Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
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
2.
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
3.
PLoS Biol ; 18(8): e3000548, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32745077

RESUMO

Sleep is vital for survival. Yet under environmentally challenging conditions, such as starvation, animals suppress their need for sleep. Interestingly, starvation-induced sleep loss does not evoke a subsequent sleep rebound. Little is known about how starvation-induced sleep deprivation differs from other types of sleep loss, or why some sleep functions become dispensable during starvation. Here, we demonstrate that down-regulation of the secreted cytokine unpaired 2 (upd2) in Drosophila flies may mimic a starved-like state. We used a genetic knockdown strategy to investigate the consequences of upd2 on visual attention and sleep in otherwise well-fed flies, thereby sidestepping the negative side effects of undernourishment. We find that knockdown of upd2 in the fat body (FB) is sufficient to suppress sleep and promote feeding-related behaviors while also improving selective visual attention. Furthermore, we show that this peripheral signal is integrated in the fly brain via insulin-expressing cells. Together, these findings identify a role for peripheral tissue-to-brain interactions in the simultaneous regulation of sleep quality and attention, to potentially promote adaptive behaviors necessary for survival in hungry animals.


Assuntos
Atenção/fisiologia , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Comportamento Alimentar/fisiologia , Inanição/genética , Percepção Visual/fisiologia , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Corpo Adiposo/metabolismo , Feminino , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Insulina/genética , Insulina/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Transdução de Sinais , Sono/fisiologia , Privação do Sono/genética , Privação do Sono/metabolismo , Inanição/metabolismo
4.
Sleep ; 42(7)2019 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-31100151

RESUMO

Sleep optimizes waking behavior, however, waking experience may also influence sleep. We used the fruit fly Drosophila melanogaster to investigate the relationship between visual experience and sleep in wild-type and mutant flies. We found that the classical visual mutant, optomotor-blind (omb), which has undeveloped horizontal system/vertical system (HS/VS) motion-processing cells and are defective in motion and visual salience perception, showed dramatically reduced and less consolidated sleep compared to wild-type flies. In contrast, optogenetic activation of the HS/VS motion-processing neurons in wild-type flies led to an increase in sleep following the activation, suggesting an increase in sleep pressure. Surprisingly, exposing wild-type flies to repetitive motion stimuli for extended periods did not increase sleep pressure. However, we observed that exposing flies to more complex image sequences from a movie led to more consolidated sleep, particularly when images were randomly shuffled through time. Our results suggest that specific forms of visual experience that involve motion circuits and complex, nonrepetitive imagery, drive sleep need in Drosophila.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/fisiologia , Proteínas do Tecido Nervoso/genética , Sono/fisiologia , Proteínas com Domínio T/genética , Percepção Visual/fisiologia , Animais , Neurônios/fisiologia , Optogenética
5.
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
6.
Front Psychiatry ; 8: 22, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28243212

RESUMO

The dopamine ontogeny hypothesis for schizophrenia proposes that transient dysregulation of the dopaminergic system during brain development increases the likelihood of this disorder in adulthood. To test this hypothesis in a high-throughput animal model, we have transiently manipulated dopamine signaling in the developing fruit fly Drosophila melanogaster and examined behavioral responsiveness in adult flies. We found that either a transient increase of dopamine neuron activity or a transient decrease of dopamine receptor expression during fly brain development permanently impairs behavioral responsiveness in adults. A screen for impaired responsiveness revealed sleep-promoting neurons in the central brain as likely postsynaptic dopamine targets modulating these behavioral effects. Transient dopamine receptor knockdown during development in a restricted set of ~20 sleep-promoting neurons recapitulated the dopamine ontogeny phenotype, by permanently reducing responsiveness in adult animals. This suggests that disorders involving impaired behavioral responsiveness might result from defective ontogeny of sleep/wake circuits.

7.
Trends Neurosci ; 38(12): 776-786, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26602764

RESUMO

Sleep is not a single state, but a complex set of brain processes that supports several physiological needs. Sleep deprivation is known to affect attention in many animals, suggesting that a key function of sleep is to regulate attention. Conversely, tasks that require more attention drive sleep need and sleep intensity. Attention involves the ability to filter incoming stimuli based on their relative salience, and this is likely to require coordinated synaptic activity across the brain. This capacity may have only become possible with the evolution of related neural mechanisms that support two key sleep functions: stimulus suppression and synaptic plasticity. We argue here that sleep and attention may have coevolved as brain states that regulate each other.


Assuntos
Atenção/fisiologia , Encéfalo/fisiologia , Sono/fisiologia , Animais , Evolução Biológica , Humanos
8.
Eur J Neurosci ; 42(7): 2455-66, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26201245

RESUMO

Neurexins are cell adhesion molecules that are important for synaptic plasticity and homeostasis, although links to sleep have not yet been investigated. We examined the effects of neurexin-1 perturbation on sleep in Drosophila, showing that neurexin-1 nulls displayed fragmented sleep and altered circadian rhythm. Conversely, the over-expression of neurexin-1 could increase and consolidate night-time sleep. This was not solely due to developmental effects as it could be induced acutely in adulthood, and was coupled with evidence of synaptic growth. The timing of over-expression could differentially impact sleep patterns, with specific night-time effects. These results show that neurexin-1 was dynamically involved in synaptic plasticity and sleep in Drosophila. Neurexin-1 and a number of its binding partners have been repeatedly associated with mental health disorders, including autism spectrum disorders, schizophrenia and Tourette syndrome, all of which are also linked to altered sleep patterns. How and when plasticity-related proteins such as neurexin-1 function during sleep can provide vital information on the interaction between synaptic homeostasis and sleep, paving the way for more informed treatments of human disorders.


Assuntos
Moléculas de Adesão Celular Neuronais/fisiologia , Ritmo Circadiano/fisiologia , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/fisiologia , Plasticidade Neuronal/fisiologia , Sono/fisiologia , Animais , Comportamento Animal/fisiologia , Moléculas de Adesão Celular Neuronais/metabolismo , Proteínas de Drosophila/metabolismo
9.
J Neurosci ; 35(28): 10304-15, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-26180205

RESUMO

A crucial function of the brain is to be able to distinguish whether or not changes in the environment are caused by one's own actions. Even the smallest brains appear to be capable of making this distinction, as has been shown by closed-loop behavioral experiments in flies controlling visual stimuli in virtual reality paradigms. We questioned whether activity in the fruit fly brain is different during such closed-loop behavior, compared with passive viewing of a stimulus. To address this question, we used a procedure to record local field potential (LFP) activity across the fly brain while flies were controlling a virtual object through their movement on an air-supported ball. The virtual object was flickered at a precise frequency (7 Hz), creating a frequency tag that allowed us to track brain responses to the object while animals were behaving. Following experiments under closed-loop control, we replayed the same stimulus to the fly in open loop, such that it could no longer control the stimulus. We found identical receptive fields and similar strength of frequency tags across the brain for the virtual object under closed loop and replay. However, when comparing central versus peripheral brain regions, we found that brain responses were differentially modulated depending on whether flies were in control or not. Additionally, coherence of LFP activity in the brain increased when flies were in control, compared with replay, even if motor behavior was similar. This suggests that processes associated with closed-loop control promote temporal coordination in the insect brain. SIGNIFICANCE STATEMENT: We show that closed-loop control of a visual stimulus promotes temporal coordination across the Drosophila brain, compared with open-loop replay of the same visual sequences. This is significant because it suggests that, to understand goal-directed behavior or visual attention in flies, it may be most informative to sample neural activity from multiple regions across the brain simultaneously, and to examine temporal relationships (e.g., coherence) between these regions.


Assuntos
Comportamento Animal/fisiologia , Mapeamento Encefálico , Encéfalo/fisiologia , Campos Visuais/fisiologia , Percepção Visual/fisiologia , Análise de Variância , Animais , Drosophila melanogaster , Potenciais Evocados Visuais/fisiologia , Feminino , Locomoção , Estimulação Luminosa
10.
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
11.
J Neurosci ; 33(16): 6917-27, 2013 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-23595750

RESUMO

How might one determine whether simple animals such as flies sleep in stages? Sleep in mammals is a dynamic process involving different stages of sleep intensity, and these are typically associated with measurable changes in brain activity (Blake and Gerard, 1937; Rechtschaffen and Kales, 1968; Webb and Agnew, 1971). Evidence for different sleep stages in invertebrates remains elusive, even though it has been well established that many invertebrate species require sleep (Campbell and Tobler, 1984; Hendricks et al., 2000; Shaw et al., 2000; Sauer et al., 2003). Here we used electrophysiology and arousal-testing paradigms to show that the fruit fly, Drosophila melanogaster, transitions between deeper and lighter sleep within extended bouts of inactivity, with deeper sleep intensities after ∼15 and ∼30 min of inactivity. As in mammals, the timing and intensity of these dynamic sleep processes in flies is homeostatically regulated and modulated by behavioral experience. Two molecules linked to synaptic plasticity regulate the intensity of the first deep sleep stage. Optogenetic upregulation of cyclic adenosine monophosphate during the day increases sleep intensity at night, whereas loss of function of a molecule involved in synaptic pruning, the fragile-X mental retardation protein, increases sleep intensity during the day. Our results show that sleep is not homogenous in insects, and suggest that waking behavior and the associated synaptic plasticity mechanisms determine the timing and intensity of deep sleep stages in Drosophila.


Assuntos
Encéfalo/fisiologia , Drosophila/fisiologia , Dinâmica não Linear , Fases do Sono/fisiologia , Adenilil Ciclases/metabolismo , Animais , Animais Geneticamente Modificados , Proteína de Ligação a CREB/metabolismo , Proteínas de Drosophila/genética , Proteínas ELAV/genética , Processamento Eletrônico de Dados , Potenciais Evocados/genética , Potenciais Evocados/fisiologia , Feminino , Proteína do X Frágil da Deficiência Intelectual/genética , Regulação da Expressão Gênica/genética , Locomoção , Masculino , Mutação/genética , Optogenética , Estimulação Física , Limiar Sensorial/fisiologia , Privação do Sono , Comportamento Social , Estatísticas não Paramétricas , Sinapses/metabolismo , Fatores de Tempo , Tropomiosina/genética , Vigília
12.
Mol Biol Cell ; 24(3): 285-96, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23223572

RESUMO

Microtubules have been known for decades to be basic elements of the cytoskeleton. They form long, dynamic, rope-like structures within the cell that are essential for mitosis, maintenance of cell shape, and intracellular transport. More recently, in vitro studies have implicated microtubules as signaling molecules that, through changes in their stability, have the potential to trigger growth of axons and dendrites in developing neurons. In this study, we show that specific mutations in the Caenorhabditis elegans mec-7/ß-tubulin gene cause ectopic axon formation in mechanosensory neurons in vivo. In mec-7 mutants, the ALM mechanosensory neuron forms a long ectopic neurite that extends posteriorly, a phenotype that can be mimicked in wild-type worms with a microtubule-stabilizing drug (paclitaxel), and suppressed by mutations in unc-33/CRMP2 and the kinesin-related gene, vab-8. Our results also reveal that these ectopic neurites contain RAB-3, a marker for presynaptic loci, suggesting that they have axon-like properties. Interestingly, in contrast with the excessive axonal growth observed during development, mec-7 mutants are inhibited in axonal regrowth and remodeling following axonal injury. Together our results suggest that MEC-7/ß-tubulin integrity is necessary for the correct number of neurites a neuron generates in vivo and for the capacity of an axon to regenerate.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiologia , Regeneração Nervosa/genética , Neuritos/fisiologia , Tubulina (Proteína)/genética , Animais , Caenorhabditis elegans/citologia , Proteínas de Caenorhabditis elegans/metabolismo , Colchicina/farmacologia , Genes Dominantes , Proteínas Quinases Ativadas por Mitógeno/genética , Mutação , Fatores de Crescimento Neural/genética , Neuritos/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Fenótipo , Polimorfismo de Nucleotídeo Único , Transporte Proteico , Análise de Sequência de DNA , Sinapses/metabolismo , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/farmacologia , Proteínas Wnt/genética
13.
Mol Microbiol ; 82(5): 1164-84, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22059885

RESUMO

In order to cause disease fungal pathogens must be capable of evading or tolerating the host immune defence system. One commonly utilized evasion mechanism is the ability to continually reside within macrophages of the innate immune system and survive subsequent phagocytic destruction. For intracellular growth to occur, fungal pathogens which typically grow in a filamentous hyphal form in the environment must be able to switch growth to a unicellular yeast growth form in a process known as dimorphic switching. The cue to undergo dimorphic switching relies on the recognition of, and response to, the intracellular host environment. Two-component signalling systems are utilized by eukaryotes to sense and respond to changes in the external environment. This study has investigated the role of the hybrid histidine kinase components encoded by drkA and slnA, in the dimorphic pathogen Penicillium marneffei. Both SlnA and DrkA are required for stress adaptation but are uniquely required for different aspects of asexual development, hyphal morphogenesis and cell wall integrity. Importantly, slnA and drkA are both essential for the generation of yeast cells in vivo, with slnA required for the germination of conidia and drkA required for dimorphic switching during macrophage infection.


Assuntos
Macrófagos/microbiologia , Penicillium/enzimologia , Penicillium/patogenicidade , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Células Cultivadas , Regulação Fúngica da Expressão Gênica , Histidina Quinase , Humanos , Microscopia , Penicillium/citologia , Penicillium/crescimento & desenvolvimento , Filogenia , Homologia de Sequência de Aminoácidos , Estresse Fisiológico , Virulência
14.
PLoS Biol ; 9(9): e1001157, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21949641

RESUMO

Nervous system function requires proper development of two functional and morphological domains of neurons, axons and dendrites. Although both these domains are equally important for signal transmission, our understanding of dendrite development remains relatively poor. Here, we show that in C. elegans the Wnt ligand, LIN-44, and its Frizzled receptor, LIN-17, regulate dendrite development of the PQR oxygen sensory neuron. In lin-44 and lin-17 mutants, PQR dendrites fail to form, display stunted growth, or are misrouted. Manipulation of temporal and spatial expression of LIN-44, combined with cell-ablation experiments, indicates that this molecule is patterned during embryogenesis and acts as an attractive cue to define the site from which the dendrite emerges. Genetic interaction between lin-44 and lin-17 suggests that the LIN-44 signal is transmitted through the LIN-17 receptor, which acts cell autonomously in PQR. Furthermore, we provide evidence that LIN-17 interacts with another Wnt molecule, EGL-20, and functions in parallel to MIG-1/Frizzled in this process. Taken together, our results reveal a crucial role for Wnt and Frizzled molecules in regulating dendrite development in vivo.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans , Dendritos/metabolismo , Receptores Frizzled/genética , Regulação da Expressão Gênica no Desenvolvimento , Glicoproteínas/genética , Larva , Neurônios/metabolismo , Receptores Acoplados a Proteínas G/genética , Animais , Axônios/metabolismo , Axônios/ultraestrutura , Padronização Corporal/genética , Caenorhabditis elegans/embriologia , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Dendritos/genética , Dendritos/ultraestrutura , Receptores Frizzled/metabolismo , Genômica , Glicoproteínas/metabolismo , Larva/citologia , Larva/genética , Larva/metabolismo , Microscopia de Fluorescência , Mutação , Neurogênese/genética , Neurônios/citologia , Oxigênio , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...