Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Nature ; 623(7987): 562-570, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37880372

ABSTRACT

Vision enables both image-forming perception, driven by a contrast-based pathway, and unconscious non-image-forming circadian photoentrainment, driven by an irradiance-based pathway1,2. Although two distinct photoreceptor populations are specialized for each visual task3-6, image-forming photoreceptors can additionally contribute to photoentrainment of the circadian clock in different species7-15. However, it is unknown how the image-forming photoreceptor pathway can functionally implement the segregation of irradiance signals required for circadian photoentrainment from contrast signals required for image perception. Here we report that the Drosophila R8 photoreceptor separates image-forming and irradiance signals by co-transmitting two neurotransmitters, histamine and acetylcholine. This segregation is further established postsynaptically by histamine-receptor-expressing unicolumnar retinotopic neurons and acetylcholine-receptor-expressing multicolumnar integration neurons. The acetylcholine transmission from R8 photoreceptors is sustained by an autocrine negative feedback of the cotransmitted histamine during the light phase of light-dark cycles. At the behavioural level, elimination of histamine and acetylcholine transmission impairs R8-driven motion detection and circadian photoentrainment, respectively. Thus, a single type of photoreceptor can achieve the dichotomy of visual perception and circadian photoentrainment as early as the first visual synapses, revealing a simple yet robust mechanism to segregate and translate distinct sensory features into different animal behaviours.


Subject(s)
Circadian Rhythm , Drosophila melanogaster , Photoreceptor Cells, Invertebrate , Visual Perception , Animals , Acetylcholine/metabolism , Biological Clocks/physiology , Biological Clocks/radiation effects , Circadian Rhythm/physiology , Circadian Rhythm/radiation effects , Drosophila melanogaster/cytology , Drosophila melanogaster/physiology , Drosophila melanogaster/radiation effects , Feedback, Physiological , Histamine/metabolism , Neurotransmitter Agents/metabolism , Photoreceptor Cells, Invertebrate/metabolism , Photoreceptor Cells, Invertebrate/radiation effects , Receptors, Cholinergic/metabolism , Receptors, Histamine/metabolism , Visual Perception/physiology , Visual Perception/radiation effects
2.
J Biomed Opt ; 28(6): 066501, 2023 06.
Article in English | MEDLINE | ID: mdl-37334209

ABSTRACT

Significance: Adaptive optics (AO) has been implemented on several microscopy setups and has proven its ability to increase both signal and resolution. However, reported configurations are not suited for fast imaging of live samples or are based on an invasive or complex implementation method. Aim: Provide a fast aberration correction method with an easy to implement AO module compatible with light-sheet fluorescence microscopy (LSFM) for enhanced imaging of live samples. Approach: Development of an AO add-on module for LSFM based on direct wavefront sensing without requiring a guide star using an extended-scene Shack-Hartmann wavefront sensor. The enhanced setup uses a two-color sample labeling strategy to optimize the photon budget. Results: Fast AO correction of in-depth aberrations in an ex-vivo adult Drosophila brain enables doubling the contrast when imaging with either cell reporters or calcium sensors for functional imaging. We quantify the gain in terms of image quality on different functional domains of sleep neurons in the Drosophila brain at various depths and discuss the optimization of key parameters driving AO. Conclusion: We developed a compact AO module that can be integrated into most of the reported light-sheet microscopy setups, provides significant improvement of image quality and is compatible with fast imaging requirements such as calcium imaging.


Subject(s)
Calcium , Drosophila melanogaster , Animals , Microscopy, Fluorescence , Drosophila , Neuroimaging , Brain/diagnostic imaging
3.
Curr Biol ; 30(15): R890-R893, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32750352

ABSTRACT

Key Drosophila clock neurons remodel their axonal arborization on a daily basis. The current view is that remodelling is part of the control of clock neuron output but new data support a major role in modulating sensory inputs.


Subject(s)
Circadian Clocks , Drosophila Proteins , Animals , Circadian Rhythm , Drosophila , Drosophila Proteins/genetics , Neurons
4.
Opt Lett ; 44(10): 2514-2517, 2019 May 15.
Article in English | MEDLINE | ID: mdl-31090720

ABSTRACT

We propose an adaptive optics light-sheet fluorescence microscope (AO-LSFM) for closed-loop aberrations' correction at the emission path, providing intrinsic instrumental simplicity and high accuracy when compared to previously reported schemes. The approach is based on direct wavefront sensing, i.e., not on time-consuming iterative algorithms, and does not require the use of any guide star, thus reducing instrumental complexity and/or sample preparation constraints. The design is based on a modified Shack-Hartmann wavefront sensor providing compatibility with extended sources such as images from optical sectioning microscopes. We report an AO-LSFM setup based on such sensors, including characterization of the sensor performance, and demonstrate for the first time to the best of our knowledge a significant contrast improvement on neuronal structures of the ex vivo adult drosophila brain in depth.

5.
Proc Natl Acad Sci U S A ; 116(12): 5721-5726, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30833404

ABSTRACT

The Drosophila circadian oscillator relies on a negative transcriptional feedback loop, in which the PERIOD (PER) and TIMELESS (TIM) proteins repress the expression of their own gene by inhibiting the activity of the CLOCK (CLK) and CYCLE (CYC) transcription factors. A series of posttranslational modifications contribute to the oscillations of the PER and TIM proteins but few posttranscriptional mechanisms have been described that affect mRNA stability. Here we report that down-regulation of the POP2 deadenylase, a key component of the CCR4-NOT deadenylation complex, alters behavioral rhythms. Down-regulating POP2 specifically increases TIM protein and tim mRNA but not tim pre-mRNA, supporting a posttranscriptional role. Indeed, reduced POP2 levels induce a lengthening of tim mRNA poly(A) tail. Surprisingly, such effects are lost in per0 mutants, supporting a PER-dependent inhibition of tim mRNA deadenylation by POP2. We report a deadenylation mechanism that controls the oscillations of a core clock gene transcript.


Subject(s)
Circadian Clocks/physiology , Drosophila Proteins/physiology , Period Circadian Proteins/physiology , ARNTL Transcription Factors/genetics , Animals , CLOCK Proteins/genetics , Circadian Clocks/genetics , Circadian Rhythm/physiology , Down-Regulation , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Gene Expression Regulation , Period Circadian Proteins/metabolism , Phosphorylation , RNA, Messenger/metabolism , Ribonucleases , Transcription, Genetic
6.
Nat Commun ; 10(1): 252, 2019 01 16.
Article in English | MEDLINE | ID: mdl-30651542

ABSTRACT

In Drosophila, the clock that controls rest-activity rhythms synchronizes with light-dark cycles through either the blue-light sensitive cryptochrome (Cry) located in most clock neurons, or rhodopsin-expressing histaminergic photoreceptors. Here we show that, in the absence of Cry, each of the two histamine receptors Ort and HisCl1 contribute to entrain the clock whereas no entrainment occurs in the absence of the two receptors. In contrast to Ort, HisCl1 does not restore entrainment when expressed in the optic lobe interneurons. Indeed, HisCl1 is expressed in wild-type photoreceptors and entrainment is strongly impaired in flies with photoreceptors mutant for HisCl1. Rescuing HisCl1 expression in the Rh6-expressing photoreceptors restores entrainment but it does not in other photoreceptors, which send histaminergic inputs to Rh6-expressing photoreceptors. Our results thus show that Rh6-expressing neurons contribute to circadian entrainment as both photoreceptors and interneurons, recalling the dual function of melanopsin-expressing ganglion cells in the mammalian retina.


Subject(s)
Chloride Channels/metabolism , Drosophila Proteins/metabolism , Drosophila/physiology , Photoreceptor Cells, Invertebrate/metabolism , Rhodopsin/metabolism , Animals , Animals, Genetically Modified , Behavior Observation Techniques/instrumentation , Behavior Observation Techniques/methods , Behavior, Animal/physiology , Chloride Channels/genetics , Compound Eye, Arthropod/cytology , Compound Eye, Arthropod/physiology , Cryptochromes/metabolism , Drosophila Proteins/genetics , Interneurons/metabolism , Male , Mutation , Optic Lobe, Nonmammalian/cytology , Optic Lobe, Nonmammalian/metabolism , Photoperiod
7.
PLoS Biol ; 16(10): e2006229, 2018 10.
Article in English | MEDLINE | ID: mdl-30278045

ABSTRACT

Exposure to man-made electromagnetic fields (EMFs), which increasingly pollute our environment, have consequences for human health about which there is continuing ignorance and debate. Whereas there is considerable ongoing concern about their harmful effects, magnetic fields are at the same time being applied as therapeutic tools in regenerative medicine, oncology, orthopedics, and neurology. This paradox cannot be resolved until the cellular mechanisms underlying such effects are identified. Here, we show by biochemical and imaging experiments that exposure of mammalian cells to weak pulsed electromagnetic fields (PEMFs) stimulates rapid accumulation of reactive oxygen species (ROS), a potentially toxic metabolite with multiple roles in stress response and cellular ageing. Following exposure to PEMF, cell growth is slowed, and ROS-responsive genes are induced. These effects require the presence of cryptochrome, a putative magnetosensor that synthesizes ROS. We conclude that modulation of intracellular ROS via cryptochromes represents a general response to weak EMFs, which can account for either therapeutic or pathological effects depending on exposure. Clinically, our findings provide a rationale to optimize low field magnetic stimulation for novel therapeutic applications while warning against the possibility of harmful synergistic effects with environmental agents that further increase intracellular ROS.


Subject(s)
Electromagnetic Fields/adverse effects , Magnetic Fields/adverse effects , Animals , Cell Enlargement , Cell Proliferation , Cryptochromes , Drosophila , HEK293 Cells , Humans , Mice , Reactive Oxygen Species/metabolism
9.
Curr Biol ; 28(13): 2007-2017.e4, 2018 07 09.
Article in English | MEDLINE | ID: mdl-29910074

ABSTRACT

The brain clock that drives circadian rhythms of locomotor activity relies on a multi-oscillator neuronal network. In addition to synchronizing the clock with day-night cycles, light also reformats the clock-driven daily activity pattern. How changes in lighting conditions modify the contribution of the different oscillators to remodel the daily activity pattern remains largely unknown. Our data in Drosophila indicate that light readjusts the interactions between oscillators through two different modes. We show that a morning s-LNv > DN1p circuit works in series, whereas two parallel evening circuits are contributed by LNds and other DN1ps. Based on the photic context, the master pacemaker in the s-LNv neurons swaps its enslaved partner-oscillator-LNd in the presence of light or DN1p in the absence of light-to always link up with the most influential phase-determining oscillator. When exposure to light further increases, the light-activated LNd pacemaker becomes independent by decoupling from the s-LNvs. The calibration of coupling by light is layered on a clock-independent network interaction wherein light upregulates the expression of the PDF neuropeptide in the s-LNvs, which inhibits the behavioral output of the DN1p evening oscillator. Thus, light modifies inter-oscillator coupling and clock-independent output-gating to achieve flexibility in the network. It is likely that the light-induced changes in the Drosophila brain circadian network could reveal general principles of adapting to varying environmental cues in any neuronal multi-oscillator system.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm/physiology , Drosophila melanogaster/physiology , Light , Animals , Brain/physiology , Male , Neurons/physiology
10.
Cell Rep ; 23(8): 2273-2282, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29791839

ABSTRACT

Circadian clocks have evolved as time-measuring molecular devices to help organisms adapt their physiology to daily changes in light and temperature. Transcriptional oscillations account for a large fraction of rhythmic protein abundance. However, cycling of various posttranslational modifications, such as ubiquitylation, also contributes to shape the rhythmic protein landscape. In this study, we used an in vivo ubiquitin labeling assay to investigate the circadian ubiquitylated proteome of Drosophila melanogaster. We find that cyclic ubiquitylation affects MEGATOR (MTOR), a chromatin-associated nucleoporin that, in turn, feeds back to regulate the core molecular oscillator. Furthermore, we show that the ubiquitin ligase subunits CULLIN-3 (CUL-3) and SUPERNUMERARY LIMBS (SLMB) cooperate for ubiquitylating the TIMELESS protein. These findings stress the importance of ubiquitylation pathways in the Drosophila circadian clock and reveal a key component of this system.


Subject(s)
Circadian Clocks , Circadian Rhythm/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Proteome/metabolism , Ubiquitination , Animals
11.
Front Neuroinform ; 12: 13, 2018.
Article in English | MEDLINE | ID: mdl-29628885

ABSTRACT

Imaging the expression patterns of reporter constructs is a powerful tool to dissect the neuronal circuits of perception and behavior in the adult brain of Drosophila, one of the major models for studying brain functions. To date, several Drosophila brain templates and digital atlases have been built to automatically analyze and compare collections of expression pattern images. However, there has been no systematic comparison of performances between alternative atlasing strategies and registration algorithms. Here, we objectively evaluated the performance of different strategies for building adult Drosophila brain templates and atlases. In addition, we used state-of-the-art registration algorithms to generate a new group-wise inter-sex atlas. Our results highlight the benefit of statistical atlases over individual ones and show that the newly proposed inter-sex atlas outperformed existing solutions for automated registration and annotation of expression patterns. Over 3,000 images from the Janelia Farm FlyLight collection were registered using the proposed strategy. These registered expression patterns can be searched and compared with a new version of the BrainBaseWeb system and BrainGazer software. We illustrate the validity of our methodology and brain atlas with registration-based predictions of expression patterns in a subset of clock neurons. The described registration framework should benefit to brain studies in Drosophila and other insect species.

12.
J Comp Neurol ; 524(14): 2828-44, 2016 10 01.
Article in English | MEDLINE | ID: mdl-26972685

ABSTRACT

Light is the major stimulus for the synchronization of circadian clocks with day-night cycles. The light-driven entrainment of the clock that controls rest-activity rhythms in Drosophila relies on different photoreceptive molecules. Cryptochrome (CRY) is expressed in most brain clock neurons, whereas six different rhodopsins (RH) are present in the light-sensing organs. The compound eye includes outer photoreceptors that express RH1 and inner photoreceptors that each express one of the four rhodopsins RH3-RH6. RH6 is also expressed in the extraretinal Hofbauer-Buchner eyelet, whereas RH2 is only found in the ocelli. In low light, the synchronization of behavioral rhythms relies on either CRY or the canonical rhodopsin phototransduction pathway, which requires the phospholipase C-ß encoded by norpA (no receptor potential A). We used norpA(P24) cry(02) double mutants that are circadianly blind in low light and restored NORPA function in each of the six types of photoreceptors, defined as expressing a particular rhodopsin. We first show that the NORPA pathway is less efficient than CRY for synchronizing rest-activity rhythms with delayed light-dark cycles but is important for proper phasing, whereas the two light-sensing pathways can mediate efficient adjustments to phase advances. Four of the six rhodopsin-expressing photoreceptors can mediate circadian entrainment, and all are more efficient for advancing than for delaying the behavioral clock. In contrast, neither RH5-expressing retinal photoreceptors nor RH2-expressing ocellar photoreceptors are sufficient to mediate synchronization through the NORPA pathway. Our results thus reveal different contributions of rhodopsin-expressing photoreceptors and suggest the existence of several circuits for rhodopsin-dependent circadian entrainment. J. Comp. Neurol. 524:2828-2844, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Circadian Rhythm/physiology , Drosophila Proteins/biosynthesis , Photic Stimulation , Photoreceptor Cells, Invertebrate/metabolism , Rhodopsin/biosynthesis , Animals , Animals, Genetically Modified , Drosophila , Drosophila Proteins/genetics , Gene Expression , Mutation/physiology , Photic Stimulation/methods , Rhodopsin/genetics
14.
Cell Rep ; 11(8): 1266-79, 2015 May 26.
Article in English | MEDLINE | ID: mdl-25981041

ABSTRACT

In the Drosophila circadian oscillator, the CLOCK/CYCLE complex activates transcription of period (per) and timeless (tim) in the evening. PER and TIM proteins then repress CLOCK (CLK) activity during the night. The pace of the oscillator depends upon post-translational regulation that affects both positive and negative components of the transcriptional loop. CLK protein is highly phosphorylated and inactive in the morning, whereas hypophosphorylated active forms are present in the evening. How this critical dephosphorylation step is mediated is unclear. We show here that two components of the STRIPAK complex, the CKA regulatory subunit of the PP2A phosphatase and its interacting protein STRIP, promote CLK dephosphorylation during the daytime. In contrast, the WDB regulatory PP2A subunit stabilizes CLK without affecting its phosphorylation state. Inhibition of the PP2A catalytic subunit and CKA downregulation affect daytime CLK similarly, suggesting that STRIPAK complexes are the main PP2A players in producing transcriptionally active hypophosphorylated CLK.


Subject(s)
CLOCK Proteins/metabolism , Circadian Rhythm/physiology , Drosophila/metabolism , Period Circadian Proteins/metabolism , Animals , Female , Male , Phosphorylation
15.
Bull Acad Natl Med ; 199(7): 1115-1131, 2015 Oct.
Article in English, French | MEDLINE | ID: mdl-29879332

ABSTRACT

The circadian clock that governs sleep-wake rhythms stems from a small set of genes, called clock genes, that are highly conserved during evolution. In insects as in mammals, a transcriptional feedback loop generates 24 h molecular oscillations. Two major transcrip- tional activators direct the expression of genes encoding repressors the accumulation of which leads afew hours later to transcriptional inhibition. This cyclic transcription is the core of the circadian oscillator and controls a large number of target genes (about 5 % of the genome), the nature of which varies from one organ to another depending on the physiology of the tissues. The period of the molecular oscillations relies on the accumulation rate of the repressors, their transfer into the cell nucleus, their ability to inhibit transcription, and their lifetime. These various parameters are largely based on post-translational regula- tions that depend on genes encoding kinases, phosphatases and ubiquitin ligases for a large fraction of them. Several syndromes that affect the sleep-wake rhythm were characterized in the human population. Inparticula; shifts of the sleep-wake rhythms compared to day-night cycles have been identifed and associated with mutations in clock genes. These mutations disrupt not only the brain clock that governs sleep-wake rhythms but also the temporal organization of many physiological processes (metabolism, detoxification etc.) through the clocks that are present in the different cell types of the body.


Subject(s)
CLOCK Proteins/genetics , Circadian Clocks/genetics , Drosophila/genetics , Evolution, Molecular , Animals , Drosophila Proteins/genetics , Humans , Light , Phylogeny
16.
PLoS Biol ; 11(8): e1001645, 2013.
Article in English | MEDLINE | ID: mdl-24013921

ABSTRACT

Phosphorylation is a pivotal regulatory mechanism for protein stability and activity in circadian clocks regardless of their evolutionary origin. It determines the speed and strength of molecular oscillations by acting on transcriptional activators and their repressors, which form negative feedback loops. In Drosophila, the CK2 kinase phosphorylates and destabilizes the PERIOD (PER) and TIMELESS (TIM) proteins, which inhibit CLOCK (CLK) transcriptional activity. Here we show that CK2 also targets the CLK activator directly. Downregulating the activity of the catalytic α subunit of CK2 induces CLK degradation, even in the absence of PER and TIM. Unexpectedly, the regulatory ß subunit of the CK2 holoenzyme is not required for the regulation of CLK stability. In addition, downregulation of CK2α activity decreases CLK phosphorylation and increases per and tim transcription. These results indicate that CK2 inhibits CLK degradation while reducing its activity. Since the CK1 kinase promotes CLK degradation, we suggest that CLK stability and transcriptional activity result from counteracting effects of CK1 and CK2.


Subject(s)
CLOCK Proteins/metabolism , Circadian Rhythm/physiology , Drosophila Proteins/metabolism , Animals , CLOCK Proteins/genetics , Circadian Rhythm/genetics , Drosophila , Drosophila Proteins/genetics , Phosphorylation
17.
PLoS Biol ; 10(8): e1001367, 2012.
Article in English | MEDLINE | ID: mdl-22879814

ABSTRACT

Eukaryotic circadian clocks rely on transcriptional feedback loops. In Drosophila, the PERIOD (PER) and TIMELESS (TIM) proteins accumulate during the night, inhibit the activity of the CLOCK (CLK)/CYCLE (CYC) transcriptional complex, and are degraded in the early morning. The control of PER and TIM oscillations largely depends on post-translational mechanisms. They involve both light-dependent and light-independent pathways that rely on the phosphorylation, ubiquitination, and proteasomal degradation of the clock proteins. SLMB, which is part of a CULLIN-1-based E3 ubiquitin ligase complex, is required for the circadian degradation of phosphorylated PER. We show here that CULLIN-3 (CUL-3) is required for the circadian control of PER and TIM oscillations. Expression of either Cul-3 RNAi or dominant negative forms of CUL-3 in the clock neurons alters locomotor behavior and dampens PER and TIM oscillations in light-dark cycles. In constant conditions, CUL-3 deregulation induces behavioral arrhythmicity and rapidly abolishes TIM cycling, with slower effects on PER. CUL-3 affects TIM accumulation more strongly in the absence of PER and forms protein complexes with hypo-phosphorylated TIM. In contrast, SLMB affects TIM more strongly in the presence of PER and preferentially associates with phosphorylated TIM. CUL-3 and SLMB show additive effects on TIM and PER, suggesting different roles for the two ubiquitination complexes on PER and TIM cycling. This work thus shows that CUL-3 is a new component of the Drosophila clock, which plays an important role in the control of TIM oscillations.


Subject(s)
Behavior, Animal/physiology , Circadian Clocks , Cullin Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Animals , Blotting, Western , Cullin Proteins/genetics , Drosophila/genetics , Drosophila/physiology , Drosophila Proteins/genetics , Gene Expression Regulation , Locomotion , Multiprotein Complexes/metabolism , Neurons/metabolism , Period Circadian Proteins/metabolism , Phosphorylation , Protein Stability , Proteolysis , RNA Interference , RNA, Messenger/analysis , RNA, Messenger/metabolism , Ubiquitination
18.
19.
PLoS One ; 7(3): e31867, 2012.
Article in English | MEDLINE | ID: mdl-22427812

ABSTRACT

Cryptochromes are conserved flavoprotein receptors found throughout the biological kingdom with diversified roles in plant development and entrainment of the circadian clock in animals. Light perception is proposed to occur through flavin radical formation that correlates with biological activity in vivo in both plants and Drosophila. By contrast, mammalian (Type II) cryptochromes regulate the circadian clock independently of light, raising the fundamental question of whether mammalian cryptochromes have evolved entirely distinct signaling mechanisms. Here we show by developmental and transcriptome analysis that Homo sapiens cryptochrome--1 (HsCRY1) confers biological activity in transgenic expressing Drosophila in darkness, that can in some cases be further stimulated by light. In contrast to all other cryptochromes, purified recombinant HsCRY1 protein was stably isolated in the anionic radical flavin state, containing only a small proportion of oxidized flavin which could be reduced by illumination. We conclude that animal Type I and Type II cryptochromes may both have signaling mechanisms involving formation of a flavin radical signaling state, and that light independent activity of Type II cryptochromes is a consequence of dark accumulation of this redox form in vivo rather than of a fundamental difference in signaling mechanism.


Subject(s)
Circadian Rhythm/physiology , Cryptochromes/metabolism , Flavins/metabolism , Metamorphosis, Biological/physiology , Signal Transduction/physiology , Animals , Animals, Genetically Modified , Cryptochromes/isolation & purification , DNA Primers/genetics , Darkness , Drosophila , Gene Expression Profiling , Humans , Microarray Analysis , Reverse Transcriptase Polymerase Chain Reaction
20.
J Neurosci ; 31(48): 17406-15, 2011 Nov 30.
Article in English | MEDLINE | ID: mdl-22131402

ABSTRACT

In Drosophila, opsin visual photopigments as well as blue-light-sensitive cryptochrome (CRY) contribute to the synchronization of circadian clocks. We focused on the relatively simple larval brain, with nine clock neurons per hemisphere: five lateral neurons (LNs), four of which express the pigment-dispersing factor (PDF) neuropeptide, and two pairs of dorsal neurons (DN1s and DN2s). CRY is present only in the PDF-expressing LNs and the DN1s. The larval visual organ expresses only two rhodopsins (RH5 and RH6) and projects onto the LNs. We recently showed that PDF signaling is required for light to synchronize the CRY(-) larval DN2s. We now show that, in the absence of functional CRY, synchronization of the DN1s also requires PDF, suggesting that these neurons have no direct connection with the visual system. In contrast, the fifth (PDF(-)) LN does not require the PDF-expressing cells to receive visual system inputs. All clock neurons are light-entrained by light-dark cycles in the rh5(2);cry(b), rh6(1) cry(b), and rh5(2);rh6(1) double mutants, whereas the triple mutant is circadianly blind. Thus, any one of the three photosensitive molecules is sufficient, and there is no other light input for the larval clock. Finally, we show that constant activation of the visual system can suppress molecular oscillations in the four PDF-expressing LNs, whereas, in the adult, this effect of constant light requires CRY. A surprising diversity and specificity of light input combinations thus exists even for this simple clock network.


Subject(s)
Brain/physiology , Drosophila/physiology , Neurons/physiology , Photoreceptor Cells, Invertebrate/physiology , Animals , Animals, Genetically Modified , Behavior, Animal/physiology , CLOCK Proteins/genetics , Drosophila Proteins/genetics , Larva/physiology , Photic Stimulation/methods , Photoperiod , Rhodopsin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...