Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Prog Biophys Mol Biol ; 163: 171-186, 2021 08.
Article in English | MEDLINE | ID: mdl-33636189

ABSTRACT

Arrival of the novel SARS-CoV-2 has launched a worldwide effort to identify both pre-approved and novel therapeutics targeting the viral proteome, highlighting the urgent need for efficient drug discovery strategies. Even with effective vaccines, infection is possible, and at-risk populations would benefit from effective drug compounds that reduce the lethality and lasting damage of COVID-19 infection. The CoV-2 MacroD-like macrodomain (Mac1) is implicated in viral pathogenicity by disrupting host innate immunity through its mono (ADP-ribosyl) hydrolase activity, making it a prime target for antiviral therapy. We therefore solved the structure of CoV-2 Mac1 from non-structural protein 3 (Nsp3) and applied structural and sequence-based genetic tracing, including newly determined A. pompejana MacroD2 and GDAP2 amino acid sequences, to compare and contrast CoV-2 Mac1 with the functionally related human DNA-damage signaling factor poly (ADP-ribose) glycohydrolase (PARG). Previously, identified targetable features of the PARG active site allowed us to develop a pharmacologically useful PARG inhibitor (PARGi). Here, we developed a focused chemical library and determined 6 novel PARGi X-ray crystal structures for comparative analysis. We applied this knowledge to discovery of CoV-2 Mac1 inhibitors by combining computation and structural analysis to identify PARGi fragments with potential to bind the distal-ribose and adenosyl pockets of the CoV-2 Mac1 active site. Scaffold development of these PARGi fragments has yielded two novel compounds, PARG-345 and PARG-329, that crystallize within the Mac1 active site, providing critical structure-activity data and a pathway for inhibitor optimization. The reported structural findings demonstrate ways to harness our PARGi synthesis and characterization pipeline to develop CoV-2 Mac1 inhibitors targeting the ADP-ribose active site. Together, these structural and computational analyses reveal a path for accelerating development of antiviral therapeutics from pre-existing drug optimization pipelines.


Subject(s)
Antiviral Agents/chemistry , Coronavirus Papain-Like Proteases/metabolism , Enzyme Inhibitors/chemistry , Glycoside Hydrolases/antagonists & inhibitors , Small Molecule Libraries/chemistry , Xanthines/chemistry , Amino Acid Sequence , Antiviral Agents/pharmacology , Catalytic Domain , Crystallography, X-Ray , Drug Discovery , Enzyme Inhibitors/pharmacology , Humans , Models, Molecular , Protein Domains , SARS-CoV-2/drug effects , SARS-CoV-2/metabolism , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Xanthines/pharmacology , COVID-19 Drug Treatment
2.
Nat Commun ; 10(1): 5654, 2019 12 11.
Article in English | MEDLINE | ID: mdl-31827085

ABSTRACT

Poly(ADP-ribose)ylation (PARylation) by PAR polymerase 1 (PARP1) and PARylation removal by poly(ADP-ribose) glycohydrolase (PARG) critically regulate DNA damage responses; yet, conflicting reports obscure PARG biology and its impact on cancer cell resistance to PARP1 inhibitors. Here, we found that PARG expression is upregulated in many cancers. We employed chemical library screening to identify and optimize methylxanthine derivatives as selective bioavailable PARG inhibitors. Multiple crystal structures reveal how substituent positions on the methylxanthine core dictate binding modes and inducible-complementarity with a PARG-specific tyrosine clasp and arginine switch, supporting inhibitor specificity and a competitive inhibition mechanism. Cell-based assays show selective PARG inhibition and PARP1 hyperPARylation. Moreover, our PARG inhibitor sensitizes cells to radiation-induced DNA damage, suppresses replication fork progression and impedes cancer cell survival. In PARP inhibitor-resistant A172 glioblastoma cells, our PARG inhibitor shows comparable killing to Nedaplatin, providing further proof-of-concept that selectively inhibiting PARG can impair cancer cell survival.


Subject(s)
DNA Replication/drug effects , Enzyme Inhibitors/pharmacology , Glycoside Hydrolases/antagonists & inhibitors , Neoplasms/genetics , Small Molecule Libraries/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Glycoside Hydrolases/genetics , Glycoside Hydrolases/metabolism , Humans , Neoplasms/enzymology , Neoplasms/metabolism , Neoplasms/physiopathology , Poly ADP Ribosylation/drug effects , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Small Molecule Libraries/chemistry
3.
Curr Biol ; 27(16): 2431-2441.e3, 2017 Aug 21.
Article in English | MEDLINE | ID: mdl-28781048

ABSTRACT

Circadian (∼24 hr) clocks regulate daily rhythms in physiology, metabolism, and behavior via cell-autonomous transcriptional feedback loops. In Drosophila, the blue-light photoreceptor CRYPTOCHROME (CRY) synchronizes these feedback loops to light:dark cycles by binding to and degrading TIMELESS (TIM) protein. CRY also acts independently of TIM in Drosophila to alter potassium channel conductance in arousal neurons after light exposure, and in many animals CRY acts independently of light to repress rhythmic transcription. CRY expression has been characterized in the Drosophila brain and eyes, but not in peripheral clock and non-clock tissues in the body. To investigate CRY expression and function in body tissues, we generated a GFP-tagged-cry transgene that rescues light-induced behavioral phase resetting in cry03 mutant flies and sensitively reports GFP-CRY expression. In bodies, CRY is detected in clock-containing tissues including Malpighian tubules, where it mediates both light-dependent TIM degradation and clock function. In larval salivary glands, which lack clock function but are amenable to electrophysiological recording, CRY prevents membrane input resistance from falling to low levels in a light-independent manner. The ability of CRY to maintain high input resistance in these non-excitable cells also requires the K+ channel subunits Hyperkinetic, Shaker, and ether-a-go-go. These findings for the first time define CRY expression in Drosophila peripheral tissues and reveal that CRY acts together with K+ channels to maintain passive membrane properties in a non-clock-containing peripheral tissue independent of light.


Subject(s)
Circadian Clocks/genetics , Cryptochromes/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/physiology , Eye Proteins/genetics , Light , Animals , Cryptochromes/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Eye Proteins/metabolism , Gene Expression Profiling , Green Fluorescent Proteins/genetics , Larva/genetics , Larva/growth & development , Larva/physiology , Transgenes/genetics
4.
J Neurosci ; 35(22): 8662-71, 2015 Jun 03.
Article in English | MEDLINE | ID: mdl-26041931

ABSTRACT

Circadian pacemaker neurons in the Drosophila brain control daily rhythms in locomotor activity. These pacemaker neurons can be subdivided into early or late groups depending on whether rhythms in period (per) and timeless (tim) expression are initiated at the first instar (L1) larval stage or during metamorphosis, respectively. Because CLOCK-CYCLE (CLK-CYC) heterodimers initiate circadian oscillator function by activating per and tim transcription, a Clk-GFP transgene was used to mark when late pacemaker neurons begin to develop. We were surprised to see that CLK-GFP was already expressed in four of five clusters of late pacemaker neurons during the third instar (L3) larval stage. CLK-GFP is only detected in postmitotic neurons from L3 larvae, suggesting that these four late pacemaker neuron clusters are formed before the L3 larval stage. A GFP-cyc transgene was used to show that CYC, like CLK, is also expressed exclusively in pacemaker neurons from L3 larval brains, demonstrating that CLK-CYC is not sufficient to activate per and tim in late pacemaker neurons at the L3 larval stage. These results suggest that most late pacemaker neurons develop days before novel factors activate circadian oscillator function during metamorphosis.


Subject(s)
Biological Clocks/physiology , Brain/cytology , Circadian Rhythm/genetics , Gene Expression Regulation, Developmental/physiology , Neurons/physiology , ARNTL Transcription Factors/genetics , Age Factors , Animals , Animals, Genetically Modified , Biological Clocks/genetics , Brain/growth & development , CLOCK Proteins/genetics , Drosophila , Drosophila Proteins/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Larva , Microscopy, Confocal , Motor Activity/genetics , Pupa
5.
Curr Biol ; 25(7): 858-67, 2015 Mar 30.
Article in English | MEDLINE | ID: mdl-25754644

ABSTRACT

Circadian neural circuits generate near 24-hr physiological rhythms that can be entrained by light to coordinate animal physiology with daily solar cycles. To examine how a circadian circuit reorganizes its activity in response to light, we imaged period (per) clock gene cycling for up to 6 days at single-neuron resolution in whole-brain explant cultures prepared from per-luciferase transgenic flies. We compared cultures subjected to a phase-advancing light pulse (LP) to cultures maintained in darkness (DD). In DD, individual neuronal oscillators in all circadian subgroups are initially well synchronized but then show monotonic decrease in oscillator rhythm amplitude and synchrony with time. The small ventral lateral neurons (s-LNvs) and dorsal lateral neurons (LNds) exhibit this decrease at a slower relative rate. In contrast, the LP evokes a rapid loss of oscillator synchrony between and within most circadian neuronal subgroups, followed by gradual phase retuning of whole-circuit oscillator synchrony. The LNds maintain high rhythmic amplitude and synchrony following the LP along with the most rapid coherent phase advance. Immunocytochemical analysis of PER shows that these dynamics in DD and LP are recapitulated in vivo. Anatomically distinct circadian neuronal subgroups vary in their response to the LP, showing differences in the degree and kinetics of their loss, recovery and/or strengthening of synchrony, and rhythmicity. Transient desynchrony appears to be an integral feature of light response of the Drosophila multicellular circadian clock. Individual oscillators in different neuronal subgroups of the circadian circuit show distinct kinetic signatures of light response and phase retuning.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm/physiology , Light , Nerve Net/physiology , Neurons/metabolism , Period Circadian Proteins/metabolism , Animals , Animals, Genetically Modified , Darkness , Drosophila/physiology , Drosophila Proteins/metabolism , Time Factors , Ventral Thalamic Nuclei/cytology
6.
Proc Natl Acad Sci U S A ; 112(7): 2245-50, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25646452

ABSTRACT

Blue light activation of the photoreceptor CRYPTOCHROME (CRY) evokes rapid depolarization and increased action potential firing in a subset of circadian and arousal neurons in Drosophila melanogaster. Here we show that acute arousal behavioral responses to blue light significantly differ in mutants lacking CRY, as well as mutants with disrupted opsin-based phototransduction. Light-activated CRY couples to membrane depolarization via a well conserved redox sensor of the voltage-gated potassium (K(+)) channel ß-subunit (Kvß) Hyperkinetic (Hk). The neuronal light response is almost completely absent in hk(-/-) mutants, but is functionally rescued by genetically targeted neuronal expression of WT Hk, but not by Hk point mutations that disable Hk redox sensor function. Multiple K(+) channel α-subunits that coassemble with Hk, including Shaker, Ether-a-go-go, and Ether-a-go-go-related gene, are ion conducting channels for CRY/Hk-coupled light response. Light activation of CRY is transduced to membrane depolarization, increased firing rate, and acute behavioral responses by the Kvß subunit redox sensor.


Subject(s)
Cryptochromes/physiology , Light Signal Transduction , Potassium Channels/physiology , Animals , Drosophila , Oxidation-Reduction
7.
J Biol Chem ; 289(28): 19681-93, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-24872414

ABSTRACT

Circadian (≅ 24 h) clocks control daily rhythms in metabolism, physiology, and behavior in animals, plants, and microbes. In Drosophila, these clocks keep circadian time via transcriptional feedback loops in which clock-cycle (CLK-CYC) initiates transcription of period (per) and timeless (tim), accumulating levels of PER and TIM proteins feed back to inhibit CLK-CYC, and degradation of PER and TIM allows CLK-CYC to initiate the next cycle of transcription. The timing of key events in this feedback loop are controlled by, or coincide with, rhythms in PER and CLK phosphorylation, where PER and CLK phosphorylation is high during transcriptional repression. PER phosphorylation at specific sites controls its subcellular localization, activity, and stability, but comparatively little is known about the identity and function of CLK phosphorylation sites. Here we identify eight CLK phosphorylation sites via mass spectrometry and determine how phosphorylation at these sites impacts behavioral and molecular rhythms by transgenic rescue of a new Clk null mutant. Eliminating phosphorylation at four of these sites accelerates the feedback loop to shorten the circadian period, whereas loss of CLK phosphorylation at serine 859 increases CLK activity, thereby increasing PER levels and accelerating transcriptional repression. These results demonstrate that CLK phosphorylation influences the circadian period by regulating CLK activity and progression through the feedback loop.


Subject(s)
CLOCK Proteins/metabolism , Circadian Rhythm/physiology , Drosophila Proteins/metabolism , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , CLOCK Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Phosphorylation/physiology
8.
Curr Biol ; 21(9): 756-61, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21514156

ABSTRACT

The Drosophila circadian oscillator is comprised of transcriptional feedback loops that are activated by CLOCK (CLK) and CYCLE (CYC) and repressed by PERIOD (PER) and TIMELESS (TIM) [1]. The timing of CLK-CYC activation and PER-TIM repression is regulated posttranslationally, in part through rhythmic phosphorylation of CLK, PER, and TIM [2-4]. Although kinases that control PER and TIM levels and subcellular localization have been identified [5-10], additional kinases are predicted to target PER, TIM, and/or CLK to promote time-specific transcriptional repression. We screened for kinases that alter circadian behavior via clock cell-directed RNA interference (RNAi) and identified the proline-directed kinase nemo (nmo) as a novel component of the circadian oscillator. Both nmo RNAi knockdown and a nmo hypomorphic mutant shorten circadian period, whereas nmo overexpression lengthens circadian period. CLK levels increase when nmo expression is knocked down in clock cells, whereas CLK levels decrease and PER and TIM accumulation are delayed when nmo is overexpressed in clock cells. These data suggest that nmo slows the pace of the circadian oscillator by altering CLK, PER, and TIM expression, thereby contributing to the generation of an ~24 hr circadian period.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , Drosophila Proteins/metabolism , Drosophila/physiology , Gene Expression Regulation/physiology , Mitogen-Activated Protein Kinases/metabolism , ARNTL Transcription Factors/metabolism , Animals , CLOCK Proteins/metabolism , Period Circadian Proteins/metabolism , Phosphorylation , RNA Interference
9.
Curr Biol ; 20(4): 300-9, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-20153192

ABSTRACT

BACKGROUND: Circadian regulation of chemosensory processes is common in animals, but little is known about how circadian clocks control chemosensory systems or the consequences of rhythms in chemosensory system function. Taste is a major chemosensory gate used to decide whether or not an animal will eat, and the main taste organ in Drosophila, the proboscis, harbors autonomous circadian oscillators. Here we examine gustatory physiology, tastant-evoked appetitive behavior, and food ingestion to understand clock-dependent regulation of the Drosophila gustatory system. RESULTS: Here we report that single-unit responses from labellar gustatory receptor neurons (GRNs) to attractive and aversive tastants show diurnal and circadian rhythms in spike amplitude, frequency, and duration across different classes of gustatory sensilla. Rhythms in electrophysiological responses parallel behavioral rhythms in proboscis extension reflex. Molecular oscillators in GRNs are necessary and sufficient for rhythms in gustatory responses and drive rhythms in G protein-coupled receptor kinase 2 (GPRK2) expression that mediate rhythms in taste sensitivity. Eliminating clock function in certain GRNs increases feeding and locomotor activity, mimicking a starvation response. CONCLUSIONS: Circadian clocks in GRNs control neuronal output and drive behavioral rhythms in taste responses that peak at a time of day when feeding is maximal in flies. Our results argue that oscillations in GPRK2 levels drive rhythms in gustatory physiology and behavior and that GRN clocks repress feeding. The similarity in gustatory system organization and feeding behavior in flies and mammals, as well as diurnal changes in taste sensitivity in humans, suggest that our results are relevant to the situation in humans.


Subject(s)
Appetitive Behavior/physiology , Chemoreceptor Cells/metabolism , Circadian Rhythm/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , G-Protein-Coupled Receptor Kinase 2/metabolism , Receptors, Cell Surface/metabolism , Taste/physiology , Analysis of Variance , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Blotting, Western , Electrophysiology , Fluorescent Antibody Technique
10.
BMC Neurosci ; 9: 119, 2008 Dec 18.
Article in English | MEDLINE | ID: mdl-19094242

ABSTRACT

BACKGROUND: The Drosophila circadian oscillator is composed of transcriptional feedback loops in which CLOCK-CYCLE (CLK-CYC) heterodimers activate their feedback regulators period (per) and timeless (tim) via E-box mediated transcription. These feedback loop oscillators are present in distinct clusters of dorsal and lateral neurons in the adult brain, but how this pattern of expression is established during development is not known. Since CLK is required to initiate feedback loop function, defining the pattern of CLK expression in embryos and larvae will shed light on oscillator neuron development. RESULTS: A novel CLK antiserum is used to show that CLK expression in the larval CNS and adult brain is limited to circadian oscillator cells. CLK is initially expressed in presumptive small ventral lateral neurons (s-LNvs), dorsal neurons 2 s (DN2s), and dorsal neuron 1 s (DN1s) at embryonic stage (ES) 16, and this CLK expression pattern persists through larval development. PER then accumulates in all CLK-expressing cells except presumptive DN2s during late ES 16 and ES 17, consistent with the delayed accumulation of PER in adult oscillator neurons and antiphase cycling of PER in larval DN2s. PER is also expressed in non-CLK-expressing cells in the embryonic CNS starting at ES 12. Although PER expression in CLK-negative cells continues in ClkJrk embryos, PER expression in cells that co-express PER and CLK is eliminated. CONCLUSION: These data demonstrate that brain oscillator neurons begin development during embryogenesis, that PER expression in non-oscillator cells is CLK-independent, and that oscillator phase is an intrinsic characteristic of brain oscillator neurons. These results define the temporal and spatial coordinates of factors that initiate Clk expression, imply that circadian photoreceptors are not activated until the end of embryogenesis, and suggest that PER functions in a different capacity before oscillator cell development is initiated.


Subject(s)
Biological Clocks/physiology , Brain/embryology , Drosophila Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Biological Clocks/genetics , Biological Clocks/radiation effects , CLOCK Proteins , Circadian Rhythm , Drosophila , Drosophila Proteins/genetics , Gene Expression , Genes, Insect , Larva/genetics , Larva/metabolism , Microscopy, Confocal , Neurogenesis , Neurons/metabolism , Period Circadian Proteins , Photoreceptor Cells, Invertebrate/physiology , Transcription Factors/genetics
11.
J Biol Rhythms ; 23(4): 296-307, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18663237

ABSTRACT

In the fruit fly Drosophila melanogaster, CRYPTOCHROME (CRY) functions as a photoreceptor to entrain circadian oscillators to light-dark cycles and as a transcription factor to maintain circadian oscillator function in certain peripheral tissues. Given the importance of CRY to circadian clock function, we expected this protein to be expressed in all oscillator cells, yet CRY cellular distribution and subcellular localization has not been firmly established. Here we investigate CRY spatial expression in the brain using a newly developed CRY antibody and a novel set of cry deletion mutants. We find that CRY is expressed in s-LNvs, l-LNvs, and a subset of LNds and DN1s, but not DN2s and DN3s. CRY is present in both the nucleus and the cytoplasm of these neurons, and its subcellular localization does not change over the circadian cycle. Although CRY is absent in DN2s and DN3s, cry promoter activity and/or cry mRNA accumulation can be detected in these neurons, suggesting that CRY levels are regulated posttranscriptionally. Oscillators in DN2s and DN3s entrain to environmental light-dark cycles, which implies that they are entrained indirectly by retinal photoreceptors, extraretinal photoreceptors, or other CRY-expressing cells.


Subject(s)
Biological Clocks/physiology , Central Nervous System/physiology , Drosophila Proteins/metabolism , Drosophila/physiology , Eye Proteins/metabolism , Photoreceptor Cells, Invertebrate/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Animals, Genetically Modified , Biological Clocks/genetics , Cell Nucleus/metabolism , Central Nervous System/metabolism , Cryptochromes , Cytoplasm/metabolism , Drosophila Proteins/genetics , Eye Proteins/genetics , Fluorescent Antibody Technique, Direct , Gene Deletion , Neurons/metabolism , Receptors, G-Protein-Coupled/genetics
12.
J Biol Rhythms ; 21(2): 93-103, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16603674

ABSTRACT

CLOCK (CLK) is a core component of the transcriptional feedback loops that comprise the circadian timekeeping mechanism in Drosophila. As a heterodimer with CYCLE (CYC), CLK binds E-boxes to activate the transcription of rhythmically expressed genes within and downstream of the circadian clock, but this activation unexpectedly occurs at times when CLK is at its lowest levels on Western blots. Recent studies demonstrate that CLK also regulates nonrhythmic gene expression and behaviors. Despite the critical roles CLK plays within and outside the circadian clock, its spatial expression pattern has not been characterized. Using a newly developed CLK antibody, the authors show that CLK is coexpressed with PERIOD (PER) in canonical oscillator cells throughout the head and body. In contrast to PER, however, the levels of CLK immunoreactivity do not cycle in intensity, CLK is detected primarily in the nucleus throughout the circadian cycle, and CLK is expressed in non-oscillator cells within the lateral and dorsal brain, including Kenyon cells, which mediate various forms of learning and memory. These results indicate that constitutive levels of nuclear CLK regulate rhythmic transcription in circadian oscillator cells and suggest that CLK contributes to other behavioral processes by regulating gene expression in non-oscillator cells.


Subject(s)
Drosophila Proteins/physiology , Gene Expression Regulation , Transcription Factors/physiology , Animals , Blotting, Western , Brain/metabolism , CLOCK Proteins , Cell Nucleus/metabolism , Circadian Rhythm , Drosophila , Drosophila Proteins/metabolism , Immunoblotting , Immunohistochemistry , Microscopy, Confocal , Microscopy, Fluorescence , Models, Biological , Nuclear Proteins/metabolism , Oscillometry , Period Circadian Proteins , Protein Binding , Time Factors , Transcription Factors/metabolism , Transcription, Genetic
13.
Genes Dev ; 20(6): 723-33, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16543224

ABSTRACT

Transcriptional activation by CLOCK-CYCLE (CLK-CYC) heterodimers and repression by PERIOD-TIMELESS (PER-TIM) heterodimers are essential for circadian oscillator function in Drosophila. PER-TIM was previously found to interact with CLK-CYC to repress transcription, and here we show that this interaction inhibits binding of CLK-CYC to E-box regulatory elements in vivo. Coincident with the interaction between PER-TIM and CLK-CYC is the hyperphosphorylation of CLK. This hyperphosphorylation occurs in parallel with the PER-dependent entry of DOUBLE-TIME (DBT) kinase into a complex with CLK-CYC, where DBT destabilizes both CLK and PER. Once PER and CLK are degraded, a novel hypophosphorylated form of CLK accumulates in parallel with E-box binding and transcriptional activation. These studies suggest that PER-dependent rhythms in CLK phosphorylation control rhythms in E-box-dependent transcription and CLK stability, thus linking PER and CLK function during the circadian cycle and distinguishing the transcriptional feedback mechanism in flies from that in mammals.


Subject(s)
Circadian Rhythm , Drosophila Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Base Sequence , CLOCK Proteins , Cells, Cultured , DNA Primers , Drosophila , Phosphorylation , Polymerase Chain Reaction
14.
Neuron ; 37(2): 249-61, 2003 Jan 23.
Article in English | MEDLINE | ID: mdl-12546820

ABSTRACT

The Drosophila circadian oscillator consists of interlocked period (per)/timeless (tim) and Clock (Clk) transcriptional/translational feedback loops. Within these feedback loops, CLK and CYCLE (CYC) activate per and tim transcription at the same time as they repress Clk transcription, thus controlling the opposite cycling phases of these transcripts. CLK-CYC directly bind E box elements to activate transcription, but the mechanism of CLK-CYC-dependent repression is not known. Here we show that a CLK-CYC-activated gene, vrille (vri), encodes a repressor of Clk transcription, thereby identifying vri as a key negative component of the Clk feedback loop in Drosophila's circadian oscillator. The blue light photoreceptor encoding cryptochrome (cry) gene is also a target for VRI repression, suggesting a broader role for VRI in the rhythmic repression of output genes that cycle in phase with Clk.


Subject(s)
Circadian Rhythm/genetics , Drosophila Proteins , Drosophila/physiology , Trans-Activators/genetics , Transcription Factors/genetics , Transcription Factors/physiology , Animals , Animals, Genetically Modified , Binding Sites , Blotting, Western , CLOCK Proteins , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/metabolism , Electrophoretic Mobility Shift Assay , Feedback/physiology , G-Box Binding Factors , Hot Temperature , Immunohistochemistry , Molecular Sequence Data , Nuclease Protection Assays , Photoreceptor Cells, Invertebrate/physiology , RNA, Messenger/biosynthesis , Transcription Factors/biosynthesis , Transcription Factors/metabolism
15.
Novartis Found Symp ; 253: 140-50; discussion 150-60, 2003.
Article in English | MEDLINE | ID: mdl-14712919

ABSTRACT

Drosophila circadian oscillators comprise interlocked period (per)/timeless (tim) and Clock (Clk) transcriptional/translational feedback loops. Within these feedback loops, CLOCK (CLK) and CYCLE (CYC) bind E-box elements to activate per and tim transcription, and we now show that at the same time CLK-CYC repress Clk by activating the transcriptional repressor vrille (vri), thus accounting for the opposite cycling phases of these transcripts and identifying vri as the negative component of the Clk-feedback-loop. The core oscillator mechanism is assumed to be the same for oscillators in different tissues. However, we have shown that CRYPTOCHROME (CRY) has a light-independent function in the oscillator that controls olfaction rhythms, suggesting that CRY may function within the oscillator mechanism itself as it does in mammals. These olfaction rhythms require the function of 'peripheral' oscillators which are distinct from the 'central' lateral neuron (LN) oscillators that mediate locomotor activity rhythms. Preliminary results show that antennal oscillator cells are sufficient and LNs are not necessary for olfaction rhythms, indicating that unlike the situation in mammals, the central oscillator has little impact on the olfaction rhythm oscillator under these conditions.


Subject(s)
Circadian Rhythm/physiology , Drosophila/physiology , Animals , CLOCK Proteins , Circadian Rhythm/genetics , Cryptochromes , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Eye Proteins/genetics , Eye Proteins/physiology , Feedback , Genes, Insect , Models, Biological , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Period Circadian Proteins , Photoreceptor Cells, Invertebrate/physiology , RNA/genetics , RNA/metabolism , Receptors, G-Protein-Coupled , Transcription Factors/genetics , Transcription Factors/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...