Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurol Neuromedicine ; 3(1): 21-25, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29682634

RESUMO

Daily rhythms in behavior and physiology are coordinated by an endogenous clock located in the suprachiasmatic nucleus (SCN) of the hypothalamus. This central pacemaker also relays day length information to allow for seasonal adaptation, a process for which melatonin signaling is essential. How the SCN encodes day length is not fully understood. MicroRNAs (miRNAs) are small, non-coding RNAs that regulate gene expression by directing target mRNAs for degradation or translational repression. The miR-132/212 cluster plays a key role in facilitating neuronal plasticity, and miR-132 has been shown previously to modulate resetting of the central clock. A recent study from our group showed that miR-132/212 in mice is required for optimal adaptation to seasons and non-24-hour light/dark cycles through regulation of its target gene, methyl CpG-binding protein (MeCP2), in the SCN and dendritic spine density of SCN neurons. Furthermore, in the seasonal rodent Mesocricetus auratus (Syrian hamster), adaptation to short photoperiods is accompanied by structural plasticity in the SCN independently of melatonin signaling, thus further supporting a key role for SCN structural and, in turn, functional plasticity in the coding of day length. In this commentary, we discuss our recent findings in context of what is known about day length encoding by the SCN, and propose future directions.

2.
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
3.
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
4.
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
5.
Proc Natl Acad Sci U S A ; 114(5): 1207-1212, 2017 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-28096421

RESUMO

Neuroendocrine mechanisms underlying social inhibition of puberty are not well understood. Here, we use a model exhibiting the most profound case of pubertal suppression among mammals to explore a role for RFamide-related peptide-3 [RFRP-3; mammalian ortholog to gonadotropin-inhibitory hormone (GnIH)] in neuroendocrine control of reproductive development. Naked mole rats (NMRs) live in sizable colonies where breeding is monopolized by two to four dominant animals, and no other members exhibit signs of puberty throughout their lives unless they are removed from the colony. Because of its inhibitory action on the reproductive axis in other vertebrates, we investigated the role of RFRP-3 in social reproductive suppression in NMRs. We report that RFRP-3 immunofluorescence expression patterns and RFRP-3/GnRH cross-talk are largely conserved in the NMR brain, with the exception of the unique presence of RFRP-3 cell bodies in the arcuate nucleus (Arc). Immunofluorescence comparisons revealed that central expression of RFRP-3 is altered by reproductive status, with RFRP-3 immunoreactivity enhanced in the paraventricular nucleus, dorsomedial nucleus, and Arc of reproductively quiescent NMRs. We further observed that exogenous RFRP-3 suppresses gonadal steroidogenesis and mating behavior in NMRs given the opportunity to undergo puberty. Together, our findings establish a role for RFRP-3 in preserving reproductive immaturity, and challenge the view that stimulatory peptides are the ultimate gatekeepers of puberty.


Assuntos
Sistema Límbico/metabolismo , Ratos-Toupeira/fisiologia , Neuropeptídeos/fisiologia , Maturidade Sexual/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Hipotalâmico Dorsomedial/metabolismo , Feminino , Hormônio Liberador de Gonadotropina/fisiologia , Injeções Intraventriculares , Kisspeptinas/metabolismo , Masculino , Neuropeptídeos/farmacologia , Ovário/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Progesterona/biossíntese , Progesterona/sangue , Comportamento Sexual Animal/efeitos dos fármacos , Comportamento Sexual Animal/fisiologia , Maturidade Sexual/efeitos dos fármacos , Isolamento Social , Testículo/metabolismo , Testosterona/biossíntese , Testosterona/sangue
6.
Receptors Clin Investig ; 3(1)2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-27088110

RESUMO

G protein-coupled receptor kinases (GRKs) are a family of serine/threonine protein kinases that terminate G protein-coupled receptor (GPCR) signaling by phosphorylating the receptor and inducing its internalization. In addition to their canonical function, some GRKs can phosphorylate non-GPCR substrates and regulate GPCR signaling in a kinase-independent manner. GPCRs are abundantly expressed in the suprachiasmatic nucleus (SCN), a structure in the mammalian brain that serves as the central circadian pacemaker. Various facets of circadian timekeeping are under the influence of GPCR signaling, and thus are potential targets for GRK regulation. Despite this, little attention has been given to the role of GRKs in circadian rhythms. In this research highlight, we discuss our latest findings on the functional involvement of GRK2 in mammalian circadian timekeeping in the SCN. Using grk2 knockout mice, we demonstrate that GRK2 is critical for maintaining proper clock speed and ensuring that the clock is appropriately synchronized to environmental light cycles. Although grk2 deficiency expectedly alters the expression of a key GPCR in the SCN, our study also reveals that GRK2 has a more direct function that touches the heart of the circadian clock.

7.
Cell Rep ; 12(8): 1272-88, 2015 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-26279567

RESUMO

The pacemaker properties of the suprachiasmatic nucleus (SCN) circadian clock are shaped by mechanisms that influence the expression and behavior of clock proteins. Here, we reveal that G-protein-coupled receptor kinase 2 (GRK2) modulates the period, amplitude, and entrainment characteristics of the SCN. Grk2-deficient mice show phase-dependent alterations in light-induced entrainment, slower recovery from jetlag, and longer behavioral rhythms. Grk2 ablation perturbs intrinsic rhythmic properties of the SCN, increasing amplitude and decreasing period. At the cellular level, GRK2 suppresses the transcription of the mPeriod1 gene and the trafficking of PERIOD1 and PERIOD2 proteins to the nucleus. Moreover, GRK2 can physically interact with PERIOD1/2 and promote PERIOD2 phosphorylation at Ser545, effects that may underlie its ability to regulate PERIOD1/2 trafficking. Together, our findings identify GRK2 as an important modulator of circadian clock speed, amplitude, and entrainment by controlling PERIOD at the transcriptional and post-translational levels.


Assuntos
Núcleo Celular/metabolismo , Relógios Circadianos/genética , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Proteínas Circadianas Period/metabolismo , Processamento de Proteína Pós-Traducional , Transporte Ativo do Núcleo Celular , Animais , Linhagem Celular , Células Cultivadas , Quinase 2 de Receptor Acoplado a Proteína G/genética , Masculino , Camundongos , Proteínas Circadianas Period/genética , Fosforilação , Ligação Proteica
8.
J Neurosci ; 35(16): 6444-51, 2015 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-25904795

RESUMO

Polyglutamine (polyQ) expansion of the androgen receptor (AR) causes Kennedy's disease/spinobulbar muscular atrophy (KD/SBMA) through poorly defined cellular mechanisms. Although KD/SBMA has been thought of as a motor neuron disease, recent evidence indicates a key role for skeletal muscle. To resolve which early aspects of the disease can be caused by neurogenic or myogenic mechanisms, we made use of the tet-On and Cre-loxP genetic systems to selectively and acutely express polyQ AR in either motor neurons (NeuroAR) or myocytes (MyoAR) of transgenic mice. After 4 weeks of transgene induction in adulthood, deficits in gross motor function were seen in NeuroAR mice, but not MyoAR mice. Conversely, reduced size of fast glycolytic fibers and alterations in expression of candidate genes were observed only in MyoAR mice. Both NeuroAR and MyoAR mice exhibited reduced oxidative capacity in skeletal muscles, as well as a shift in fast fibers from oxidative to glycolytic. Markers of oxidative stress were increased in the muscle of NeuroAR mice and were reduced in motor neurons of both NeuroAR and MyoAR mice. Despite secondary pathology in skeletal muscle and behavioral deficits, no pathological signs were observed in motor neurons of NeuroAR mice, possibly due to relatively low levels of polyQ AR expression. These results indicate that polyQ AR in motor neurons can produce secondary pathology in muscle. Results also support both neurogenic and myogenic contributions of polyQ AR to several acute aspects of pathology and provide further evidence for disordered cellular respiration in KD/SBMA skeletal muscle.


Assuntos
Modelos Animais de Doenças , Neurônios Motores/patologia , Células Musculares/patologia , Transtornos Musculares Atróficos/patologia , Animais , Expressão Gênica , Masculino , Camundongos , Camundongos Transgênicos , Neurônios Motores/metabolismo , Destreza Motora , Células Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Transtornos Musculares Atróficos/genética , Estresse Oxidativo/genética , Receptores Androgênicos/genética
9.
PLoS Genet ; 10(10): e1004695, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25330117

RESUMO

The suprachiasmatic nucleus (SCN) acts as the central clock to coordinate circadian oscillations in mammalian behavior, physiology and gene expression. Despite our knowledge of the circadian transcriptome of the SCN, how it impacts genome-wide protein expression is not well understood. Here, we interrogated the murine SCN proteome across the circadian cycle using SILAC-based quantitative mass spectrometry. Of the 2112 proteins that were accurately quantified, 20% (421 proteins) displayed a time-of-day-dependent expression profile. Within this time-of-day proteome, 11% (48 proteins) were further defined as circadian based on a sinusoidal expression pattern with a ∼24 h period. Nine circadianly expressed proteins exhibited 24 h rhythms at the transcript level, with an average time lag that exceeded 8 h. A substantial proportion of the time-of-day proteome exhibited abrupt fluctuations at the anticipated light-to-dark and dark-to-light transitions, and was enriched for proteins involved in several key biological pathways, most notably, mitochondrial oxidative phosphorylation. Additionally, predicted targets of miR-133ab were enriched in specific hierarchical clusters and were inversely correlated with miR133ab expression in the SCN. These insights into the proteomic landscape of the SCN will facilitate a more integrative understanding of cellular control within the SCN clock.


Assuntos
Ritmo Circadiano/fisiologia , Proteoma/metabolismo , Núcleo Supraquiasmático/metabolismo , Animais , Regulação da Expressão Gênica , Luz , Masculino , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Proteínas Mitocondriais/metabolismo , Fosforilação Oxidativa , Mapas de Interação de Proteínas , Proteoma/análise , Proteômica/instrumentação , Proteômica/métodos , Transcriptoma
10.
PLoS One ; 9(8): e103103, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25084275

RESUMO

Circadian rhythms of behavior and physiology are driven by the biological clock that operates endogenously but can also be entrained to the light-dark cycle of the environment. In mammals, the master circadian pacemaker is located in the suprachiasmatic nucleus (SCN), which is composed of individual cellular oscillators that are driven by a set of core clock genes interacting in transcriptional/translational feedback loops. Light signals can trigger molecular events in the SCN that ultimately impact on the phase of expression of core clock genes to reset the master pacemaker. While transcriptional regulation has received much attention in the field of circadian biology in the past, other mechanisms including targeted protein degradation likely contribute to the clock timing and entrainment process. In the present study, proteome-wide screens of the murine SCN led to the identification of ubiquitin protein ligase E3 component N-recognin 4 (UBR4), a novel E3 ubiquitin ligase component of the N-end rule pathway, as a time-of-day-dependent and light-inducible protein. The spatial and temporal expression pattern of UBR4 in the SCN was subsequently characterized by immunofluorescence microscopy. UBR4 is expressed across the entire rostrocaudal extent of the SCN in a time-of-day-dependent fashion. UBR4 is localized exclusively to arginine vasopressin (AVP)-expressing neurons of the SCN shell. Upon photic stimulation in the early subjective night, the number of UBR4-expressing cells within the SCN increases. This study is the first to identify a novel E3 ubiquitin ligase component, UBR4, in the murine SCN and to implicate the N-end rule degradation pathway as a potential player in regulating core clock mechanisms and photic entrainment.


Assuntos
Relógios Circadianos/genética , Regulação da Expressão Gênica , Luz , Proteínas Associadas aos Microtúbulos/genética , Núcleo Supraquiasmático/metabolismo , Ubiquitina-Proteína Ligases/genética , Animais , Proteínas de Ligação a Calmodulina , Linhagem Celular , Ritmo Circadiano/genética , Masculino , Camundongos , Proteínas Associadas aos Microtúbulos/química , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/metabolismo , Peptídeos/genética , Peptídeos/metabolismo , Fotoperíodo , Ligação Proteica , Transporte Proteico , Proteoma , Proteômica/métodos , Reprodutibilidade dos Testes , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo
11.
Crit Rev Oncog ; 19(6): 505-16, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25597360

RESUMO

In 1984, a cytosolic protein was isolated from bovine brain and coined phosphatidylethanolamine binding protein (PEBP) to describe its phospholipid-binding potential. Its cellular function remained elusive for more than a decade until it was discovered that PEBP had the ability to suppress the Raf1-mitogen activated protein kinase (MAPK) pathway, earning it the new name of Raf1 kinase inhibitory protein (RKIP). This milestone discovery has paved the way for numerous studies that have now extended the reach of RKIP's function to other signaling cascades, within the context of various physiological and pathophysiological systems. This review will summarize our current knowledge of the neurophysiological roles of RKIP in the mammalian brain, including its function in the circadian clock and synaptic plasticity. It will also discuss evidence for an involvement of RKIP and its derived neuropeptide, hippocampal cholinergic neurostimulating peptide (HCNP), in neural development and differentiation. Implications in certain pathologies such as Alzheimer's disease and brain cancer will be highlighted. By chronicling the diverse functions of RKIP in the brain, we hope that this review will serve as a timely resource that ignites future studies on this versatile, multifaceted protein in the nervous system.


Assuntos
Encéfalo/metabolismo , Mamíferos , Proteína de Ligação a Fosfatidiletanolamina/fisiologia , Animais , Bovinos , Expressão Gênica , Humanos , Mamíferos/fisiologia , Proteína de Ligação a Fosfatidiletanolamina/química , Conformação Proteica , Distribuição Tecidual
12.
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
13.
J Mol Biol ; 425(19): 3609-24, 2013 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-23142644

RESUMO

Evolved under the selective pressures of a 24-h world, circadian timekeeping mechanisms are present in virtually all living organisms to coordinate daily rhythms in physiology and behavior. Until recently, the circadian clock was modeled as simple, interlocked transcription-translation feedback loops driving rhythms in gene expression of a handful of core clock genes. However, it has become evident that circadian clock regulation is immensely more complex than once thought and involves posttranscriptional, translational and posttranslational mechanisms. In particular, there has been a growing awareness of the vital role played by microRNAs (miRNAs) in regulating various aspects of circadian clock function. In this review, we will summarize our current knowledge of miRNA-dependent regulation of the circadian timing system in multiple organisms, including flies, mammals and higher plants. We will also discuss future perspectives for research on the role of miRNAs and noncoding RNAs in circadian regulation of health and disease.


Assuntos
Relógios Circadianos/genética , MicroRNAs/genética , Animais , Ritmo Circadiano/genética , Drosophila/fisiologia , Expressão Gênica , Humanos , MicroRNAs/metabolismo , Plantas/genética , RNA não Traduzido/genética , RNA não Traduzido/metabolismo , Transcrição Gênica
14.
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
15.
Nature ; 480(7376): 209-14, 2011 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-22080954

RESUMO

Murine epidermal stem cells undergo alternate cycles of dormancy and activation, fuelling tissue renewal. However, only a subset of stem cells becomes active during each round of morphogenesis, indicating that stem cells coexist in heterogeneous responsive states. Using a circadian-clock reporter-mouse model, here we show that the dormant hair-follicle stem cell niche contains coexisting populations of cells at opposite phases of the clock, which are differentially predisposed to respond to homeostatic cues. The core clock protein Bmal1 modulates the expression of stem cell regulatory genes in an oscillatory manner, to create populations that are either predisposed, or less prone, to activation. Disrupting this clock equilibrium, through deletion of Bmal1 (also known as Arntl) or Per1/2, resulted in a progressive accumulation or depletion of dormant stem cells, respectively. Stem cell arrhythmia also led to premature epidermal ageing, and a reduction in the development of squamous tumours. Our results indicate that the circadian clock fine-tunes the temporal behaviour of epidermal stem cells, and that its perturbation affects homeostasis and the predisposition to tumorigenesis.


Assuntos
Relógios Circadianos/fisiologia , Ritmo Circadiano/fisiologia , Folículo Piloso/citologia , Células-Tronco/citologia , Fatores de Transcrição ARNTL/deficiência , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Animais , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Adesão Celular/genética , Ciclo Celular/genética , Células Cultivadas , Senescência Celular , Relógios Circadianos/genética , Ritmo Circadiano/genética , Sinais (Psicologia) , Feminino , Regulação da Expressão Gênica/genética , Homeostase/genética , Homeostase/fisiologia , Masculino , Camundongos , Camundongos Knockout , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Nicho de Células-Tronco , Células-Tronco/metabolismo , Fator de Crescimento Transformador beta/genética , Via de Sinalização Wnt/genética
16.
Chronobiol Int ; 28(7): 553-62, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21834641

RESUMO

Loss of Dexras1 in gene-targeted mice impairs circadian entrainment to light cycles and produces complex changes to phase-dependent resetting responses (phase shifts) to light. The authors now describe greatly enhanced and phase-specific nonphotic responses induced by arousal in dexras1(-/-) mice. In constant conditions, mutant mice exhibited significant arousal-induced phase shifts throughout the subjective day. Unusual phase advances in the late subjective night were also produced when arousal has little effect in mice. Bilateral lesions of the intergeniculate leaflet (IGL) completely eliminated both the nonphotic as well as the light-induced phase shifts of circadian locomotor rhythms during the subjective day, but had no effect on nighttime phase shifts. The expression of FOS-like protein in the suprachiasmatic nucleus (SCN) was not affected by either photic or nonphotic stimulation in the subjective day in either genotype. Therefore, the loss of Dexras1 (1) enhances nonphotic phase shifts in a phase-dependent manner, and (2) demonstrates that the IGL in mice is a primary mediator of circadian phase-resetting responses to both photic and nonphotic events during the subjective day, but plays a different functional role in the subjective night. Furthermore, (3) the change in FOS level does not appear to be a critical step in the entrainment pathways for either light or arousal during the subjective day. The cumulative evidence suggests that Dexras1 regulates multiple photic and nonphotic signal-transduction pathways, thereby playing an essential role modulating species-specific characteristics of circadian entrainment.


Assuntos
Relógios Biológicos/fisiologia , Ritmo Circadiano/fisiologia , Corpos Geniculados/anatomia & histologia , Corpos Geniculados/fisiologia , Proteínas ras/genética , Proteínas ras/metabolismo , Animais , Nível de Alerta , Comportamento Animal/fisiologia , Marcação de Genes , Corpos Geniculados/patologia , Luz , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/fisiologia , Neuropeptídeo Y/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Núcleo Supraquiasmático/citologia , Núcleo Supraquiasmático/fisiologia
17.
Mol Cell Proteomics ; 10(11): M110.007252, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21859948

RESUMO

In mammals, the suprachiasmatic nucleus (SCN) is the central circadian pacemaker that governs rhythmic fluctuations in behavior and physiology in a 24-hr cycle and synchronizes them to the external environment by daily resetting in response to light. The bilateral SCN is comprised of a mere ~20,000 neurons serving as cellular oscillators, a fact that has, until now, hindered the systematic study of the SCN on a global proteome level. Here we developed a fully automated and integrated proteomics platform, termed AutoProteome system, for an in-depth analysis of the light-responsive proteome of the murine SCN. All requisite steps for a large-scale proteomic study, including preconcentration, buffer exchanging, reduction, alkylation, digestion and online two-dimensional liquid chromatography-tandem MS analysis, are performed automatically on a standard liquid chromatography-MS system. As low as 2 ng of model protein bovine serum albumin and up to 20 µg and 200 µg of SCN proteins can be readily processed and analyzed by this system. From the SCN tissue of a single mouse, we were able to confidently identify 2131 proteins, of which 387 were light-regulated based on a spectral counts quantification approach. Bioinformatics analysis of the light-inducible proteins reveals their diverse distribution in different canonical pathways and their heavy connection in 19 protein interaction networks. The AutoProteome system identified vasopressin-neurophysin 2-copeptin and casein kinase 1 delta, both of which had been previously implicated in clock timing processes, as light-inducible proteins in the SCN. Ras-specific guanine nucleotide-releasing factor 1, ubiquitin protein ligase E3A, and X-linked ubiquitin specific protease 9, none of which had previously been implicated in SCN clock timing processes, were also identified in this study as light-inducible proteins. The AutoProteome system opens a new avenue to systematically explore the proteome-wide events that occur in the SCN, either in response to light or other stimuli, or as a consequence of its intrinsic pacemaker capacity.


Assuntos
Automação Laboratorial , Relógios Circadianos , Luz , Proteoma/metabolismo , Núcleo Supraquiasmático/metabolismo , Animais , Bovinos , Cromatografia Líquida/normas , Expressão Gênica/efeitos da radiação , Masculino , Redes e Vias Metabólicas , Camundongos , Camundongos Endogâmicos C57BL , Mapas de Interação de Proteínas , Proteoma/genética , Proteoma/isolamento & purificação , Soroalbumina Bovina/normas , Núcleo Supraquiasmático/efeitos da radiação , Espectrometria de Massas em Tandem/normas
18.
Hum Mol Genet ; 20(4): 731-51, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21118894

RESUMO

Mammalian circadian rhythms are synchronized to the external time by daily resetting of the suprachiasmatic nucleus (SCN) in response to light. As the master circadian pacemaker, the SCN coordinates the timing of diverse cellular oscillators in multiple tissues. Aberrant regulation of clock timing is linked to numerous human conditions, including cancer, cardiovascular disease, obesity, various neurological disorders and the hereditary disorder familial advanced sleep phase syndrome. Additionally, mechanisms that underlie clock resetting factor into the sleep and physiological disturbances experienced by night-shift workers and travelers with jet lag. The Ca(2+)/cAMP response element-binding protein-regulated microRNA, miR-132, is induced by light within the SCN and attenuates its capacity to reset, or entrain, the clock. However, the specific targets that are regulated by miR-132 and underlie its effects on clock entrainment remained elusive until now. Here, we show that genes involved in chromatin remodeling (Mecp2, Ep300, Jarid1a) and translational control (Btg2, Paip2a) are direct targets of miR-132 in the mouse SCN. Coordinated regulation of these targets underlies miR-132-dependent modulation of Period gene expression and clock entrainment: the mPer1 and mPer2 promoters are bound to and transcriptionally activated by MeCP2, whereas PAIP2A and BTG2 suppress the translation of the PERIOD proteins by enhancing mRNA decay. We propose that miR-132 is selectively enriched for chromatin- and translation-associated target genes and is an orchestrator of chromatin remodeling and protein translation within the SCN clock, thereby fine-tuning clock entrainment. These findings will further our understanding of mechanisms governing clock entrainment and its involvement in human diseases.


Assuntos
Montagem e Desmontagem da Cromatina/efeitos dos fármacos , Montagem e Desmontagem da Cromatina/genética , Ritmo Circadiano/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Animais , Biologia Computacional , Proteínas de Ligação a DNA , Proteína p300 Associada a E1A/metabolismo , Regulação da Expressão Gênica , Células HEK293 , Humanos , Proteínas Imediatamente Precoces/metabolismo , Histona Desmetilases com o Domínio Jumonji , Luz , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células NIH 3T3 , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Estabilidade de RNA , Proteína 2 de Ligação ao Retinoblastoma/metabolismo , Transdução de Sinais , Núcleo Supraquiasmático/metabolismo , Proteínas Supressoras de Tumor/metabolismo
19.
J Biol Rhythms ; 26(6): 561-4, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22215614

RESUMO

Recent demonstration of the histone acetyltransferase activity of the Clock gene greatly expanded the regulatory role of circadian clocks in gene transcription. Clock and its partner Bmal1 are responsible for the generation of circadian oscillations that are synchronized (entrained) to the external light cycle. Entraining light often produces long-lasting changes in the endogenous period called aftereffects. Aftereffects are light-dependent alterations in the speed of free-running rhythms that persist for several weeks upon termination of light exposure. How light causes such long-lasting changes is unknown. However, the persistent nature of circadian aftereffects in conjunction with the long-term effects of epigenetic modifications on development and various aspects of brain physiology prompted us to hypothesize that the histone acetyltransferase CLOCK was required for circadian aftereffects. The authors exposed Clock knockout mice to 25-hour light cycles and report that these mice retain the ability to display circadian aftereffects, indicating that Clock is dispensable for this form of circadian plasticity.


Assuntos
Acetiltransferases/metabolismo , Relógios Biológicos/fisiologia , Proteínas CLOCK/metabolismo , Ritmo Circadiano/fisiologia , Acetiltransferases/genética , Animais , Comportamento Animal/fisiologia , Proteínas CLOCK/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/fisiologia , Núcleo Supraquiasmático/citologia , Núcleo Supraquiasmático/metabolismo
20.
Hum Mol Genet ; 18(16): 3110-24, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19477955

RESUMO

The suprachiasmatic nuclei (SCN) of the mammalian hypothalamus function as the master circadian clock, coordinating the timing of diverse cell populations and organ systems. Dysregulation of clock timing is linked to a broad range of human conditions, including obesity, cardiovascular disease and a wide spectrum of neurological disorders. Aberrant regulation of expression of the PERIOD genes has been associated with improper cell division and human cancers, while the autosomal dominant disorder familial advanced sleep phase syndrome has been mapped to a single missense mutation within the critical clock gene hPERIOD2. An essential tool to begin to dissect the inherent molecular timing process is the clock gene reporter. Here, we functionally characterize two new mouse transgenic clock reporters, mPeriod1-Venus and mPeriod2-DsRED. Venus and DsRED are fluorescent proteins that can be used to monitor transcription in individual cells in real-time. Imaging of the SCN revealed oscillations, as well as light inducibility, in Venus and DsRED expression. Rhythmic Venus and DsRED expression was observed in distinct SCN cell populations, suggesting the existence of discrete cellular SCN clocks. Outside of the SCN, mPeriod1-Venus expression was broadly expressed in neuronal and non-neuronal populations. Conversely, mPeriod2-DsRED was expressed in glial populations and progenitor cells of the dentate gyrus; limited expression was detected in neurons. This distinct expression pattern of the two reporters reveals that the central nervous system possesses mechanistically distinct subpopulations of neuronal and non-neuronal cellular clocks. These novel mouse models will facilitate our understanding of clock timing and its role in human diseases.


Assuntos
Encéfalo/metabolismo , Proteínas de Ciclo Celular/genética , Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neuroglia/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Animais , Encéfalo/citologia , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Ritmo Circadiano/efeitos da radiação , Expressão Gênica/efeitos da radiação , Genes Reporter , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Luz , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Nucleares/metabolismo , Proteínas Circadianas Period , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...