Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 May 22.
Article in English | MEDLINE | ID: mdl-38826334

ABSTRACT

Knowing how neural circuits change with neuronal plasticity and differ between individuals is important to fully understand behavior. Connectomes are typically assembled using electron microscopy, but this is low throughput and impractical for analyzing plasticity or mutations. Here, we modified the trans-Tango genetic circuit-tracing technique to identify neurons synaptically downstream of Drosophila s-LNv clock neurons, which show 24hr plasticity rhythms. s-LNv target neurons were labeled specifically in adult flies using a nuclear reporter gene, which facilitated their purification and then single cell sequencing. We call this Tango-seq, and it allows transcriptomic data - and thus cell identity - to be overlayed on top of anatomical data. We found that s-LNvs preferentially make synaptic connections with a subset of the CNMa+ DN1p clock neurons, and that these are likely plastic connections. We also identified synaptic connections between s-LNvs and mushroom body Kenyon cells. Tango-seq should be a useful addition to the connectomics toolkit.

2.
bioRxiv ; 2024 May 26.
Article in English | MEDLINE | ID: mdl-38826237

ABSTRACT

Drosophila s-LNv circadian pacemaker neurons show dramatic structural plasticity, with their projections expanded at dawn and then retracted by dusk. This predictable plasticity makes s-LNvs ideal to study molecular mechanisms of plasticity. Although s-LNv plasticity is controlled by their molecular clock, changing s-LNv excitability also regulates plasticity. Here, we tested the idea that s-LNvs use activity-regulated genes to control plasticity. We found that inducing expression of either of the activity-regulated transcription factors Hr38 or Sr (orthologs of mammalian Nr4a1 and Egr1) is sufficient to rapidly expand s-LNv projections. Conversely, transiently knocking down expression of either Hr38 or sr blocks expansion of s-LNv projections at dawn. We show that Hr38 rapidly induces transcription of sif, which encodes a Rac1 GEF required for s-LNv plasticity rhythms. We conclude that the s-LNv molecular clock controls s-LNv excitability, which couples to an activity-regulated gene expression program to control s-LNv plasticity.

3.
bioRxiv ; 2023 Sep 03.
Article in English | MEDLINE | ID: mdl-37693612

ABSTRACT

Fragile X syndrome (FXS) is a neuro-developmental disorder caused by silencing Fmr1, which encodes the RNA-binding protein FMRP. Although Fmr1 is expressed in adult neurons, it has been challenging to separate acute from chronic effects of loss of Fmr1 in models of FXS. We have used the precision of Drosophila genetics to test if Fmr1 acutely affects adult neuronal plasticity in vivo, focusing on the s-LNv circadian pacemaker neurons that show 24 hour rhythms in structural plasticity. We found that over-expressing Fmr1 for only 4 hours blocks the activity-dependent expansion of s-LNv projections without altering the circadian clock or activity-regulated gene expression. Conversely, acutely reducing Fmr1 expression prevented s-LNv projections from retracting. One FMRP target that we identified in s-LNvs is sif, which encodes a Rac1 GEF. Our data indicate that FMRP normally reduces sif mRNA translation at dusk to reduce Rac1 activity. Overall, our data reveal a previously unappreciated rapid and direct role for FMRP in acutely regulating neuronal plasticity in adult neurons, and underscore the importance of RNA-binding proteins in this process.

4.
Sci Signal ; 11(520)2018 03 06.
Article in English | MEDLINE | ID: mdl-29511120

ABSTRACT

Genetically encoded fluorescent sensors enable cell-specific measurements of ions and small molecules in real time. Cyclic adenosine monophosphate (cAMP) is one of the most important signaling molecules in virtually all cell types and organisms. We describe cAMPr, a new single-wavelength cAMP sensor. We developed cAMPr in bacteria and embryonic stem cells and validated the sensor in mammalian neurons in vitro and in Drosophila circadian pacemaker neurons in intact brains. Comparison with other single-wavelength cAMP sensors showed that cAMPr improved the quantitative detection of cAMP abundance. In addition, cAMPr is compatible with both single-photon and two-photon imaging. This enabled us to use cAMPr together with the red fluorescent Ca2+ sensor RCaMP1h to simultaneously monitor Ca2+ and cAMP in Drosophila brains. Thus, cAMPr is a new and versatile genetically encoded cAMP sensor.


Subject(s)
Biosensing Techniques/methods , Calcium/analysis , Cyclic AMP/analysis , Neurons/metabolism , Animals , Bacteria/genetics , Bacteria/metabolism , Brain/cytology , Brain/metabolism , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Drosophila , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fluorescence Resonance Energy Transfer/methods , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Time-Lapse Imaging/methods
5.
J Biol Rhythms ; 32(5): 380-393, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29098954

ABSTRACT

Genome biology approaches have made enormous contributions to our understanding of biological rhythms, particularly in identifying outputs of the clock, including RNAs, proteins, and metabolites, whose abundance oscillates throughout the day. These methods hold significant promise for future discovery, particularly when combined with computational modeling. However, genome-scale experiments are costly and laborious, yielding "big data" that are conceptually and statistically difficult to analyze. There is no obvious consensus regarding design or analysis. Here we discuss the relevant technical considerations to generate reproducible, statistically sound, and broadly useful genome-scale data. Rather than suggest a set of rigid rules, we aim to codify principles by which investigators, reviewers, and readers of the primary literature can evaluate the suitability of different experimental designs for measuring different aspects of biological rhythms. We introduce CircaInSilico, a web-based application for generating synthetic genome biology data to benchmark statistical methods for studying biological rhythms. Finally, we discuss several unmet analytical needs, including applications to clinical medicine, and suggest productive avenues to address them.


Subject(s)
Circadian Rhythm/genetics , Genome , Genomics , Statistics as Topic/methods , Biostatistics , Computational Biology/methods , Genomics/statistics & numerical data , Humans , Metabolomics , Proteomics , Software , Systems Biology
6.
Cell ; 165(6): 1310-1311, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27259141

ABSTRACT

The drive to sleep increases the longer that we stay awake, but this process is poorly understood at the cellular level. Now, Liu et al. show that the plasticity of a small group of neurons in the Drosophila central brain is a key component of the sleep homeostat.


Subject(s)
Drosophila melanogaster/metabolism , Receptors, N-Methyl-D-Aspartate , Animals , Drosophila/metabolism , Drosophila Proteins/metabolism , Sheep , Sleep
7.
Nat Neurosci ; 19(4): 587-95, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26928065

ABSTRACT

Twenty-four hour rhythms in behavior are organized by a network of circadian pacemaker neurons. Rhythmic activity in this network is generated by intrinsic rhythms in clock neuron physiology and communication between clock neurons. However, it is poorly understood how the activity of a small number of pacemaker neurons is translated into rhythmic behavior of the whole animal. To understand this, we screened for signals that could identify circadian output circuits in Drosophila melanogaster. We found that leucokinin neuropeptide (LK) and its receptor (LK-R) were required for normal behavioral rhythms. This LK/LK-R circuit connects pacemaker neurons to brain areas that regulate locomotor activity and sleep. Our experiments revealed that pacemaker neurons impose rhythmic activity and excitability on LK- and LK-R-expressing neurons. We also found pacemaker neuron-dependent activity rhythms in a second circadian output pathway controlled by DH44 neuropeptide-expressing neurons. We conclude that rhythmic clock neuron activity propagates to multiple downstream circuits to orchestrate behavioral rhythms.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , Nerve Net/physiology , Neurons/physiology , Animals , Animals, Genetically Modified , Drosophila Proteins/analysis , Drosophila Proteins/biosynthesis , Drosophila melanogaster , Male , Motor Activity/physiology , Nerve Net/chemistry , Neurons/chemistry
8.
Cell ; 162(4): 823-35, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26234154

ABSTRACT

Neuronal plasticity helps animals learn from their environment. However, it is challenging to link specific changes in defined neurons to altered behavior. Here, we focus on circadian rhythms in the structure of the principal s-LNv clock neurons in Drosophila. By quantifying neuronal architecture, we observed that s-LNv structural plasticity changes the amount of axonal material in addition to cycles of fasciculation and defasciculation. We found that this is controlled by rhythmic Rho1 activity that retracts s-LNv axonal termini by increasing myosin phosphorylation and simultaneously changes the balance of pre-synaptic and dendritic markers. This plasticity is required to change clock network hierarchy and allow seasonal adaptation. Rhythms in Rho1 activity are controlled by clock-regulated transcription of Puratrophin-1-like (Pura), a Rho1 GEF. Since spinocerebellar ataxia is associated with mutations in human Puratrophin-1, our data support the idea that defective actin-related plasticity underlies this ataxia.


Subject(s)
Circadian Rhythm , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , rho GTP-Binding Proteins/metabolism , Animals , Axons/metabolism , Biological Clocks , Guanine Nucleotide Exchange Factors/metabolism , Myosins/metabolism , Neuronal Plasticity , Phosphorylation , Seasons , Signal Transduction , Spectrin/metabolism , Synapses/metabolism
9.
PLoS Biol ; 12(9): e1001959, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25268747

ABSTRACT

Synchronized neuronal activity is vital for complex processes like behavior. Circadian pacemaker neurons offer an unusual opportunity to study synchrony as their molecular clocks oscillate in phase over an extended timeframe (24 h). To identify where, when, and how synchronizing signals are perceived, we first studied the minimal clock neural circuit in Drosophila larvae, manipulating either the four master pacemaker neurons (LNvs) or two dorsal clock neurons (DN1s). Unexpectedly, we found that the PDF Receptor (PdfR) is required in both LNvs and DN1s to maintain synchronized LNv clocks. We also found that glutamate is a second synchronizing signal that is released from DN1s and perceived in LNvs via the metabotropic glutamate receptor (mGluRA). Because simultaneously reducing Pdfr and mGluRA expression in LNvs severely dampened Timeless clock protein oscillations, we conclude that the master pacemaker LNvs require extracellular signals to function normally. These two synchronizing signals are released at opposite times of day and drive cAMP oscillations in LNvs. Finally we found that PdfR and mGluRA also help synchronize Timeless oscillations in adult s-LNvs. We propose that differentially timed signals that drive cAMP oscillations and synchronize pacemaker neurons in circadian neural circuits will be conserved across species.


Subject(s)
Circadian Clocks/genetics , Cyclic AMP/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Neurons/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Metabotropic Glutamate/genetics , Animals , Circadian Rhythm/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Regulation , Glutamic Acid/metabolism , Larva/genetics , Larva/metabolism , Neurons/cytology , Neuropeptides/metabolism , Photoperiod , Receptors, G-Protein-Coupled/metabolism , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction
11.
Curr Biol ; 22(20): 1871-80, 2012 Oct 23.
Article in English | MEDLINE | ID: mdl-22940468

ABSTRACT

BACKGROUND: Circadian (∼24 hr) rhythms offer one of the best examples of how gene expression is tied to behavior. Circadian pacemaker neurons contain molecular clocks that control 24 hr rhythms in gene expression that in turn regulate electrical activity rhythms to control behavior. RESULTS: Here we demonstrate the inverse relationship: there are broad transcriptional changes in Drosophila clock neurons (LN(v)s) in response to altered electrical activity, including a large set of circadian genes. Hyperexciting LN(v)s creates a morning-like expression profile for many circadian genes while hyperpolarization leads to an evening-like transcriptional state. The electrical effects robustly persist in per(0) mutant LN(v)s but not in cyc(0) mutant LN(v)s, suggesting that neuronal activity interacts with the transcriptional activators of the core circadian clock. Bioinformatic and immunocytochemical analyses suggest that CREB family transcription factors link LN(v) electrical state to circadian gene expression. CONCLUSIONS: The electrical state of a clock neuron can impose time of day to its transcriptional program. We propose that this acts as an internal zeitgeber to add robustness and precision to circadian behavioral rhythms.


Subject(s)
Activating Transcription Factor 2/physiology , Circadian Clocks/physiology , Circadian Rhythm , Drosophila Proteins/physiology , Drosophila/physiology , Neurons/physiology , Transcriptome , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Behavior, Animal , Circadian Rhythm/genetics , Circadian Rhythm/physiology , Cyclic AMP Response Element-Binding Protein/physiology , Drosophila/genetics , Gene Expression Regulation , Genes, Insect , Oligonucleotide Array Sequence Analysis , Sodium Channels/genetics , Sodium Channels/metabolism , Transcription, Genetic , Transcriptional Activation
12.
J Biol Rhythms ; 27(5): 353-64, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23010658

ABSTRACT

Although the intracellular molecular clocks that regulate circadian (~24 h) behavioral rhythms are well understood, it remains unclear how molecular clock information is transduced into rhythmic neuronal activity that in turn drives behavioral rhythms. To identify potential clock outputs, the authors generated expression profiles from a homogeneous population of purified pacemaker neurons (LN(v)s) from wild-type and clock mutant Drosophila. They identified a group of genes with enriched expression in LN(v)s and a second group of genes rhythmically expressed in LN(v)s in a clock-dependent manner. Only 10 genes fell into both groups: 4 core clock genes, including period (per) and timeless (tim), and 6 genes previously unstudied in circadian rhythms. The authors focused on one of these 6 genes, Ir, which encodes an inward rectifier K(+) channel likely to regulate resting membrane potential, whose expression peaks around dusk. Reducing Ir expression in LN(v)s increased larval light avoidance and lengthened the period of adult locomotor rhythms, consistent with increased LN(v) excitability. In contrast, increased Ir expression made many adult flies arrhythmic and dampened PER protein oscillations. The authors propose that rhythmic Ir expression contributes to daily rhythms in LN(v) neuronal activity, which in turn feed back to regulate molecular clock oscillations.


Subject(s)
Circadian Rhythm , Neurons/physiology , ARNTL Transcription Factors/biosynthesis , Animals , Basic-Leucine Zipper Transcription Factors/biosynthesis , Behavior, Animal/physiology , Circadian Rhythm/genetics , Cryptochromes/biosynthesis , Darkness , Drosophila/metabolism , Drosophila Proteins/biosynthesis , Eye Proteins/biosynthesis , Larva , Light Signal Transduction , Period Circadian Proteins/biosynthesis , Transcriptome
13.
Neuron ; 74(4): 706-18, 2012 May 24.
Article in English | MEDLINE | ID: mdl-22632728

ABSTRACT

Circadian rhythms offer an excellent opportunity to dissect the neural circuits underlying innate behavior because the genes and neurons involved are relatively well understood. We first sought to understand how Drosophila clock neurons interact in the simple circuit that generates circadian rhythms in larval light avoidance. We used genetics to manipulate two groups of clock neurons, increasing or reducing excitability, stopping their molecular clocks, and blocking neurotransmitter release and reception. Our results revealed that lateral neurons (LN(v)s) promote and dorsal clock neurons (DN(1)s) inhibit light avoidance, these neurons probably signal at different times of day, and both signals are required for rhythmic behavior. We found that similar principles apply in the more complex adult circadian circuit that generates locomotor rhythms. Thus, the changing balance in activity between clock neurons with opposing behavioral effects generates robust circadian behavior and probably helps organisms transition between discrete behavioral states, such as sleep and wakefulness.


Subject(s)
Circadian Rhythm/physiology , Drosophila Proteins/metabolism , Glutamic Acid/metabolism , Neurons/physiology , Period Circadian Proteins/metabolism , Animals , Behavior, Animal/physiology , Drosophila , Drosophila Proteins/genetics , Motor Activity/physiology , Period Circadian Proteins/genetics , Photic Stimulation
14.
J Neurosci ; 31(17): 6527-34, 2011 Apr 27.
Article in English | MEDLINE | ID: mdl-21525293

ABSTRACT

Visual organs perceive environmental stimuli required for rapid initiation of behaviors and can also entrain the circadian clock. The larval eye of Drosophila is capable of both functions. Each eye contains only 12 photoreceptors (PRs), which can be subdivided into two subtypes. Four PRs express blue-sensitive rhodopsin5 (rh5) and eight express green-sensitive rhodopsin6 (rh6). We found that either PR-subtype is sufficient to entrain the molecular clock by light, while only the Rh5-PR subtype is essential for light avoidance. Acetylcholine released from PRs confers both functions. Both subtypes of larval PRs innervate the main circadian pacemaker neurons of the larva, the neuropeptide PDF (pigment-dispersing factor)-expressing lateral neurons (LNs), providing sensory input to control circadian rhythms. However, we show that PDF-expressing LNs are dispensable for light avoidance, and a distinct set of three clock neurons is required. Thus we have identified distinct sensory and central circuitry regulating light avoidance behavior and clock entrainment. Our findings provide insights into the coding of sensory information for distinct behavioral functions and the underlying molecular and neuronal circuitry.


Subject(s)
Circadian Clocks/physiology , Escape Reaction/physiology , Larva/physiology , Light/adverse effects , Photoreceptor Cells, Invertebrate/physiology , Visual Pathways/physiology , Acetylcholine/metabolism , Analysis of Variance , Animals , Animals, Genetically Modified , Behavior, Animal/physiology , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Eye Proteins/metabolism , Light Signal Transduction , Luminescent Proteins/genetics , Microscopy, Confocal/methods , Mutation/genetics , Neurons/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Neuropil/physiology , Photoreceptor Cells, Invertebrate/classification , Rhodopsin/genetics
15.
Neuron ; 68(5): 964-77, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-21145008

ABSTRACT

Intercellular signaling is important for accurate circadian rhythms. In Drosophila, the small ventral lateral neurons (s-LN(v)s) are the dominant pacemaker neurons and set the pace of most other clock neurons in constant darkness. Here we show that two distinct G protein signaling pathways are required in LN(v)s for 24 hr rhythms. Reducing signaling in LN(v)s via the G alpha subunit Gs, which signals via cAMP, or via the G alpha subunit Go, which we show signals via Phospholipase 21c, lengthens the period of behavioral rhythms. In contrast, constitutive Gs or Go signaling makes most flies arrhythmic. Using dissociated LN(v)s in culture, we found that Go and the metabotropic GABA(B)-R3 receptor are required for the inhibitory effects of GABA on LN(v)s and that reduced GABA(B)-R3 expression in vivo lengthens period. Although no clock neurons produce GABA, hyperexciting GABAergic neurons disrupts behavioral rhythms and s-LN(v) molecular clocks. Therefore, s-LN(v)s require GABAergic inputs for 24 hr rhythms.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Neurons/metabolism , Receptors, GABA-B/metabolism , Animals , Drosophila/cytology , Drosophila/physiology , Neural Pathways/cytology , Neural Pathways/metabolism , Signal Transduction/physiology
16.
Curr Biol ; 20(13): 1209-15, 2010 Jul 13.
Article in English | MEDLINE | ID: mdl-20541409

ABSTRACT

Neural systems controlling the vital functions of sleep and feeding in mammals are tightly interconnected: sleep deprivation promotes feeding, whereas starvation suppresses sleep. Here we show that starvation in Drosophila potently suppresses sleep, suggesting that these two homeostatically regulated behaviors are also integrated in flies. The sleep-suppressing effect of starvation is independent of the mushroom bodies, a previously identified sleep locus in the fly brain, and therefore is regulated by distinct neural circuitry. The circadian clock genes Clock (Clk) and cycle (cyc) are critical for proper sleep suppression during starvation. However, the sleep suppression is independent of light cues and of circadian rhythms as shown by the fact that starved period mutants sleep like wild-type flies. By selectively targeting subpopulations of Clk-expressing neurons, we localize the observed sleep phenotype to the dorsally located circadian neurons. These findings show that Clk and cyc act during starvation to modulate the conflict of whether flies sleep or search for food.


Subject(s)
ARNTL Transcription Factors/physiology , CLOCK Proteins/physiology , Drosophila Proteins/physiology , Drosophila/physiology , Sleep Deprivation , Animals , Feeding Behavior , Female , Male , Sleep
17.
J Neurosci ; 30(17): 5855-65, 2010 Apr 28.
Article in English | MEDLINE | ID: mdl-20427646

ABSTRACT

The transcription factor Mef2 has well established roles in muscle development in Drosophila and in the differentiation of many cell types in mammals, including neurons. Here, we describe a role for Mef2 in the Drosophila pacemaker neurons that regulate circadian behavioral rhythms. We found that Mef2 is normally produced in all adult clock neurons and that Mef2 overexpression in clock neurons leads to long period and complex rhythms of adult locomotor behavior. Knocking down Mef2 expression via RNAi or expressing a repressor form of Mef2 caused flies to lose circadian behavioral rhythms. These behavioral changes are correlated with altered molecular clocks in pacemaker neurons: Mef2 overexpression causes the oscillations in individual pacemaker neurons to become desynchronized, while Mef2 knockdown strongly dampens molecular rhythms. Thus, a normal level of Mef2 activity is required in clock neurons to maintain robust and accurate circadian behavioral rhythms.


Subject(s)
Circadian Rhythm/physiology , Drosophila Proteins/metabolism , Drosophila/physiology , Myogenic Regulatory Factors/metabolism , Neurons/physiology , Animals , Animals, Genetically Modified , Brain/physiology , Circadian Rhythm/genetics , Drosophila/genetics , Drosophila Proteins/genetics , Gene Expression , Gene Knockdown Techniques , Immunohistochemistry , Microscopy, Confocal , Motor Activity/genetics , Motor Activity/physiology , Myogenic Regulatory Factors/genetics , Periodicity , Phenotype , RNA Interference , Time Factors
18.
J Neurosci ; 29(4): 1152-62, 2009 Jan 28.
Article in English | MEDLINE | ID: mdl-19176824

ABSTRACT

The ubiquitin-proteasome system plays a major role in the rhythmic accumulation and turnover of molecular clock components. In turn, these approximately 24 h molecular rhythms drive circadian rhythms of behavior and physiology. In Drosophila, the ubiquitin-proteasome system also plays a critical role in light-dependent degradation of the clock protein Timeless (TIM), a key step in the entrainment of the molecular clocks to light-dark cycles. Here, we investigated the role of the COP9 signalosome (CSN), a general regulator of protein degradation, in fly circadian rhythms. We found that null mutations in the genes encoding the CSN4 and CSN5 subunits prevent normal TIM degradation by light in the pacemaker lateral neurons (LNs) as does LN-specific expression of a dominant-negative CSN5 transgene. These defects are accompanied by strong reductions in behavioral phase shifts of adult flies lacking normal CSN5 activity in LNs. Defects in TIM degradation and resetting of behavioral phases were rescued by overexpression of Jetlag (JET), the F-box protein required for light-mediated TIM degradation. Flies lacking normal CSN activity in all clock neurons are rhythmic in constant light, a phenotype previously associated with jet mutants. Together, these data indicate that JET and the CSN lie in a common pathway leading to light-dependent TIM degradation. Surprisingly, we found that manipulations that strongly inhibit CSN activity had minimal effects on circadian rhythms in constant darkness, indicating a specific role for the CSN in light-mediated TIM degradation.


Subject(s)
Circadian Rhythm/physiology , Drosophila Proteins/metabolism , F-Box Proteins/metabolism , Gene Expression Regulation/physiology , Light , Nuclear Proteins/physiology , Adaptor Proteins, Signal Transducing , Animals , Animals, Genetically Modified , Behavior, Animal/physiology , Brain/cytology , COP9 Signalosome Complex , Circadian Rhythm/genetics , Drosophila , Drosophila Proteins/genetics , F-Box Proteins/genetics , Gene Expression Regulation/genetics , Larva , Motor Activity/genetics , Mutation/genetics , Neurons/metabolism , Nuclear Proteins/classification , Nuclear Proteins/genetics , Oscillometry , Peptide Hydrolases/genetics , Peptide Hydrolases/physiology , Photoreceptor Cells, Invertebrate , Time Factors
19.
Genes Dev ; 22(13): 1737-40, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18593875

ABSTRACT

Period (PER) proteins are essential parts of the molecular clocks that control circadian rhythms in flies and mammals. Phosphorylation regulates PER's stability and subcellular localization; however, the physiologically relevant sites have been difficult to identify in spite of knowing the relevant kinase. In this issue of Genes & Development, Chiu and colleagues (1758-1772) identify a key phosphorylation site on PER that recruits the F-box protein Slimb to trigger PER degradation and set clock speed.


Subject(s)
Circadian Rhythm/physiology , Drosophila melanogaster/physiology , Nuclear Proteins/metabolism , Animals , Binding Sites , Cell Cycle Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Mutation , Period Circadian Proteins , Phosphorylation , Ubiquitin-Protein Ligases/metabolism
20.
Pflugers Arch ; 454(5): 857-67, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17226053

ABSTRACT

"Even a stopped clock tells the right time twice a day, and for once I'm inclined to believe Withnail is right. We are indeed drifting into the arena of the unwell... What we need is harmony. Fresh air. Stuff like that" "Bruce Robinson (1986, ref. 1)". Although a stopped Drosophila clock probably does not tell the right time even once a day, recent findings have demonstrated that accurate circadian time-keeping is dependent on harmony between groups of clock neurons within the brain. Furthermore, when harmony between the environment and the endogenous clock is lost, as during jet lag, we definitely feel unwell. In this review, we provide an overview of the current understanding of circadian rhythms in Drosophila, focussing on recent discoveries that demonstrate how approximately 100 neurons within the Drosophila brain control the behaviour of the whole fly, and how these rhythms respond to the environment.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , Drosophila/physiology , Adaptation, Biological/genetics , Adaptation, Biological/physiology , Adaptation, Biological/radiation effects , Animals , Biological Clocks/radiation effects , Circadian Rhythm/radiation effects , Drosophila/anatomy & histology , Drosophila Proteins/physiology , Drosophila Proteins/radiation effects , Feedback, Physiological , Genes, Insect/physiology , Light , Models, Neurological , Mutagenesis, Site-Directed , Nerve Net/physiology , Nerve Net/radiation effects , Photoreceptor Cells, Invertebrate/cytology , Photoreceptor Cells, Invertebrate/physiology , Photoreceptor Cells, Invertebrate/radiation effects , Thermosensing/genetics , Thermosensing/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...