Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Front Cell Dev Biol ; 10: 1011109, 2022.
Article in English | MEDLINE | ID: mdl-36263009

ABSTRACT

Early forebrain patterning entails the correct regional designation of the neuroepithelium, and appropriate specification, generation, and distribution of neural cells during brain development. Specific signaling and transcription factors are known to tightly regulate patterning of the dorsal telencephalon to afford proper structural/functional cortical arealization and morphogenesis. Nevertheless, whether and how changes of the chromatin structure link to the transcriptional program(s) that control cortical patterning remains elusive. Here, we report that the BAF chromatin remodeling complex regulates the spatiotemporal patterning of the mouse dorsal telencephalon. To determine whether and how the BAF complex regulates cortical patterning, we conditionally deleted the BAF complex scaffolding subunits BAF155 and BAF170 in the mouse dorsal telencephalic neuroepithelium. Morphological and cellular changes in the BAF mutant forebrain were examined using immunohistochemistry and in situ hybridization. RNA sequencing, Co-immunoprecipitation, and mass spectrometry were used to investigate the molecular basis of BAF complex involvement in forebrain patterning. We found that conditional ablation of BAF complex in the dorsal telencephalon neuroepithelium caused expansion of the cortical hem and medial cortex beyond their developmental boundaries. Consequently, the hippocampal primordium is not specified, the mediolateral cortical patterning is compromised, and the cortical identity is disturbed in the absence of BAF complex. The BAF complex was found to interact with the cortical hem suppressor LHX2. The BAF complex suppresses cortical hem fate to permit proper forebrain patterning. We provide evidence that BAF complex modulates mediolateral cortical patterning possibly by interacting with the transcription factor LHX2 to drive the LHX2-dependent transcriptional program essential for dorsal telencephalon patterning. Our data suggest a putative mechanistic synergy between BAF chromatin remodeling complex and LHX2 in regulating forebrain patterning and ontogeny.

2.
Sci Adv ; 7(38): eabc6792, 2021 Sep 17.
Article in English | MEDLINE | ID: mdl-34524839

ABSTRACT

Increase in the size of human neocortex­acquired in evolution­accounts for the unique cognitive capacity of humans. This expansion reflects the evolutionarily enhanced proliferative ability of basal progenitors (BPs), including the basal radial glia and basal intermediate progenitors (bIPs) in mammalian cortex, which may have been acquired through epigenetic alterations in BPs. However, how the epigenome in BPs differs across species is not known. Here, we report that histone H3 acetylation is a key epigenetic regulation in bIP amplification and cortical expansion. Through epigenetic profiling of sorted bIPs, we show that histone H3 lysine 9 acetylation (H3K9ac) is low in murine bIPs and high in human bIPs. Elevated H3K9ac preferentially increases bIP proliferation, increasing the size and folding of the normally smooth mouse neocortex. H3K9ac drives bIP amplification by increasing expression of the evolutionarily regulated gene, Trnp1, in developing cortex. Our findings demonstrate a previously unknown mechanism that controls cortical architecture.

3.
PLoS Genet ; 17(3): e1009446, 2021 03.
Article in English | MEDLINE | ID: mdl-33750945

ABSTRACT

The BAF complex plays an important role in the development of a wide range of tissues by modulating gene expression programs at the chromatin level. However, its role in neural crest development has remained unclear. To determine the role of the BAF complex, we deleted BAF155/BAF170, the core subunits required for the assembly, stability, and functions of the BAF complex in neural crest cells (NCCs). Neural crest-specific deletion of BAF155/BAF170 leads to embryonic lethality due to a wide range of developmental defects including craniofacial, pharyngeal arch artery, and OFT defects. RNAseq and transcription factor enrichment analysis revealed that the BAF complex modulates the expression of multiple signaling pathway genes including Hippo and Notch, essential for the migration, proliferation, and differentiation of the NCCs. Furthermore, we demonstrated that the BAF complex is essential for the Brg1-Yap-Tead-dependent transcription of target genes in NCCs. Together, our results demonstrate an important role of the BAF complex in modulating the gene regulatory network essential for neural crest development.


Subject(s)
Chromatin Assembly and Disassembly , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Neural Crest/embryology , Neural Crest/metabolism , Neurogenesis/genetics , Animals , Cell Differentiation/genetics , Cell Proliferation , DNA-Binding Proteins/metabolism , Embryonic Development/genetics , Gene Deletion , Gene Regulatory Networks , Genes, Reporter , Mice , Mice, Transgenic , Organ Specificity , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
4.
Front Cell Dev Biol ; 8: 584314, 2020.
Article in English | MEDLINE | ID: mdl-33344448

ABSTRACT

The main stem cell niche for neurogenesis in the adult mammalian brain is the subventricular zone (SVZ) that extends along the cerebral lateral ventricles. We aimed at characterizing the initial molecular responses of the macaque monkey SVZ to transient, global cerebral ischemia. We microdissected tissue lining the anterior horn of the lateral ventricle (SVZa) from 7 day post-ischemic and sham-operated monkeys. Transcriptomics shows that in ischemic SVZa, 541 genes were upregulated and 488 genes were down-regulated. The transcription data encompassing the upregulated genes revealed a profile typical for quiescent stem cells and astrocytes. In the primate brain the SVZ is morphologically subdivided in distinct and separate ependymal and subependymal regions. The subependymal contains predominantly neural stem cells (NSC) and differentiated progenitors. To determine in which SVZa region ischemia had evoked transcriptional upregulation, sections through control and ischemic SVZa were analyzed by high-throughput in situ hybridization for a total of 150 upregulated genes shown in the www.monkey-niche.org image database. The majority of the differentially expressed genes mapped to the subependymal layers on the striatal or callosal aspect of the SVZa. Moreover, a substantial number of upregulated genes was expressed in the ependymal layer, implicating a contribution of the ependyma to stem cell biology. The transcriptome analysis yielded several novel gene markers for primate SVZa including the apelin receptor that is strongly expressed in the primate SVZa niche upon ischemic insult.

5.
Mol Neurobiol ; 56(1): 567-582, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29748916

ABSTRACT

The transcription factor (TF) Zbtb20 is important for the hippocampal specification and the regulation of neurogenesis of neocortical projection neurons. Herein, we show a critical involvement of the TF Zbtb20 in the neurogenesis of both projection neurons and interneurons of the olfactory bulb during embryonic stages. Our data indicate that the lack of Zbtb20 significantly diminishes the generation of a set of early-born Tbr2+ neurons during embryogenesis. Furthermore, we provide evidence that Zbtb20 regulates the transition between neurogenesis to gliogenesis in cortical radial glial progenitor cells at the perinatal (E18.5) stage. In the adult mammalian brain, Zbtb20 is expressed by GFAP+ neural progenitor cells (NPCs) located in the forebrain neurogenic niche, i.e., the subventricular zone (SVZ) of the lateral ventricles. Upon induction of cerebral ischemia, we found that Zbtb20 expression is upregulated in astrocytic-like cells, whereas diminishing the expression levels of Zbtb20 significantly reduces the ischemia-induced astrocytic reaction as observed in heterozygous Zbtb20 loss-of-function mice. Altogether, these results highlight the important role of the TF Zbtb20 as a temporal regulator of neurogenesis or gliogenesis, depending on the developmental context.


Subject(s)
Aging/metabolism , Brain Injuries/metabolism , Neurogenesis , Neuroglia/metabolism , Olfactory Bulb/metabolism , Transcription Factors/metabolism , Animals , Animals, Newborn , Astrocytes/metabolism , Brain Injuries/pathology , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Olfactory Bulb/pathology , Phenotype , Stem Cell Niche , Stem Cells/metabolism , Stroke/metabolism , Stroke/pathology
6.
iScience ; 4: 109-126, 2018 Jun 29.
Article in English | MEDLINE | ID: mdl-30240734

ABSTRACT

The abundance of basal progenitors (BPs), basal radial glia progenitors (bRGs) and basal intermediate progenitors (bIPs), in primate brain has been correlated to the high degree of cortical folding. Here we examined the role of BAF155, a subunit of the chromatin remodeling BAF complex, in generation of cortical progenitor heterogeneity. The conditional deletion of BAF155 led to diminished bIP pool and increased number of bRGs, due to delamination of apical RGs. We found that BAF155 is required for normal activity of neurogenic transcription factor PAX6, thus controlling the expression of genes that are involved in bIP specification, cell-cell interaction, and establishment of adherens junction. In a PAX6-dependent manner, BAF155 regulates the expression of the CDC42 effector protein CEP4, thereby controlling progenitor delamination. Furthermore, BAF155-dependent chromatin remodeling seems to exert a specific role in the genesis of BPs through the regulation of human RG-specific genes (such as Foxn4) that possibly acquired evolutionary significance.

7.
Stem Cell Reports ; 10(6): 1734-1750, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29779894

ABSTRACT

During early cortical development, neural stem cells (NSCs) divide symmetrically to expand the progenitor pool, whereas, in later stages, NSCs divide asymmetrically to self-renew and produce other cell types. The timely switch from such proliferative to differentiative division critically determines progenitor and neuron numbers. However, the mechanisms that limit proliferative division in late cortical development are not fully understood. Here, we show that the BAF (mSWI/SNF) complexes restrict proliferative competence and promote neuronal differentiation in late corticogenesis. Inactivation of BAF complexes leads to H3K27me3-linked silencing of neuronal differentiation-related genes, with concurrent H3K4me2-mediated activation of proliferation-associated genes via de-repression of Wnt signaling. Notably, the deletion of BAF complexes increased proliferation of neuroepithelial cell-like NSCs, impaired neuronal differentiation, and exerted a Wnt-dependent effect on neocortical and hippocampal development. Thus, these results demonstrate that BAF complexes act as both activators and repressors to control global epigenetic and gene expression programs in late corticogenesis.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Embryonic Development/genetics , Epigenesis, Genetic , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Ribonucleoproteins/metabolism , Wnt Signaling Pathway , Animals , Cell Differentiation , Cell Proliferation , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/genetics , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Hippocampus/embryology , Hippocampus/metabolism , Mice , Neurogenesis , Neurons/cytology , Neurons/metabolism , Protein Binding , Ribonucleoproteins/genetics
8.
Oncol Rep ; 37(3): 1579-1592, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28184912

ABSTRACT

A misbalance between proliferation and differentiation of neural stem cells in niches for adult brain neurogenesis is a key mechanism in glioma pathogenesis. In the adult brain, the expression of Pax6 marks stem cells in the forebrain neurogenic niche. We analyzed the expression profile of the two active in vertebrates Pax6 isoforms, Pax6 and Pax6-5a, along with the expression of microRNA cluster miR-183-96-182 in a large set of glioma patient specimens and glioma cell lines which showed opposite expression level, low and high, respectively, with the progression of tumor malignancy. Our results from biochemical and in vitro studies in glioma cell lines disclosed a specific regulation of the PAX6-5a isoform by miR-183. Mechanistically, we show that the downregulation of the lipid kinase SPHK1 by both PAX6 isoforms and the simultaneous induction of CTNDD2 expression, specifically by PAX6-5a, results in reduced glioma cell survival, decreased migration and invasion and increased cell death, in glioma cell lines. Taken together, our findings point towards the important role of PAX6 and define PAX6-5a as a new essential player in glioma development. Finally, we propose that the expression level of TFs PAX6/PAX6-5a and miR-183-96-182 may potentially serve as prognostic markers for the progression of glioma tumors from low- to high-grade with a potential to identify new therapeutic approaches.


Subject(s)
Brain Neoplasms/pathology , Glioma/pathology , MicroRNAs/metabolism , PAX6 Transcription Factor/metabolism , Adult , Algorithms , Apoptosis , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Proliferation , Disease Progression , Female , Fluorescent Antibody Technique , Glioma/genetics , Glioma/metabolism , Humans , Male , MicroRNAs/genetics , PAX6 Transcription Factor/genetics , Protein Isoforms , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
9.
Mol Neurobiol ; 54(6): 4618-4635, 2017 08.
Article in English | MEDLINE | ID: mdl-27392482

ABSTRACT

The BAF chromatin remodeling complex plays an essential role in brain development. However its function in postnatal neurogenesis in hippocampus is still unknown. Here, we show that in postnatal dentate gyrus (DG), the BAF170 subunit of the complex is expressed in radial glial-like (RGL) progenitors and in cell types involved in subsequent steps of adult neurogenesis including mature astrocytes. Conditional deletion of BAF170 during cortical late neurogenesis as well as during adult brain neurogenesis depletes the pool of RGL cells in DG, and promotes terminal astrocyte differentiation. These derangements are accompanied by distinct behavioral deficits, as reflected by an impaired accuracy of place responding in the Morris water maze test, during both hidden platform as well as reversal learning. Inducible deletion of BAF170 in DG during adult brain neurogenesis resulted in mild spatial learning deficits, having a more pronounced effect on spatial learning during the reversal test. These findings demonstrate involvement of BAF170-dependent chromatin remodeling in hippocampal neurogenesis and cognition and suggest a specific role of adult neurogenesis in DG in adaptive behavior.


Subject(s)
Cell Differentiation , Chromosomal Proteins, Non-Histone/deficiency , Dentate Gyrus/cytology , Dentate Gyrus/growth & development , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Spatial Learning , Aging/metabolism , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins , Integrases/metabolism , Maze Learning/drug effects , Mice, Inbred C57BL , Mice, Knockout , Nestin/metabolism , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Neuroglia/drug effects , Neuroglia/metabolism , Spatial Learning/drug effects , Tamoxifen/pharmacology , Transcription Factors
10.
PLoS Genet ; 12(9): e1006274, 2016 09.
Article in English | MEDLINE | ID: mdl-27611684

ABSTRACT

Neurogenesis is a key developmental event through which neurons are generated from neural stem/progenitor cells. Chromatin remodeling BAF (mSWI/SNF) complexes have been reported to play essential roles in the neurogenesis of the central nervous system. However, whether BAF complexes are required for neuron generation in the olfactory system is unknown. Here, we identified onscBAF and ornBAF complexes, which are specifically present in olfactory neural stem cells (oNSCs) and olfactory receptor neurons (ORNs), respectively. We demonstrated that BAF155 subunit is highly expressed in both oNSCs and ORNs, whereas high expression of BAF170 subunit is observed only in ORNs. We report that conditional deletion of BAF155, a core subunit in both onscBAF and ornBAF complexes, causes impaired proliferation of oNSCs as well as defective maturation and axonogenesis of ORNs in the developing olfactory epithelium (OE), while the high expression of BAF170 is important for maturation of ORNs. Interestingly, in the absence of BAF complexes in BAF155/BAF170 double-conditional knockout mice (dcKO), OE is not specified. Mechanistically, BAF complex is required for normal activation of Pax6-dependent transcriptional activity in stem cells/progenitors of the OE. Our findings unveil a novel mechanism mediated by the mSWI/SNF complex in OE neurogenesis and development.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Neurogenesis , Olfactory Mucosa/metabolism , Transcription Factors/genetics , Animals , Cells, Cultured , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins , Gene Deletion , Mice , Mice, Inbred C57BL , Olfactory Mucosa/cytology , Olfactory Mucosa/embryology , Olfactory Receptor Neurons/cytology , Olfactory Receptor Neurons/metabolism , Transcription Factors/metabolism
11.
Mol Brain ; 9(1): 65, 2016 06 09.
Article in English | MEDLINE | ID: mdl-27282384

ABSTRACT

BACKGROUND: During corticogenesis, genetic programs encoded in progenitor cells at different developmental stages and inherited in postmitotic neurons specify distinct layer and area identities. Transcription factor Zbtb20 has been shown to play a role for hippocampal development but whether it is implicated in mammalian neocortical morphogenesis remains unknown. RESULTS: Here, we report that during embyogenesis transcription factor Zbtb20 has a dynamic spatio-temporal expression pattern in mitotic cortical progenitors through which it modulates the sequential generation of cortical neuronal layer identities. Zbtb20 knock out mice exhibited enhanced populations of early born L6-L4 neuronal subtypes and a dramatic reduction of the late born L3/L2 neurons. This defect was due to a temporal misbalance in the production of earlier versus later born neurons, leading to a progressive diminishing of the progenitor pool for the generation of L3-L2 neurons. Zbtb20 implements these temporal effects in part by binding to promoter of the orphan nuclear receptor CoupTF1/Nr2f1. In addition to its effects exerted in cortical progenitors, the postmitotic expression of Zbtb20 in L3/L2 neurons starting at birth may contribute to their proper differentiation and migration. CONCLUSIONS: Our findings reveal Zbtb20 as a novel temporal regulator for the generation of layer-specific neuronal identities.


Subject(s)
Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Neurons/metabolism , Transcription Factors/metabolism , Animals , Cell Cycle , Cell Lineage , Cell Movement , Mice, Transgenic , Models, Biological , Neurogenesis , Neurons/cytology , Stem Cells/metabolism , Transcription Factors/deficiency
12.
Cell Cycle ; 15(10): 1317-24, 2016 05 18.
Article in English | MEDLINE | ID: mdl-26986003

ABSTRACT

The multi-subunit chromatin-remodeling SWI/SNF (known as BAF for Brg/Brm-associated factor) complexes play essential roles in development. Studies have shown that the loss of individual BAF subunits often affects local chromatin structure and specific transcriptional programs. However, we do not fully understand how BAF complexes function in development because no animal mutant had been engineered to lack entire multi-subunit BAF complexes. Importantly, we recently reported that double conditional knock-out (dcKO) of the BAF155 and BAF170 core subunits in mice abolished the presence of the other BAF subunits in the developing cortex. The generated dcKO mutant provides a novel and powerful tool for investigating how entire BAF complexes affect cortical development. Using this model, we found that BAF complexes globally control the key heterochromatin marks, H3K27me2 and -3, by directly modulating the enzymatic activity of the H3K27 demethylases, Utx and Jmjd3. Here, we present further insights into how the scaffolding ability of the BAF155 and BAF170 core subunits maintains the stability of BAF complexes in the forebrain and throughout the embryo during development. Furthermore, we show that the loss of BAF complexes in the above-described model up-regulates H3K27me3 and impairs forebrain development and embryogenesis. These findings improve our understanding of epigenetic mechanisms and their modulation by the chromatin-remodeling SWI/SNF complexes that control embryonic development.


Subject(s)
Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Epigenesis, Genetic , Animals , Brain/growth & development , Brain/metabolism , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/deficiency , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins , Embryonic Development , Histone Demethylases/metabolism , Histones/metabolism , Immunohistochemistry , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Transcription Factors/deficiency , Transcription Factors/genetics
13.
Cell Rep ; 13(9): 1842-54, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26655900

ABSTRACT

BAF (Brg/Brm-associated factors) complexes play important roles in development and are linked to chromatin plasticity at selected genomic loci. Nevertheless, a full understanding of their role in development and chromatin remodeling has been hindered by the absence of mutants completely lacking BAF complexes. Here, we report that the loss of BAF155/BAF170 in double-conditional knockout (dcKO) mice eliminates all known BAF subunits, resulting in an overall reduction in active chromatin marks (H3K9Ac), a global increase in repressive marks (H3K27me2/3), and downregulation of gene expression. We demonstrate that BAF complexes interact with H3K27 demethylases (JMJD3 and UTX) and potentiate their activity. Importantly, BAF complexes are indispensable for forebrain development, including proliferation, differentiation, and cell survival of neural progenitor cells. Our findings reveal a molecular mechanism mediated by BAF complexes that controls the global transcriptional program and chromatin state in development.


Subject(s)
Chromatin/metabolism , Chromosomal Proteins, Non-Histone/genetics , Transcription Factors/genetics , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cerebellar Cortex/metabolism , Chromosomal Proteins, Non-Histone/deficiency , Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins , Down-Regulation , Embryo, Mammalian/metabolism , Histone Demethylases/genetics , Histone Demethylases/metabolism , Histones/genetics , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Nuclear Proteins/metabolism , Transcription Factors/deficiency , Transcription Factors/metabolism
14.
Cell Mol Life Sci ; 72(9): 1795-809, 2015 May.
Article in English | MEDLINE | ID: mdl-25352170

ABSTRACT

Cortical glutamatergic neurons are generated by radial glial cells (RGCs), specified by the expression of transcription factor (TF) Pax6, in the germinative zones of the dorsal telencephalon. Here, we demonstrate that Pax6 regulates the structural assembly of the interphase centrosomes. In the cortex of the Pax6-deficient Small eye (Sey/Sey) mutant, we find a defect of the appendages of the mother centrioles, indicating incomplete centrosome maturation. Consequently, RGCs fail to generate primary cilia, and instead of staying in the germinative zone for renewal, RGCs detach from the ventricular surface thus affecting the interkinetic nuclear migration and they exit prematurely from mitosis. Mechanistically, we show that TF Pax6 directly regulates the activity of the Odf2 gene encoding for the appendage-specific protein Odf2 with a role for the assembly of mother centriole. Our findings demonstrate a molecular mechanism that explains important characteristics of the centrosome disassembly and malfunctioning in developing cortex lacking Pax6.


Subject(s)
Centrioles/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Eye Proteins/metabolism , Heat-Shock Proteins/metabolism , Homeodomain Proteins/metabolism , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , Animals , Base Sequence , Centrioles/ultrastructure , Eye Proteins/analysis , Eye Proteins/genetics , Female , Gene Expression Regulation, Developmental , HEK293 Cells , Heat-Shock Proteins/analysis , Heat-Shock Proteins/genetics , Homeodomain Proteins/analysis , Homeodomain Proteins/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , NIH 3T3 Cells , PAX6 Transcription Factor , Paired Box Transcription Factors/analysis , Paired Box Transcription Factors/genetics , Promoter Regions, Genetic , Repressor Proteins/analysis , Repressor Proteins/genetics
15.
Cell Mol Life Sci ; 71(17): 3199-218, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24614969

ABSTRACT

The mammalian neocortex is a sheet of cells covering the cerebrum that provides the structural basis for the perception of sensory inputs, motor output responses, cognitive function, and mental capacity of primates. Recent discoveries promote the concept that increased cortical surface size and thickness in phylogenetically advanced species is a result of an increased generation of neurons, a process that underlies higher cognitive and intellectual performance in higher primates and humans. Here, we review some of the advances in the field, focusing on the diversity of neocortical progenitors in different species and the cellular mechanisms of neurogenesis. We discuss recent views on intrinsic and extrinsic molecular determinants, including the role of epigenetic chromatin modifiers and microRNA, in the control of neuronal output in developing cortex and in the establishment of normal cortical architecture.


Subject(s)
Neocortex/growth & development , Animals , Cell Division , Cell Polarity , Chromatin Assembly and Disassembly , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Humans , Mammals/growth & development , MicroRNAs/physiology , Neocortex/anatomy & histology , Nerve Tissue Proteins/physiology , Neural Stem Cells/cytology , Neurogenesis , Neurons/cytology , Organ Size , RNA Processing, Post-Transcriptional , Signal Transduction , Species Specificity
16.
Cereb Cortex ; 24(3): 754-72, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23180754

ABSTRACT

Scratch genes (Scrt) are neural-specific zinc-finger transcription factors (TFs) with an unknown function in the developing brain. Here, we show that, in addition to the reported expression of mammalian Scrt2 in postmitotic differentiating and mature neurons in the developing and early postnatal brain, Scrt2 is also localized in subsets of mitotic and neurogenic radial glial (RGP) and intermediate (IP) progenitors, as well as in their descendants-postmitotic IPs and differentiating neurons at the border subventricular/intermediate zone. Conditional activation of transgenic Scrt2 in cortical progenitors in mice promotes neuronal differentiation by favoring the direct mode of neurogenesis of RGPs at the onset of neurogenesis, at the expense of IP generation. Neuronal amplification via indirect IP neurogenesis is thereby extenuated, leading to a mild postnatal reduction of cortical thickness. Forced in vivo overexpression of Scrt2 suppressed the generation of IPs from RGPs and caused a delay in the radial migration of upper layer neurons toward the cortical plate. Mechanistically, our results indicate that Scrt2 negatively regulates the transcriptional activation of the basic helix loop helix TFs Ngn2/NeuroD1 on E-box containing common target genes, including Rnd2, a well-known major effector for migrational defects in developing cortex. Altogether, these findings reveal a modulatory role of Scrt2 protein in cortical neurogenesis and neuronal migration.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Movement/genetics , Neocortex/physiology , Neurogenesis/genetics , Neurons/physiology , Transcription Factors/genetics , Animals , Animals, Newborn , Cell Line, Transformed , Cells, Cultured , Embryo, Mammalian , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Mice, Transgenic , Neocortex/cytology , Nerve Tissue Proteins/metabolism , Transcription Factors/metabolism , Xenopus , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
17.
Cell Cycle ; 12(18): 2953-9, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23974113

ABSTRACT

The multi-subunit chromatin remodeling BAF complex controls different developmental processes. Using cortex-specific conditional knockout and overexpression mouse models, we have recently reported that BAF170, a subunit of the vertebrate BAF chromatin remodeling complex, interacts with transcription factor (TF) Pax6 to control cortical size and volume. The mechanistic basis includes suppression of the expression of Pax6 target genes, which are required for genesis of cortical intermediate progenitors (IPs) and specification of late neuronal subtype identity. In addition, we showed that a dynamic competition between BAF170 and BAF155 subunits within the BAF complex during progression of neurogenesis is a primary event in modulating the size of the mammalian cortex. Here, we present additional insights into the interaction between the BAF complex and TF Pax6 in the genesis of IPs of the developing cortex. Furthermore, we show that such competition between BAF170 and BAF155 is involved as well in the determination of the size of the embryonic body. Our results add new insights into a cell-intrinsic mechanism, mediated by the chromatin remodeling BAF complex that controls vertebrate body shape and size.


Subject(s)
Chromatin Assembly and Disassembly/physiology , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Transcription Factors/metabolism , Animals , Body Size , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins , Embryo, Mammalian/metabolism , Embryonic Development , Eye Proteins/genetics , Eye Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , PAX6 Transcription Factor , Paired Box Transcription Factors/deficiency , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Repressor Proteins/deficiency , Repressor Proteins/genetics , Repressor Proteins/metabolism
18.
Nat Neurosci ; 16(8): 1060-7, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23831966

ABSTRACT

The primary somatosensory cortex (S1) contains a complete body map that mirrors the subcortical maps developed by peripheral sensory input projecting to the sensory hindbrain, the thalamus and then S1. Peripheral changes during development alter these maps through 'bottom-up' plasticity. Unknown is how S1 size influences map organization and whether an altered S1 map feeds back to affect subcortical maps. We show that the size of S1 in mice is significantly reduced by cortex-specific deletion of Pax6, resulting in a reduced body map and loss of body representations by an exclusion of later-differentiating sensory thalamocortical input. An initially normal sensory thalamus was repatterned to match the aberrant S1 map by apoptotic deletion of thalamic neurons representing body parts with axons excluded from S1. Deleted representations were rescued by altering competition between thalamocortical axons using sensory deprivation or increasing the size of S1. Thus, S1 size determined the resolution and completeness of body maps and engaged 'top-down' plasticity that repatterned the sensory thalamus to match S1.


Subject(s)
Neuronal Plasticity/physiology , Posterior Thalamic Nuclei/physiology , Somatosensory Cortex/physiology , Animals , Apoptosis , Axons/physiology , Body Image , Eye Proteins/genetics , Eye Proteins/physiology , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neural Pathways/physiology , Neurons/physiology , Organ Specificity , PAX6 Transcription Factor , Paired Box Transcription Factors/deficiency , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/physiology , Posterior Thalamic Nuclei/growth & development , Recombinant Fusion Proteins/physiology , Repressor Proteins/deficiency , Repressor Proteins/genetics , Repressor Proteins/physiology , Rhombencephalon/physiology , Sensation/physiology , Somatosensory Cortex/pathology , Vibrissae/innervation
19.
Dev Cell ; 25(3): 256-69, 2013 May 13.
Article in English | MEDLINE | ID: mdl-23643363

ABSTRACT

Increased cortical size is essential to the enhanced intellectual capacity of primates during mammalian evolution. The mechanisms that control cortical size are largely unknown. Here, we show that mammalian BAF170, a subunit of the chromatin remodeling complex mSWI/SNF, is an intrinsic factor that controls cortical size. We find that conditional deletion of BAF170 promotes indirect neurogenesis by increasing the pool of intermediate progenitors (IPs) and results in an enlarged cortex, whereas cortex-specific BAF170 overexpression results in the opposite phenotype. Mechanistically, BAF170 competes with BAF155 subunit in the BAF complex, affecting euchromatin structure and thereby modulating the binding efficiency of the Pax6/REST-corepressor complex to Pax6 target genes that regulate the generation of IPs and late cortical progenitors. Our findings reveal a molecular mechanism mediated by the mSWI/SNF chromatin-remodeling complex that controls cortical architecture.


Subject(s)
Cerebral Cortex/metabolism , Chromatin/metabolism , Neurogenesis , Transcription Factors/metabolism , Animals , Cerebral Cortex/pathology , Chromatin/genetics , Chromatin Assembly and Disassembly , DNA Methylation , Epigenesis, Genetic , Eye Proteins/genetics , Eye Proteins/metabolism , Female , HeLa Cells , Histones/genetics , Histones/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Mice , Mice, Transgenic/genetics , Mice, Transgenic/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Neurons/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Organ Size , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Interaction Mapping , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics
20.
Development ; 140(5): 1123-36, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23404109

ABSTRACT

To achieve adequate organ development and size, cell proliferation and differentiation have to be tightly regulated and coordinated. The transcription factor Pax6 regulates patterning, neurogenesis and proliferation in forebrain development. The molecular basis of this regulation is not well understood. As the bipartite DNA-binding paired domain of Pax6 regulates forebrain development, we examined mice with point mutations in its individual DNA-binding subdomains PAI (Pax6(Leca4), N50K) and RED (Pax6(Leca2), R128C). This revealed distinct roles in regulating proliferation in the developing cerebral cortex, with the PAI and RED subdomain mutations reducing and increasing, respectively, the number of mitoses. Conversely, neurogenesis was affected only by the PAI subdomain mutation, phenocopying the neurogenic defects observed in full Pax6 mutants. Genome-wide expression profiling identified molecularly discrete signatures of Pax6(Leca4) and Pax6(Leca2) mutations. Comparison to Pax6 targets identified by chromatin immunoprecipitation led to the identification and functional characterization of distinct DNA motifs in the promoters of target genes dysregulated in the Pax6(Leca2) or Pax6(Leca4) mutants, further supporting the distinct regulatory functions of the DNA-binding subdomains. Thus, Pax6 achieves its key roles in the developing forebrain by utilizing particular subdomains to coordinate patterning, neurogenesis and proliferation simultaneously.


Subject(s)
Cell Proliferation , Eye Proteins/chemistry , Eye Proteins/genetics , Eye Proteins/physiology , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Neurogenesis/genetics , Paired Box Transcription Factors/chemistry , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/physiology , Repressor Proteins/chemistry , Repressor Proteins/genetics , Repressor Proteins/physiology , Animals , Animals, Genetically Modified , Body Patterning/genetics , Body Patterning/physiology , Embryo, Mammalian , Eye Proteins/metabolism , Homeodomain Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Models, Biological , Mutagenesis, Site-Directed , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , Prosencephalon/embryology , Prosencephalon/metabolism , Protein Interaction Mapping , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , Repressor Proteins/metabolism , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...