Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
EMBO Rep ; 24(8): e56233, 2023 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-37382163

RESUMO

Cortical neurogenesis depends on the balance between self-renewal and differentiation of apical progenitors (APs). Here, we study the epigenetic control of AP's division mode by focusing on the enzymatic activity of the histone methyltransferase DOT1L. Combining lineage tracing with single-cell RNA sequencing of clonally related cells, we show at the cellular level that DOT1L inhibition increases neurogenesis driven by a shift of APs from asymmetric self-renewing to symmetric neurogenic consumptive divisions. At the molecular level, DOT1L activity prevents AP differentiation by promoting transcription of metabolic genes. Mechanistically, DOT1L inhibition reduces activity of an EZH2/PRC2 pathway, converging on increased expression of asparagine synthetase (ASNS), a microcephaly associated gene. Overexpression of ASNS in APs phenocopies DOT1L inhibition, and also increases neuronal differentiation of APs. Our data suggest that DOT1L activity/PRC2 crosstalk controls AP lineage progression by regulating asparagine metabolism.


Assuntos
Aspartato-Amônia Ligase , Células-Tronco Neurais , Aspartato-Amônia Ligase/metabolismo , Diferenciação Celular/genética , Células-Tronco Neurais/metabolismo , Neurogênese/genética
2.
EMBO J ; 36(17): 2642-2658, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28765163

RESUMO

The generation of neocortical neurons from neural progenitor cells (NPCs) is primarily controlled by transcription factors binding to DNA in the context of chromatin. To understand the complex layer of regulation that orchestrates different NPC types from the same DNA sequence, epigenome maps with cell type resolution are required. Here, we present genomewide histone methylation maps for distinct neural cell populations in the developing mouse neocortex. Using different chromatin features, we identify potential novel regulators of cortical NPCs. Moreover, we identify extensive H3K27me3 changes between NPC subtypes coinciding with major developmental and cell biological transitions. Interestingly, we detect dynamic H3K27me3 changes on promoters of several crucial transcription factors, including the basal progenitor regulator Eomes We use catalytically inactive Cas9 fused with the histone methyltransferase Ezh2 to edit H3K27me3 at the Eomes locus in vivo, which results in reduced Tbr2 expression and lower basal progenitor abundance, underscoring the relevance of dynamic H3K27me3 changes during neocortex development. Taken together, we provide a rich resource of neocortical histone methylation data and outline an approach to investigate its contribution to the regulation of selected genes during neocortical development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Histonas/genética , Neocórtex/metabolismo , Células-Tronco Neurais/fisiologia , Animais , Epigênese Genética , Perfilação da Expressão Gênica , Genoma , Metilação , Camundongos Transgênicos , Neurogênese/fisiologia
3.
EMBO Rep ; 20(10): e47880, 2019 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-31469223

RESUMO

Microinjection into single cells in brain tissue is a powerful technique to study and manipulate neural stem cells. However, such microinjection requires expertise and is a low-throughput process. We developed the "Autoinjector", a robot that utilizes images from a microscope to guide a microinjection needle into tissue to deliver femtoliter volumes of liquids into single cells. The Autoinjector enables microinjection of hundreds of cells within a single organotypic slice, resulting in an overall yield that is an order of magnitude greater than manual microinjection. The Autoinjector successfully targets both apical progenitors (APs) and newborn neurons in the embryonic mouse and human fetal telencephalon. We used the Autoinjector to systematically study gap-junctional communication between neural progenitors in the embryonic mouse telencephalon and found that apical contact is a characteristic feature of the cells that are part of a gap junction-coupled cluster. The throughput and versatility of the Autoinjector will render microinjection an accessible high-performance single-cell manipulation technique and will provide a powerful new platform for performing single-cell analyses in tissue for bioengineering and biophysics applications.


Assuntos
Encéfalo/citologia , Microinjeções , Robótica , Análise de Célula Única , Animais , Automação , Comunicação Celular , Linhagem da Célula , Humanos , Processamento de Imagem Assistida por Computador , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/citologia , Neurogênese , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Telencéfalo/citologia , Telencéfalo/embriologia
4.
PLoS Biol ; 13(8): e1002217, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26252244

RESUMO

The evolutionary expansion of the neocortex in mammals has been linked to enlargement of the subventricular zone (SVZ) and increased proliferative capacity of basal progenitors (BPs), notably basal radial glia (bRG). The transcription factor Pax6 is known to be highly expressed in primate, but not mouse, BPs. Here, we demonstrate that sustaining Pax6 expression selectively in BP-genic apical radial glia (aRG) and their BP progeny of embryonic mouse neocortex suffices to induce primate-like progenitor behaviour. Specifically, we conditionally expressed Pax6 by in utero electroporation using a novel, Tis21-CreERT2 mouse line. This expression altered aRG cleavage plane orientation to promote bRG generation, increased cell-cycle re-entry of BPs, and ultimately increased upper-layer neuron production. Upper-layer neuron production was also increased in double-transgenic mouse embryos with sustained Pax6 expression in the neurogenic lineage. Strikingly, increased BPs existed not only in the SVZ but also in the intermediate zone of the neocortex of these double-transgenic mouse embryos. In mutant mouse embryos lacking functional Pax6, the proportion of bRG among BPs was reduced. Our data identify specific Pax6 effects in BPs and imply that sustaining this Pax6 function in BPs could be a key aspect of SVZ enlargement and, consequently, the evolutionary expansion of the neocortex.


Assuntos
Proteínas do Olho/metabolismo , Proteínas de Homeodomínio/metabolismo , Neocórtex/citologia , Células-Tronco Neurais/metabolismo , Neuroglia/metabolismo , Fatores de Transcrição Box Pareados/metabolismo , Proteínas Repressoras/metabolismo , Animais , Evolução Biológica , Feminino , Imunofluorescência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neocórtex/embriologia , Neocórtex/metabolismo , Neurônios/metabolismo , Fator de Transcrição PAX6 , Primatas
5.
Sci Adv ; 8(30): eabn7702, 2022 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-35905187

RESUMO

Since the ancestors of modern humans separated from those of Neanderthals, around 100 amino acid substitutions spread to essentially all modern humans. The biological significance of these changes is largely unknown. Here, we examine all six such amino acid substitutions in three proteins known to have key roles in kinetochore function and chromosome segregation and to be highly expressed in the stem cells of the developing neocortex. When we introduce these modern human-specific substitutions in mice, three substitutions in two of these proteins, KIF18a and KNL1, cause metaphase prolongation and fewer chromosome segregation errors in apical progenitors of the developing neocortex. Conversely, the ancestral substitutions cause shorter metaphase length and more chromosome segregation errors in human brain organoids, similar to what we find in chimpanzee organoids. These results imply that the fidelity of chromosome segregation during neocortex development improved in modern humans after their divergence from Neanderthals.


Assuntos
Hominidae , Homem de Neandertal , Animais , Encéfalo , Segregação de Cromossomos/genética , Humanos , Cinesinas , Metáfase , Camundongos , Homem de Neandertal/genética
6.
Bio Protoc ; 11(10): e4031, 2021 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-34150938

RESUMO

The size of the neocortex and its morphology are highly divergent across mammalian species. Several approaches have been utilized for the analysis of neocortical development and comparison among different species. In the present protocol (Note: This protocol requires basic knowledge of brain anatomy), we describe three ex vivo neocortical slice/tissue culture methods: (i) organotypic slice culture (mouse, ferret, human); (ii) hemisphere rotation culture (mouse, ferret); and (iii) free-floating tissue culture (mouse, ferret, human). Each of these three culture methods offers distinct features with regard to the analyses to be performed and can be combined with genetic manipulation by electroporation and treatment with specific inhibitors. These three culture methods are therefore powerful techniques to examine the function of genes involved in neocortical development.

7.
J Vis Exp ; (167)2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33554975

RESUMO

A central question in developmental neurobiology is how neural stem and progenitor cells form the brain. To answer this question, one needs to label, manipulate, and follow single cells in the brain tissue with high resolution over time. This task is extremely challenging due to the complexity of tissues in the brain. We have recently developed a robot, that guide a microinjection needle into brain tissue upon utilizing images acquired from a microscope to deliver femtoliter volumes of solution into single cells. The robotic operation increases resulting an overall yield that is an order of magnitude greater than manual microinjection and allows for precise labeling and flexible manipulation of single cells in living tissue. With this, one can microinject hundreds of cells within a single organotypic slice. This article demonstrates the use of the microinjection robot for automated microinjection of neural progenitor cells and neurons in the brain tissue slices. More broadly, it can be used on any epithelial tissue featuring a surface that can be reached by the pipette. Once set up, the microinjection robot can execute 15 or more microinjections per minute. The microinjection robot because of its throughput and versality will make microinjection a broadly straightforward high-performance cell manipulation technique to be used in bioengineering, biotechnology, and biophysics for performing single-cell analyses in organotypic brain slices.


Assuntos
Encéfalo/citologia , Microinjeções , Células-Tronco Neurais/citologia , Neurônios/citologia , Robótica , Análise de Célula Única , Animais , Automação , Imunofluorescência , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Tecidos
8.
Science ; 369(6503): 546-550, 2020 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-32554627

RESUMO

The neocortex has expanded during mammalian evolution. Overexpression studies in developing mouse and ferret neocortex have implicated the human-specific gene ARHGAP11B in neocortical expansion, but the relevance for primate evolution has been unclear. Here, we provide functional evidence that ARHGAP11B causes expansion of the primate neocortex. ARHGAP11B expressed in fetal neocortex of the common marmoset under control of the gene's own (human) promoter increased the numbers of basal radial glia progenitors in the marmoset outer subventricular zone, increased the numbers of upper-layer neurons, enlarged the neocortex, and induced its folding. Thus, the human-specific ARHGAP11B drives changes in development in the nonhuman primate marmoset that reflect the changes in evolution that characterize human neocortical development.


Assuntos
Proteínas Ativadoras de GTPase/metabolismo , Neocórtex/embriologia , Animais , Animais Geneticamente Modificados , Callithrix , Feto , Proteínas Ativadoras de GTPase/genética , Humanos , Ventrículos Laterais/embriologia , Ventrículos Laterais/metabolismo , Neocórtex/anatomia & histologia , Neocórtex/metabolismo , Células-Tronco Neurais/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Tamanho do Órgão , Regiões Promotoras Genéticas
9.
Neuron ; 97(6): 1299-1314.e8, 2018 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-29503187

RESUMO

Delamination of neural progenitor cells (NPCs) from the ventricular surface is a crucial prerequisite to form the subventricular zone, the germinal layer linked to the expansion of the mammalian neocortex in development and evolution. Here, we dissect the molecular mechanism by which the transcription factor Insm1 promotes the generation of basal progenitors (BPs). Insm1 protein is most highly expressed in newborn BPs in mouse and human developing neocortex. Forced Insm1 expression in embryonic mouse neocortex causes NPC delamination, converting apical to basal radial glia. Insm1 represses the expression of the apical adherens junction belt-specific protein Plekha7. CRISPR/Cas9-mediated disruption of Plekha7 expression suffices to cause NPC delamination. Plekha7 overexpression impedes the intrinsic and counteracts the Insm1-induced, NPC delamination. Our findings uncover a novel molecular mechanism underlying NPC delamination in which a BP-genic transcription factor specifically targets the integrity of the apical adherens junction belt, rather than adherens junction components as such.


Assuntos
Junções Aderentes/metabolismo , Proteínas de Transporte/biossíntese , Proteínas de Ligação a DNA/biossíntese , Regulação para Baixo/fisiologia , Neocórtex/metabolismo , Células-Tronco Neurais/metabolismo , Fatores de Transcrição/biossíntese , Animais , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neocórtex/citologia , Neocórtex/crescimento & desenvolvimento , Técnicas de Cultura de Órgãos , Gravidez , Proteínas Repressoras
10.
J Neurosci ; 25(28): 6533-8, 2005 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-16014714

RESUMO

During embryonic development of the mammalian brain, the average cell-cycle length of progenitor cells in the ventricular zone is known to increase. However, for any given region of the developing cortex and stage of neurogenesis, the length of the cell cycle is thought to be similar in the two coexisting subpopulations of progenitors [i.e., those undergoing (symmetric) proliferative divisions and those undergoing (either asymmetric or symmetric) neuron-generating divisions]. Using cumulative bromodeoxyuridine labeling of Tis21-green fluorescent protein knock-in mouse embryos, in which these two subpopulations of progenitors can be distinguished in vivo, we now show that at the onset as well as advanced stages of telencephalic neurogenesis, progenitors undergoing neuron-generating divisions are characterized by a significantly longer cell cycle than progenitors undergoing proliferative divisions. In addition, we find that the recently characterized neuronal progenitors dividing at the basal side of the ventricular zone and in the subventricular zone have a longer G(2) phase than those dividing at the ventricular surface. These findings are consistent with the hypothesis (Calegari and Huttner, 2003) that cell-cycle lengthening can causally contribute to neural progenitors switching from proliferative to neuron-generating divisions and may have important implications for the expansion of somatic stem cells in general.


Assuntos
Encéfalo/embriologia , Ciclo Celular , Neuroglia/citologia , Neurônios/citologia , Células-Tronco/citologia , Animais , Encéfalo/citologia , Diferenciação Celular , Divisão Celular , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Genes Supressores de Tumor , Idade Gestacional , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Proteínas Imediatamente Precoces/análise , Proteínas Imediatamente Precoces/biossíntese , Proteínas Imediatamente Precoces/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/biossíntese , Telencéfalo/citologia , Telencéfalo/embriologia , Fatores de Tempo , Proteínas Supressoras de Tumor
11.
Sci Rep ; 6: 21206, 2016 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-26879757

RESUMO

Apical radial glia (aRG), the stem cells in developing neocortex, are unique bipolar epithelial cells, extending an apical process to the ventricle and a basal process to the basal lamina. Here, we report novel features of the Golgi apparatus, a central organelle for cell polarity, in mouse aRGs. The Golgi was confined to the apical process but not associated with apical centrosome(s). In contrast, in aRG-derived, delaminating basal progenitors that lose apical polarity, the Golgi became pericentrosomal. The aRG Golgi underwent evolutionarily conserved, accordion-like compression and extension concomitant with cell cycle-dependent nuclear migration. Importantly, in line with endoplasmic reticulum but not Golgi being present in the aRG basal process, its plasma membrane contained glycans lacking Golgi processing, consistent with direct ER-to-cell surface membrane traffic. Our study reveals hitherto unknown complexity of neural stem cell polarity, differential Golgi contribution to their specific architecture, and fundamental Golgi re-organization upon cell fate change.


Assuntos
Complexo de Golgi/metabolismo , Células-Tronco Neurais/metabolismo , Animais , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Expressão Gênica , Genes Reporter , Complexo de Golgi/ultraestrutura , Camundongos , Camundongos Transgênicos , Mitose , Células-Tronco Neurais/ultraestrutura , Polissacarídeos/metabolismo , Transporte Proteico
12.
Front Cell Neurosci ; 9: 325, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26379497

RESUMO

Midbody release from proliferative neural progenitor cells is tightly associated with the neuronal commitment of neural progenitor cells during the progression of neurogenesis in the mammalian cerebral cortex. While the central portion of the midbody, a cytoplasmic bridge between nascent daughter cells, is engulfed by one of the daughter cell by most cells in vitro, it is shown to be released into the extracellular cerebrospinal fluid (CF) in vivo in mouse embryos. Several proteins have been involved in midbody release; however, few studies have addressed the participation of the plasma membrane's lipids in this process. Here, we show by Shotgun Lipidomic analysis that phosphatydylserine (PS), among other lipids, is enriched in the released midbodies compared to lipoparticles and cellular membranes, both collected from the CF of the developing mouse embryos. Moreover, the developing mouse embryo neural progenitor cells released two distinct types of midbodies carrying either internalized PS or externalized PS on their membrane. This strongly suggests that phagocytosis and an alternative fate of released midbodies exists. HeLa cells, which are known to mainly engulf the midbody show almost no PS exposure, if any, on the outer leaflet of the midbody membrane. These results point toward that PS exposure might be involved in the selection of recipients of released midbodies, either to be engulfed by daughter cells or phagocytosed by non-daughter cells or another cell type in the developing cerebral cortex.

13.
Science ; 347(6229): 1465-70, 2015 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-25721503

RESUMO

Evolutionary expansion of the human neocortex reflects increased amplification of basal progenitors in the subventricular zone, producing more neurons during fetal corticogenesis. In this work, we analyze the transcriptomes of distinct progenitor subpopulations isolated by a cell polarity-based approach from developing mouse and human neocortex. We identify 56 genes preferentially expressed in human apical and basal radial glia that lack mouse orthologs. Among these, ARHGAP11B has the highest degree of radial glia-specific expression. ARHGAP11B arose from partial duplication of ARHGAP11A (which encodes a Rho guanosine triphosphatase-activating protein) on the human lineage after separation from the chimpanzee lineage. Expression of ARHGAP11B in embryonic mouse neocortex promotes basal progenitor generation and self-renewal and can increase cortical plate area and induce gyrification. Hence, ARHGAP11B may have contributed to evolutionary expansion of human neocortex.


Assuntos
Proteínas Ativadoras de GTPase/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Neocórtex/embriologia , Células-Tronco Neurais/citologia , Neurogênese/genética , Animais , Separação Celular , Proteínas Ativadoras de GTPase/química , Proteínas Ativadoras de GTPase/genética , Duplicação Gênica , Humanos , Ventrículos Laterais/citologia , Camundongos , Neocórtex/citologia , Neocórtex/metabolismo , Células-Tronco Neurais/metabolismo , Neuroglia/citologia , Neuroglia/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Estrutura Terciária de Proteína , Transcriptoma
14.
Cell Rep ; 7(2): 398-411, 2014 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-24726360

RESUMO

Mammalian neocortex size primarily reflects the number and mode of divisions of neural stem and progenitor cells. Cortical stem cells (apical progenitors) switching from symmetric divisions, which expand their population, to asymmetric divisions, which generate downstream neuronal progenitors (basal progenitors), start expressing Tis21, a so-called antiproliferative/prodifferentiative gene. Tis21 encodes a small (17.5 kDa), functionally poorly characterized protein and a relatively large (2 kb), highly conserved 3' UTR. Here, we show that mice lacking the Tis21 3' UTR develop a microcephalic neocortex with fewer neurons, notably in the upper layers. This reflects a progressive decrease in basal progenitors, which in turn is due to a fraction of apical progenitors prematurely switching from asymmetric self-renewing to symmetric self-consuming divisions. This switch is caused by the markedly increased Tis21 protein level resulting from lack of microRNA-, notably miR-92-, dependent restriction of Tis21 expression. Our data show that a premature onset of consumptive neural stem cell divisions can lead to microcephaly.


Assuntos
Regiões 3' não Traduzidas , Divisão Celular Assimétrica , Proteínas Imediatamente Precoces/metabolismo , MicroRNAs/genética , Neocórtex/metabolismo , Células-Tronco Neurais/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Regulação para Baixo , Proteínas Imediatamente Precoces/genética , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Microcefalia/genética , Neocórtex/embriologia , Neocórtex/patologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/fisiologia , Neurogênese , Tamanho do Órgão , Proteínas Supressoras de Tumor/genética
15.
Nat Protoc ; 9(5): 1170-82, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24762784

RESUMO

This microinjection protocol allows the manipulation and tracking of neural stem and progenitor cells in tissue at single-cell resolution. We demonstrate how to apply microinjection to organotypic brain slices obtained from mice and ferrets; however, our technique is not limited to mouse and ferret embryos, but provides a means of introducing a wide variety of membrane-impermeable molecules (e.g., nucleic acids, proteins, hydrophilic compounds) into neural stem and progenitor cells of any developing mammalian brain. Microinjection experiments are conducted by using a phase-contrast microscope equipped with epifluorescence, a transjector and a micromanipulator. The procedure normally takes ∼2 h for an experienced researcher, and the entire protocol, including tissue processing, can be performed within 1 week. Thus, microinjection is a unique and versatile method for changing and tracking the fate of a cell in organotypic slice culture.


Assuntos
Encéfalo/citologia , Microinjeções/métodos , Células-Tronco Neurais/metabolismo , Animais , Furões , Fluorescência , Camundongos , Micromanipulação/métodos , Microscopia de Contraste de Fase
16.
Nat Neurosci ; 15(2): 329-37, 2011 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-22179113

RESUMO

A challenge in the field of neural stem cell biology is the mechanistic dissection of single stem cell behavior in tissue. Although such behavior can be tracked by sophisticated imaging techniques, current methods of genetic manipulation do not allow researchers to change the level of a defined gene product on a truly acute time scale and are limited to very few genes at a time. To overcome these limitations, we established microinjection of neuroepithelial/radial glial cells (apical progenitors) in organotypic slice culture of embryonic mouse brain. Microinjected apical progenitors showed cell cycle parameters that were indistinguishable to apical progenitors in utero, underwent self-renewing divisions and generated neurons. Microinjection of single genes, recombinant proteins or complex mixtures of RNA was found to elicit acute and defined changes in apical progenitor behavior and progeny fate. Thus, apical progenitor microinjection provides a new approach to acutely manipulating single neural stem and progenitor cells in tissue.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Neurais/fisiologia , Animais , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Diferenciação Celular/genética , Embrião de Mamíferos , Proteínas Imediatamente Precoces/genética , Técnicas In Vitro , Proteínas Luminescentes/administração & dosagem , Proteínas Luminescentes/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microinjeções/métodos , Mutação/genética , Células-Tronco Neurais/efeitos dos fármacos , RNA Mensageiro/farmacologia , Rombencéfalo/citologia , Rombencéfalo/embriologia , Fatores de Tempo , Proteínas Supressoras de Tumor/genética , Proteína cdc42 de Ligação ao GTP/administração & dosagem , Proteína cdc42 de Ligação ao GTP/metabolismo
17.
Nat Commun ; 2: 154, 2011 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-21224845

RESUMO

During mammalian cerebral cortex development, the G1-phase of the cell cycle is known to lengthen, but it has been unclear which neural stem and progenitor cells are affected. In this paper, we develop a novel approach to determine cell-cycle parameters in specific classes of neural stem and progenitor cells, identified by molecular markers rather than location. We found that G1 lengthening was associated with the transition from stem cell-like apical progenitors to fate-restricted basal (intermediate) progenitors. Unexpectedly, expanding apical and basal progenitors exhibit a substantially longer S-phase than apical and basal progenitors committed to neuron production. Comparative genome-wide gene expression analysis of expanding versus committed progenitor cells revealed changes in key factors of cell-cycle regulation, DNA replication and repair and chromatin remodelling. Our findings suggest that expanding neural stem and progenitor cells invest more time during S-phase into quality control of replicated DNA than those committed to neuron production.

18.
Development ; 135(23): 3911-21, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18997113

RESUMO

Neurogenesis during the development of the mammalian cerebral cortex involves a switch of neural stem and progenitor cells from proliferation to differentiation. To explore the possible role of microRNAs (miRNAs) in this process, we conditionally ablated Dicer in the developing mouse neocortex using Emx1-Cre, which is specifically expressed in the dorsal telencephalon as early as embryonic day (E) 9.5. Dicer ablation in neuroepithelial cells, which are the primary neural stem and progenitor cells, and in the neurons derived from them, was evident from E10.5 onwards, as ascertained by the depletion of the normally abundant miRNAs miR-9 and miR-124. Dicer ablation resulted in massive hypotrophy of the postnatal cortex and death of the mice shortly after weaning. Analysis of the cytoarchitecture of the Dicer-ablated cortex revealed a marked reduction in radial thickness starting at E13.5, and defective cortical layering postnatally. Whereas the former was due to neuronal apoptosis starting at E12.5, which was the earliest detectable phenotype, the latter reflected dramatic impairment of neuronal differentiation. Remarkably, the primary target cells of Dicer ablation, the neuroepithelial cells, and the neurogenic progenitors derived from them, were unaffected by miRNA depletion with regard to cell cycle progression, cell division, differentiation and viability during the early stage of neurogenesis, and only underwent apoptosis starting at E14.5. Our results support the emerging concept that progenitors are less dependent on miRNAs than their differentiated progeny, and raise interesting perspectives as to the expansion of somatic stem cells.


Assuntos
Diferenciação Celular/genética , MicroRNAs/metabolismo , Neocórtex/citologia , Neocórtex/embriologia , Neurogênese/genética , Neurônios/citologia , Células-Tronco/citologia , Animais , Animais Recém-Nascidos , Apoptose , Contagem de Células , Linhagem da Célula , Proliferação de Células , Sobrevivência Celular , RNA Helicases DEAD-box/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário , Endorribonucleases/metabolismo , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Interneurônios/citologia , Interneurônios/metabolismo , Camundongos , Mitose , Neocórtex/metabolismo , Neurônios/enzimologia , Ribonuclease III , Células-Tronco/enzimologia
19.
Neuron ; 60(1): 40-55, 2008 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-18940587

RESUMO

Basal (intermediate) progenitors are the major source of neurons in the mammalian neocortex. The molecular machinery governing basal progenitor biogenesis is unknown. Here, we show that the zinc-finger transcription factor Insm1 (insulinoma-associated 1) is expressed specifically in progenitors undergoing neurogenic divisions, has a panneurogenic role throughout the brain, and promotes basal progenitor formation in the neocortex. Mouse embryos lacking Insm1 contained half the number of basal progenitors and showed a marked reduction in cortical plate radial thickness. Forced premature expression of Insm1 in neuroepithelial cells resulted in their mitosis occurring at the basal (rather than apical) side of the ventricular zone and induced expression of the basal progenitor marker Tbr2. Remarkably, these cells remained negative for Tis21, a marker of neurogenic progenitors, and did not generate neurons but underwent self-amplification. Our data imply that Insm1 is involved in the generation and expansion of basal progenitors, a hallmark of neocortex evolution.


Assuntos
Diferenciação Celular/fisiologia , Proteínas de Ligação a DNA/fisiologia , Neocórtex/embriologia , Neocórtex/metabolismo , Neurônios/metabolismo , Células-Tronco/metabolismo , Fatores de Transcrição/fisiologia , Animais , Diferenciação Celular/genética , Células Cultivadas , Proteínas de Ligação a DNA/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neocórtex/citologia , Neurônios/citologia , Gravidez , Proteínas Repressoras , Células-Tronco/citologia , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA