Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Cell ; 163(5): 1214-1224, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-26590423

ABSTRACT

Circadian clocks integrate light and temperature input to remain synchronized with the day/night cycle. Although light input to the clock is well studied, the molecular mechanisms by which circadian clocks respond to temperature remain poorly understood. We found that temperature phase shifts Drosophila circadian clocks through degradation of the pacemaker protein TIM. This degradation is mechanistically distinct from photic CRY-dependent TIM degradation. Thermal TIM degradation is triggered by cytosolic calcium increase and CALMODULIN binding to TIM and is mediated by the atypical calpain protease SOL. This thermal input pathway and CRY-dependent light input thus converge on TIM, providing a molecular mechanism for the integration of circadian light and temperature inputs. Mammals use body temperature cycles to keep peripheral clocks synchronized with their brain pacemaker. Interestingly, downregulating the mammalian SOL homolog SOLH blocks thermal mPER2 degradation and phase shifts. Thus, we propose that circadian thermosensation in insects and mammals share common principles.


Subject(s)
Circadian Clocks , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Nerve Tissue Proteins/metabolism , Animals , Biological Clocks , Calcium Signaling , Calmodulin/metabolism , Calpain , Circadian Rhythm , Male , Mammals/physiology , Proteolysis
2.
Trends Genet ; 40(5): 387-397, 2024 May.
Article in English | MEDLINE | ID: mdl-38336520

ABSTRACT

The coastline is a particularly challenging environment for its inhabitants. Not only do they have to cope with the solar day and the passing of seasons, but they must also deal with tides. In addition, many marine species track the phase of the moon, especially to coordinate reproduction. Marine animals show remarkable behavioral and physiological adaptability, using biological clocks to anticipate specific environmental cycles. Presently, we lack a basic understanding of the molecular mechanisms underlying circatidal and circalunar clocks. Recent advances in genome engineering and the development of genetically tractable marine model organisms are transforming how we study these timekeeping mechanisms and opening a novel era in marine chronobiology.


Subject(s)
Aquatic Organisms , Gene Editing , Animals , Aquatic Organisms/genetics , Genome/genetics , Biological Clocks/genetics , Circadian Rhythm/genetics
4.
Mol Psychiatry ; 26(12): 7793-7802, 2021 12.
Article in English | MEDLINE | ID: mdl-34471250

ABSTRACT

Dopamine (DA) is required for movement, sleep, and reward, and DA signaling is tightly controlled by the presynaptic DA transporter (DAT). Therapeutic and addictive psychostimulants, including methylphenidate (Ritalin; MPH), cocaine, and amphetamine (AMPH), markedly elevate extracellular DA via their actions as competitive DAT inhibitors (MPH, cocaine) and substrates (AMPH). DAT silencing in mice and invertebrates results in hyperactivity, reduced sleep, and blunted psychostimulant responses, highlighting DAT's essential role in DA-dependent behaviors. DAT surface expression is not static; rather it is dynamically regulated by endocytic trafficking. PKC-stimulated DAT endocytosis requires the neuronal GTPase, Rit2, and Rit2 silencing in mouse DA neurons impacts psychostimulant sensitivity. However, it is unknown whether or not Rit2-mediated changes in psychostimulant sensitivity are DAT-dependent. Here, we leveraged Drosophila melanogaster to test whether the Drosophila Rit2 ortholog, Ric, impacts dDAT function, trafficking, and DA-dependent behaviors. Orthologous to hDAT and Rit2, dDAT and Ric directly interact, and the constitutively active Ric mutant Q117L increased dDAT surface levels and function in cell lines and ex vivo Drosophila brains. Moreover, DAergic RicQ117L expression caused sleep fragmentation in a DAT-dependent manner but had no effect on total sleep and daily locomotor activity. Importantly, we found that Rit2 is required for AMPH-stimulated DAT internalization in mouse striatum, and that DAergic RicQ117L expression significantly increased Drosophila AMPH sensitivity in a DAT-dependent manner, suggesting a conserved impact of Ric-dependent DAT trafficking on AMPH sensitivity. These studies support that the DAT/Rit2 interaction impacts both baseline behaviors and AMPH sensitivity, potentially by regulating DAT trafficking.


Subject(s)
Dopamine Plasma Membrane Transport Proteins/metabolism , Drosophila Proteins/metabolism , Monomeric GTP-Binding Proteins , ras Proteins/metabolism , Amphetamine/pharmacology , Animals , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/genetics , Dopaminergic Neurons/metabolism , Drosophila melanogaster , GTP Phosphohydrolases/metabolism , Mice , Monomeric GTP-Binding Proteins/metabolism , Sleep Quality
5.
Proc Natl Acad Sci U S A ; 114(32): E6669-E6677, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28743754

ABSTRACT

The physiology and behavior of many organisms are subject to daily cycles. In Drosophila melanogaster the daily locomotion patterns of single flies are characterized by bursts of activity at dawn and dusk. Two distinct clusters of clock neurons-morning oscillators (M cells) and evening oscillators (E cells)-are largely responsible for these activity bursts. In contrast, male-female pairs of flies follow a distinct pattern, most notably characterized by an activity trough at dusk followed by a high level of male courtship during the night. This male sex drive rhythm (MSDR) is mediated by the M cells along with DN1 neurons, a cluster of clock neurons located in the dorsal posterior region of the brain. Here we report that males lacking Salt-inducible kinase 3 (SIK3) expression in M cells exhibit a short period of MSDR but a long period of single-fly locomotor rhythm (SLR). Moreover, lack of Sik3 in M cells decreases the amplitude of PERIOD (PER) cycling in DN1 neurons, suggesting that SIK3 non-cell-autonomously regulates DN1 neurons' molecular clock. We also show that Sik3 reduction interferes with circadian nucleocytoplasmic shuttling of Histone deacetylase 4 (HDAC4), a SIK3 phosphorylation target, in clock neurons and that constitutive HDAC4 localization in the nucleus shortens the period of MSDR. Taking these findings together, we conclude that SIK3-HDAC4 signaling in M cells regulates MSDR by regulating the molecular oscillation in DN1 neurons.


Subject(s)
Circadian Rhythm/physiology , Drosophila Proteins/metabolism , Histone Deacetylases/metabolism , Neurons/enzymology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Histone Deacetylases/genetics , Male , Neurons/cytology , Protein Serine-Threonine Kinases/genetics
6.
J Neurosci ; 38(27): 6161-6171, 2018 07 04.
Article in English | MEDLINE | ID: mdl-29875268

ABSTRACT

Light is one of the chief environmental cues that reset circadian clocks. In Drosophila, CRYPTOCHROME (CRY) mediates acute photic resetting of circadian clocks by promoting the degradation of TIMELESS in a cell-autonomous manner. Thus, even circadian oscillators in peripheral organs can independently perceive light in Drosophila However, there is substantial evidence for nonautonomous mechanisms of circadian photoreception in the brain. We have previously shown that the morning (M) and evening (E) oscillators are critical light-sensing neurons that cooperate to shift the phase of circadian behavior in response to light input. We show here that light can efficiently phase delay or phase advance circadian locomotor behavior in male Drosophila even when either the M- or the E-oscillators are ablated, suggesting that behavioral phase shifts and their directionality are largely a consequence of the cell-autonomous nature of CRY-dependent photoreception. Our observation that the phase response curves of brain and peripheral oscillators are remarkably similar further supports this idea. Nevertheless, the neural network modulates circadian photoresponses. We show that the M-oscillator neurotransmitter pigment dispersing factor plays a critical role in the coordination between M- and E-oscillators after light exposure, and we uncover a potential role for a subset of dorsal neurons in the control of phase advances. Thus, neural modulation of autonomous light detection might play an important role in the plasticity of circadian behavior.SIGNIFICANCE STATEMENT Input pathways provide circadian rhythms with the flexibility needed to harmonize their phase with environmental cycles. Light is the chief environmental cue that synchronizes circadian clocks. In Drosophila, the photoreceptor CRYPTOCHROME resets circadian clocks cell-autonomously. However, recent studies indicate that, in the brain, interactions between clock neurons are critical to reset circadian locomotor behavior. We present evidence supporting the idea that the ability of flies to advance or delay their rhythmic behavior in response to light input essentially results from cell-autonomous photoreception. However, because of their networked organization, we find that circadian neurons have to cooperate to reset the phase of circadian behavior in response to photic cues. Our work thus helps to reconcile cell-autonomous and non-cell-autonomous models of circadian entrainment.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm/physiology , Cryptochromes/metabolism , Drosophila Proteins/metabolism , Eye Proteins/metabolism , Locomotion/physiology , Nerve Net/physiology , Animals , Drosophila , Light , Male , Photoreceptor Cells, Invertebrate/physiology
7.
J Neurosci ; 36(6): 2007-13, 2016 Feb 10.
Article in English | MEDLINE | ID: mdl-26865623

ABSTRACT

Animals use circadian rhythms to anticipate daily environmental changes. Circadian clocks have a profound effect on behavior. In Drosophila, for example, brain pacemaker neurons dictate that flies are mostly active at dawn and dusk. miRNAs are small, regulatory RNAs (≈22 nt) that play important roles in posttranscriptional regulation. Here, we identify miR-124 as an important regulator of Drosophila circadian locomotor rhythms. Under constant darkness, flies lacking miR-124 (miR-124(KO)) have a dramatically advanced circadian behavior phase. However, whereas a phase defect is usually caused by a change in the period of the circadian pacemaker, this is not the case in miR-124(KO) flies. Moreover, the phase of the circadian pacemaker in the clock neurons that control rhythmic locomotion is not altered either. Therefore, miR-124 modulates the output of circadian clock neurons rather than controlling their molecular pacemaker. Circadian phase is also advanced under temperature cycles, but a light/dark cycle partially corrects the defects in miR-124(KO) flies. Indeed, miR-124(KO) shows a normal evening phase under the latter conditions, but morning behavioral activity is suppressed. In summary, miR-124 controls diurnal activity and determines the phase of circadian locomotor behavior without affecting circadian pacemaker function. It thus provides a potent entry point to elucidate the mechanisms by which the phase of circadian behavior is determined. SIGNIFICANCE STATEMENT: In animals, molecular circadian clocks control the timing of behavioral activities to optimize them with the day/night cycle. This is critical for their fitness and survival. The mechanisms by which the phase of circadian behaviors is determined downstream of the molecular pacemakers are not yet well understood. Recent studies indicate that miRNAs are important regulators of circadian outputs. We found that miR-124 shapes diurnal behavioral activity and has a striking impact on the phase of circadian locomotor behavior. Surprisingly, the period and phase of the neural circadian pacemakers driving locomotor rhythms are unaffected. Therefore, miR-124 is a critical modulator of the circadian output pathways that control circadian behavioral rhythms.


Subject(s)
Circadian Rhythm/genetics , Circadian Rhythm/physiology , MicroRNAs/genetics , MicroRNAs/physiology , Motor Activity/genetics , Motor Activity/physiology , Animals , Biological Clocks , Darkness , Drosophila melanogaster , Light , Male , Mutation/genetics , Mutation/physiology , Nerve Net/abnormalities , Photoreceptor Cells, Invertebrate/physiology , Temperature
8.
Methods ; 68(1): 140-50, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24412370

ABSTRACT

Circadian rhythms have a profound influence on most bodily functions: from metabolism to complex behaviors. They ensure that all these biological processes are optimized with the time-of-day. They are generated by endogenous molecular oscillators that have a period that closely, but not exactly, matches day length. These molecular clocks are synchronized by environmental cycles such as light intensity and temperature. Drosophila melanogaster has been a model organism of choice to understand genetically, molecularly and at the level of neural circuits how circadian rhythms are generated, how they are synchronized by environmental cues, and how they drive behavioral cycles such as locomotor rhythms. This review will cover a wide range of techniques that have been instrumental to our understanding of Drosophila circadian rhythms, and that are essential for current and future research.


Subject(s)
Biological Assay/methods , Circadian Rhythm/genetics , Drosophila melanogaster/growth & development , Animals , Behavior, Animal/physiology , Brain/physiology , Circadian Rhythm/physiology , Drosophila melanogaster/genetics , Neurons/metabolism , Temperature
9.
Curr Opin Insect Sci ; 64: 101219, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38848811

ABSTRACT

Sleep is conserved across the animal kingdom, and Drosophila melanogaster is a prime model to understand its intricate circadian and homeostatic control. GABA (gamma-aminobutyric acid), the brain's main inhibitory neurotransmitter, plays a central role in sleep. This review delves into GABA's complex mechanisms of actions within Drosophila's sleep-regulating neural networks. We discuss how GABA promotes sleep, both by inhibiting circadian arousal neurons and by being a key neurotransmitter in sleep homeostatic circuits. GABA's impact on sleep is modulated by glia through astrocytic GABA recapture and metabolism. Interestingly, GABA can be coexpressed with other neurotransmitters in sleep-regulating neurons, which likely contributes to context-based sleep plasticity.


Subject(s)
Drosophila melanogaster , Sleep , gamma-Aminobutyric Acid , Animals , Sleep/physiology , gamma-Aminobutyric Acid/metabolism , Drosophila melanogaster/physiology , Neurons/physiology , Neurons/metabolism , Circadian Rhythm/physiology , Brain/physiology , Brain/metabolism
10.
Elife ; 132024 May 08.
Article in English | MEDLINE | ID: mdl-38716806

ABSTRACT

Studies of the starlet sea anemone provide important insights into the early evolution of the circadian clock in animals.


Subject(s)
Circadian Clocks , Sea Anemones , Animals , Biological Evolution , Circadian Clocks/physiology , Circadian Rhythm/physiology , Cnidaria/physiology , Sea Anemones/physiology
11.
J Neurosci ; 32(47): 16959-70, 2012 Nov 21.
Article in English | MEDLINE | ID: mdl-23175847

ABSTRACT

Circadian rhythms are generated by well-conserved interlocked transcriptional feedback loops in animals. In Drosophila, the dimeric transcription factor CLOCK/CYCLE (CLK/CYC) promotes period (per), timeless (tim), vrille (vri), and PAR-domain protein 1 (Pdp1) transcription. PER and TIM negatively feed back on CLK/CYC transcriptional activity, whereas VRI and PDP1 negatively and positively regulate Clk transcription, respectively. Here, we show that the α isoform of the Drosophila FOS homolog KAYAK (KAY) is required for normal circadian behavior. KAY-α downregulation in circadian pacemaker neurons increases period length by 1.5 h. This behavioral phenotype is correlated with decreased expression of several circadian proteins. The strongest effects are on CLK and the neuropeptide PIGMENT DISPERSING FACTOR, which are both under VRI and PDP1 control. Consistently, KAY-α can bind to VRI and inhibit its interaction with the Clk promoter. Interestingly, KAY-α can also repress CLK activity. Hence, in flies with low KAY-α levels, CLK derepression would partially compensate for increased VRI repression, thus attenuating the consequences of KAY-α downregulation on CLK targets. We propose that the double role of KAY-α in the two transcriptional loops controlling Drosophila circadian behavior brings precision and stability to their oscillations.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , Drosophila Proteins/physiology , Drosophila/physiology , Feedback, Physiological/physiology , Neurons/physiology , Transcription, Genetic/physiology , Animals , Animals, Genetically Modified/physiology , Biological Clocks/genetics , Cells, Cultured , DNA/genetics , Drosophila Proteins/genetics , HEK293 Cells , Humans , Immunohistochemistry , Motor Activity/physiology , Neuropeptides/genetics , Neuropeptides/physiology , Plasmids/genetics , RNA, Double-Stranded/biosynthesis , RNA, Double-Stranded/genetics , Transfection
12.
Mol Cell Proteomics ; 10(6): M110.003822, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21447708

ABSTRACT

A new result report for Mascot search results is described. A greedy set cover algorithm is used to create a minimal set of proteins, which is then grouped into families on the basis of shared peptide matches. Protein families with multiple members are represented by dendrograms, generated by hierarchical clustering using the score of the nonshared peptide matches as a distance metric. The peptide matches to the proteins in a family can be compared side by side to assess the experimental evidence for each protein. If the evidence for a particular family member is considered inadequate, the dendrogram can be cut to reduce the number of distinct family members.


Subject(s)
Algorithms , Cluster Analysis , Proteomics/methods , Sequence Analysis, Protein/methods , Amino Acid Sequence , Computer Simulation , Databases, Protein , Models, Molecular , Molecular Sequence Data , Molecular Weight , Peptide Fragments/chemistry , Tandem Mass Spectrometry/methods
14.
PLoS One ; 18(12): e0296085, 2023.
Article in English | MEDLINE | ID: mdl-38128014

ABSTRACT

Spinocerebellar ataxia 2 (SCA2) is a neurodegenerative disorder caused by the expansion of the poly-glutamine (polyQ) tract of Ataxin-2 (ATXN2). Other polyQ-containing proteins such as ATXN7 and huntingtin are associated with the development of neurodegenerative diseases when their N-terminal polyQ domains are expanded. Furthermore, they undergo proteolytic processing events that produce N-terminal fragments that include the polyQ stretch, which are implicated in pathogenesis. Interestingly, N-terminal ATXN2 fragments were reported in a brain extract from a SCA2 patient, but it is currently unknown whether an expanded polyQ domain contributes to ATXN2 proteolytic susceptibility. Here, we used transient expression in HEK293 cells to determine whether ATXN2 is a target for specific N-terminal proteolysis. We found that ATXN2 proteins with either normal or expanded polyQ stretches undergo proteolytic cleavage releasing an N-terminal polyQ-containing fragment. We identified a short amino acid sequence downstream of the polyQ domain that is necessary for N-terminal cleavage of full-length ATXN2 and sufficient to induce proteolysis of a heterologous protein. However, this sequence is not required for cleavage of a short ATXN2 isoform produced from an alternative start codon located just upstream of the CAG repeats encoding the polyQ domain. Our study extends our understanding of ATXN2 posttranslational regulation by revealing that this protein can be the target of specific proteolytic cleavage events releasing polyQ-containing products that are modulated by the N-terminal domain of ATXN2. N-terminal ATXN2 proteolysis of expanded polyQ domains might contribute to SCA2 pathology, as observed in other neurodegenerative disorders caused by polyQ domain expansion.


Subject(s)
Ataxin-2 , Spinocerebellar Ataxias , Humans , Ataxin-2/genetics , Ataxin-2/metabolism , Proteolysis , HEK293 Cells , Spinocerebellar Ataxias/pathology , Amino Acid Sequence
15.
J Biol Rhythms ; 38(3): 245-258, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37226809

ABSTRACT

The origin of experimental chronobiology can be traced to observations made in the 18th and 19th centuries on the sensitive plant Mimosa, which were described in two seminal reports: Jean-Jacques d'Ortous de Mairan's "Observation Botanique" (A Botanical Observation) and Augustin Pyramus de Candolle's "Du sommeil des feuilles" (On the sleep of leaves). Both report observations of the striking daily closing and opening of Mimosa leaves in controlled environments. This review presents translations of both texts with the aim of staying as faithful as possible to the original French texts. We also present the historical context in which these texts were written and link them to subsequent experiments that aimed at testing the veracity of their central conclusions. In particular, we definitely establish that Mairan himself presented his work to the French Royal Academy of Sciences, while the published report of his observation was authored by Fontenelle, the Secretary of the Academy. In addition, we offer a translation of Mairan's own presentation, based on the hand-written minutes of the academy. Finally, we discuss the decades of work on plant rhythms that laid the foundation for modern experimental chronobiology, including translations and discussion of the insightful and prescient reports by Charles François de Cisternay Dufay, Henri Louis Duhamel du Monceau, Johann Gottfried Zinn, and Wilhelm Pfeffer, which describe their efforts to reproduce and extend Mairan's pioneering observations.


Subject(s)
Circadian Rhythm , Mimosa , Sleep , Plant Leaves
16.
Curr Biol ; 33(10): 1867-1882.e5, 2023 05 22.
Article in English | MEDLINE | ID: mdl-36977416

ABSTRACT

Organisms living in the intertidal zone are exposed to a particularly challenging environment. In addition to daily changes in light intensity and seasonal changes in photoperiod and weather patterns, they experience dramatic oscillations in environmental conditions due to the tides. To anticipate tides, and thus optimize their behavior and physiology, animals occupying intertidal ecological niches have acquired circatidal clocks. Although the existence of these clocks has long been known, their underlying molecular components have proven difficult to identify, in large part because of the lack of an intertidal model organism amenable to genetic manipulation. In particular, the relationship between the circatidal and circadian molecular clocks, and the possibility of shared genetic components, has been a long-standing question. Here, we introduce the genetically tractable crustacean Parhyale hawaiensis as a system for the study of circatidal rhythms. First, we show that P. hawaiensis exhibits robust 12.4-h rhythms of locomotion that can be entrained to an artificial tidal regimen and are temperature compensated. Using CRISPR-Cas9 genome editing, we then demonstrate that the core circadian clock gene Bmal1 is required for circatidal rhythms. Our results thus demonstrate that Bmal1 is a molecular link between circatidal and circadian clocks and establish P. hawaiensis as a powerful system to study the molecular mechanisms underlying circatidal rhythms and their entrainment.


Subject(s)
Amphipoda , Circadian Clocks , Animals , Circadian Rhythm/physiology , Circadian Clocks/genetics , Photoperiod , Locomotion
17.
PLoS Genet ; 5(12): e1000787, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20041201

ABSTRACT

Circadian pacemakers are essential to synchronize animal physiology and behavior with the dayrationight cycle. They are self-sustained, but the phase of their oscillations is determined by environmental cues, particularly light intensity and temperature cycles. In Drosophila, light is primarily detected by a dedicated blue-light photoreceptor: CRYPTOCHROME (CRY). Upon light activation, CRY binds to the pacemaker protein TIMELESS (TIM) and triggers its proteasomal degradation, thus resetting the circadian pacemaker. To understand further the CRY input pathway, we conducted a misexpression screen under constant light based on the observation that flies with a disruption in the CRY input pathway remain robustly rhythmic instead of becoming behaviorally arrhythmic. We report the identification of more than 20 potential regulators of CRY-dependent light responses. We demonstrate that one of them, the chromatin-remodeling enzyme KISMET (KIS), is necessary for normal circadian photoresponses, but does not affect the circadian pacemaker. KIS genetically interacts with CRY and functions in PDF-negative circadian neurons, which play an important role in circadian light responses. It also affects daily CRY-dependent TIM oscillations in a peripheral tissue: the eyes. We therefore conclude that KIS is a key transcriptional regulator of genes that function in the CRY signaling cascade, and thus it plays an important role in the synchronization of circadian rhythms with the dayrationight cycle.


Subject(s)
Circadian Rhythm/genetics , Circadian Rhythm/radiation effects , DNA Helicases/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Genetic Testing , Homeodomain Proteins/genetics , Light , Animals , Behavior, Animal/radiation effects , Cryptochromes/genetics , Cryptochromes/metabolism , DNA Helicases/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/radiation effects , Gene Expression Regulation/radiation effects , Genes, Insect/genetics , Homeodomain Proteins/metabolism , Neurons/metabolism , Neurons/radiation effects , Protein Processing, Post-Translational/radiation effects , RNA, Double-Stranded/metabolism
18.
Curr Biol ; 32(9): 1895-1908.e5, 2022 05 09.
Article in English | MEDLINE | ID: mdl-35303417

ABSTRACT

A precise balance between sleep and wakefulness is essential to sustain a good quality of life and optimal brain function. GABA is known to play a key and conserved role in sleep control, and GABAergic tone should, therefore, be tightly controlled in sleep circuits. Here, we examined the role of the astrocytic GABA transporter (GAT) in sleep regulation using Drosophila melanogaster. We found that a hypomorphic gat mutation (gat33-1) increased sleep amount, decreased sleep latency, and increased sleep consolidation at night. Interestingly, sleep defects were suppressed when gat33-1 was combined with a mutation disrupting wide-awake (wake), a gene that regulates the cell-surface levels of the GABAA receptor resistance to dieldrin (RDL) in the wake-promoting large ventral lateral neurons (l-LNvs). Moreover, RNAi knockdown of rdl and its modulators dnlg4 and wake in these circadian neurons also suppressed gat33-1 sleep phenotypes. Brain immunohistochemistry showed that GAT-expressing astrocytes were located near RDL-positive l-LNv cell bodies and dendritic processes. We concluded that astrocytic GAT decreases GABAergic tone and RDL activation in arousal-promoting LNvs, thus determining proper sleep amount and quality in Drosophila.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Astrocytes/metabolism , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , GABA Plasma Membrane Transport Proteins , GABAergic Neurons/metabolism , Neurons/physiology , Quality of Life , Receptors, GABA-A , Sleep/physiology
19.
Front Physiol ; 13: 888262, 2022.
Article in English | MEDLINE | ID: mdl-35721569

ABSTRACT

Ambient temperature varies constantly. However, the period of circadian pacemakers is remarkably stable over a wide-range of ecologically- and physiologically-relevant temperatures, even though the kinetics of most biochemical reactions accelerates as temperature rises. This thermal buffering phenomenon, called temperature compensation, is a critical feature of circadian rhythms, but how it is achieved remains elusive. Here, we uncovered the important role played by the Drosophila PERIOD (PER) phosphodegron in temperature compensation. This phosphorylation hotspot is crucial for PER proteasomal degradation and is the functional homolog of mammalian PER2 S478 phosphodegron, which also impacts temperature compensation. Using CRISPR-Cas9, we introduced a series of mutations that altered three Serines of the PER phosphodegron. While all three Serine to Alanine substitutions lengthened period at all temperatures tested, temperature compensation was differentially affected. S44A and S45A substitutions caused undercompensation, while S47A resulted in overcompensation. These results thus reveal unexpected functional heterogeneity of phosphodegron residues in thermal compensation. Furthermore, mutations impairing phosphorylation of the per s phosphocluster showed undercompensation, consistent with its inhibitory role on S47 phosphorylation. We observed that S47A substitution caused increased accumulation of hyper-phosphorylated PER at warmer temperatures. This finding was corroborated by cell culture assays in which S47A slowed down phosphorylation-dependent PER degradation at high temperatures, causing PER degradation to be excessively temperature-compensated. Thus, our results point to a novel role of the PER phosphodegron in temperature compensation through temperature-dependent modulation of the abundance of hyper-phosphorylated PER. Our work reveals interesting mechanistic convergences and differences between mammalian and Drosophila temperature compensation of the circadian clock.

20.
Neuron ; 55(3): 337-9, 2007 Aug 02.
Article in English | MEDLINE | ID: mdl-17678846

ABSTRACT

Whether CNS glial cells play an important role in the regulation of complex behaviors has been a longstanding question. In this issue of Neuron, Suh and Jackson demonstrate a circadian rhythmicity in glial expression of ebony, an N-beta-alanyl-biogenic amine synthase, and show that Ebony activity in glia is essential for the proper regulation of Drosophila circadian behavior.


Subject(s)
Circadian Rhythm/physiology , DNA-Binding Proteins/physiology , Drosophila Proteins/physiology , Drosophila/physiology , Motor Activity/physiology , Neuroglia/metabolism , Animals , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL