Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Cell ; 157(3): 689-701, 2014 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-24766812

RESUMO

Though much is known about the cellular and molecular components of the circadian clock, output pathways that couple clock cells to overt behaviors have not been identified. We conducted a screen for circadian-relevant neurons in the Drosophila brain and report here that cells of the pars intercerebralis (PI), a functional homolog of the mammalian hypothalamus, comprise an important component of the circadian output pathway for rest:activity rhythms. GFP reconstitution across synaptic partners (GRASP) analysis demonstrates that PI cells are connected to the clock through a polysynaptic circuit extending from pacemaker cells to PI neurons. Molecular profiling of relevant PI cells identified the corticotropin-releasing factor (CRF) homolog, DH44, as a circadian output molecule that is specifically expressed by PI neurons and is required for normal rest:activity rhythms. Notably, selective activation or ablation of just six DH44+ PI cells causes arrhythmicity. These findings delineate a circuit through which clock cells can modulate locomotor rhythms.


Assuntos
Relógios Circadianos , Drosophila/fisiologia , Neurônios/fisiologia , Animais , Animais Geneticamente Modificados , Encéfalo/citologia , Encéfalo/fisiologia , Ritmo Circadiano , Drosophila/citologia , Neurônios/citologia , Análise de Célula Única , Transcriptoma
2.
PLoS Genet ; 9(9): e1003749, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24086144

RESUMO

Circadian rhythms in Drosophila rely on cyclic regulation of the period (per) and timeless (tim) clock genes. The molecular cycle requires rhythmic phosphorylation of PER and TIM proteins, which is mediated by several kinases and phosphatases such as Protein Phosphatase-2A (PP2A) and Protein Phosphatase-1 (PP1). Here, we used mass spectrometry to identify 35 "phospho-occupied" serine/threonine residues within PER, 24 of which are specifically regulated by PP1/PP2A. We found that cell culture assays were not good predictors of protein function in flies and so we generated per transgenes carrying phosphorylation site mutations and tested for rescue of the per(01) arrhythmic phenotype. Surprisingly, most transgenes restore wild type rhythms despite carrying mutations in several phosphorylation sites. One particular transgene, in which T610 and S613 are mutated to alanine, restores daily rhythmicity, but dramatically lengthens the period to ~ 30 hrs. Interestingly, the single S613A mutation extends the period by 2-3 hours, while the single T610A mutation has a minimal effect, suggesting these phospho-residues cooperate to control period length. Conservation of S613 from flies to humans suggests that it possesses a critical clock function, and mutational analysis of residues surrounding T610/S613 implicates the entire region in determining circadian period. Biochemical and immunohistochemical data indicate defects in overall phosphorylation and altered timely degradation of PER carrying the double or single S613A mutation(s). The PER-T610A/S613A mutant also alters CLK phosphorylation and CLK-mediated output. Lastly, we show that a mutation at a previously identified site, S596, is largely epistatic to S613A, suggesting that S613 negatively regulates phosphorylation at S596. Together these data establish functional significance for a new domain of PER, demonstrate that cooperativity between phosphorylation sites maintains PER function, and support a model in which specific phosphorylated regions regulate others to control circadian period.


Assuntos
Ritmo Circadiano/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteínas Circadianas Period/genética , Fosforilação/genética , Animais , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/fisiologia , Humanos , Mutação , Proteínas Circadianas Period/fisiologia , Fenótipo , Proteína Fosfatase 1/genética , Proteína Fosfatase 1/metabolismo , Proteína Fosfatase 2/genética , Proteína Fosfatase 2/metabolismo
3.
Cells ; 13(14)2024 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-39056774

RESUMO

Studies on numerous species have demonstrated strikingly conserved mechanisms that determine the aging process, from yeasts to worms, flies, zebrafish, mice, and humans. The fruit fly Drosophila melanogaster is an excellent model organism for studying the biological basis of normal aging and etiology of age-related diseases. Since its inception in 1967, the Bloomington Drosophila Stock Center (BDSC) has grown into the largest collection of documented D. melanogaster strains (currently > 91,000). This paper aims to briefly review conserved mechanisms of aging and provides a guide to help users understand the organization of stock listings on the BDSC website and familiarize themselves with the search functions on BDSC and FlyBase, with an emphasis on using genes in conserved pathways as examples to find stocks for aging studies.


Assuntos
Envelhecimento , Drosophila melanogaster , Animais , Drosophila melanogaster/genética , Bases de Dados Genéticas , Humanos
4.
Sleep ; 2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38629438

RESUMO

The nuclear factor binding the κ light chain in B-cells (NFκB) is involved in a wide range of cellular processes including development, growth, innate immunity, and sleep. However, genetic studies of the role of specific NFκB transcription factors in sleep have been limited. Drosophila fruit flies carry three genes encoding NFκB transcription factors, Dorsal, Dorsal Immunity Factor (Dif), and Relish. We previously found that loss of the Relish gene from fat body suppressed daily nighttime sleep, and abolished infection-induced sleep. Here we show that Dif regulates daily sleep and recovery sleep following prolonged wakefulness. Mutants of Dif showed reduced daily sleep and suppressed recovery in response to sleep deprivation. Pan-neuronal knockdown of Dif strongly suppressed daily sleep, indicating that in contrast to Relish, Dif functions from the central nervous system to regulate sleep. Based on the unique expression pattern of a Dif- GAL4 driver, we hypothesized that its effects on sleep were mediated by the pars intercerebralis (PI). While RNAi knock-down of Dif in the PI reduced daily sleep, it had no effect on the recovery response to sleep deprivation. However, recovery sleep was suppressed when RNAi knock-down of Dif was distributed across a wider range of neurons. Induction of the nemuri (nur) antimicrobial peptide by sleep deprivation was reduced in Dif mutants and pan-neuronal over-expression of nur also suppressed the Dif mutant phenotype by significantly increasing sleep and reducing nighttime arousability. Together, these findings indicate that Dif functions from brain to target nemuri and to promote deep sleep.

5.
Acta Biochim Biophys Sin (Shanghai) ; 45(1): 61-9, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23257295

RESUMO

An endogenous circadian (∼24 h) clock regulates rhythmic processes of physiology, metabolism and behavior in most living organisms. While able to free-run under constant conditions, the circadian clock is coupled to day : night cycles to increase its amplitude and align the phase of circadian rhythms to the right time of the day. Disruptions of the circadian clock are correlated with brain dysfunctions, cardiovascular diseases and metabolic disorders. In this review, we focus on the interactions between the circadian clock and metabolism. We discuss recent findings on circadian clock regulation of feeding behavior and rhythmic expression of metabolic genes, and present evidence of metabolic input to the circadian clock. We emphasize how misalignment of circadian clocks within the body and with environmental cycles or daily schedules leads to the increasing prevalence of metabolic syndromes in modern society.


Assuntos
Relógios Circadianos , Metabolismo , Animais , Humanos
6.
J Neurosci ; 29(35): 10920-7, 2009 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-19726650

RESUMO

The Drosophila PAR domain protein 1 (Pdp1) gene encodes a transcription factor with multiple functions. One isoform, PDP1epsilon, was proposed to be an essential activator of the core clock gene, Clock (Clk). However, a central clock function for PDP1epsilon was recently disputed, and genetic analysis has been difficult due to developmental lethality of Pdp1-null mutants. Here we report the discovery of a mutation that specifically disrupts the Pdp1epsilon isoform. Homozygous Pdp1epsilon mutants are viable and exhibit arrhythmic circadian behavior in constant darkness and also in the presence of light:dark cycles. Importantly, the mutants show diminished expression of CLK and PERIOD (PER) in the central clock cells. In addition, expression of PDF (pigment-dispersing factor) is reduced in a subset of the central clock cells. Loss of Pdp1epsilon also alters the phosphorylation status of the CLK protein and disrupts cyclic expression of a per-luciferase reporter in peripheral clocks under free-running conditions. Transgenic expression of PDP1epsilon in clock neurons of Pdp1epsilon mutants can restore rhythmic circadian behavior. However, transgenic expression of CLK in these mutants rescues the expression of PER in the central clock, but fails to restore behavioral rhythms, suggesting that PDP1epsilon has effects outside the core molecular clock. Together, these data support a model in which PDP1epsilon functions in the central circadian oscillator as well as in the output pathway.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/fisiologia , Ritmo Circadiano/genética , Proteínas de Drosophila/fisiologia , Animais , Animais Geneticamente Modificados , Fatores de Transcrição de Zíper de Leucina Básica/genética , Ritmo Circadiano/fisiologia , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Masculino , Atividade Motora/genética , Atividade Motora/fisiologia , Mutação/genética , Mutação/fisiologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia
7.
Neuron ; 47(1): 115-27, 2005 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-15996552

RESUMO

Entrainment of the Drosophila circadian clock to light involves the light-induced degradation of the clock protein timeless (TIM). We show here that this entrainment mechanism is inhibited by serotonin, acting through the Drosophila serotonin receptor 1B (d5-HT1B). d5-HT1B is expressed in clock neurons, and alterations of its levels affect molecular and behavioral responses of the clock to light. Effects of d5-HT1B are synergistic with a mutation in the circadian photoreceptor cryptochrome (CRY) and are mediated by SHAGGY (SGG), Drosophila glycogen synthase kinase 3beta (GSK3beta), which phosphorylates TIM. Levels of serotonin are decreased in flies maintained in extended constant darkness, suggesting that modulation of the clock by serotonin may vary under different environmental conditions. These data identify a molecular connection between serotonin signaling and the central clock component TIM and suggest a homeostatic mechanism for the regulation of circadian photosensitivity in Drosophila.


Assuntos
Ritmo Circadiano/fisiologia , Drosophila/fisiologia , Serotonina/fisiologia , 5-Hidroxitriptofano/farmacologia , Animais , Animais Geneticamente Modificados , Química Encefálica , Ritmo Circadiano/efeitos dos fármacos , Ritmo Circadiano/genética , Citalopram/farmacologia , Fluoxetina/farmacologia , Quinase 3 da Glicogênio Sintase/fisiologia , Imuno-Histoquímica , Luz , Mutação/fisiologia , Fosforilação , Interferência de RNA , Receptor 5-HT1A de Serotonina/efeitos dos fármacos , Receptor 5-HT1B de Serotonina/efeitos dos fármacos , Serotonina/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia
8.
Genetics ; 178(3): 1147-55, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18385110

RESUMO

Circadian ( approximately 24 hr) rhythms of behavior and physiology are driven by molecular clocks that are endogenous to most organisms. The mechanisms underlying these clocks are remarkably conserved across evolution and typically consist of auto-regulatory loops in which specific proteins (clock proteins) rhythmically repress expression of their own genes. Such regulation maintains 24-hr cycles of RNA and protein expression. Despite the conservation of these mechanisms, however, questions are now being raised about the relevance of different molecular oscillations. Indeed, several studies have demonstrated that oscillations of some critical clock genes can be eliminated without loss of basic clock function. Here, we describe the multiple levels at which clock gene/protein expression and function can be rhythmically regulated-transcription, protein expression, post-translational modification, and localization-and speculate as to which aspect of this regulation is most critical. While the review is focused on Drosophila, we include some discussion of mammalian clocks to indicate the extent to which the questions concerning clock mechanisms are similar, regardless of the organism under study.


Assuntos
Relógios Biológicos/fisiologia , Ritmo Circadiano/fisiologia , Animais , Relógios Biológicos/efeitos da radiação , Ritmo Circadiano/efeitos da radiação , Retroalimentação Fisiológica/efeitos da radiação , Luz , Processamento de Proteína Pós-Traducional/efeitos da radiação
9.
PLoS One ; 14(6): e0217111, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31211778

RESUMO

The pathophysiology of refractive errors is poorly understood. Myopia (nearsightedness) in particular both blurs vision and predisposes the eye to many blinding diseases during adulthood. Based on past findings of diurnal variations in the dimensions of the eyes of humans and other vertebrates, altered diurnal rhythms of these ocular dimensions with experimentally induced myopia, and evolving evidence that ambient light exposures influence refractive development, we assessed whether disturbances in circadian signals might alter the refractive development of the eye. In mice, retinal-specific knockout of the clock gene Bmal1 induces myopia and elongates the vitreous chamber, the optical compartment separating the lens and the retina. These alterations simulate common ocular findings in clinical myopia. In Drosophila melanogaster, knockouts of the clock genes cycle or period lengthen the pseudocone, the optical component of the ommatidium that separates the facet lens from the photoreceptors. Disrupting circadian signaling thus alters optical development of the eye in widely separated species. We propose that mechanisms of myopia include circadian dysregulation, a frequent occurrence in modern societies where myopia also is both highly prevalent and increasing at alarming rates. Addressing circadian dysregulation may improve understanding of the pathogenesis of refractive errors and introduce novel therapeutic approaches to ameliorate myopia development in children.


Assuntos
Relógios Circadianos/genética , Olho/fisiopatologia , Animais , Ritmo Circadiano/genética , Drosophila melanogaster , Camundongos , Miopia/genética , Miopia/fisiopatologia , Retina/fisiopatologia
10.
Cell Rep ; 29(7): 1778-1788.e4, 2019 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-31722196

RESUMO

Drosophila Myc (dMyc) is highly conserved and functions as a transcription factor similar to mammalian Myc. We previously found that oncogenic Myc disrupts the molecular clock in cancer cells. Here, we demonstrate that misregulation of dMyc expression affects Drosophila circadian behavior. dMyc overexpression results in a high percentage of arrhythmic flies, concomitant with increases in the expression of clock genes cyc, tim, cry, and cwo. Conversely, flies with hypomorphic mutations in dMyc exhibit considerable arrhythmia, which can be rescued by loss of dMnt, a suppressor of dMyc activity. Metabolic profiling of fly heads revealed that loss of dMyc and its overexpression alter steady-state metabolite levels and have opposing effects on histidine, the histamine precursor, which is rescued in dMyc mutants by ablation of dMnt and could contribute to effects of dMyc on locomotor behavior. Our results demonstrate a role of dMyc in modulating Drosophila circadian clock, behavior, and metabolism.


Assuntos
Comportamento Animal , Ritmo Circadiano , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Fatores de Transcrição/genética
11.
Cell Rep ; 22(13): 3416-3426, 2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29590612

RESUMO

Loss of the Neurofibromatosis 1 (Nf1) protein, neurofibromin, in Drosophila disrupts circadian rhythms of locomotor activity without impairing central clock function, suggesting effects downstream of the clock. However, the relevant cellular mechanisms are not known. Leveraging the discovery of output circuits for locomotor rhythms, we dissected cellular actions of neurofibromin in recently identified substrates. Herein, we show that neurofibromin affects the levels and cycling of calcium in multiple circadian peptidergic neurons. A prominent site of action is the pars intercerebralis (PI), the fly equivalent of the hypothalamus, with cell-autonomous effects of Nf1 in PI cells that secrete DH44. Nf1 interacts genetically with peptide signaling to affect circadian behavior. We extended these studies to mammals to demonstrate that mouse astrocytes exhibit a 24-hr rhythm of calcium levels, which is also attenuated by lack of neurofibromin. These findings establish a conserved role for neurofibromin in intracellular signaling rhythms within the nervous system.


Assuntos
Ritmo Circadiano/genética , Proteínas de Drosophila/genética , Genes da Neurofibromatose 1 , Proteínas do Tecido Nervoso/genética , Proteínas Ativadoras de ras GTPase/genética , Animais , Animais Geneticamente Modificados , Linhagem Celular , Drosophila , Masculino , Neurofibromatose 1/genética
12.
Cell Metab ; 23(1): 143-54, 2016 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-26626459

RESUMO

Endogenous circadian clocks orchestrate several metabolic and signaling pathways that are known to modulate lifespan, suggesting clocks as potential targets for manipulation of metabolism and lifespan. We report here that the core circadian clock genes, timeless (tim) and period (per), are required for the metabolic and lifespan responses to DR in Drosophila. Consistent with the involvement of a circadian mechanism, DR enhances the amplitude of cycling of most circadian clock genes, including tim, in peripheral tissues. Mass-spectrometry-based lipidomic analysis suggests a role of tim in cycling of specific medium chain triglycerides under DR. Furthermore, overexpression of tim in peripheral tissues improves its oscillatory amplitude and extends lifespan under ad libitum conditions. Importantly, effects of tim on lifespan appear to be mediated through enhanced fat turnover. These findings identify a critical role for specific clock genes in modulating the effects of nutrient manipulation on fat metabolism and aging.


Assuntos
Proteínas CLOCK/metabolismo , Relógios Circadianos , Proteínas de Drosophila/metabolismo , Metabolismo dos Lipídeos , Longevidade , Animais , Proteínas CLOCK/genética , Restrição Calórica , Proteínas de Drosophila/genética , Drosophila melanogaster , Feminino , Expressão Gênica , Regulação da Expressão Gênica , Masculino , Transdução de Sinais
13.
BMC Neurosci ; 5: 3, 2004 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-15005796

RESUMO

BACKGROUND: SLOB binds to and modulates the activity of the Drosophila Slowpoke (dSlo) calcium activated potassium channel. Recent microarray analyses demonstrated circadian cycling of slob mRNA. RESULTS: We report the mRNA and protein expression pattern of slob in Drosophila heads. slob transcript is present in the photoreceptors, optic lobe, pars intercerebralis (PI) neurons and surrounding brain cortex. SLOB protein exhibits a similar distribution pattern, and we show that it cycles in Drosophila heads, in photoreceptor cells and in neurosecretory cells of the PI. The cycling of SLOB is altered in various clock gene mutants, and SLOB is expressed in ectopic locations in tim01 flies. We also demonstrate that SLOB no longer cycles in the PI neurons of Clkjrk flies, and that SLOB expression is reduced in the PI neurons of flies that lack pigment dispersing factor (PDF), a neuropeptide secreted by clock cells. CONCLUSIONS: These data are consistent with the idea that SLOB may participate in one or more circadian pathways in Drosophila.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Canais de Potássio/genética , Canais de Potássio/metabolismo , RNA Mensageiro/metabolismo , Animais , Proteínas CLOCK , Ritmo Circadiano/fisiologia , Gânglios dos Invertebrados/metabolismo , Regulação da Expressão Gênica , Hormônios de Invertebrado/genética , Hormônios de Invertebrado/metabolismo , Mutação , Neurônios/metabolismo , Lobo Óptico de Animais não Mamíferos/metabolismo , Periodicidade , Células Fotorreceptoras de Invertebrados/metabolismo , Fatores de Transcrição/genética
14.
Mol Cell Biol ; 34(14): 2682-94, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24820422

RESUMO

Casein kinase 1, known as DOUBLETIME (DBT) in Drosophila melanogaster, is a critical component of the circadian clock that phosphorylates and promotes degradation of the PERIOD (PER) protein. However, other functions of DBT in circadian regulation are not clear, in part because severe reduction of dbt causes preadult lethality. Here we report the molecular and behavioral phenotype of a viable dbt(EY02910) loss-of-function mutant. We found that DBT protein levels are dramatically reduced in adult dbt(EY02910) flies, and the majority of mutant flies display arrhythmic behavior, with a few showing weak, long-period (∼32 h) rhythms. Peak phosphorylation of PER is delayed, and both hyper- and hypophosphorylated forms of the PER and CLOCK proteins are present throughout the day. In addition, molecular oscillations of the circadian clock are dampened. In the central brain, PER and TIM expression is heterogeneous and decoupled in the canonical clock neurons of the dbt(EY02910) mutants. We also report an interaction between dbt and the signaling pathway involving pigment dispersing factor (PDF), a synchronizing peptide in the clock network. These data thus demonstrate that overall reduction of DBT causes long and arrhythmic behavior, and they reveal an unexpected role of DBT in promoting synchrony of the circadian clock network.


Assuntos
Caseína Quinase 1 épsilon/metabolismo , Relógios Circadianos , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiologia , Animais , Encéfalo/metabolismo , Proteínas CLOCK/metabolismo , Caseína Quinase 1 épsilon/genética , Ritmo Circadiano , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Regulação da Expressão Gênica , Mutação , Proteínas Circadianas Period/metabolismo , Fosforilação , Transdução de Sinais
15.
Nat Commun ; 5: 5697, 2014 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-25511299

RESUMO

Little is known about molecular links between circadian clocks and steroid hormone signalling, although both are important for normal physiology. Here we report a circadian function for a nuclear receptor, ecdysone-induced protein 75 (Eip75/E75), which we identified through a gain-of-function screen for circadian genes in Drosophila melanogaster. Overexpression or knockdown of E75 in clock neurons disrupts rest:activity rhythms and dampens molecular oscillations. E75 represses expression of the gene encoding the transcriptional activator, CLOCK (CLK), and may also affect circadian output. PER inhibits the activity of E75 on the Clk promoter, thereby providing a mechanism for a previously proposed de-repressor effect of PER on Clk transcription. The ecdysone receptor is also expressed in central clock cells and manipulations of its expression produce effects similar to those of E75 on circadian rhythms. We find that E75 protects rhythms under stressful conditions, suggesting a function for steroid signalling in the maintenance of circadian rhythms in Drosophila.


Assuntos
Proteínas CLOCK/genética , Relógios Circadianos/genética , Ritmo Circadiano/genética , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Receptores de Esteroides/genética , Fatores de Transcrição/genética , Animais , Proteínas CLOCK/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Regulação da Expressão Gênica , Neurônios/citologia , Neurônios/metabolismo , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores de Esteroides/antagonistas & inibidores , Receptores de Esteroides/metabolismo , Transdução de Sinais , Estresse Fisiológico , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo , Transcrição Gênica
16.
Trends Neurosci ; 35(9): 574-85, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22748426

RESUMO

In most organisms, an intrinsic circadian (~24-h) timekeeping system drives rhythms of physiology and behavior. Within cells that contain a circadian clock, specific transcriptional activators and repressors reciprocally regulate each other to generate a basic molecular oscillator. A mismatch of the period generated by this oscillator with the external environment creates circadian disruption, which can have adverse effects on neural function. Although several clock genes have been extensively characterized, a fundamental question remains: how do these genes work together to generate a ~24-h period? Period-altering mutations in clock genes can affect any of multiple regulated steps in the molecular oscillator. In this review, we examine the regulatory mechanisms that contribute to setting the pace of the circadian oscillator.


Assuntos
Relógios Biológicos/fisiologia , Relógios Circadianos/fisiologia , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/metabolismo , Retroalimentação Fisiológica/fisiologia , Núcleo Supraquiasmático/fisiopatologia , Ativação Transcricional/fisiologia , Animais , Humanos
17.
Aging Cell ; 11(3): 428-38, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22268765

RESUMO

Sleep-wake cycles break down with age, but the causes of this degeneration are not clear. Using a Drosophila model, we addressed the contribution of circadian mechanisms to this age-induced deterioration. We found that in old flies, free-running circadian rhythms (behavioral rhythms assayed in constant darkness) have a longer period and an unstable phase before they eventually degenerate. Surprisingly, rhythms are weaker in light-dark cycles and the circadian-regulated morning peak of activity is diminished under these conditions. On a molecular level, aging results in reduced amplitude of circadian clock gene expression in peripheral tissues. However, oscillations of the clock protein PERIOD (PER) are robust and synchronized among different clock neurons, even in very old, arrhythmic flies. To improve rhythms in old flies, we manipulated environmental conditions, which can have direct effects on behavior, and also tested a role for molecules that act downstream of the clock. Coupling temperature cycles with a light-dark schedule or reducing expression of protein kinase A (PKA) improved behavioral rhythms and consolidated sleep. Our data demonstrate that a robust molecular timekeeping mechanism persists in the central pacemaker of aged flies, and reducing PKA can strengthen behavioral rhythms.


Assuntos
Comportamento Animal/fisiologia , Ritmo Circadiano/fisiologia , Drosophila/fisiologia , Fatores Etários , Animais , Ritmo Circadiano/genética , Drosophila/metabolismo , Meio Ambiente , Feminino , Masculino , Atividade Motora/genética , Atividade Motora/fisiologia , Fotoperíodo
18.
Curr Biol ; 20(13): 1203-8, 2010 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-20619819

RESUMO

The circadian clock coordinates cellular and organismal energy metabolism. The importance of this circadian timing system is underscored by findings that defects in the clock cause deregulation of metabolic physiology and result in metabolic disorders. On the other hand, metabolism also influences the circadian clock, such that circadian gene expression in peripheral tissues is affected in mammalian models of obesity and diabetes. However, to date there is little to no information on the effect of metabolic genes on the central brain pacemaker which drives behavioral rhythms. We have found that the AKT and TOR-S6K pathways, which are major regulators of nutrient metabolism, cell growth, and senescence, impact the brain circadian clock that drives behavioral rhythms in Drosophila. Elevated AKT or TOR activity lengthens circadian period, whereas reduced AKT signaling shortens it. Effects of TOR-S6K appear to be mediated by SGG/GSK3beta, a known kinase involved in clock regulation. Like SGG, TOR signaling affects the timing of nuclear accumulation of the circadian clock protein TIMELESS. Given that activities of AKT and TOR pathways are affected by nutrient/energy levels and endocrine signaling, these data suggest that metabolic disorders caused by nutrient and energy imbalance are associated with altered rest:activity behavior.


Assuntos
Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/metabolismo , Drosophila/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Animais , Animais Geneticamente Modificados , Western Blotting , Núcleo Celular/metabolismo , Proteínas de Drosophila , Imuno-Histoquímica , Microscopia Confocal , Proteínas Quinases , Serina-Treonina Quinases TOR
19.
Cell Metab ; 8(4): 289-300, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18840359

RESUMO

Studies in mammals have indicated a connection between circadian clocks and feeding behavior, but the nature of the interaction and its relationship to nutrient metabolism are not understood. In Drosophila, clock proteins are expressed in many metabolically important tissues but have not been linked to metabolic processes. Here we demonstrate that Drosophila feeding behavior displays a 24 hr circadian rhythm that is regulated by clocks in digestive/metabolic tissues. Flies lacking clocks in these tissues, in particular in the fat body, also display increased food consumption but have decreased levels of glycogen and a higher sensitivity to starvation. Interestingly, glycogen levels and starvation sensitivity are also affected by clocks in neuronal cells, but the effects of neuronal clocks generally oppose those of the fat body. We propose that the input of neuronal clocks and clocks in metabolic tissues is coordinated to provide effective energy homeostasis.


Assuntos
Relógios Biológicos/fisiologia , Ritmo Circadiano/fisiologia , Drosophila melanogaster/fisiologia , Ingestão de Alimentos/fisiologia , Comportamento Alimentar/fisiologia , Neurônios/fisiologia , Fatores de Transcrição ARNTL , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas CLOCK , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/anatomia & histologia , Metabolismo Energético , Corpo Adiposo/fisiologia , Genótipo , Glicogênio/metabolismo , Homeostase , Humanos , Masculino , Inanição , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
20.
Genes Dev ; 22(11): 1522-33, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18519643

RESUMO

Circadian clocks regulate many different physiological processes and synchronize these to environmental light:dark cycles. In Drosophila, light is transmitted to the clock by a circadian blue light photoreceptor CRYPTOCHROME (CRY). In response to light, CRY promotes the degradation of the circadian clock protein TIMELESS (TIM) and then is itself degraded. To identify novel genes involved in circadian entrainment, we performed an unbiased genome-wide screen in Drosophila cells using a sensitive and quantitative assay that measures light-induced degradation of CRY. We systematically knocked down the expression of approximately 21,000 genes and identified those that regulate CRY stability. These genes include ubiquitin ligases, signal transduction molecules, and redox molecules. Many of the genes identified in the screen are specific for CRY degradation and do not affect degradation of the TIM protein in response to light, suggesting that, for the most part, these two pathways are distinct. We further validated the effect of three candidate genes on CRY stability in vivo by assaying flies mutant for each of these genes. This work identifies a novel regulatory network involved in light-dependent CRY degradation and demonstrates the power of a genome-wide RNAi approach for understanding circadian biology.


Assuntos
Ritmo Circadiano/genética , Luz , Células Fotorreceptoras de Invertebrados , Transativadores/metabolismo , Animais , Proteínas CLOCK , Células Cultivadas , Drosophila/genética , Interferência de RNA
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa