Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Curr Biol ; 34(11): 2487-2501.e3, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38772361

ABSTRACT

Sleep is broadly conserved across the animal kingdom but can vary widely between species. It is currently unclear which selective pressures and regulatory mechanisms influence differences in sleep between species. The fruit fly Drosophila melanogaster has become a successful model system for examining sleep regulation and function, but little is known about the sleep patterns in many related fly species. Here, we find that fly species with adaptations to extreme desert environments, including D. mojavensis, exhibit strong increases in baseline sleep compared with D. melanogaster. Long-sleeping D. mojavensis show intact homeostasis, indicating that desert flies carry an elevated drive for sleep. In addition, D. mojavensis exhibit altered abundance or distribution of several sleep/wake-related neuromodulators and neuropeptides that are consistent with their reduced locomotor activity and increased sleep. Finally, we find that in a nutrient-deprived environment, the sleep patterns of individual D. mojavensis are strongly correlated with their survival time and that disrupting sleep via constant light stimulation renders D. mojavensis more sensitive to starvation. Our results demonstrate that D. mojavensis is a novel model for studying organisms with high sleep drive and for exploring sleep strategies that provide resilience in extreme environments.


Subject(s)
Drosophila , Sleep , Animals , Sleep/physiology , Drosophila/physiology , Drosophila melanogaster/physiology , Stress, Physiological , Female , Male , Desert Climate , Species Specificity
2.
Proc Natl Acad Sci U S A ; 121(13): e2312664121, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38498719

ABSTRACT

Sleep is an evolutionarily conserved state that supports brain functions, including synaptic plasticity, in species across the animal kingdom. Here, we examine the neuroanatomical and cell-type distribution of presynaptic scaling in the fly brain after sleep loss. We previously found that sleep loss drives accumulation of the active zone scaffolding protein Bruchpilot (BRP) within cholinergic Kenyon cells of the Drosophila melanogaster mushroom body (MB), but not in other classes of MB neurons. To test whether similar cell type-specific trends in plasticity occur broadly across the brain, we used a flp-based genetic reporter to label presynaptic BRP in cholinergic, dopaminergic, GABAergic, or glutamatergic neurons. We then collected whole-brain confocal image stacks of BRP intensity to systematically quantify BRP, a marker of presynapse abundance, across 37 neuropil regions of the central fly brain. Our results indicate that sleep loss, either by overnight (12-h) mechanical stimulation or chronic sleep disruption in insomniac mutants, broadly elevates cholinergic synapse abundance across the brain, while synapse abundance in neurons that produce other neurotransmitters undergoes weaker, if any, changes. Extending sleep deprivation to 24 h drives brain-wide upscaling in glutamatergic, but not other, synapses. Finally, overnight male-male social pairings induce increased BRP in excitatory synapses despite male-female pairings eliciting more waking activity, suggesting experience-specific plasticity. Within neurotransmitter class and waking context, BRP changes are similar across the 37 neuropil domains, indicating that similar synaptic scaling rules may apply across the brain during acute sleep loss and that sleep need may broadly alter excitatory-inhibitory balance in the central brain.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Animals , Female , Male , Drosophila melanogaster/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Sleep Deprivation/metabolism , Synapses/metabolism , Brain/metabolism , Cholinergic Agents
3.
eNeuro ; 10(9)2023 09.
Article in English | MEDLINE | ID: mdl-37679041

ABSTRACT

Recent work in Drosophila has uncovered several neighboring classes of sleep-regulatory neurons within the central complex. However, the logic of connectivity and network motifs remains limited by the incomplete examination of relevant cell types. Using a recent genetic-anatomic classification of ellipsoid body ring neurons, we conducted a thermogenetic screen in female flies to assess sleep/wake behavior and identified two wake-promoting drivers that label ER3d neurons and two sleep-promoting drivers that express in ER3m cells. We then used intersectional genetics to refine driver expression patterns. Activation of ER3d cells shortened sleep bouts, suggesting a key role in sleep maintenance. While sleep-promoting drivers from our mini-screen label overlapping ER3m neurons, intersectional strategies cannot rule out sleep regulatory roles for additional neurons in their expression patterns. Suppressing GABA synthesis in ER3m neurons prevents postinjury sleep, and GABAergic ER3d cells are required for thermogenetically induced wakefulness. Finally, we use an activity-dependent fluorescent reporter for putative synaptic contacts to embed these neurons within the known sleep-regulatory network. ER3m and ER3d neurons may receive connections from wake-active Helicon/ExR1 cells, and ER3m neurons likely inhibit ER3d neurons. Together, these data suggest a neural mechanism by which previously uncharacterized circuit elements stabilize sleep-wake states.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Female , Sleep/physiology , Neurons/physiology , Wakefulness/physiology , Drosophila melanogaster/physiology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism
4.
Cell ; 186(13): 2730-2732, 2023 06 22.
Article in English | MEDLINE | ID: mdl-37352834

ABSTRACT

In this issue of Cell, we see first evidence of sleep-dependent circuit remodeling alongside behavioral memory consolidation in C. elegans. Examining memory of a never-rewarded odor during post-training sleep from synapse to behavior all in one organism opens the opportunity to use this well-mapped nervous system to study mechanisms of sleep-dependent memory consolidation.


Subject(s)
Caenorhabditis elegans , Memory Consolidation , Animals , Sleep/physiology , Memory Consolidation/physiology
5.
bioRxiv ; 2023 May 28.
Article in English | MEDLINE | ID: mdl-37292829

ABSTRACT

Sleep is broadly conserved across the animal kingdom, but can vary widely between species. It is currently unclear which types of selective pressures and sleep regulatory mechanisms influence differences in sleep between species. The fruit fly Drosophila melanogaster has become a successful model system for examining sleep regulation and function, but little is known about the sleep patterns and need for sleep in many related fly species. Here, we find that Drosophila mojavensis, a fly species that has adapted to extreme desert environments, exhibits strong increases in sleep compared to D. melanogaster. Long-sleeping D. mojavensis show intact sleep homeostasis, indicating that these flies carry an elevated need for sleep. In addition, D. mojavensis exhibit altered abundance or distribution of several sleep/wake related neuromodulators and neuropeptides that are consistent with their reduced locomotor activity, and increased sleep. Finally, we find that in a nutrient-deprived environment, the sleep responses of individual D. mojavensis are correlated with their survival time. Our results demonstrate that D. mojavensis is a novel model for studying organisms with high sleep need, and for exploring sleep strategies that provide resilience in extreme environments.

6.
PLoS Genet ; 18(11): e1010289, 2022 11.
Article in English | MEDLINE | ID: mdl-36409783

ABSTRACT

The Serotonin Transporter (SERT) regulates extracellular serotonin levels and is the target of most current drugs used to treat depression. The mechanisms by which inhibition of SERT activity influences behavior are poorly understood. To address this question in the model organism Drosophila melanogaster, we developed new loss of function mutations in Drosophila SERT (dSERT). Previous studies in both flies and mammals have implicated serotonin as an important neuromodulator of sleep, and our newly generated dSERT mutants show an increase in total sleep and altered sleep architecture that is mimicked by feeding the SSRI citalopram. Differences in daytime versus nighttime sleep architecture as well as genetic rescue experiments unexpectedly suggest that distinct serotonergic circuits may modulate daytime versus nighttime sleep. dSERT mutants also show defects in copulation and food intake, akin to the clinical side effects of SSRIs and consistent with the pleomorphic influence of serotonin on the behavior of D. melanogaster. Starvation did not overcome the sleep drive in the mutants and in male dSERT mutants, the drive to mate also failed to overcome sleep drive. dSERT may be used to further explore the mechanisms by which serotonin regulates sleep and its interplay with other complex behaviors.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Animals , Male , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Serotonin , Courtship , Drosophila/metabolism , Sleep/genetics , Mutation , Feeding Behavior , Mammals/metabolism
7.
Curr Biol ; 31(15): 3248-3261.e3, 2021 08 09.
Article in English | MEDLINE | ID: mdl-34107302

ABSTRACT

Sleep is essential for a variety of plastic processes, including learning and memory. However, the consequences of insufficient sleep on circuit connectivity remain poorly understood. To better appreciate the effects of sleep loss on synaptic connectivity across a memory-encoding circuit, we examined changes in the distribution of synaptic markers in the Drosophila mushroom body (MB). Protein-trap tags for active zone components indicate that recent sleep time is inversely correlated with Bruchpilot (BRP) abundance in the MB lobes; sleep loss elevates BRP while sleep induction reduces BRP across the MB. Overnight sleep deprivation also elevated levels of dSyd-1 and Cacophony, but not other pre-synaptic proteins. Cell-type-specific genetic reporters show that MB-intrinsic Kenyon cells (KCs) exhibit increased pre-synaptic BRP throughout the axonal lobes after sleep deprivation; similar increases were not detected in projections from large interneurons or dopaminergic neurons that innervate the MB. These results indicate that pre-synaptic plasticity in KCs is responsible for elevated levels of BRP in the MB lobes of sleep-deprived flies. Because KCs provide synaptic inputs to several classes of post-synaptic partners, we next used a fluorescent reporter for synaptic contacts to test whether each class of KC output connections is scaled uniformly by sleep loss. The KC output synapses that we observed here can be divided into three classes: KCs to MB interneurons; KCs to dopaminergic neurons; and KCs to MB output neurons. No single class showed uniform scaling across each constituent member, indicating that different rules may govern plasticity during sleep loss across cell types.


Subject(s)
Drosophila Proteins , Mushroom Bodies , Animals , Dopaminergic Neurons , Drosophila/physiology , Drosophila Proteins/genetics , Mushroom Bodies/physiology , Sleep Deprivation , Synapses/physiology
8.
Front Behav Neurosci ; 15: 777799, 2021.
Article in English | MEDLINE | ID: mdl-35126067

ABSTRACT

Sleep is a vital physiological state that has been broadly conserved across the evolution of animal species. While the precise functions of sleep remain poorly understood, a large body of research has examined the negative consequences of sleep loss on neural and behavioral plasticity. While sleep disruption generally results in degraded neural plasticity and cognitive function, the impact of sleep loss can vary widely with age, between individuals, and across physiological contexts. Additionally, several recent studies indicate that sleep loss differentially impacts distinct neuronal populations within memory-encoding circuitry. These findings indicate that the negative consequences of sleep loss are not universally shared, and that identifying conditions that influence the resilience of an organism (or neuron type) to sleep loss might open future opportunities to examine sleep's core functions in the brain. Here, we discuss the functional roles for sleep in adaptive plasticity and review factors that can contribute to individual variations in sleep behavior and responses to sleep loss.

9.
Curr Biol ; 30(6): 1063-1076.e3, 2020 03 23.
Article in English | MEDLINE | ID: mdl-32142703

ABSTRACT

Following acute neural injury, severed axons undergo programmed Wallerian degeneration over several following days. While sleep has been linked with synaptic reorganization under other conditions, the role of sleep in responses to neural injuries remains poorly understood. To study the relationship between sleep and neural injury responses, we examined Drosophila melanogaster following the removal of antennae or other sensory tissues. Daytime sleep is elevated after antennal or wing injury, but sleep returns to baseline levels within 24 h after injury. Similar increases in sleep are not observed when olfactory receptor neurons are silenced or when other sensory organs are severed, suggesting that increased sleep after injury is not attributed to sensory deprivation, nociception, or generalized inflammatory responses. Neuroprotective disruptions of the E3 ubiquitin ligase highwire and c-Jun N-terminal kinase basket in olfactory receptor neurons weaken the sleep-promoting effects of antennal injury, suggesting that post-injury sleep may be influenced by the clearance of damaged neurons. Finally, we show that pre-synaptic active zones are preferentially removed from severed axons within hours after injury and that depriving recently injured flies of sleep slows the removal of both active zones and damaged axons. These data support a bidirectional interaction between sleep and synapse pruning after antennal injury: locally increasing the need to clear neural debris is associated with increased sleep, which is required for efficient active zone removal after injury.


Subject(s)
Arthropod Antennae/physiopathology , Drosophila melanogaster/physiology , Sleep/physiology , Synapses/physiology , Wings, Animal/physiopathology , Animals , Arthropod Antennae/injuries , Disease Models, Animal , Female , Olfactory Receptor Neurons/physiology , Wings, Animal/injuries
10.
Curr Opin Neurobiol ; 54: 120-126, 2019 02.
Article in English | MEDLINE | ID: mdl-30366270

ABSTRACT

Sleep has been universally conserved across animal species. The basic functions of sleep remain unclear, but insufficient sleep impairs memory acquisition and retention in both vertebrates and invertebrates. Sleep is also a homeostatic process that is influenced not only by the amount of time awake, but also by neural activity and plasticity. Because of the breadth and precision of available genetic tools, the fruit fly has become a powerful model system to understand sleep regulation and function. Importantly, these tools enable the dissection of memory-encoding circuits at the level of individual neurons, and have allowed the development of genetic tools to induce sleep on-demand. This review describes recent investigations of the role for sleep in memory using Drosophila and current hypotheses of sleep's functions for supporting plasticity, learning, and memory.


Subject(s)
Diptera/physiology , Memory/physiology , Sleep/physiology , Animals , Brain/cytology , Neurons/physiology , Synapses/physiology
11.
Neuron ; 97(2): 378-389.e4, 2018 01 17.
Article in English | MEDLINE | ID: mdl-29307711

ABSTRACT

Sleep-promoting neurons in the dorsal fan-shaped body (dFB) of Drosophila are integral to sleep homeostasis, but how these cells impose sleep on the organism is unknown. We report that dFB neurons communicate via inhibitory transmitters, including allatostatin-A (AstA), with interneurons connecting the superior arch with the ellipsoid body of the central complex. These "helicon cells" express the galanin receptor homolog AstA-R1, respond to visual input, gate locomotion, and are inhibited by AstA, suggesting that dFB neurons promote rest by suppressing visually guided movement. Sleep changes caused by enhanced or diminished allatostatinergic transmission from dFB neurons and by inhibition or optogenetic stimulation of helicon cells support this notion. Helicon cells provide excitation to R2 neurons of the ellipsoid body, whose activity-dependent plasticity signals rising sleep pressure to the dFB. By virtue of this autoregulatory loop, dFB-mediated inhibition interrupts processes that incur a sleep debt, allowing restorative sleep to rebalance the books. VIDEO ABSTRACT.


Subject(s)
Drosophila melanogaster/physiology , Interneurons/physiology , Sleep/physiology , Animals , Brain/physiology , Circadian Rhythm , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Excitatory Postsynaptic Potentials/physiology , Female , Homeostasis , Insect Hormones/physiology , Light , Locomotion/radiation effects , Male , Membrane Potentials , Nerve Tissue Proteins/physiology , Neurons/physiology , Optogenetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/physiology , Recombinant Fusion Proteins/metabolism , Vision, Ocular
12.
Curr Opin Insect Sci ; 24: 51-57, 2017 12.
Article in English | MEDLINE | ID: mdl-29208223

ABSTRACT

Sleep is necessary for survival, and prolonged waking causes a homeostatic increase in the need for recovery sleep. Homeostasis is a core component of sleep regulation and has been tightly conserved across evolution from invertebrates to man. Homeostatic sleep regulation was first identified among insects in cockroaches several decades ago, but the characterization of sleep rebound in Drosophila melanogaster opened the use of insect model species to understand homeostatic functions and regulation of sleep. This review describes circuits in two neuropil structures, the central complex and mushroom bodies, that influence sleep homeostasis and neuromodulatory systems that influence the accrual of homeostatic sleep need.


Subject(s)
Drosophila melanogaster/physiology , Homeostasis/physiology , Mushroom Bodies/physiology , Sleep/physiology , Animals , Neuropil/physiology , Neurotransmitter Agents/metabolism , Stress, Physiological
13.
Curr Opin Neurobiol ; 44: 228-235, 2017 06.
Article in English | MEDLINE | ID: mdl-28628804

ABSTRACT

Sleep homeostasis is a fundamental property of vigilance state regulation that is highly conserved across species. Neuronal systems and circuits that underlie sleep homeostasis are not well understood. In Drosophila, a neuronal circuit involving neurons in the ellipsoid body and in the dorsal Fan-shaped body is a candidate for both tracing sleep need during waking and translating it to increased sleep drive and expression. Sleep homeostasis in rats and mice involves multiple neuromodulators acting on multiple wake- and sleep-promoting neuronal systems. A functional central homeostat emerges from A1 receptor mediated actions of adenosine on wake-promoting neurons in the basal forebrain and hypothalamus, and A2A adenosine receptor-mediated actions on sleep-promoting neurons in the preoptic hypothalamus and nucleus accumbens.


Subject(s)
Homeostasis/physiology , Neurons/physiology , Sleep/physiology , Adenosine/metabolism , Animals , Wakefulness/physiology
14.
Nature ; 536(7616): 333-337, 2016 08 18.
Article in English | MEDLINE | ID: mdl-27487216

ABSTRACT

Sleep disconnects animals from the external world, at considerable risks and costs that must be offset by a vital benefit. Insight into this mysterious benefit will come from understanding sleep homeostasis: to monitor sleep need, an internal bookkeeper must track physiological changes that are linked to the core function of sleep. In Drosophila, a crucial component of the machinery for sleep homeostasis is a cluster of neurons innervating the dorsal fan-shaped body (dFB) of the central complex. Artificial activation of these cells induces sleep, whereas reductions in excitability cause insomnia. dFB neurons in sleep-deprived flies tend to be electrically active, with high input resistances and long membrane time constants, while neurons in rested flies tend to be electrically silent. Correlative evidence thus supports the simple view that homeostatic sleep control works by switching sleep-promoting neurons between active and quiescent states. Here we demonstrate state switching by dFB neurons, identify dopamine as a neuromodulator that operates the switch, and delineate the switching mechanism. Arousing dopamine caused transient hyperpolarization of dFB neurons within tens of milliseconds and lasting excitability suppression within minutes. Both effects were transduced by Dop1R2 receptors and mediated by potassium conductances. The switch to electrical silence involved the downregulation of voltage-gated A-type currents carried by Shaker and Shab, and the upregulation of voltage-independent leak currents through a two-pore-domain potassium channel that we term Sandman. Sandman is encoded by the CG8713 gene and translocates to the plasma membrane in response to dopamine. dFB-restricted interference with the expression of Shaker or Sandman decreased or increased sleep, respectively, by slowing the repetitive discharge of dFB neurons in the ON state or blocking their entry into the OFF state. Biophysical changes in a small population of neurons are thus linked to the control of sleep-wake state.


Subject(s)
Drosophila melanogaster/physiology , Homeostasis , Sleep/physiology , Animals , Cell Membrane/metabolism , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/cytology , Electric Conductivity , Female , Male , Neurotransmitter Agents/metabolism , Optogenetics , Potassium/metabolism , Potassium Channels/chemistry , Potassium Channels/metabolism , Protein Transport , Receptors, Dopamine/metabolism , Shaker Superfamily of Potassium Channels/metabolism , Sleep Deprivation , Sleep Initiation and Maintenance Disorders/physiopathology , Time Factors , Wakefulness/physiology
15.
Sleep ; 37(9): 1427-37, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25142573

ABSTRACT

STUDY OBJECTIVES: Aging has been linked with decreased neural plasticity and memory formation in humans and in laboratory model species such as the fruit fly, Drosophila melanogaster. Here, we examine plastic responses following social experience in Drosophila as a high-throughput method to identify interventions that prevent these impairments. PATIENTS OR PARTICIPANTS: Wild-type and transgenic Drosophila melanogaster. DESIGN AND INTERVENTIONS: Young (5-day old) or aged (20-day old) adult female Drosophila were housed in socially enriched (n = 35-40) or isolated environments, then assayed for changes in sleep and for structural markers of synaptic terminal growth in the ventral lateral neurons (LNVs) of the circadian clock. MEASUREMENTS AND RESULTS: When young flies are housed in a socially enriched environment, they exhibit synaptic elaboration within a component of the circadian circuitry, the LNVs, which is followed by increased sleep. Aged flies, however, no longer exhibit either of these plastic changes. Because of the tight correlation between neural plasticity and ensuing increases in sleep, we use sleep after enrichment as a high-throughput marker for neural plasticity to identify interventions that prolong youthful plasticity in aged flies. To validate this strategy, we find three independent genetic manipulations that delay age-related losses in plasticity: (1) elevation of dopaminergic signaling, (2) over-expression of the transcription factor blistered (bs) in the LNVs, and (3) reduction of the Imd immune signaling pathway. These findings provide proof-of-principle evidence that measuring changes in sleep in flies after social enrichment may provide a highly scalable assay for the study of age-related deficits in synaptic plasticity. CONCLUSIONS: These studies demonstrate that Drosophila provides a promising model for the study of age-related loss of neural plasticity and begin to identify genes that might be manipulated to delay the onset of functional senescence.


Subject(s)
Aging/genetics , Aging/physiology , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Social Behavior , Animals , Animals, Genetically Modified , Biological Assay , Biomarkers , Carrier Proteins/genetics , Carrier Proteins/metabolism , Circadian Rhythm/physiology , Dopaminergic Neurons/physiology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Female , High-Throughput Screening Assays , Immunity/genetics , Male , Memory/physiology , Models, Animal , Reproducibility of Results , Signal Transduction , Sleep/physiology , Social Isolation , Synapses/physiology , Time Factors
16.
Neuron ; 81(4): 860-72, 2014 02 19.
Article in English | MEDLINE | ID: mdl-24559676

ABSTRACT

Sleep is under homeostatic control, but the mechanisms that sense sleep need and correct sleep deficits remain unknown. Here, we report that sleep-promoting neurons with projections to the dorsal fan-shaped body (FB) form the output arm of Drosophila's sleep homeostat. Homeostatic sleep control requires the Rho-GTPase-activating protein encoded by the crossveinless-c (cv-c) gene in order to transduce sleep pressure into increased electrical excitability of dorsal FB neurons. cv-c mutants exhibit decreased sleep time, diminished sleep rebound, and memory deficits comparable to those after sleep loss. Targeted ablation and rescue of Cv-c in sleep-control neurons of the dorsal FB impair and restore, respectively, normal sleep patterns. Sleep deprivation increases the excitability of dorsal FB neurons, but this homeostatic adjustment is disrupted in short-sleeping cv-c mutants. Sleep pressure thus shifts the input-output function of sleep-promoting neurons toward heightened activity by modulating ion channel function in a mechanism dependent on Cv-c.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , GTPase-Activating Proteins/genetics , Homeostasis/physiology , Mutation/genetics , Neurons/metabolism , Sleep/genetics , Animals , GTPase-Activating Proteins/metabolism , Sleep Deprivation/genetics , Sleep Deprivation/metabolism
17.
Neuron ; 81(6): 1442, 2014 Mar 19.
Article in English | MEDLINE | ID: mdl-28898631
18.
Science ; 332(6037): 1571-6, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21700877

ABSTRACT

Sleep is believed to play an important role in memory consolidation. We induced sleep on demand by expressing the temperature-gated nonspecific cation channel Transient receptor potential cation channel (UAS-TrpA1) in neurons, including those with projections to the dorsal fan-shaped body (FB). When the temperature was raised to 31°C, flies entered a quiescent state that meets the criteria for identifying sleep. When sleep was induced for 4 hours after a massed-training protocol for courtship conditioning that is not capable of inducing long-term memory (LTM) by itself, flies develop an LTM. Activating the dorsal FB in the absence of sleep did not result in the formation of LTM after massed training.


Subject(s)
Drosophila/physiology , Memory, Long-Term/physiology , Neurons/physiology , Sleep/physiology , Animals , Conditioning, Psychological , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Female , Models, Animal , Motor Activity , Presynaptic Terminals/physiology , Social Isolation , Temperature , Transcription Factors/genetics , Transcription Factors/metabolism , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism
19.
Science ; 324(5923): 105-8, 2009 Apr 03.
Article in English | MEDLINE | ID: mdl-19342592

ABSTRACT

Sleep is important for memory consolidation and is responsive to waking experience. Clock circuitry is uniquely positioned to coordinate interactions between processes underlying memory and sleep need. Flies increase sleep both after exposure to an enriched social environment and after protocols that induce long-term memory. We found that flies mutant for rutabaga, period, and blistered were deficient for experience-dependent increases in sleep. Rescue of each of these genes within the ventral lateral neurons (LNVs) restores increased sleep after social enrichment. Social experiences that induce increased sleep were associated with an increase in the number of synaptic terminals in the LNV projections into the medulla. The number of synaptic terminals was reduced during sleep and this decline was prevented by sleep deprivation.


Subject(s)
Drosophila melanogaster/physiology , Neuronal Plasticity , Neurons/physiology , Sleep/physiology , Synapses/physiology , Adenylyl Cyclases/genetics , Adenylyl Cyclases/physiology , Animals , Biological Clocks/genetics , Brain/physiology , Circadian Rhythm/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Genes, Insect , Male , Memory , Models, Animal , Mutation , Neurons/ultrastructure , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Period Circadian Proteins , Presynaptic Terminals/physiology , Presynaptic Terminals/ultrastructure , Receptors, Invertebrate Peptide/genetics , Receptors, Invertebrate Peptide/metabolism , Serum Response Factor/genetics , Serum Response Factor/physiology , Sleep Deprivation , Social Behavior
20.
Adv Genet ; 68: 57-81, 2009.
Article in English | MEDLINE | ID: mdl-20109659

ABSTRACT

Social experience alters the expression of genes related to synaptic function and plasticity, induces elaborations in the morphology of neural structures throughout the brain (Volkmar and Greenough, 1972; Greenough et al., 1978; Technau, 2007), improves cognitive and behavioral performance (Pham et al., 1999a; Toscano et al., 2006) and alters subsequent sleep (Ganguly-Fitzgerald et al., 2006). In this review, we discuss the plastic mechanisms that are induced in response to social experience and how social enrichment can provide insight into the biological functions of sleep.


Subject(s)
Behavior, Animal , Neuronal Plasticity , Sleep , Animals , Social Behavior
SELECTION OF CITATIONS
SEARCH DETAIL
...