Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Elife ; 122023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37910019

RESUMO

Sleep in mammals can be broadly classified into two different physiological categories: rapid eye movement (REM) sleep and slow-wave sleep (SWS), and accordingly REM and SWS are thought to achieve a different set of functions. The fruit fly Drosophila melanogaster is increasingly being used as a model to understand sleep functions, although it remains unclear if the fly brain also engages in different kinds of sleep as well. Here, we compare two commonly used approaches for studying sleep experimentally in Drosophila: optogenetic activation of sleep-promoting neurons and provision of a sleep-promoting drug, gaboxadol. We find that these different sleep-induction methods have similar effects on increasing sleep duration, but divergent effects on brain activity. Transcriptomic analysis reveals that drug-induced deep sleep ('quiet' sleep) mostly downregulates metabolism genes, whereas optogenetic 'active' sleep upregulates a wide range of genes relevant to normal waking functions. This suggests that optogenetics and pharmacological induction of sleep in Drosophila promote different features of sleep, which engage different sets of genes to achieve their respective functions.


Assuntos
Drosophila melanogaster , Drosophila , Animais , Drosophila melanogaster/genética , Sono/genética , Sono REM , Encéfalo , Mamíferos
2.
Front Neurosci ; 17: 1175478, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37274220

RESUMO

Despite the fact that sleep deprivation substantially affects the way animals regulate their body temperature, the specific mechanisms behind this phenomenon are not well understood. In both mammals and flies, neural circuits regulating sleep and thermoregulation overlap, suggesting an interdependence that may be relevant for sleep function. To investigate this relationship further, we exposed flies to 12 h of sleep deprivation, or 48 h of sleep fragmentation and evaluated temperature preference in a thermal gradient. Flies exposed to 12 h of sleep deprivation chose warmer temperatures after sleep deprivation. Importantly, sleep fragmentation, which prevents flies from entering deeper stages of sleep, but does not activate sleep homeostatic mechanisms nor induce impairments in short-term memory also resulted in flies choosing warmer temperatures. To identify the underlying neuronal circuits, we used RNAi to knock down the receptor for Pigment dispersing factor, a peptide that influences circadian rhythms, temperature preference and sleep. Expressing UAS-PdfrRNAi in subsets of clock neurons prevented sleep fragmentation from increasing temperature preference. Finally, we evaluated temperature preference after flies had undergone a social jet lag protocol which is known to disrupt clock neurons. In this protocol, flies experience a 3 h light phase delay on Friday followed by a 3 h light advance on Sunday evening. Flies exposed to social jet lag exhibited an increase in temperature preference which persisted for several days. Our findings identify specific clock neurons that are modulated by sleep disruption to increase temperature preference. Moreover, our data indicate that temperature preference may be a more sensitive indicator of sleep disruption than learning and memory.

3.
bioRxiv ; 2023 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-37066182

RESUMO

Sleep in mammals can be broadly classified into two different physiological categories: rapid eye movement (REM) sleep and slow wave sleep (SWS), and accordingly REM and SWS are thought to achieve a different set of functions. The fruit fly Drosophila melanogaster is increasingly being used as a model to understand sleep functions, although it remains unclear if the fly brain also engages in different kinds of sleep as well. Here, we compare two commonly used approaches for studying sleep experimentally in Drosophila: optogenetic activation of sleep-promoting neurons and provision of a sleep-promoting drug, Gaboxadol. We find that these different sleep-induction methods have similar effects on increasing sleep duration, but divergent effects on brain activity. Transcriptomic analysis reveals that drug-induced deep sleep ('quiet' sleep) mostly downregulates metabolism genes, whereas optogenetic 'active' sleep upregulates a wide range of genes relevant to normal waking functions. This suggests that optogenetics and pharmacological induction of sleep in Drosophila promote different features of sleep, which engage different sets of genes to achieve their respective functions.

4.
PLoS Biol ; 20(9): e3001797, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36173939

RESUMO

Falling asleep at the wrong time can place an individual at risk of immediate physical harm. However, not sleeping degrades cognition and adaptive behavior. To understand how animals match sleep need with environmental demands, we used live-brain imaging to examine the physiological response properties of the dorsal fan-shaped body (dFB) following interventions that modify sleep (sleep deprivation, starvation, time-restricted feeding, memory consolidation) in Drosophila. We report that dFB neurons change their physiological response-properties to dopamine (DA) and allatostatin-A (AstA) in response to different types of waking. That is, dFB neurons are not simply passive components of a hard-wired circuit. Rather, the dFB neurons intrinsically regulate their response to the activity from upstream circuits. Finally, we show that the dFB appears to contain a memory trace of prior exposure to metabolic challenges induced by starvation or time-restricted feeding. Together, these data highlight that the sleep homeostat is plastic and suggests an underlying mechanism.


Assuntos
Dopamina , Inanição , Animais , Drosophila , Neurônios , Plásticos , Sono , Privação do Sono
5.
PLoS Biol ; 19(6): e3001324, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34191802

RESUMO

Circadian rhythms help animals synchronize motivated behaviors to match environmental demands. Recent evidence indicates that clock neurons influence the timing of behavior by differentially altering the activity of a distributed network of downstream neurons. Downstream circuits can be remodeled by Hebbian plasticity, synaptic scaling, and, under some circumstances, activity-dependent addition of cell surface receptors; the role of this receptor respecification phenomena is not well studied. We demonstrate that high sleep pressure quickly reprograms the wake-promoting large ventrolateral clock neurons to express the pigment dispersing factor receptor (PDFR). The addition of this signaling input into the circuit is associated with increased waking and early mating success. The respecification of PDFR in both young and adult large ventrolateral neurons requires 2 dopamine (DA) receptors and activation of the transcriptional regulator nejire (cAMP response element-binding protein [CREBBP]). These data identify receptor respecification as an important mechanism to sculpt circuit function to match sleep levels with demand.


Assuntos
Adaptação Psicológica , Comportamento Animal/fisiologia , Relógios Biológicos/fisiologia , Drosophila melanogaster/fisiologia , Sono/fisiologia , Vigília/fisiologia , Envelhecimento/fisiologia , Animais , Proteínas de Drosophila/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Comportamento Sexual Animal , Fatores de Transcrição de p300-CBP/metabolismo
6.
J Neurosci ; 41(24): 5173-5189, 2021 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-33931552

RESUMO

We developed a method for single-cell resolution longitudinal bioluminescence imaging of PERIOD (PER) protein and TIMELESS (TIM) oscillations in cultured male adult Drosophila brains that captures circadian circuit-wide cycling under simulated day/night cycles. Light input analysis confirms that CRYPTOCHROME (CRY) is the primary circadian photoreceptor and mediates clock disruption by constant light (LL), and that eye light input is redundant to CRY; 3-h light phase delays (Friday) followed by 3-h light phase advances (Monday morning) simulate the common practice of staying up later at night on weekends, sleeping in later on weekend days then returning to standard schedule Monday morning [weekend light shift (WLS)]. PER and TIM oscillations are highly synchronous across all major circadian neuronal subgroups in unshifted light schedules for 11 d. In contrast, WLS significantly dampens PER oscillator synchrony and rhythmicity in most circadian neurons during and after exposure. Lateral ventral neuron (LNv) oscillations are the first to desynchronize in WLS and the last to resynchronize in WLS. Surprisingly, the dorsal neuron group-3 (DN3s) increase their within-group synchrony in response to WLS. In vivo, WLS induces transient defects in sleep stability, learning, and memory that temporally coincide with circuit desynchrony. Our findings suggest that WLS schedules disrupt circuit-wide circadian neuronal oscillator synchrony for much of the week, thus leading to observed behavioral defects in sleep, learning, and memory.


Assuntos
Encéfalo/fisiopatologia , Ritmo Circadiano/fisiologia , Criptocromos/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas do Olho/metabolismo , Rede Nervosa/fisiopatologia , Proteínas Circadianas Period/metabolismo , Animais , Encéfalo/metabolismo , Drosophila , Aprendizagem/fisiologia , Masculino , Memória/fisiologia , Rede Nervosa/metabolismo , Sono/fisiologia
7.
Sleep ; 44(3)2021 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-32959053

RESUMO

Sleep loss and aging impair hippocampus-dependent Spatial Learning in mammalian systems. Here we use the fly Drosophila melanogaster to investigate the relationship between sleep and Spatial Learning in healthy and impaired flies. The Spatial Learning assay is modeled after the Morris Water Maze. The assay uses a "thermal maze" consisting of a 5 × 5 grid of Peltier plates maintained at 36-37°C and a visual panorama. The first trial begins when a single tile that is associated with a specific visual cue is cooled to 25°C. For subsequent trials, the cold tile is heated, the visual panorama is rotated and the flies must find the new cold tile by remembering its association with the visual cue. Significant learning was observed with two different wild-type strains-Cs and 2U, validating our design. Sleep deprivation prior to training impaired Spatial Learning. Learning was also impaired in the classic learning mutant rutabaga (rut); enhancing sleep restored learning to rut mutants. Further, we found that flies exhibited a dramatic age-dependent cognitive decline in Spatial Learning starting at 20-24 days of age. These impairments could be reversed by enhancing sleep. Finally, we find that Spatial Learning requires dopaminergic signaling and that enhancing dopaminergic signaling in aged flies restored learning. Our results are consistent with the impairments seen in rodents and humans. These results thus demonstrate a critical conserved role for sleep in supporting Spatial Learning, and suggest potential avenues for therapeutic intervention during aging.


Assuntos
Drosophila melanogaster , Drosophila , Animais , Aprendizagem em Labirinto , Sono , Privação do Sono , Aprendizagem Espacial
8.
Curr Biol ; 31(3): 578-590.e6, 2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33238155

RESUMO

The dynamic nature of sleep in many animals suggests distinct stages that serve different functions. Genetic sleep induction methods in animal models provide a powerful way to disambiguate these stages and functions, although behavioral methods alone are insufficient to accurately identify what kind of sleep is being engaged. In Drosophila, activation of the dorsal fan-shaped body (dFB) promotes sleep, but it remains unclear what kind of sleep this is, how the rest of the fly brain is behaving, or if any specific sleep functions are being achieved. Here, we developed a method to record calcium activity from thousands of neurons across a volume of the fly brain during spontaneous sleep and compared this to dFB-induced sleep. We found that spontaneous sleep typically transitions from an active "wake-like" stage to a less active stage. In contrast, optogenetic activation of the dFB promotes sustained wake-like levels of neural activity even though flies become unresponsive to mechanical stimuli. When we probed flies with salient visual stimuli, we found that the activity of visually responsive neurons in the central brain was blocked by transient dFB activation, confirming an acute disconnect from the external environment. Prolonged optogenetic dFB activation nevertheless achieved a key sleep function by correcting visual attention defects brought on by sleep deprivation. These results suggest that dFB activation promotes a distinct form of sleep in Drosophila, where brain activity appears similar to wakefulness, but responsiveness to external sensory stimuli is profoundly suppressed.


Assuntos
Drosophila melanogaster , Sono , Animais , Drosophila melanogaster/genética , Privação do Sono , Vigília
9.
J Vis Exp ; (166)2020 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-33369606

RESUMO

Sleep homeostasis, the increase in sleep observed following sleep loss, is one of the defining criteria used to identify sleep throughout the animal kingdom. As a consequence, sleep deprivation and sleep restriction are powerful tools that are commonly used to provide insight into sleep function. Nonetheless, sleep deprivation experiments are inherently problematic in that the deprivation stimulus itself may be the cause of observed changes in physiology and behavior. Accordingly, successful sleep deprivation techniques should keep animals awake and, ideally, result in a robust sleep rebound without also inducing a large number of unintended consequences. Here, we describe a sleep deprivation technique for Drosophila melanogaster. The Sleep Nullifying Apparatus (SNAP) administers a stimulus every 10s to induce negative geotaxis. Although the stimulus is predictable, the SNAP effectively prevents >95% of nighttime sleep even in flies with high sleep drive. Importantly, the subsequent homeostatic response is very similar to that achieved using hand-deprivation. The timing and spacing of the stimuli can be modified to minimize sleep loss and thus examine non-specific effects of the stimulus on physiology and behavior. The SNAP can also be used for sleep restriction and to assess arousal thresholds. The SNAP is a powerful sleep disruption technique that can be used to better understand sleep function.


Assuntos
Drosophila melanogaster/fisiologia , Polissonografia/métodos , Privação do Sono/fisiopatologia , Animais , Homeostase/fisiologia , Sono/fisiologia , Inquéritos e Questionários
10.
J Exp Biol ; 221(Pt 24)2018 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-30355611

RESUMO

Although sleep deprivation is known to impair attention in humans and other mammals, the underlying reasons are not well understood, and whether similar effects are present in non-mammalian species is not known. We therefore sought to investigate whether sleep is important for optimizing attention in an invertebrate species, the genetic model Drosophila melanogaster We developed a high-throughput paradigm to measure visual attention in freely walking Drosophila, using competing foreground/background visual stimuli. We found that whereas sleep-deprived flies could respond normally to either stimulus alone, they were more distracted by background cues in a visual competition task. Other stressful manipulations such as starvation, heat exposure and mechanical stress had no effects on visual attention in this paradigm. In contrast to sleep deprivation, providing additional sleep using the GABA-A agonist 4,5,6,7-tetrahydroisoxazolo-[5,4-c]pyridine-3-ol (THIP) did not affect attention in wild-type flies, but specifically improved attention in the learning mutant dunce Our results reveal a key function of sleep in optimizing attention processes in Drosophila, and establish a behavioral paradigm that can be used to explore the molecular mechanisms involved.


Assuntos
Atenção/fisiologia , Drosophila melanogaster/fisiologia , Privação do Sono/fisiopatologia , Sono/fisiologia , Percepção Visual/fisiologia , Animais , Feminino , Masculino
11.
Elife ; 72018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-30109983

RESUMO

Sleep is a dynamic process in most animals, involving distinct stages that probably perform multiple functions for the brain. Before sleep functions can be initiated, it is likely that behavioral responsiveness to the outside world needs to be reduced, even while the animal is still awake. Recent work in Drosophila has uncovered a sleep switch in the dorsal fan-shaped body (dFB) of the fly's central brain, but it is not known whether these sleep-promoting neurons also govern the acute need to ignore salient stimuli in the environment during sleep transitions. We found that optogenetic activation of the sleep switch suppressed behavioral responsiveness to mechanical stimuli, even in awake flies, indicating a broader role for these neurons in regulating arousal. The dFB-mediated suppression mechanism and its associated neural correlates requires innexin6 expression, suggesting that the acute need to reduce sensory perception when flies fall asleep is mediated in part by electrical synapses.


Assuntos
Encéfalo/metabolismo , Conexinas/genética , Sono/genética , Vigília/genética , Animais , Encéfalo/fisiologia , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/fisiologia , Sinapses Elétricas/genética , Sinapses Elétricas/fisiologia , Regulação da Expressão Gênica/genética , Homeostase , Neurônios/metabolismo , Optogenética , Sono/fisiologia , Vigília/fisiologia
12.
Nat Commun ; 8(1): 1815, 2017 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-29180766

RESUMO

Sleep is a dynamic process comprising multiple stages, each associated with distinct electrophysiological properties and potentially serving different functions. While these phenomena are well described in vertebrates, it is unclear if invertebrates have distinct sleep stages. We perform local field potential (LFP) recordings on flies spontaneously sleeping, and compare their brain activity to flies induced to sleep using either genetic activation of sleep-promoting circuitry or the GABAA agonist Gaboxadol. We find a transitional sleep stage associated with a 7-10 Hz oscillation in the central brain during spontaneous sleep. Oscillatory activity is also evident when we acutely activate sleep-promoting neurons in the dorsal fan-shaped body (dFB) of Drosophila. In contrast, sleep following Gaboxadol exposure is characterized by low-amplitude LFPs, during which dFB-induced effects are suppressed. Sleep in flies thus appears to involve at least two distinct stages: increased oscillatory activity, particularly during sleep induction, followed by desynchronized or decreased brain activity.


Assuntos
Encéfalo/fisiologia , Dípteros/fisiologia , Eletrofisiologia/métodos , Fases do Sono/fisiologia , Animais , Drosophila melanogaster/fisiologia , Feminino , Isoxazóis/metabolismo , Fenômenos Fisiológicos do Sistema Nervoso , Neurônios/fisiologia , Sono/fisiologia , Fatores de Tempo
13.
Front Neural Circuits ; 11: 79, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29109678

RESUMO

Although patients with primary insomnia experience sleep disruption, they are able to maintain normal performance on a variety of cognitive tasks. This observation suggests that insomnia may be a condition where predisposing factors simultaneously increase the risk for insomnia and also mitigate against the deleterious consequences of waking. To gain insight into processes that might regulate sleep and buffer neuronal circuits during sleep loss, we manipulated three genes, fat facet (faf), highwire (hiw) and the GABA receptor Resistance to dieldrin (Rdl), that were differentially modulated in a Drosophila model of insomnia. Our results indicate that increasing faf and decreasing hiw or Rdl within wake-promoting large ventral lateral clock neurons (lLNvs) induces sleep loss. As expected, sleep loss induced by decreasing hiw in the lLNvs results in deficits in short-term memory and increases of synaptic growth. However, sleep loss induced by knocking down Rdl in the lLNvs protects flies from sleep-loss induced deficits in short-term memory and increases in synaptic markers. Surprisingly, decreasing hiw and Rdl within the Mushroom Bodies (MBs) protects against the negative effects of sleep deprivation (SD) as indicated by the absence of a subsequent homeostatic response, or deficits in short-term memory. Together these results indicate that specific genes are able to disrupt sleep and protect against the negative consequences of waking in a circuit dependent manner.


Assuntos
Proteínas de Drosophila/metabolismo , Endopeptidases/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Plasticidade Neuronal/fisiologia , Receptores de GABA-A/metabolismo , Privação do Sono/metabolismo , Distúrbios do Início e da Manutenção do Sono/metabolismo , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Drosophila , Proteínas de Drosophila/genética , Endopeptidases/genética , Homeostase/genética , Homeostase/fisiologia , Aprendizagem , Memória de Curto Prazo/fisiologia , Atividade Motora/genética , Atividade Motora/fisiologia , Corpos Pedunculados/metabolismo , Corpos Pedunculados/patologia , Proteínas do Tecido Nervoso/genética , Plasticidade Neuronal/genética , Neurônios/metabolismo , Neurônios/patologia , Receptores de GABA-A/genética , Privação do Sono/genética , Privação do Sono/patologia , Distúrbios do Início e da Manutenção do Sono/genética , Distúrbios do Início e da Manutenção do Sono/patologia , Sinapses/genética , Sinapses/metabolismo , Sinapses/patologia
14.
Artigo em Inglês | MEDLINE | ID: mdl-29094110

RESUMO

To test the hypothesis that sleep can reverse cognitive impairment during Alzheimer's disease, we enhanced sleep in flies either co-expressing human amyloid precursor protein and Beta-secretase (APP:BACE), or in flies expressing human tau. The ubiquitous expression of APP:BACE or human tau disrupted sleep. The sleep deficits could be reversed and sleep could be enhanced when flies were administered the GABA-A agonist 4,5,6,7-tetrahydroisoxazolo-[5,4-c]pyridine-3-ol (THIP). Expressing APP:BACE disrupted both Short-term memory (STM) and Long-term memory (LTM) as assessed using Aversive Phototaxic Suppression (APS) and courtship conditioning. Flies expressing APP:BACE also showed reduced levels of the synaptic protein discs large (DLG). Enhancing sleep in memory-impaired APP:BACE flies fully restored both STM and LTM and restored DLG levels. Sleep also restored STM to flies expressing human tau. Using live-brain imaging of individual clock neurons expressing both tau and the cAMP sensor Epac1-camps, we found that tau disrupted cAMP signaling. Importantly, enhancing sleep in flies expressing human tau restored proper cAMP signaling. Thus, we demonstrate that sleep can be used as a therapeutic to reverse deficits that accrue during the expression of toxic peptides associated with Alzheimer's disease.

15.
Nature ; 536(7616): 278-80, 2016 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-27487223

Assuntos
Neurociências , Sono , Humanos
16.
Curr Biol ; 25(21): R1040-R1041, 2015 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-26528745

RESUMO

In mammals, evidence for memory reactivation during sleep highlighted the important role that sleep plays in memory consolidation. A new study reports that memory reactivation is evolutionarily conserved and can also be found in the honeybee.


Assuntos
Sonhos , Memória , Animais , Abelhas , Humanos , Aprendizagem , Sono
17.
Curr Sleep Med Rep ; 1(1): 47-54, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26120553

RESUMO

Drosophila has proven to be a powerful model to identify genes and circuits that impact sleep. While the majority of studies have primarily been interested in identifying manipulations that alter sleep time, a growing body of work has begun to focus on how changing sleep influences functional outcomes such as cognitive performance, structural plasticity, and metabolism to name a few. Evaluating sleep time provides an appropriate entry point into elucidating sleep function. However, it is not possible to fully understand how a manipulation has impacted sleep regulation without first establishing how it has affected the animals' well-being. Synaptic plasticity and memory are important functional outcomes that can be used to asses an animal's status. In this manuscript, we review recent advances in studies examining sleep, memory, and performance. We conclude that as Drosophila sleep researchers expand their analysis beyond sleep time, the opportunities to discover the function of sleep will be enhanced.

18.
Curr Biol ; 25(10): 1270-81, 2015 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-25913403

RESUMO

Given the role that sleep plays in modulating plasticity, we hypothesized that increasing sleep would restore memory to canonical memory mutants without specifically rescuing the causal molecular lesion. Sleep was increased using three independent strategies: activating the dorsal fan-shaped body, increasing the expression of Fatty acid binding protein (dFabp), or by administering the GABA-A agonist 4,5,6,7-tetrahydroisoxazolo-[5,4-c]pyridine-3-ol (THIP). Short-term memory (STM) or long-term memory (LTM) was evaluated in rutabaga (rut) and dunce (dnc) mutants using aversive phototaxic suppression and courtship conditioning. Each of the three independent strategies increased sleep and restored memory to rut and dnc mutants. Importantly, inducing sleep also reverses memory defects in a Drosophila model of Alzheimer's disease. Together, these data demonstrate that sleep plays a more fundamental role in modulating behavioral plasticity than previously appreciated and suggest that increasing sleep may benefit patients with certain neurological disorders.


Assuntos
Adenilil Ciclases/genética , Comportamento Animal/fisiologia , Proteínas de Drosophila/genética , Drosophila melanogaster/fisiologia , Sono/fisiologia , Doença de Alzheimer/fisiopatologia , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Drosophila melanogaster/genética , Proteínas de Ligação a Ácido Graxo/genética , Feminino , Isoxazóis/farmacologia , Masculino , Memória de Longo Prazo/fisiologia , Memória de Curto Prazo/efeitos dos fármacos , Memória de Curto Prazo/fisiologia , Mutação , Compostos Organofosforados/farmacologia , Receptores de GABA/genética , Reserpina/farmacologia , Sono/efeitos dos fármacos
19.
Mediators Inflamm ; 2015: 539627, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25873764

RESUMO

Excessive daytime sleepiness (EDS) is a ubiquitous problem that affects public health and safety. A test that can reliably identify individuals that suffer from EDS is needed. In contrast to other methods, salivary biomarkers are an objective, inexpensive, and noninvasive method to identify individuals with inadequate sleep. Although we have previously shown that inflammatory genes are elevated in saliva samples taken from sleep deprived individuals, it is unclear if inflammatory genes will be elevated in clinical populations with EDS. In this study, salivary samples from individuals with sleep apnea were evaluated using the Taqman low density inflammation array. Transcript levels for 3 genes, including prostaglandin-endoperoxide synthase 2 (PTGS2), were elevated in patients with sleep apnea. Interestingly, PTGS2 was also elevated in patients with EDS but who did not have sleep apnea. These data demonstrate the feasibility of using salivary transcript levels to identify individuals that self-report excessive daytime sleepiness.


Assuntos
Ciclo-Oxigenase 2/genética , Distúrbios do Sono por Sonolência Excessiva/metabolismo , Saliva/metabolismo , Adulto , Idoso , Biomarcadores , Índice de Massa Corporal , Caspase 1/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/análise , Síndromes da Apneia do Sono/metabolismo
20.
Sleep ; 38(5): 801-14, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25409104

RESUMO

BACKGROUND AND STUDY OBJECTIVES: Flies mutant for the canonical clock protein cycle (cyc(01)) exhibit a sleep rebound that is ∼10 times larger than wild-type flies and die after only 10 h of sleep deprivation. Surprisingly, when starved, cyc(01) mutants can remain awake for 28 h without demonstrating negative outcomes. Thus, we hypothesized that identifying transcripts that are differentially regulated between waking induced by sleep deprivation and waking induced by starvation would identify genes that underlie the deleterious effects of sleep deprivation and/or protect flies from the negative consequences of waking. DESIGN: We used partial complementary DNA microarrays to identify transcripts that are differentially expressed between cyc(01) mutants that had been sleep deprived or starved for 7 h. We then used genetics to determine whether disrupting genes involved in lipid metabolism would exhibit alterations in their response to sleep deprivation. SETTING: Laboratory. PATIENTS OR PARTICIPANTS: Drosophila melanogaster. INTERVENTIONS: Sleep deprivation and starvation. MEASUREMENTS AND RESULTS: We identified 84 genes with transcript levels that were differentially modulated by 7 h of sleep deprivation and starvation in cyc(01) mutants and were confirmed in independent samples using quantitative polymerase chain reaction. Several of these genes were predicted to be lipid metabolism genes, including bubblegum, cueball, and CG4500, which based on our data we have renamed heimdall (hll). Using lipidomics we confirmed that knockdown of hll using RNA interference significantly decreased lipid stores. Importantly, genetically modifying bubblegum, cueball, or hll resulted in sleep rebound alterations following sleep deprivation compared to genetic background controls. CONCLUSIONS: We have identified a set of genes that may confer resilience/vulnerability to sleep deprivation and demonstrate that genes involved in lipid metabolism modulate sleep homeostasis.


Assuntos
Adaptação Fisiológica/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/fisiologia , Predisposição Genética para Doença/genética , Privação do Sono/genética , Inanição/genética , Animais , Feminino , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Homeostase/genética , Metabolismo dos Lipídeos/genética , Masculino , Mutação/genética , Análise de Sequência com Séries de Oligonucleotídeos , Interferência de RNA , RNA Mensageiro/análise , RNA Mensageiro/genética , Sono/fisiologia , Privação do Sono/fisiopatologia , Inanição/fisiopatologia , Fatores de Tempo , Vigília/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...