Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35064085

RESUMEN

Transcriptional repression drives feedback loops that are central to the generation of circadian (∼24-h) rhythms. In mammals, circadian repression of circadian locomotor output cycles kaput, and brain and muscle ARNT-like 1 (CLOCK:BMAL1)-mediated transcription is provided by a complex formed by PERIOD (PER) and CRYPTOCHROME (CRY) proteins. PER initiates transcriptional repression by binding CLK:BMAL1, which ultimately results in their removal from DNA. Although PER's ability to repress transcription is widely recognized, how PER binding triggers repression by removing CLK:BMAL1 from DNA is not known. Here, we use the monarch butterfly as a model system to address this problem because it harbors a simplified version of the CLK:BMAL1-activated circadian clock present in mammals. We report that an intact CLOCK mouse exon 19 homologous region (CLKe19r) and the histone methyltransferase TRITHORAX (TRX) are both necessary for monarch CLK:BMAL1-mediated transcriptional activation, CLK-PER interaction, and PER repression. Our results show that TRX catalytic activity is essential for CLK-PER interaction and PER repression via the methylation of a single arginine methylation site (R45) on heat shock protein 68 (HSP68). Our study reveals TRX and HSP68 as essential links between circadian activation and PER-mediated repression and suggests a potential conserved clock function for HSPs in eukaryotes.


Asunto(s)
Arginina/metabolismo , Mariposas Diurnas/fisiología , Proteínas Cromosómicas no Histona/metabolismo , Ritmo Circadiano , Proteínas de Choque Térmico/metabolismo , Proteínas Circadianas Period/metabolismo , Secuencia de Aminoácidos , Animales , Ritmo Circadiano/genética , Secuencia Conservada , Exones , Proteínas de Choque Térmico/genética , Péptidos y Proteínas de Señalización Intracelular , Metilación , Modelos Biológicos , Activación Transcripcional
2.
Proc Natl Acad Sci U S A ; 114(41): 10972-10977, 2017 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-28973907

RESUMEN

The Drosophila circadian clock keeps time via transcriptional feedback loops. These feedback loops are initiated by CLOCK-CYCLE (CLK-CYC) heterodimers, which activate transcription of genes encoding the feedback repressors PERIOD and TIMELESS. Circadian clocks normally operate in ∼150 brain pacemaker neurons and in many peripheral tissues in the head and body, but can also be induced by expressing CLK in nonclock cells. These ectopic clocks also require cyc, yet CYC expression is restricted to canonical clock cells despite evidence that cyc mRNA is widely expressed. Here we show that CLK binds to and stabilizes CYC in cell culture and in nonclock cells in vivo. Ectopic clocks also require the blue light photoreceptor CRYPTOCHROME (CRY), which is required for both light entrainment and clock function in peripheral tissues. These experiments define the genetic architecture required to initiate circadian clock function in Drosophila, reveal mechanisms governing circadian activator stability that are conserved in perhaps all eukaryotes, and suggest that Clk, cyc, and cry expression is sufficient to drive clock expression in naive cells.


Asunto(s)
Factores de Transcripción ARNTL/química , Animales Modificados Genéticamente/metabolismo , Proteínas CLOCK/metabolismo , Relojes Circadianos , Criptocromos/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Neuronas/fisiología , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Animales , Animales Modificados Genéticamente/genética , Proteínas CLOCK/genética , Células Cultivadas , Ritmo Circadiano , Proteínas de Drosophila/genética , Neuronas/citología
3.
Proc Natl Acad Sci U S A ; 114(36): E7516-E7525, 2017 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-28831003

RESUMEN

Circadian repression of CLOCK-BMAL1 by PERIOD and CRYPTOCHROME (CRY) in mammals lies at the core of the circadian timekeeping mechanism. CRY repression of CLOCK-BMAL1 and regulation of circadian period are proposed to rely primarily on competition for binding with coactivators on an α-helix located within the transactivation domain (TAD) of the BMAL1 C terminus. This model has, however, not been tested in vivo. Here, we applied CRISPR/Cas9-mediated mutagenesis in the monarch butterfly (Danaus plexippus), which possesses a vertebrate-like CRY (dpCRY2) and an ortholog of BMAL1, to show that insect CRY2 regulates circadian repression through TAD α-helix-dependent and -independent mechanisms. Monarch mutants lacking the BMAL1 C terminus including the TAD exhibited arrhythmic eclosion behavior. In contrast, mutants lacking the TAD α-helix but retaining the most distal C-terminal residues exhibited robust rhythms during the first day of constant darkness (DD1), albeit with a delayed peak of eclosion. Phase delay in this mutant on DD1 was exacerbated in the presence of a single functional allele of dpCry2, and rhythmicity was abolished in the absence of dpCRY2. Reporter assays in Drosophila S2 cells further revealed that dpCRY2 represses through two distinct mechanisms: a TAD-dependent mechanism that involves the dpBMAL1 TAD α-helix and dpCLK W328 and a TAD-independent mechanism involving dpCLK E333. Together, our results provide evidence for independent mechanisms of vertebrate-like CRY circadian regulation on the BMAL1 C terminus and the CLK PAS-B domain and demonstrate the importance of a BMAL1 TAD-independent mechanism for generating circadian rhythms in vivo.


Asunto(s)
Factores de Transcripción ARNTL/genética , Mariposas Diurnas/genética , Proteínas CLOCK/genética , Ritmo Circadiano/genética , Criptocromos/genética , Transcripción Genética/genética , Vertebrados/genética , Secuencia de Aminoácidos , Animales , Células Cultivadas , Drosophila/genética , Proteínas de Drosophila/genética , Ratones , Proteínas Circadianas Period/genética
4.
PLoS Genet ; 12(11): e1006430, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27814361

RESUMEN

The Drosophila circadian oscillator controls daily rhythms in physiology, metabolism and behavior via transcriptional feedback loops. CLOCK-CYCLE (CLK-CYC) heterodimers initiate feedback loop function by binding E-box elements to activate per and tim transcription. PER-TIM heterodimers then accumulate, bind CLK-CYC to inhibit transcription, and are ultimately degraded to enable the next round of transcription. The timing of transcriptional events in this feedback loop coincide with, and are controlled by, rhythms in CLK-CYC binding to E-boxes. PER rhythmically binds CLK-CYC to initiate transcriptional repression, and subsequently promotes the removal of CLK-CYC from E-boxes. However, little is known about the mechanism by which CLK-CYC is removed from DNA. Previous studies demonstrated that the transcription repressor CLOCKWORK ORANGE (CWO) contributes to core feedback loop function by repressing per and tim transcription in cultured S2 cells and in flies. Here we show that CWO rhythmically binds E-boxes upstream of core clock genes in a reciprocal manner to CLK, thereby promoting PER-dependent removal of CLK-CYC from E-boxes, and maintaining repression until PER is degraded and CLK-CYC displaces CWO from E-boxes to initiate transcription. These results suggest a model in which CWO co-represses CLK-CYC transcriptional activity in conjunction with PER by competing for E-box binding once CLK-CYC-PER complexes have formed. Given that CWO orthologs DEC1 and DEC2 also target E-boxes bound by CLOCK-BMAL1, a similar mechanism may operate in the mammalian clock.


Asunto(s)
Factores de Transcripción ARNTL/genética , Proteínas CLOCK/genética , Ritmo Circadiano/genética , Proteínas de Drosophila/genética , Proteínas Circadianas Period/genética , Factores de Transcripción ARNTL/metabolismo , Animales , Proteínas CLOCK/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Elementos E-Box , Regulación de la Expresión Génica , Proteínas Circadianas Period/metabolismo , Proteínas Represoras , Transcripción Genética
5.
J Neurosci ; 36(13): 3860-70, 2016 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-27030770

RESUMEN

InDrosophila, a transcriptional feedback loop that is activated by CLOCK-CYCLE (CLK-CYC) complexes and repressed by PERIOD-TIMELESS (PER-TIM) complexes keeps circadian time. The timing of CLK-CYC activation and PER-TIM repression is regulated post-translationally, in part through rhythmic phosphorylation of CLK, PER, and TIM. Although kinases that control PER, TIM, and CLK levels, activity, and/or subcellular localization have been identified, less is known about phosphatases that control clock protein dephosphorylation. To identify clock-relevant phosphatases, clock-cell-specific RNAi knockdowns ofDrosophilaphosphatases were screened for altered activity rhythms. One phosphatase that was identified, the receptor protein tyrosine phosphatase leukocyte-antigen-related (LAR), abolished activity rhythms in constant darkness (DD) without disrupting the timekeeping mechanism in brain pacemaker neurons. However, expression of the neuropeptide pigment-dispersing factor (PDF), which mediates pacemaker neuron synchrony and output, is eliminated in the dorsal projections from small ventral lateral (sLNv) pacemaker neurons whenLarexpression is knocked down during development, but not in adults. Loss ofLarfunction eliminates sLNvdorsal projections, but PDF expression persists in sLNvand large ventral lateral neuron cell bodies and their remaining projections. In contrast to the defects in lights-on and lights-off anticipatory activity seen in flies that lack PDF,LarRNAi knockdown flies anticipate the lights-on and lights-off transition normally. Our results demonstrate thatLaris required for sLNvdorsal projection development and suggest that PDF expression in LNvcell bodies and their remaining projections mediate anticipation of the lights-on and lights-off transitions during a light/dark cycle. SIGNIFICANCE STATEMENT: In animals, circadian clocks drive daily rhythms in physiology, metabolism, and behavior via transcriptional feedback loops. Because key circadian transcriptional activators and repressors are regulated by phosphorylation, we screened for phosphatases that alter activity rhythms when their expression was reduced. One such phosphatase, leukocyte-antigen-related (LAR), abolishes activity rhythms, but does not disrupt feedback loop function. Rather,Lardisrupts clock output by eliminating axonal processes from clock neurons that release pigment-dispersing factor (PDF) neuropeptide into the dorsal brain, but PDF expression persists in their cell bodies and remaining projections. In contrast to flies that lack PDF, flies that lackLaranticipate lights-on and lights-off transitions normally, which suggests that the remaining PDF expression mediates activity during light/dark cycles.


Asunto(s)
Ritmo Circadiano/genética , Oscuridad , Proteínas de Drosophila/metabolismo , Drosophila/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Neuronas/fisiología , Proteínas Tirosina Fosfatasas Similares a Receptores/metabolismo , Animales , Animales Modificados Genéticamente , Relojes Biológicos/genética , Encéfalo/metabolismo , ADN sin Sentido/farmacología , Proteínas de Drosophila/genética , Embrión no Mamífero , Larva , Masculino , Actividad Motora/genética , Mutación/genética , Neuropéptidos/genética , Neuropéptidos/metabolismo , Interferencia de ARN/fisiología , Proteínas Tirosina Fosfatasas Similares a Receptores/genética
6.
PLoS Genet ; 10(8): e1004545, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25121504

RESUMEN

Transcriptional/translational feedback loops drive daily cycles of expression in clock genes and clock-controlled genes, which ultimately underlie many of the overt circadian rhythms manifested by organisms. Moreover, phosphorylation of clock proteins plays crucial roles in the temporal regulation of clock protein activity, stability and subcellular localization. dCLOCK (dCLK), the master transcription factor driving cyclical gene expression and the rate-limiting component in the Drosophila circadian clock, undergoes daily changes in phosphorylation. However, the physiological role of dCLK phosphorylation is not clear. Using a Drosophila tissue culture system, we identified multiple phosphorylation sites on dCLK. Expression of a mutated version of dCLK where all the mapped phospho-sites were switched to alanine (dCLK-15A) rescues the arrythmicity of Clk(out) flies, yet with an approximately 1.5 hr shorter period. The dCLK-15A protein attains substantially higher levels in flies compared to the control situation, and also appears to have enhanced transcriptional activity, consistent with the observed higher peak values and amplitudes in the mRNA rhythms of several core clock genes. Surprisingly, the clock-controlled daily activity rhythm in dCLK-15A expressing flies does not synchronize properly to daily temperature cycles, although there is no defect in aligning to light/dark cycles. Our findings suggest a novel role for clock protein phosphorylation in governing the relative strengths of entraining modalities by adjusting the dynamics of circadian gene expression.


Asunto(s)
Proteínas CLOCK/genética , Ritmo Circadiano/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Alanina/genética , Animales , Proteínas CLOCK/biosíntesis , Proteínas de Drosophila/biosíntesis , Drosophila melanogaster/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Mutación , Fosforilación/genética , ARN Mensajero/biosíntesis
7.
J Neurosci ; 35(22): 8662-71, 2015 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-26041931

RESUMEN

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.


Asunto(s)
Relojes Biológicos/fisiología , Encéfalo/citología , Ritmo Circadiano/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Neuronas/fisiología , Factores de Transcripción ARNTL/genética , Factores de Edad , Animales , Animales Modificados Genéticamente , Relojes Biológicos/genética , Encéfalo/crecimiento & desarrollo , Proteínas CLOCK/genética , Drosophila , Proteínas de Drosophila/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Larva , Microscopía Confocal , Actividad Motora/genética , Pupa
8.
J Biol Chem ; 289(28): 19681-93, 2014 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-24872414

RESUMEN

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.


Asunto(s)
Proteínas CLOCK/metabolismo , Ritmo Circadiano/fisiología , Proteínas de Drosophila/metabolismo , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Animales , Proteínas CLOCK/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Fosforilación/fisiología
9.
Proc Natl Acad Sci U S A ; 107(6): 2544-9, 2010 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-20133786

RESUMEN

The circadian clock controls many circadian outputs. Although a large number of transcripts are affected by the circadian oscillator, very little is known about their regulation and function. We show here that the Drosophila takeout gene, one of the output genes of the circadian oscillator, is regulated similarly to the circadian clock genes Clock (Clk) and cry. takeout RNA levels are at constant high levels in Clk(JRK) mutants. The circadian transcription factor PAR domain protein 1 (Pdp1epsilon) is a transcription factor that had previously been postulated to control clock output genes, particularly genes regulated similarly to Clk. In agreement with this, we show here that Pdp1epsilon is a regulator of takeout. Takeout levels are low in flies with reduced Pdp1epsilon and high in flies with increased amounts of Pdp1epsilon. Furthermore, flies with reduced or elevated Pdp1epsilon levels in the fat body display courtship defects, identifying Pdp1epsilon as an important transcriptional regulator in that tissue.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Ritmo Circadiano/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Animales , Animales Modificados Genéticamente , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Northern Blotting , Western Blotting , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Cortejo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Drosophila melanogaster/fisiología , Cuerpo Adiposo/metabolismo , Femenino , Regulación de la Expresión Génica , Masculino , Mutación , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Conducta Sexual Animal/fisiología
10.
Front Syst Neurosci ; 17: 1129152, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37034015

RESUMEN

The role of the circadian system in memory formation is an important question in neurobiology. Despite this hypothesis being intuitively appealing, the existing data is confusing. Recent work in Drosophila has helped to clarify certain aspects of the problem, but the emerging sense is that the likely mechanisms are more complex than originally conceptualized. In this report, we identify a post-training window of time (during consolidation) when the circadian clock and its components are involved in memory formation. In the broader context, our data suggest that circadian biology might have multiple roles during memory formation. Testing for its roles at multiple timepoints, and in different cells, will be necessary to resolve some of the conflicting data.

11.
Curr Biol ; 18(11): 787-94, 2008 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-18499458

RESUMEN

BACKGROUND: The Drosophila circadian clock controls rhythms in the amplitude of odor-induced electrophysiological responses that peak during the middle of night. These rhythms are dependent on clocks in olfactory sensory neurons (OSNs), suggesting that odorant receptors (ORs) or OR-dependent processes are under clock control. Because responses to odors are initiated by heteromeric OR complexes that form odor-gated and cyclic-nucleotide-activated cation channels, we tested whether regulators of ORs were under circadian-clock control. RESULTS: The levels of G protein-coupled receptor kinase 2 (Gprk2) messenger RNA and protein cycle in a circadian-clock-dependent manner with a peak around the middle of the night in antennae. Gprk2 overexpression in OSNs from wild-type or cyc(01) flies elicits constant high-amplitude electroantennogram (EAG) responses to ethyl acetate, whereas Gprk2 mutants produce constant low-amplitude EAG responses. ORs accumulate to high levels in the dendrites of OSNs around the middle of the night, and this dendritic localization of ORs is enhanced by GPRK2 overexpression at times when ORs are primarily localized in the cell body. CONCLUSIONS: These results support a model in which circadian-clock-dependent rhythms in GPRK2 abundance control the rhythmic accumulation of ORs in OSN dendrites, which in turn control rhythms in olfactory responses. The enhancement of OR function by GPRK2 contrasts with the traditional role of GPRKs in desensitizing activated receptors and suggests that GPRK2 functions through a fundamentally different mechanism to modulate OR activity.


Asunto(s)
Ritmo Circadiano/fisiología , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Receptores Odorantes/metabolismo , Olfato/fisiología , Acetatos/metabolismo , Animales , Relojes Biológicos/fisiología , Dendritas/metabolismo , Quinasa 4 del Receptor Acoplado a Proteína-G , Humanos , Neuronas Aferentes/metabolismo , Isoformas de Proteínas/metabolismo
12.
Curr Biol ; 18(11): 803-7, 2008 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-18499459

RESUMEN

Circadian changes in membrane potential and spontaneous firing frequency have been observed in microbial systems, invertebrates, and mammals. Oscillators in olfactory sensory neurons (OSNs) from Drosophila are both necessary and sufficient to sustain rhythms in electroanntenogram (EAG) responses, suggesting that odorant receptors (ORs) and/or OR-dependent processes are under clock control. We measured single-unit responses in different antennal sensillae from wild-type, clock mutant, odorant-receptor mutant, and G protein-coupled receptor kinase 2 (Gprk2) mutant flies to examine the cellular and molecular mechanisms that drive rhythms in olfaction. Spontaneous spike amplitude, but not spontaneous or odor-induced firing frequency, is under clock control in ab1 and ab3 basiconic sensillae and T2 trichoid sensillae. Mutants lacking odorant receptors in dendrites display constant low spike amplitudes, and the reduction or increase of levels of GPRK2 in OSNs results in constant low or constant high spontaneous spike amplitudes, respectively. We conclude that spike amplitude is controlled by circadian clocks in basiconic and trichoid sensillae and requires GPRK2 expression and the presence of functional ORs in dendrites. These results argue that rhythms in GPRK2 levels control OR localization and OR-dependent ion channel activity and/or composition to mediate rhythms in spontaneous spike amplitude.


Asunto(s)
Potenciales de Acción/fisiología , Relojes Biológicos/fisiología , Ritmo Circadiano/fisiología , Drosophila/fisiología , Neuronas Aferentes/fisiología , Animales , Dendritas/metabolismo , Proteínas de Drosophila/metabolismo , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Receptores Odorantes/metabolismo
13.
Curr Biol ; 31(19): 4207-4218.e4, 2021 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-34331859

RESUMEN

The Drosophila circadian clock is driven by a transcriptional feedback loop in which CLOCK-CYCLE (CLK-CYC) binds E-boxes to transcribe genes encoding the PERIOD-TIMELESS (PER-TIM) repressor, which releases CLK-CYC from E-boxes to inhibit transcription. CLOCKWORK ORANGE (CWO) reinforces PER-TIM repression by binding E-boxes to maintain PER-TIM bound CLK-CYC off DNA, but also promotes CLK-CYC transcription through an unknown mechanism. To determine how CWO activates CLK-CYC transcription, we identified CWO target genes that are upregulated in the absence of CWO repression, conserved in mammals, and preferentially expressed in brain pacemaker neurons. Among the genes identified was a putative ortholog of mouse Clock Interacting Protein Circadian (Cipc), which represses CLOCK-BMAL1 transcription. Reducing or eliminating Drosophila Cipc expression shortens period, while overexpressing Cipc lengthens period, which is consistent with previous work showing that Drosophila Cipc represses CLK-CYC transcription in S2 cells. Cipc represses CLK-CYC transcription in vivo, but not uniformly, as per is strongly repressed, tim less so, and vri hardly at all. Long period rhythms in cwo mutant flies are largely rescued when Cipc expression is reduced or eliminated, indicating that increased Cipc expression mediates the period lengthening of cwo mutants. Consistent with this behavioral rescue, eliminating Cipc rescues the decreased CLK-CYC transcription in cwo mutant flies, where per is strongly rescued, tim is moderately rescued, and vri shows little rescue. These results suggest a mechanism for CWO-dependent CLK-CYC activation: CWO inhibition of CIPC repression promotes CLK-CYC transcription. This mechanism may be conserved since cwo and Cipc perform analogous roles in the mammalian circadian clock.


Asunto(s)
Proteínas de Drosophila , Drosophila , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Animales , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Clorprofam/metabolismo , Ritmo Circadiano/genética , Drosophila/fisiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Regulación de la Expresión Génica , Mamíferos/genética , Ratones , Transcripción Genética
14.
iScience ; 24(1): 101893, 2021 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-33364582

RESUMEN

The vrille (vri) gene encodes a transcriptional repressor required for Drosophila development as well as circadian behavior in adults. Alternate first exons produce vri transcripts predicted to produce a short VRI isoform during development and long VRI in adults. A vri mutant (vri Δ679) lacking long VRI transcripts is viable, confirming that short VRI is sufficient for developmental functions, yet behavioral rhythms in vri Δ679 flies persist, showing that short VRI is sufficient for clock output. E-box regulatory elements that drive rhythmic long VRI transcript expression are required for developmental expression of short VRI transcripts. Surprisingly, long VRI transcripts primarily produce short VRI in adults, apparently due to a poor Kozak sequence context, demonstrating that short VRI drives circadian behavior. Thus, E-box-driven long VRI transcripts primarily control circadian rhythms via short VRI, whereas the same E-boxes drive short VRI transcripts that control developmental functions using short VRI.

15.
Curr Biol ; 17(3): 244-51, 2007 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-17276917

RESUMEN

Many behaviors and physiological processes including locomotor activity, feeding, sleep, mating, and migration are dependent on daily or seasonally reoccurring, external stimuli. In D. melanogaster, one of the best-studied circadian behaviors is locomotion. The fruit fly is considered a diurnal (day active/night inactive) insect, based on locomotor-activity recordings of single, socially naive flies. We developed a new circadian paradigm that can simultaneously monitor two flies in simple social contexts. We find that heterosexual couples exhibit a drastically different locomotor-activity pattern than individual males, females, or homosexual couples. Specifically, male-female couples exhibit a brief rest phase around dusk but are highly active throughout the night and early morning. This distinct locomotor-activity rhythm is dependent on the clock genes and synchronized with close-proximity encounters, which reflect courtship, between the male and female. The close-proximity rhythm is dependent on the male and not the female and requires circadian oscillators in the brain and the antenna. Taken together, our data show that constant exposure to stimuli emanating from the female and received by the male olfactory and other sensory systems is responsible for the significant shift in intrinsic locomotor output of socially interacting flies.


Asunto(s)
Ritmo Circadiano , Drosophila/fisiología , Conducta Sexual Animal , Animales , Animales Modificados Genéticamente , Relojes Biológicos , Encéfalo/fisiología , Cortejo , Proteínas de Drosophila , Femenino , Masculino , Actividad Motora , Proteínas Nucleares/genética , Proteínas Circadianas Period , Olfato
16.
Mol Cell Biol ; 27(13): 5014-28, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17452449

RESUMEN

A common feature of animal circadian clocks is the progressive phosphorylation of PERIOD (PER) proteins from hypo- to hyperphosphorylated species, events that are highly dependent on casein kinase 1 epsilon (termed DOUBLETIME [DBT] in Drosophila melanogaster) and necessary for normal clock progression. Drosophila PER (dPER) functions in the negative limb of the clockworks by presumably binding to the transcription factor CLOCK (CLK) and inhibiting its transactivation activity. Here, we identify a small region on dPER that is conserved with mammalian PERs and contains the major in vivo DBT binding domain, termed dPDBD (for dPER DBT binding domain). This domain is required for the manifestation of molecular and behavioral rhythms in vivo. In the absence of the dPDBD, the dPER protein is present at constant high levels throughout a daily cycle, undergoes little phosphorylation, and is severely impaired in its ability to function as a transcriptional repressor. Our findings indicate that the binding of dPER to CLK is not sufficient for transcriptional inhibition, implicating a more indirect mode of action whereby dPER acts as a molecular bridge to "deliver" DBT and/or other factors that directly repress CLK-dependent gene expression.


Asunto(s)
Relojes Biológicos , Caseína Cinasa 1 épsilon/metabolismo , Ritmo Circadiano , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Transcripción Genética , Secuencia de Aminoácidos , Animales , Proteínas de Ciclo Celular/metabolismo , Drosophila melanogaster/fisiología , Retroalimentación Fisiológica , Modelos Biológicos , Datos de Secuencia Molecular , Actividad Motora , Proteínas Mutantes/metabolismo , Proteínas Circadianas Period , Fosforilación , Unión Proteica , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Proteínas Represoras/metabolismo , Eliminación de Secuencia , Relación Estructura-Actividad , Activación Transcripcional/genética , Ubiquitina-Proteína Ligasas/metabolismo
17.
PLoS Genet ; 3(1): e16, 2007 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-17257054

RESUMEN

Mating behavior in Drosophila depends critically on the sexual identity of specific regions in the brain, but several studies have identified courtship genes that express products only outside the nervous system. Although these genes are each active in a variety of non-neuronal cell types, they are all prominently expressed in the adult fat body, suggesting an important role for this tissue in behavior. To test its role in male courtship, fat body was feminized using the highly specific Larval serum protein promoter. We report here that the specific feminization of this tissue strongly reduces the competence of males to perform courtship. This effect is limited to the fat body of sexually mature adults as the feminization of larval fat body that normally persists in young adults does not affect mating. We propose that feminization of fat body affects the synthesis of male-specific secreted circulating proteins that influence the central nervous system. In support of this idea, we demonstrate that Takeout, a protein known to influence mating, is present in the hemolymph of adult males but not females and acts as a secreted protein.


Asunto(s)
Conducta Animal , Drosophila melanogaster/fisiología , Cuerpo Adiposo/fisiología , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Cromatografía de Afinidad , Cartilla de ADN , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Femenino , Masculino , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Maduración Sexual
18.
Sci Rep ; 10(1): 17951, 2020 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-33087840

RESUMEN

Circadian clocks keep time via ~ 24 h transcriptional feedback loops. In Drosophila, CLOCK-CYCLE (CLK-CYC) activators and PERIOD-TIMELESS (PER-TIM) repressors are feedback loop components whose transcriptional status varies over a circadian cycle. Although changes in the state of activators and repressors has been characterized, how their status is translated to transcriptional activity is not understood. We used mass spectrometry to identify proteins that interact with GFP-tagged CLK (GFP-CLK) in fly heads at different times of day. Many expected and novel interacting proteins were detected, of which several interacted rhythmically and were potential regulators of protein levels, activity or transcriptional output. Genes encoding these proteins were tested to determine if they altered circadian behavior via RNAi knockdown in clock cells. The NIPPED-A protein, a scaffold for the SAGA and Tip60 histone modifying complexes, interacts with GFP-CLK as transcription is activated, and reducing Nipped-A expression lengthens circadian period. RNAi analysis of other SAGA complex components shows that the SAGA histone deubiquitination (DUB) module lengthened period similarly to Nipped-A RNAi knockdown and weakened rhythmicity, whereas reducing Tip60 HAT expression drastically weakened rhythmicity. These results suggest that CLK-CYC binds NIPPED-A early in the day to promote transcription through SAGA DUB and Tip60 HAT activity.


Asunto(s)
Proteínas CLOCK/fisiología , Relojes Circadianos/genética , Relojes Circadianos/fisiología , Ritmo Circadiano/genética , Ritmo Circadiano/fisiología , Proteínas de Drosophila/fisiología , Drosophila/genética , Drosophila/fisiología , Histona Acetiltransferasas/fisiología , Proteómica , Factores de Transcripción/fisiología , Animales , Proteínas de Drosophila/genética , Expresión Génica , Histona Acetiltransferasas/genética , Unión Proteica , Interferencia de ARN , Factores de Transcripción/genética , Transcripción Genética
19.
BMC Physiol ; 9: 22, 2009 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-19943954

RESUMEN

BACKGROUND: Seven-transmembrane receptors typically mediate olfactory signal transduction by coupling to G-proteins. Although insect odorant receptors have seven transmembrane domains like G-protein coupled receptors, they have an inverted membrane topology and function as ligand-gated cation channels. Consequently, the involvement of cyclic nucleotides and G proteins in insect odor reception is controversial. Since the heterotrimeric Goalpha subunit is expressed in Drosophila olfactory receptor neurons, we reasoned that Go acts together with insect odorant receptor cation channels to mediate odor-induced physiological responses. RESULTS: To test whether Go dependent signaling is involved in mediating olfactory responses in Drosophila, we analyzed electroantennogram and single-sensillum recording from flies that conditionally express pertussis toxin, a specific inhibitor of Go in Drosophila. Pertussis toxin expression in olfactory receptor neurons reversibly reduced the amplitude and hastened the termination of electroantennogram responses induced by ethyl acetate. The frequency of odor-induced spike firing from individual sensory neurons was also reduced by pertussis toxin. These results demonstrate that Go signaling is involved in increasing sensitivity of olfactory physiology in Drosophila. The effect of pertussis toxin was independent of odorant identity and intensity, indicating a generalized involvement of Go in olfactory reception. CONCLUSION: These results demonstrate that Go is required for maximal physiological responses to multiple odorants in Drosophila, and suggest that OR channel function and G-protein signaling are required for optimal physiological responses to odors.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Neuronas Receptoras Olfatorias/fisiología , Receptores Odorantes/fisiología , Olfato/fisiología , Análisis de Varianza , Animales , Animales Modificados Genéticamente , Drosophila melanogaster , Electrofisiología , Odorantes , Vías Olfatorias/efectos de los fármacos , Vías Olfatorias/fisiología , Neuronas Receptoras Olfatorias/efectos de los fármacos , Toxina del Pertussis/genética , Toxina del Pertussis/farmacología , Receptores Odorantes/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Olfato/efectos de los fármacos
20.
J Biol Rhythms ; 23(4): 283-95, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18663236

RESUMEN

In Drosophila, cryptochrome (cry) encodes a blue-light photoreceptor that mediates light input to circadian oscillators and sustains oscillator function in peripheral tissues. The levels of cry mRNA cycle with a peak at approximately ZT5, which is similar to the phase of Clock (Clk) mRNA cycling in Drosophila. To understand how cry spatial and circadian expression is regulated, a series of cry-Gal4 trans-genes containing different portions of cry upstream and intron 1 sequences were tested for spatial and circadian expression. In fly heads, cry upstream sequences drive constitutive expression in brain oscillator neurons, a novel group of nonoscillator cells in the optic lobe, and peripheral oscillator cells in eyes and antennae. In contrast, cry intron 1 drives rhythmic expression in eyes and antennae, but not brain oscillator neurons. These results demonstrate that intron 1 is sufficient for high-amplitude cry mRNA cycling, show that cry upstream sequences are sufficient for expression in brain oscillator neurons, and suggest that cry spatial and circadian expression are regulated by different elements.


Asunto(s)
Relojes Biológicos/fisiología , Ritmo Circadiano/fisiología , Proteínas de Drosophila/fisiología , Drosophila/fisiología , Proteínas del Ojo/fisiología , Regulación de la Expresión Génica , Receptores Acoplados a Proteínas G/fisiología , Animales , Animales Modificados Genéticamente , Criptocromos , Proteínas de Drosophila/genética , Proteínas del Ojo/genética , Técnica del Anticuerpo Fluorescente Indirecta , Intrones , Células Fotorreceptoras de Invertebrados/metabolismo , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G/genética , Transgenes , beta-Galactosidasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA