novel and unexpected activity of the proneural gene Ascl1, and revealed a direct molecular link between the phase of expansion of neural progenitors and the subsequent phases of cell cycle exit and neuronal differentiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis , Telencephalon/cytology , Telencephalon/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Cell Line , Cell Proliferation , Cells, Cultured , Female , Gene Expression Profiling , Gene Knockdown Techniques , Genome-Wide Association Study , Mice , Pregnancy
19.
Neural Dev ; 6: 9, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21418559

ABSTRACT

BACKGROUND: The transcription factor Foxg1 is an important regulator of telencephalic cell cycles. Its inactivation causes premature lengthening of telencephalic progenitor cell cycles and increased neurogenic divisions, leading to severe hypoplasia of the telencephalon. These proliferation defects could be a secondary consequence of the loss of Foxg1 caused by the abnormal expression of several morphogens (Fibroblast growth factor 8, bone morphogenetic proteins) in the telencephalon of Foxg1 null mutants. Here we investigated whether Foxg1 has a cell autonomous role in the regulation of telencephalic progenitor proliferation. We analysed Foxg1+/+↔Foxg1-/- chimeras, in which mutant telencephalic cells have the potential to interact with, and to have any cell non-autonomous defects rescued by, normal wild-type cells. RESULTS: Our analysis showed that the Foxg1-/- cells are under-represented in the chimeric telencephalon and the proportion of them in S-phase is significantly smaller than that of their wild-type neighbours, indicating that their under-representation is caused by a cell autonomous reduction in their proliferation. We then analysed the expression of the cell-cycle regulator Pax6 and found that it is cell-autonomously downregulated in Foxg1-/- dorsal telencephalic cells. We went on to show that the introduction into Foxg1-/- embryos of a transgene designed to reverse Pax6 expression defects resulted in a partial rescue of the telencephalic progenitor proliferation defects. CONCLUSIONS: We conclude that Foxg1 exerts control over telencephalic progenitor proliferation by cell autonomous mechanisms that include the regulation of Pax6, which itself is known to regulate proliferation cell autonomously in a regional manner.


Subject(s)
Eye Proteins/biosynthesis , Eye Proteins/genetics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/physiology , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Neural Stem Cells/physiology , Paired Box Transcription Factors/biosynthesis , Paired Box Transcription Factors/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Telencephalon/cytology , Animals , Antimetabolites , Bromodeoxyuridine , Cell Count , Cell Proliferation , Chimera , Down-Regulation/genetics , Down-Regulation/physiology , Female , Gene Expression Regulation , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Knockout , PAX6 Transcription Factor , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction
20.
Cell Mol Life Sci ; 68(10): 1769-83, 2011 May.
Article in English | MEDLINE | ID: mdl-20981563

ABSTRACT

Neural stem (NS) cells are a self-renewing population of symmetrically dividing multipotent radial glia-like stem cells, characterized by homogeneous expansion in monolayer. Here we report that fetal NS cells isolated from different regions of the developing mouse nervous system behave in a similar manner with respect to self-renewal and neuropotency, but exhibit distinct positional identities. For example, NS cells from the neocortex maintain the expression of anterior transcription factors, including Otx2 and Foxg1, while Hoxb4 and Hoxb9 are uniquely found in spinal cord-derived NS cells. This molecular signature was stable for over 20 passages and was strictly linked to the developmental stage of the donor, because only NS cells derived from E14.5 cortex, and not those derived from E12.5 cortex, carried a consistent transcription factor profile. We also showed that traits of this positional code are maintained during neuronal differentiation, leading to the generation of electrophysiologically active neurons, even if they do not acquire a complete neurochemical identity.


Subject(s)
Fetus/cytology , Neural Stem Cells/metabolism , Animals , Cell Differentiation , Cells, Cultured , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Neocortex/cytology , Neocortex/embryology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Otx Transcription Factors/genetics , Otx Transcription Factors/metabolism , Patch-Clamp Techniques , Spinal Cord/cytology , Spinal Cord/embryology , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...