Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
3.
Cereb Cortex ; 27(6): 3378-3396, 2017 06 01.
Article in English | MEDLINE | ID: mdl-27600842

ABSTRACT

The T-box containing Tbr2 gene encodes for a transcription factor essential for the specification of the intermediate neural progenitors (INPs) originating the excitatory neurons of the cerebral cortex. However, its overall mechanism of action, direct target genes and cofactors remain unknown. Herein, we carried out global gene expression profiling combined with genome-wide binding site identification to determine the molecular pathways regulated by TBR2 in INPs. This analysis led to the identification of novel protein-protein interactions that control multiple features of INPs including cell-type identity, morphology, proliferation and migration dynamics. In particular, NEUROG2 and JMJD3 were found to associate with TBR2 revealing unexplored TBR2-dependent mechanisms. These interactions can explain, at least in part, the role of this transcription factor in the implementation of the molecular program controlling developmental milestones during corticogenesis. These data identify TBR2 as a major determinant of the INP-specific traits by regulating both genetic and epigenetic pathways.


Subject(s)
Cell Differentiation/genetics , Cerebral Cortex/cytology , Gene Expression Regulation, Developmental/genetics , Neural Stem Cells/physiology , Neurons/physiology , T-Box Domain Proteins/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle/genetics , Cell Movement/genetics , Cell Polarity/genetics , Embryo, Mammalian , Gene Expression Regulation, Developmental/physiology , Gene Regulatory Networks/genetics , Hippocampus/cytology , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , T-Box Domain Proteins/metabolism , Transcription Factors/metabolism
5.
Cell Stem Cell ; 17(1): 74-88, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26119235

ABSTRACT

Direct lineage reprogramming induces dramatic shifts in cellular identity, employing poorly understood mechanisms. Recently, we demonstrated that expression of Neurog2 or Ascl1 in postnatal mouse astrocytes generates glutamatergic or GABAergic neurons. Here, we take advantage of this model to study dynamics of neuronal cell fate acquisition at the transcriptional level. We found that Neurog2 and Ascl1 rapidly elicited distinct neurogenic programs with only a small subset of shared target genes. Within this subset, only NeuroD4 could by itself induce neuronal reprogramming in both mouse and human astrocytes, while co-expression with Insm1 was required for glutamatergic maturation. Cultured astrocytes gradually became refractory to reprogramming, in part by the repressor REST preventing Neurog2 from binding to the NeuroD4 promoter. Notably, in astrocytes refractory to Neurog2 activation, the underlying neurogenic program remained amenable to reprogramming by exogenous NeuroD4. Our findings support a model of temporal hierarchy for cell fate change during neuronal reprogramming.


Subject(s)
Astrocytes/cytology , Astrocytes/metabolism , Cellular Reprogramming/physiology , Repressor Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cells, Cultured , Cellular Reprogramming/genetics , DNA-Binding Proteins/metabolism , Humans , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Promoter Regions, Genetic , Repressor Proteins/deficiency , Repressor Proteins/genetics , Transcription Factors/metabolism , Transcription, Genetic
6.
Cell Rep ; 10(9): 1544-1556, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25753420

ABSTRACT

The proneural transcription factor Ascl1 coordinates gene expression in both proliferating and differentiating progenitors along the neuronal lineage. Here, we used a cellular model of neurogenesis to investigate how Ascl1 interacts with the chromatin landscape to regulate gene expression when promoting neuronal differentiation. We find that Ascl1 binding occurs mostly at distal enhancers and is associated with activation of gene transcription. Surprisingly, the accessibility of Ascl1 to its binding sites in neural stem/progenitor cells remains largely unchanged throughout their differentiation, as Ascl1 targets regions of both readily accessible and closed chromatin in proliferating cells. Moreover, binding of Ascl1 often precedes an increase in chromatin accessibility and the appearance of new regions of open chromatin, associated with de novo gene expression during differentiation. Our results reveal a function of Ascl1 in promoting chromatin accessibility during neurogenesis, linking the chromatin landscape at Ascl1 target regions with the temporal progression of its transcriptional program.

7.
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
8.
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
9.
Nucleic Acids Res ; 41(11): 5555-68, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23595148

ABSTRACT

Accurately characterizing transcription factor (TF)-DNA affinity is a central goal of regulatory genomics. Although thermodynamics provides the most natural language for describing the continuous range of TF-DNA affinity, traditional motif discovery algorithms focus instead on classification paradigms that aim to discriminate 'bound' and 'unbound' sequences. Moreover, these algorithms do not directly model the distribution of tags in ChIP-seq data. Here, we present a new algorithm named Thermodynamic Modeling of ChIP-seq (TherMos), which directly estimates a position-specific binding energy matrix (PSEM) from ChIP-seq/exo tag profiles. In cross-validation tests on seven genome-wide TF-DNA binding profiles, one of which we generated via ChIP-seq on a complex developing tissue, TherMos predicted quantitative TF-DNA binding with greater accuracy than five well-known algorithms. We experimentally validated TherMos binding energy models for Klf4 and Esrrb, using a novel protocol to measure PSEMs in vitro. Strikingly, our measurements revealed strong non-additivity at multiple positions within the two PSEMs. Among the algorithms tested, only TherMos was able to model the entire binding energy landscape of Klf4 and Esrrb. Our study reveals new insights into the energetics of TF-DNA binding in vivo and provides an accurate first-principles approach to binding energy inference from ChIP-seq and ChIP-exo data.


Subject(s)
Algorithms , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Animals , Chromatin Immunoprecipitation , High-Throughput Nucleotide Sequencing , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Mice , Mutation , Protein Binding , Receptors, Estrogen/metabolism , Sequence Analysis, DNA , Thermodynamics
10.
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
11.
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
12.
Nat Neurosci ; 12(10): 1229-37, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19749747

ABSTRACT

An important feature of the cerebral cortex is its layered organization, which is modulated in an area-specific manner. We found that the transcription factor AP2gamma regulates laminar fate in a region-specific manner. Deletion of AP2gamma (also known as Tcfap2c) during development resulted in a specific reduction of upper layer neurons in the occipital cortex, leading to impaired function and enhanced plasticity of the adult visual cortex. AP2gamma functions in apical progenitors, and its absence resulted in mis-specification of basal progenitors in the occipital cortex at the time at which upper layer neurons were generated. AP2gamma directly regulated the basal progenitor fate determinants Math3 (also known as Neurod4) and Tbr2, and its overexpression promoted the generation of layer II/III neurons in a time- and region-specific manner. Thus, AP2gamma acts as a regulator of basal progenitor fate, linking regional and laminar specification in the mouse developing cerebral cortex.


Subject(s)
Cell Differentiation/physiology , Cerebral Cortex , Embryonic Stem Cells/physiology , Neurogenesis/physiology , Transcription Factor AP-2/physiology , Adult , Animals , Bromodeoxyuridine/metabolism , Cell Count/methods , Cell Line, Transformed , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Embryo, Mammalian , Evoked Potentials, Visual/genetics , Evoked Potentials, Visual/physiology , Eye Proteins/genetics , Eye Proteins/metabolism , Fetus , Gene Deletion , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Green Fluorescent Proteins/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immediate-Early Proteins/genetics , Ki-67 Antigen/metabolism , Macaca fascicularis , Mice , Mice, Inbred C57BL , Mice, Transgenic , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Photic Stimulation/methods , RNA, Messenger/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , T-Box Domain Proteins/metabolism , Transcription Factor AP-2/genetics , Transcription Factors/genetics , Transfection/methods , Tumor Suppressor Proteins/genetics
13.
Mol Cell Neurosci ; 38(1): 15-42, 2008 May.
Article in English | MEDLINE | ID: mdl-18372191

ABSTRACT

Since the discovery of radial glia as the source of neurons, their heterogeneity in regard to neurogenesis has been described by clonal and time-lapse analysis in vitro. However, the molecular determinants specifying neurogenic radial glia differently from radial glia that mostly self-renew remain ill-defined. Here, we isolated two radial glial subsets that co-exist at mid-neurogenesis in the developing cerebral cortex and their immediate progeny. While one subset generates neurons directly, the other is largely non-neurogenic but also gives rise to Tbr2-positive basal precursors, thereby contributing indirectly to neurogenesis. Isolation of these distinct radial glia subtypes allowed determining interesting differences in their transcriptome. These transcriptomes were also strikingly different from the transcriptome of radial glia isolated at the end of neurogenesis. This analysis therefore identifies, for the first time, the lineage origin of basal progenitors and the molecular differences of this lineage in comparison to directly neurogenic and gliogenic radial glia.


Subject(s)
Cell Lineage/genetics , Gene Expression Profiling/methods , Neuroglia/classification , Neuroglia/physiology , Animals , Cell Separation , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroglia/cytology , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...