Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Data Brief ; 24: 103909, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31061857

RESUMO

SOX2 is a stem cell-associated pluripotency transcription factor whose role in neuronal populations is undefined. Here we present the RNA-sequencing based transcriptome profiles of control (Sox2 fl/fl ) and SOX2 conditional knock-out (Vgat-cre;Sox2 fl/fl ) mice at four time points in one 24-h circadian cycle. The raw sequencing data were deposited to ArrayExpress database at EMBL-EBI (https://www.ebi.ac.uk/arrayexpress) under the accession number E-MTAB-7496. Results of rhythmicity analysis, differential expression analysis, network prediction, and potential target identification stemming from the RNA-sequencing dataset are also given in this article. The interpretation and discussion of these data can be found in the related research article entitled "SOX2-dependent transcription in clock neurons promotes the robustness of the central circadian pacemaker." Cheng et al. 2019.

2.
Cell Rep ; 26(12): 3191-3202.e8, 2019 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-30893593

RESUMO

Clock neurons within the mammalian suprachiasmatic nuclei (SCN) encode circadian time using interlocked transcription-translation feedback loops (TTFLs) that drive rhythmic gene expression. However, the contributions of other transcription factors outside of the circadian TTFLs to the functionality of the SCN remain obscure. Here, we report that the stem and progenitor cell transcription factor, sex-determining region Y-box 2 (SOX2), is expressed in adult SCN neurons and positively regulates transcription of the core clock gene, Period2. Mice lacking SOX2 selectively in SCN neurons display imprecise, poorly consolidated behavioral rhythms that do not entrain efficiently to environmental light cycles and that are highly susceptible to constant light-induced arrhythmicity. RNA sequencing revealed that Sox2 deficiency alters the SCN transcriptome, reducing the expression of core clock genes and neuropeptide-receptor systems. By defining the transcriptional landscape within SCN neurons, SOX2 enables the generation of robust, entrainable circadian rhythms that accurately reflect environmental time.


Assuntos
Relógios Circadianos/fisiologia , Proteínas Circadianas Period/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Núcleo Supraquiasmático/metabolismo , Transcrição Gênica , Animais , Camundongos , Camundongos Transgênicos , Proteínas Circadianas Period/genética , Fatores de Transcrição SOXB1/genética , Núcleo Supraquiasmático/citologia
3.
Sci Rep ; 8(1): 5294, 2018 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-29593295

RESUMO

Adult hippocampal neurogenesis is highly responsive to exercise, which promotes the proliferation of neural progenitor cells and the integration of newborn granule neurons in the dentate gyrus. Here we show that genetic ablation of the small GTPase, Dexras1, suppresses exercise-induced proliferation of neural progenitors, alters survival of mitotic and post-mitotic cells in a stage-specific manner, and increases the number of mature newborn granule neurons. Dexras1 is required for exercise-triggered recruitment of quiescent neural progenitors into the cell cycle. Pharmacological inhibition of NMDA receptors enhances SGZ cell proliferation in wild-type but not dexras1-deficient mice, suggesting that NMDA receptor-mediated signaling is dependent on Dexras1. At the molecular level, the absence of Dexras1 abolishes exercise-dependent activation of ERK/MAPK and CREB, and inhibits the upregulation of NMDA receptor subunit NR2A, bdnf, trkB and vegf-a expression in the dentate gyrus. Our study reveals Dexras1 as an important stage-specific regulator of exercise-induced neurogenesis in the adult hippocampus by enhancing pro-mitogenic signaling to neural progenitor cells and modulating cell survival.


Assuntos
Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Proteínas ras/metabolismo , Animais , Animais Recém-Nascidos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Ciclo Celular , Diferenciação Celular , Proliferação de Células/fisiologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/efeitos dos fármacos , Giro Denteado/metabolismo , Hipocampo/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Condicionamento Físico Animal/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais , Lobo Temporal/metabolismo , Proteínas ras/genética
4.
Cell Rep ; 22(5): 1124-1131, 2018 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-29386101

RESUMO

Histone variants were recently discovered to regulate neural plasticity, with H2A.Z emerging as a memory suppressor. Using whole-genome sequencing of the mouse hippocampus, we show that basal H2A.Z occupancy is positively associated with steady-state transcription, whereas learning-induced H2A.Z removal is associated with learning-induced gene expression. AAV-mediated H2A.Z depletion enhanced fear memory and resulted in gene-specific alterations of learning-induced transcription, reinforcing the role of H2A.Z as a memory suppressor. H2A.Z accumulated with age, although it remained sensitive to learning-induced eviction. Learning-related H2A.Z removal occurred at largely distinct genes in young versus aged mice, suggesting that H2A.Z is subject to regulatory shifts in the aged brain despite similar memory performance. When combined with prior evidence of H3.3 accumulation in neurons, our data suggest that nucleosome composition in the brain is reorganized with age.


Assuntos
Envelhecimento/metabolismo , Hipocampo/metabolismo , Histonas/metabolismo , Aprendizagem/fisiologia , Animais , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nucleossomos/metabolismo
5.
Cell Rep ; 19(3): 505-520, 2017 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-28423315

RESUMO

The central circadian pacemaker, the suprachiasmatic nucleus (SCN), encodes day length information by mechanisms that are not well understood. Here, we report that genetic ablation of miR-132/212 alters entrainment to different day lengths and non-24 hr day-night cycles, as well as photoperiodic regulation of Period2 expression in the SCN. SCN neurons from miR-132/212-deficient mice have significantly reduced dendritic spine density, along with altered methyl CpG-binding protein (MeCP2) rhythms. In Syrian hamsters, a model seasonal rodent, day length regulates spine density on SCN neurons in a melatonin-independent manner, as well as expression of miR-132, miR-212, and their direct target, MeCP2. Genetic disruption of Mecp2 fully restores the level of dendritic spines of miR-132/212-deficient SCN neurons. Our results reveal that, by regulating the dendritic structure of SCN neurons through a MeCP2-dependent mechanism, miR-132/212 affects the capacity of the SCN to encode seasonal time.


Assuntos
Adaptação Fisiológica/genética , Relógios Circadianos/genética , Dendritos/metabolismo , MicroRNAs/metabolismo , Estações do Ano , Adaptação Fisiológica/efeitos da radiação , Animais , Comportamento Animal , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Relógios Circadianos/efeitos da radiação , Dendritos/efeitos da radiação , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/efeitos da radiação , Feminino , Deleção de Genes , Regulação da Expressão Gênica/efeitos da radiação , Luz , Masculino , Mesocricetus , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , Neurônios/metabolismo , Fotoperíodo , Proteoma/metabolismo , Transdução de Sinais/efeitos da radiação , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/efeitos da radiação , Serina-Treonina Quinases TOR/metabolismo , Fatores de Tempo
6.
Cell Mol Life Sci ; 74(6): 1035-1059, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27689221

RESUMO

Circadian timekeeping is a ubiquitous mechanism that enables organisms to maintain temporal coordination between internal biological processes and time of the local environment. The molecular basis of circadian rhythms lies in a set of transcription-translation feedback loops (TTFLs) that drives the rhythmic transcription of core clock genes, whose level and phase of expression serve as the marker of circadian time. However, it has become increasingly evident that additional regulatory mechanisms impinge upon the TTFLs to govern the properties and behavior of the circadian clock. Such mechanisms include changes in chromatin architecture, interactions with other transcription factor networks, post-transcriptional control by RNA modifications, alternative splicing and microRNAs, and post-translational regulation of subcellular trafficking and protein degradation. In this review, we will summarize the current knowledge of circadian clock regulation-from transcriptional to post-translational-drawing from literature pertaining to the Drosophila and murine circadian systems.


Assuntos
Relógios Circadianos/genética , Drosophila melanogaster/genética , Animais , Regulação da Expressão Gênica , Camundongos , Modelos Genéticos , Biossíntese de Proteínas/genética , Transcrição Gênica
7.
Cell Rep ; 5(4): 961-73, 2013 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-24268780

RESUMO

The subgranular zone (SGZ) of the adult hippocampus contains a pool of quiescent neural progenitor cells (QNPs) that are capable of entering the cell cycle and producing newborn neurons. The mechanisms that control the timing and extent of adult neurogenesis are not well understood. Here, we show that QNPs of the adult SGZ express molecular-clock components and proliferate in a rhythmic fashion. The clock proteins PERIOD2 and BMAL1 are critical for proper control of neurogenesis. The absence of PERIOD2 abolishes the gating of cell-cycle entrance of QNPs, whereas genetic ablation of bmal1 results in constitutively high levels of proliferation and delayed cell-cycle exit. We use mathematical model simulations to show that these observations may arise from clock-driven expression of a cell-cycle inhibitor that targets the cyclin D/Cdk4-6 complex. Our findings may have broad implications for the circadian clock in timing cell-cycle events of other stem cell populations throughout the body.


Assuntos
Fatores de Transcrição ARNTL/genética , Relógios Circadianos/genética , Hipocampo/crescimento & desenvolvimento , Neurogênese/genética , Proteínas Circadianas Period/genética , Fatores de Transcrição ARNTL/biossíntese , Animais , Ciclo Celular/genética , Proliferação de Células , Ciclina D/antagonistas & inibidores , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Regulação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Neurais , Neurônios/citologia , Proteínas Circadianas Period/biossíntese , Regiões Promotoras Genéticas
8.
Chronobiol Int ; 29(8): 965-81, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22928915

RESUMO

Restricted feeding (RF) schedules are potent zeitgebers capable of entraining metabolic and hormonal rhythms in peripheral oscillators in anticipation of food. Behaviorally, this manifests in the form of food anticipatory activity (FAA) in the hours preceding food availability. Circadian rhythms of FAA are thought to be controlled by a food-entrainable oscillator (FEO) outside of the suprachiasmatic nucleus (SCN), the central circadian pacemaker in mammals. Although evidence suggests that the FEO and the SCN are capable of interacting functionally under RF conditions, the genetic basis of these interactions remains to be defined. In this study, using dexras1-deficient (dexras1(-/-)) mice, the authors examined whether Dexras1, a modulator of multiple inputs to the SCN, plays a role in regulating the effects of RF on activity rhythms and gene expression in the SCN. Daytime RF under 12L:12D or constant darkness (DD) resulted in potentiated (but less stable) FAA expression in dexras1(-/-) mice compared with wild-type (WT) controls. Under these conditions, the magnitude and phase of the SCN-driven activity component were greatly perturbed in the mutants. Restoration to ad libitum (AL) feeding revealed a stable phase displacement of the SCN-driven activity component of dexras1(-/-) mice by ~2 h in advance of the expected time. RF in the late night/early morning induced a long-lasting increase in the period of the SCN-driven activity component in the mutants but not the WT. At the molecular level, daytime RF advanced the rhythm of PER1, PER2, and pERK expression in the mutant SCN without having any effect in the WT. Collectively, these results indicate that the absence of Dexras1 sensitizes the SCN to perturbations resulting from restricted feeding.


Assuntos
Criação de Animais Domésticos , Relógios Circadianos/fisiologia , Núcleo Supraquiasmático/fisiologia , Proteínas ras/metabolismo , Animais , Regulação da Expressão Gênica/fisiologia , Masculino , Camundongos , Camundongos Knockout , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Fatores de Tempo , Proteínas ras/genética
9.
J Neurosci ; 32(14): 4867-77, 2012 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-22492043

RESUMO

Activation of the MAPK/ERK signaling cascade in the suprachiasmatic nucleus (SCN) is a key event that couples light to circadian clock entrainment. However, we do not fully understand the mechanisms that shape the properties of MAPK/ERK signaling in the SCN, and how these mechanisms may influence overt circadian rhythms. Here we show that Raf kinase inhibitor protein (RKIP) controls the kinetics of light-induced MAPK/ERK activity in the SCN and photic entrainment of behavioral rhythms. Light triggers robust phosphorylation of RKIP in the murine SCN and dissociation of RKIP and c-Raf. Overexpression of a nonphosphorylatable form of RKIP in the SCN of transgenic mice blocks light-induced ERK1/2 activation in the SCN and severely dampens light-induced phase delays in behavioral rhythms. Conversely, in RKIP knock-out (RKIP(-/-)) mice, light-induced ERK1/2 activity in the SCN is prolonged in the early and late subjective night, resulting in augmentation of the phase-delaying and -advancing effects of light. Reentrainment to an advancing light cycle was also accelerated in RKIP(-/-) mice. In relation to the molecular clockwork, genetic deletion of RKIP potentiated light-evoked PER1 and PER2 protein expression in the SCN in the early night. Additionally, RKIP(-/-) mice displayed enhanced transcriptional activation of mPeriod1 and the immediate early gene c-Fos in the SCN in response to a phase-delaying light pulse. Collectively, our data reveal an important role of RKIP in the regulation of MAPK/ERK signaling in the SCN and photic entrainment of the SCN clock.


Assuntos
Relógios Circadianos/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Proteína de Ligação a Fosfatidiletanolamina/fisiologia , Estimulação Luminosa/métodos , Núcleo Supraquiasmático/fisiologia , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Dados de Sequência Molecular , Núcleo Supraquiasmático/enzimologia
10.
J Mol Cell Cardiol ; 52(5): 1191-202, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22366713

RESUMO

Calcific aortic valve disease (CAVD) is a disorder related to progressive mineralization of valvular tissue that is a leading cause of heart disease. Thus far, there is no medical treatment to prevent the mineralization of aortic valves. It is generally thought that pathologic mineralization is linked to apoptosis of vascular cells. However, the role of apoptosis during mineralization as well as the survival signals for valvular interstitial cells (VICs), the main cellular component of aortic valves, remains to be identified. Here, through several lines of evidence, we show that bioavailability of extracellular ATP is a signal which determines survival or apoptosis of VICs and, in doing so, plays a major role in the development of CAVD. Specifically, in CAVD and in VIC cultures undergoing mineralization, we found a high level of the ectonucleotidase ENPP1. In addition, a genetic polymorphism in the intron 9 of the ENPP1 gene was associated with CAVD in a case-control cohort as well as with mRNA expression levels of ENPP1 in aortic valves. A high level of ENPP1 in CAVD promoted apoptosis-mediated mineralization of VICs by depleting the extracellular pool of ATP. We then documented that release of ATP by VICs promoted cell survival via the P2Y(2) receptor and the PI3K/Akt signaling pathway. Hence, our results show that level of ENPP1 modulates extracellular concentration of ATP, which is an important survival signal for VICs. These findings may help to develop novel pharmacological treatment for CAVD.


Assuntos
Trifosfato de Adenosina/fisiologia , Valva Aórtica/patologia , Calcinose/metabolismo , Cardiomiopatias/metabolismo , Células Epiteliais/metabolismo , Diester Fosfórico Hidrolases/genética , Pirofosfatases/genética , Trifosfato de Adenosina/metabolismo , Valva Aórtica/metabolismo , Apoptose , Calcinose/patologia , Cardiomiopatias/patologia , Estudos de Casos e Controles , Células Cultivadas , Perfilação da Expressão Gênica , Estudos de Associação Genética , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Fosfatos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Diester Fosfórico Hidrolases/metabolismo , Polimorfismo de Nucleotídeo Único , Pirofosfatases/metabolismo , Receptores Purinérgicos P2X/genética , Receptores Purinérgicos P2X/metabolismo , Receptores Purinérgicos P2Y/genética , Receptores Purinérgicos P2Y/metabolismo , Transdução de Sinais , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/metabolismo , Análise Serial de Tecidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA