Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
Nature ; 536(7616): 278-80, 2016 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-27487223

Assuntos
Neurociências , Sono , Humanos
6.
Brain Behav Immun ; 47: 75-85, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25451614

RESUMO

Individuals frequently find themselves confronted with a variety of challenges that threaten their wellbeing. While some individuals face these challenges efficiently and thrive (resilient) others are unable to cope and may suffer persistent consequences (vulnerable). Resilience/vulnerability to sleep disruption may contribute to the vulnerability of individuals exposed to challenging conditions. With that in mind we exploited individual differences in a fly's ability to form short-term memory (STM) following 3 different types of sleep disruption to identify the underlying genes. Our analysis showed that in each category of flies examined, there are individuals that form STM in the face of sleep loss (resilient) while other individuals show dramatic declines in cognitive behavior (vulnerable). Molecular genetic studies revealed that Antimicrobial Peptides, factors important for innate immunity, were candidates for conferring resilience/vulnerability to sleep deprivation. Specifically, Metchnikowin (Mtk), drosocin (dro) and Attacin (Att) transcript levels seemed to be differentially increased by sleep deprivation in glia (Mtk), neurons (dro) or primarily in the head fat body (Att). Follow-up genetic studies confirmed that expressing Mtk in glia but not neurons, and expressing dro in neurons but not glia, disrupted memory while modulating sleep in opposite directions. These data indicate that various factors within glia or neurons can contribute to individual differences in resilience/vulnerability to sleep deprivation.


Assuntos
Neuroglia/imunologia , Neurônios/imunologia , Privação do Sono/imunologia , Sono/imunologia , Animais , Comportamento Animal/fisiologia , Drosophila , Individualidade , Memória de Curto Prazo/fisiologia
7.
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
8.
Proc Natl Acad Sci U S A ; 109(7): 2613-8, 2012 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-22308351

RESUMO

Recent human studies suggest that genetic polymorphisms allow an individual to maintain optimal cognitive functioning during sleep deprivation. If such polymorphisms were not associated with additional costs, selective pressures would allow these alleles to spread through the population such that an evolutionary alternative to sleep would emerge. To determine whether there are indeed costs associated with resiliency to sleep loss, we challenged natural allelic variants of the foraging gene (for) with either sleep deprivation or starvation. Flies with high levels of Protein Kinase G (PKG) (for(R)) do not display deficits in short-term memory following 12 h of sleep deprivation. However, short-term memory is significantly disrupted when for(R) flies are starved overnight. In contrast, flies with low levels of PKG (for(s), for(s2)) show substantial deficits in short-term memory following sleep deprivation but retain their ability to learn after 12 h of starvation. We found that for(R) phenotypes could be largely recapitulated in for(s) flies by selectively increasing the level of PKG in the α/ß lobes of the mushroom bodies, a structure known to regulate both sleep and memory. Together, these data indicate that whereas the expression of for may appear to provide resilience in one environmental context, it may confer an unexpected vulnerability in other situations. Understanding how these tradeoffs confer resilience or vulnerability to specific environmental challenges may provide additional clues as to why an evolutionary alternative to sleep has not emerged.


Assuntos
Comportamento Animal , Drosophila/fisiologia , Comportamento Alimentar , Sono , Inanição , Animais
9.
PLoS Biol ; 8(8)2010 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-20824166

RESUMO

Extended periods of waking result in physiological impairments in humans, rats, and flies. Sleep homeostasis, the increase in sleep observed following sleep loss, is believed to counter the negative effects of prolonged waking by restoring vital biological processes that are degraded during sleep deprivation. Sleep homeostasis, as with other behaviors, is influenced by both genes and environment. We report here that during periods of starvation, flies remain spontaneously awake but, in contrast to sleep deprivation, do not accrue any of the negative consequences of prolonged waking. Specifically, the homeostatic response and learning impairments that are a characteristic of sleep loss are not observed following prolonged waking induced by starvation. Recently, two genes, brummer (bmm) and Lipid storage droplet 2 (Lsd2), have been shown to modulate the response to starvation. bmm mutants have excess fat and are resistant to starvation, whereas Lsd2 mutants are lean and sensitive to starvation. Thus, we hypothesized that bmm and Lsd2 may play a role in sleep regulation. Indeed, bmm mutant flies display a large homeostatic response following sleep deprivation. In contrast, Lsd2 mutant flies, which phenocopy aspects of starvation as measured by low triglyceride stores, do not exhibit a homeostatic response following sleep loss. Importantly, Lsd2 mutant flies are not learning impaired after sleep deprivation. These results provide the first genetic evidence, to our knowledge, that lipid metabolism plays an important role in regulating the homeostatic response and can protect against neuronal impairments induced by prolonged waking.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiologia , Homeostase , Aprendizagem/efeitos dos fármacos , Sono , Animais , Proteínas de Transporte , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Aprendizagem/fisiologia , Metabolismo dos Lipídeos , Mutação , Perilipina-1 , Fosfoproteínas/química , Sono/fisiologia , Privação do Sono , Triglicerídeos/metabolismo
10.
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
11.
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.

12.
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.

13.
Curr Biol ; 18(15): 1110-7, 2008 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-18674913

RESUMO

BACKGROUND: Extended wakefulness disrupts acquisition of short-term memories in mammals. However, the underlying molecular mechanisms triggered by extended waking and restored by sleep are unknown. Moreover, the neuronal circuits that depend on sleep for optimal learning remain unidentified. RESULTS: Learning was evaluated with aversive phototaxic suppression. In this task, flies learn to avoid light that is paired with an aversive stimulus (quinine-humidity). We demonstrate extensive homology in sleep-deprivation-induced learning impairment between flies and humans. Both 6 hr and 12 hr of sleep deprivation are sufficient to impair learning in Canton-S (Cs) flies. Moreover, learning is impaired at the end of the normal waking day in direct correlation with time spent awake. Mechanistic studies indicate that this task requires intact mushroom bodies (MBs) and requires the dopamine D1-like receptor (dDA1). Importantly, sleep-deprivation-induced learning impairments could be rescued by targeted gene expression of the dDA1 receptor to the MBs. CONCLUSIONS: These data provide direct evidence that extended wakefulness disrupts learning in Drosophila. These results demonstrate that it is possible to prevent the effects of sleep deprivation by targeting a single neuronal structure and identify cellular and molecular targets adversely affected by extended waking in a genetically tractable model organism.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila/fisiologia , Aprendizagem/fisiologia , Corpos Pedunculados/fisiologia , Receptores de Dopamina D1/fisiologia , Privação do Sono , Animais , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Corpos Pedunculados/metabolismo , Receptores de Dopamina D1/metabolismo , Transdução de Sinais
14.
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
15.
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
16.
J Neurosci ; 29(22): 7148-57, 2009 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-19494137

RESUMO

Although it is widely accepted that sleep must serve an essential biological function, little is known about molecules that underlie sleep regulation. Given that insomnia is a common sleep disorder that disrupts the ability to initiate and maintain restorative sleep, a better understanding of its molecular underpinning may provide crucial insights into sleep regulatory processes. Thus, we created a line of flies using laboratory selection that share traits with human insomnia. After 60 generations, insomnia-like (ins-l) flies sleep 60 min a day, exhibit difficulty initiating sleep, difficulty maintaining sleep, and show evidence of daytime cognitive impairment. ins-l flies are also hyperactive and hyperresponsive to environmental perturbations. In addition, they have difficulty maintaining their balance, have elevated levels of dopamine, are short-lived, and show increased levels of triglycerides, cholesterol, and free fatty acids. Although their core molecular clock remains intact, ins-l flies lose their ability to sleep when placed into constant darkness. Whole-genome profiling identified genes that are modified in ins-l flies. Among those differentially expressed transcripts, genes involved in metabolism, neuronal activity, and sensory perception constituted over-represented categories. We demonstrate that two of these genes are upregulated in human subjects after acute sleep deprivation. Together, these data indicate that the ins-l flies are a useful tool that can be used to identify molecules important for sleep regulation and may provide insights into both the causes and long-term consequences of insomnia.


Assuntos
Proteínas de Drosophila/genética , Regulação da Expressão Gênica/fisiologia , Distúrbios do Início e da Manutenção do Sono/genética , Sono/genética , Análise de Variância , Animais , Animais Geneticamente Modificados , Aprendizagem da Esquiva/fisiologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Comportamento Animal , Colesterol/metabolismo , Ritmo Circadiano/genética , Proteínas Contráteis/metabolismo , Modelos Animais de Doenças , Dopamina/metabolismo , Drosophila , Ácidos Graxos não Esterificados/metabolismo , Feminino , Filaminas , Perfilação da Expressão Gênica/métodos , Humanos , Lipídeos , Locomoção/genética , Malato Desidrogenase/metabolismo , Masculino , Proteínas dos Microfilamentos/metabolismo , Neurotransmissores/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Hormônios Peptídicos/genética , Hormônios Peptídicos/metabolismo , Fenótipo , Privação do Sono/fisiopatologia , Estatísticas não Paramétricas , Estresse Psicológico/genética , Triglicerídeos/metabolismo , Vigília
17.
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
18.
Sleep ; 32(8): 984-92, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19725249

RESUMO

STUDY OBJECTIVES: Parkinson disease (PD) is the second most common neurodegenerative disorder in the United States. It is associated with motor deficits, sleep disturbances, and cognitive impairment. The pathology associated with PD and the effects of sleep deprivation impinge, in part, upon common molecular pathways suggesting that sleep loss may be particularly deleterious to the degenerating brain. Thus we investigated the long-term consequences of sleep deprivation on shortterm memory using a Drosophila model of Parkinson disease. PARTICIPANTS: Transgenic strains of Drosophila melanogaster. DESIGN: Using the GAL4-UAS system, human alpha-synuclein was expressed throughout the nervous system of adult flies. Alpha-synuclein expressing flies (alpha S flies) and the corresponding genetic background controls were sleep deprived for 12 h at age 16 days and allowed to recover undisturbed for at least 3 days. Short-term memory was evaluated using aversive phototaxis suppression. Dopaminergic systems were assessed using mRNA profiling and immunohistochemistry. MEASURMENTS AND RESULTS: When sleep deprived at an intermediate stage of the pathology, alpha S flies showed persistent short-term memory deficits that lasted > or = 3 days. Cognitive deficits were not observed in younger alpha S flies nor in genetic background controls. Long-term impairments were not associated with accelerated loss of dopaminergic neurons. However mRNA expression of the dopamine receptors dDA1 and DAMB were significantly increased in sleep deprived alpha S flies. Blocking D1-like receptors during sleep deprivation prevented persistent shortterm memory deficits. Importantly, feeding flies the polyphenolic compound curcumin blocked long-term learning deficits. CONCLUSIONS: These data emphasize the importance of sleep in a degenerating/reorganizing brain and shows that pathological processes induced by sleep deprivation can be dissected at the molecular and cellular level using Drosophila genetics.


Assuntos
Drosophila melanogaster/genética , Memória de Curto Prazo , Transtornos Parkinsonianos/psicologia , Privação do Sono/psicologia , Fatores Etários , Animais , Animais Geneticamente Modificados , Aprendizagem da Esquiva , Comportamento de Escolha/efeitos dos fármacos , Curcumina/farmacologia , Proteínas de Drosophila/genética , Drosophila melanogaster/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Perfilação da Expressão Gênica , Humanos , Inibição Psicológica , Luz , Aprendizagem em Labirinto/efeitos dos fármacos , Memória de Curto Prazo/efeitos dos fármacos , Motivação , Neurotoxinas/antagonistas & inibidores , Oxidopamina/antagonistas & inibidores , Transtornos Parkinsonianos/genética , RNA Mensageiro/genética , Receptores Dopaminérgicos/genética , Receptores de Dopamina D2/genética , alfa-Sinucleína/genética
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA