Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neurosci ; 43(7): 1154-1165, 2023 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-36596698

RESUMO

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


Assuntos
Neocórtex , Células-Tronco Neurais , Neurogênese , Animais , Camundongos , Fosfatases cdc25/genética , Fosfatases cdc25/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese/genética , Neurônios/metabolismo
2.
Elife ; 102021 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-34607629

RESUMO

Although cell-to-cell heterogeneity in gene and protein expression within cell populations has been widely documented, we know little about its biological functions. By studying progenitors of the posterior region of bird embryos, we found that expression levels of transcription factors Sox2 and Bra, respectively involved in neural tube (NT) and mesoderm specification, display a high degree of cell-to-cell heterogeneity. By combining forced expression and downregulation approaches with time-lapse imaging, we demonstrate that Sox2-to-Bra ratio guides progenitor's motility and their ability to stay in or exit the progenitor zone to integrate neural or mesodermal tissues. Indeed, high Bra levels confer high motility that pushes cells to join the paraxial mesoderm, while high levels of Sox2 tend to inhibit cell movement forcing cells to integrate the NT. Mathematical modeling captures the importance of cell motility regulation in this process and further suggests that randomness in Sox2/Bra cell-to-cell distribution favors cell rearrangements and tissue shape conservation.


Assuntos
Diferenciação Celular/genética , Embrião não Mamífero/fisiologia , Proteínas Fetais/genética , Regulação da Expressão Gênica no Desenvolvimento , Expressão Gênica , Fatores de Transcrição SOXB1/genética , Proteínas com Domínio T/genética , Animais , Diferenciação Celular/fisiologia , Linhagem da Célula , Codorniz
3.
Dev Biol ; 436(2): 94-107, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29486153

RESUMO

Proper embryonic development relies on a tight control of spatial and temporal gene expression profiles in a highly regulated manner. One good example is the ON/OFF switching of the transcription factor PAX6 that governs important steps of neurogenesis. In the neural tube PAX6 expression is initiated in neural progenitors through the positive action of retinoic acid signaling and downregulated in neuronal precursors by the bHLH transcription factor NEUROG2. How these two regulatory inputs are integrated at the molecular level to properly fine tune temporal PAX6 expression is not known. In this study we identified and characterized a 940-bp long distal cis-regulatory module (CRM), located far away from the PAX6 transcription unit and which conveys positive input from RA signaling pathway and indirect repressive signal(s) from NEUROG2. These opposing regulatory signals are integrated through HOMZ, a 94 bp core region within E940 which is evolutionarily conserved in distant organisms such as the zebrafish. We show that within HOMZ, NEUROG2 and RA exert their opposite temporal activities through a short 60 bp region containing a functional RA-responsive element (RARE). We propose a model in which retinoic acid receptors (RARs) and NEUROG2 repressive target(s) compete on the same DNA motif to fine tune temporal PAX6 expression during the course of spinal neurogenesis.


Assuntos
Elementos Facilitadores Genéticos/genética , Tubo Neural/metabolismo , Neurogênese/genética , Fator de Transcrição PAX6/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Embrião de Galinha , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Tubo Neural/embriologia , Receptores do Ácido Retinoico/metabolismo , Transdução de Sinais/fisiologia , Peixe-Zebra
4.
Development ; 139(6): 1095-104, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22318230

RESUMO

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


Assuntos
Fase G2 , Sistema Nervoso/embriologia , Células-Tronco Neurais/citologia , Neurogênese , Medula Espinal/embriologia , Fosfatases cdc25/metabolismo , Animais , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Embrião de Galinha , Células-Tronco Neurais/fisiologia , Neurônios/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Medula Espinal/citologia , Fosfatases cdc25/biossíntese
5.
Dev Biol ; 305(2): 659-73, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17399698

RESUMO

During neurogenesis, complex networks of genes act sequentially to control neuronal differentiation. In the neural tube, the expression of Pax6, a paired-box-containing gene, just precedes the appearance of the first post-mitotic neurons. So far, its only reported function in the spinal cord is in specifying subsets of neurons. Here we address its possible function in controlling the balance between proliferation and commitment of neural progenitors. We report that increasing Pax6 level is sufficient to push neural progenitors toward cell cycle exit and neuronal commitment via Neurogenin 2 (Ngn2) upregulation. However, neuronal precursors maintaining Pax6(On) fail to perform neuronal differentiation. Conversely, turning off Pax6 function in these precursors is sufficient to provoke premature differentiation and the number of differentiated neurons depends of the amount of Pax6 protein. Moreover, we found that Pax6 expression involves negative feedback regulation by Ngn2 and this repression is critical for the proneural activity of Ngn2. We present a model in which the level of Pax6 activity first conditions the moment when a given progenitor will leave the cell cycle and second, the moment when a selected neuronal precursor will irreversibly differentiate.


Assuntos
Diferenciação Celular/fisiologia , Proteínas do Olho/fisiologia , Proteínas de Homeodomínio/fisiologia , Neurônios/citologia , Fatores de Transcrição Box Pareados/fisiologia , Proteínas Repressoras/fisiologia , Medula Espinal/embriologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/antagonistas & inibidores , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Ciclo Celular/fisiologia , Diferenciação Celular/genética , Embrião de Galinha , Proteínas do Olho/antagonistas & inibidores , Proteínas do Olho/biossíntese , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/antagonistas & inibidores , Proteínas de Homeodomínio/biossíntese , Camundongos , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/antagonistas & inibidores , Fatores de Transcrição Box Pareados/biossíntese , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/biossíntese , Medula Espinal/citologia , Medula Espinal/fisiologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Tempo
6.
Dev Biol ; 294(1): 133-47, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16564519

RESUMO

Sonic hedgehog (Shh) signaling controls numerous aspects of vertebrate development, including proliferation of precursors in different organs. Identification of molecules that link the Shh pathway to cell cycle machinery is therefore of major importance for an understanding of the mechanisms underlying Shh-dependent proliferation. Here, we show that an actor in the control of entry into mitosis, the phosphatase CDC25B, is transcriptionally upregulated by the Shh/Gli pathway. Unlike other G2/M regulators, CDC25B is highly expressed in domains of Shh activity, including the ventral neural tube and the posterior limb bud. Loss- and gain-of-function experiments reveal that Shh contributes to CDC25B transcriptional activation in the neural tube both of chick and mouse embryos. Moreover, CDC25B transcripts are absent from the posterior limb bud of Shh-/- mice, while anterior grafts of Shh-expressing cells in the chicken limb bud induce ectopic CDC25B expression. Arresting the cell cycle does not reduce the level of CDC25B expression in the neural tube strongly suggesting that the upregulation of CDC25B is not an indirect consequence of the Shh-dependent proliferation. These data reveal an unexpected developmental link between the Shh pathway and a participant in G2/M control.


Assuntos
Proteínas de Ciclo Celular/genética , Divisão Celular/fisiologia , Fase G2/fisiologia , Transativadores/metabolismo , Fosfatases cdc25/genética , Animais , Proliferação de Células , Sistema Nervoso Central/embriologia , Embrião de Galinha , Embrião de Mamíferos , Proteínas Hedgehog , Botões de Extremidades , Camundongos , Camundongos Knockout , Transativadores/fisiologia , Transcrição Gênica , Regulação para Cima
7.
Dev Biol ; 273(2): 195-209, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15328007

RESUMO

In the vertebrate embryo, spinal cord elongation requires FGF signaling that promotes the continuous development of the posterior nervous system by maintaining a stem zone of proliferating neural progenitors. Those escaping the caudal neural stem zone, which is expressed to Shh signal, initiate ventral patterning in the neural groove before starting neuronal differentiation in the neural tube. Here we investigated the integration of D-type cyclins, known to govern cell cycle progression under the control of extracellular signals, in the program of spinal cord maturation. In chicken embryo, we find that cyclin D2 is preferentially expressed in the posterior neural plate, whereas cyclin D1 appears in the neural groove. We demonstrated by loss- and gain-of-function experiments that FGF signaling maintains cyclin D2 in the immature caudal neural epithelium, while Shh activates cyclin D1 in the neural groove. Moreover, forced maintenance of cyclin D1 or D2 in the neural tube favors proliferation at the expense of neuronal differentiation. These results contribute to our understanding of how the cell cycle control can be linked to the patterning programs to influence the balance between proliferation and neuronal differentiation in discrete progenitors domains.


Assuntos
Ciclina D1/metabolismo , Ciclinas/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Medula Espinal/embriologia , Medula Espinal/metabolismo , Transativadores/metabolismo , Animais , Animais Geneticamente Modificados , Sequência de Bases , Padronização Corporal , Ciclo Celular , Diferenciação Celular , Embrião de Galinha , Ciclina D1/genética , Ciclinas/genética , Primers do DNA/genética , Fatores de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog , Hibridização In Situ , Modelos Neurológicos , Transdução de Sinais , Medula Espinal/citologia , Transativadores/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA