Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Neurosci ; 44(5)2024 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-38296648

RESUMEN

The molecular clock that generates daily rhythms of behavior and physiology consists of interlocked transcription-translation feedback loops. In Drosophila, the primary feedback loop involving the CLOCK-CYCLE transcriptional activators and the PERIOD-TIMELESS transcriptional repressors is interlocked with a secondary loop involving VRILLE (VRI) and PAR DOMAIN PROTEIN 1 (PDP1), a repressor and activator of Clock transcription, respectively. Whereas extensive studies have found numerous transcriptional, translational, and posttranslational modulators of the primary loop, relatively little is known about the secondary loop. In this study, using male and female flies as well as cultured cells, we demonstrate that TARANIS (TARA), a Drosophila homolog of the TRIP-Br/SERTAD family of transcriptional coregulators, functions with VRI and PDP1 to modulate the circadian period and rhythm strength. Knocking down tara reduces rhythm amplitude and can shorten the period length, while overexpressing TARA lengthens the circadian period. Additionally, tara mutants exhibit reduced rhythmicity and lower expression of the PDF neuropeptide. We find that TARA can form a physical complex with VRI and PDP1, enhancing their repressor and activator functions, respectively. The conserved SERTA domain of TARA is required to regulate the transcriptional activity of VRI and PDP1, and its deletion leads to reduced locomotor rhythmicity. Consistent with TARA's role in enhancing VRI and PDP1 activity, overexpressing tara has a similar effect on the circadian period and rhythm strength as simultaneously overexpressing vri and Pdp1 Together, our results suggest that TARA modulates circadian behavior by enhancing the transcriptional activity of VRI and PDP1.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Masculino , Femenino , Drosophila/fisiología , Retroalimentación , Proteínas de Drosophila/metabolismo , Ritmo Circadiano/genética , Proteínas CLOCK/genética , Drosophila melanogaster/metabolismo
2.
bioRxiv ; 2023 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-38076905

RESUMEN

The molecular clock that generates daily rhythms of behavior and physiology consists of interlocked transcription-translation feedback loops. In Drosophila, the primary feedback loop involving the CLOCK-CYCLE transcriptional activators and the PERIOD-TIMELESS transcriptional repressors is interlocked with a secondary loop involving VRILLE (VRI) and PAR DOMAIN PROTEIN 1 (PDP1), a repressor and activator of Clock transcription, respectively. Whereas extensive studies have found numerous transcriptional, translational, and post-translational modulators of the primary loop, relatively little is known about the secondary loop. In this study, using male and female flies as well as cultured cells, we demonstrate that TARANIS (TARA), a Drosophila homolog of the TRIP-Br/SERTAD family of transcriptional coregulators, functions with VRI and PDP1 to modulate the circadian period and rhythm strength. Knocking down tara reduces rhythm amplitude and can shorten the period length, while overexpressing TARA lengthens the circadian period. Additionally, tara mutants exhibit reduced rhythmicity and lower expression of the PDF neuropeptide. We find that TARA can form a physical complex with VRI and PDP1, enhancing their repressor and activator functions, respectively. The conserved SERTA domain of TARA is required to regulate the transcriptional activity of VRI and PDP1, and its deletion leads to reduced locomotor rhythmicity. Consistent with TARA's role in enhancing VRI and PDP1 activity, overexpressing tara has a similar effect on the circadian period and rhythm strength as simultaneously overexpressing vri and Pdp1. Together, our results suggest that TARA modulates circadian behavior by enhancing the transcriptional activity of VRI and PDP1.

3.
Curr Biol ; 33(13): 2702-2716.e3, 2023 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-37352854

RESUMEN

Sleep is essential, but animals may forgo sleep to engage in other critical behaviors, such as feeding and reproduction. Previous studies have shown that female flies exhibit decreased sleep after mating, but our understanding of the process is limited. Here, we report that postmating nighttime sleep loss is modulated by diet and sleep deprivation, demonstrating a complex interaction among sleep, reproduction, and diet. We also find that female-specific pC1 neurons and sleep-promoting dorsal fan-shaped body (dFB) neurons are required for postmating sleep plasticity. Activating pC1 neurons leads to sleep suppression on standard fly culture media but has little sleep effect on sucrose-only food. Published connectome data suggest indirect, inhibitory connections among pC1 subtypes. Using calcium imaging, we show that activating the pC1e subtype inhibits dFB neurons. We propose that pC1 and dFB neurons integrate the mating status, food context, and sleep drive to modulate postmating sleep plasticity.


Asunto(s)
Proteínas de Drosophila , Trastornos del Inicio y del Mantenimiento del Sueño , Animales , Femenino , Drosophila/fisiología , Proteínas de Drosophila/fisiología , Sueño/fisiología , Privación de Sueño , Drosophila melanogaster/fisiología
5.
FEBS J ; 290(4): 931-950, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-34908236

RESUMEN

The two-process model of sleep regulation posits two main processes regulating sleep: the circadian process controlled by the circadian clock and the homeostatic process that depends on the history of sleep and wakefulness. The model has provided a dominant conceptual framework for sleep research since its publication ~ 40 years ago. The time of day and prior wake time are the primary factors affecting the circadian and homeostatic processes, respectively. However, it is critical to consider other factors influencing sleep. Since sleep is incompatible with other behaviors, it is affected by the need for essential behaviors such as eating, foraging, mating, caring for offspring, and avoiding predators. Sleep is also affected by sensory inputs, sickness, increased need for memory consolidation after learning, and other factors. Here, we review multiple factors influencing sleep and discuss recent insights into the mechanisms balancing competing needs.


Asunto(s)
Relojes Circadianos , Sueño , Sueño/fisiología , Ritmo Circadiano , Vigilia/fisiología , Homeostasis
6.
Cell Rep ; 33(9): 108462, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33264620

RESUMEN

People tend to fall asleep when gently rocked or vibrated. Experimental studies have shown that rocking promotes sleep in humans and mice. However, the mechanisms underlying the phenomenon are not well understood. A habituation model proposes that habituation, a form of non-associative learning, mediates sleep induction by monotonous stimulation. Here, we show that gentle vibration promotes sleep in Drosophila in part through habituation. Vibration-induced sleep (VIS) leads to increased homeostatic sleep credit and reduced arousability, and can be suppressed by heightened arousal or reduced GABA signaling. Multiple mechanosensory organs mediate VIS, and the magnitude of VIS depends on vibration frequency and genetic background. Sleep induction improves over successive blocks of vibration. Furthermore, training with continuous vibration does not generalize to intermittent vibration, demonstrating stimulus specificity, a characteristic of habituation. Our findings suggest that habituation plays a significant role in sleep induction by vibration.


Asunto(s)
Habituación Psicofisiológica/fisiología , Fármacos Inductores del Sueño/uso terapéutico , Sueño/fisiología , Animales , Drosophila , Fármacos Inductores del Sueño/farmacología
7.
Elife ; 92020 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-33084567

RESUMEN

Sleep is essential but incompatible with other behaviors, and thus sleep drive competes with other motivations. We previously showed Drosophila males balance sleep and courtship via octopaminergic neurons that act upstream of courtship-regulating P1 neurons (Machado et al., 2017). Here, we show nutrition modulates the sleep-courtship balance and identify sleep-regulatory neurons downstream of P1 neurons. Yeast-deprived males exhibited attenuated female-induced nighttime sleep loss yet normal daytime courtship, which suggests male flies consider nutritional status in deciding whether the potential benefit of pursuing female partners outweighs the cost of losing sleep. Trans-synaptic tracing and calcium imaging identified dopaminergic neurons projecting to the protocerebral bridge (DA-PB) as postsynaptic partners of P1 neurons. Activation of DA-PB neurons led to reduced sleep in normally fed but not yeast-deprived males. Additional PB-projecting neurons regulated male sleep, suggesting several groups of PB-projecting neurons act downstream of P1 neurons to mediate nutritional modulation of the sleep-courtship balance.


Asunto(s)
Cortejo , Drosophila melanogaster/fisiología , Estado Nutricional/fisiología , Sueño/fisiología , Animales , Drosophila melanogaster/metabolismo , Femenino , Privación de Alimentos/fisiología , Masculino , Neuronas/fisiología
8.
iScience ; 23(2): 100845, 2020 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-32058958

RESUMEN

Circadian output genes act downstream of the clock to promote rhythmic changes in behavior and physiology, yet their molecular and cellular functions are not well understood. Here we characterize an interaction between regulators of circadian entrainment, output, and synaptic development in Drosophila that influences clock-driven anticipatory increases in morning and evening activity. We previously showed the JETLAG (JET) E3 ubiquitin ligase resets the clock upon light exposure, whereas the PDZ protein DYSCHRONIC (DYSC) regulates circadian locomotor output and synaptic development. Surprisingly, we find that JET and DYSC antagonistically regulate synaptic development at the larval neuromuscular junction, and reduced JET activity rescues arrhythmicity of dysc mutants. Consistent with our prior finding that DYSC regulates SLOWPOKE (SLO) potassium channel expression, jet mutations also rescue circadian and synaptic phenotypes in slo mutants. Collectively, our data suggest that JET, DYSC, and SLO promote circadian output in part by regulating synaptic morphology.

9.
Elife ; 62017 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-28510528

RESUMEN

Molecular and circuit mechanisms for balancing competing drives are not well understood. While circadian and homeostatic mechanisms generally ensure sufficient sleep at night, other pressing needs can overcome sleep drive. Here, we demonstrate that the balance between sleep and sex drives determines whether male flies sleep or court, and identify a subset of octopaminergic neurons (MS1) that regulate sleep specifically in males. When MS1 neurons are activated, isolated males sleep less, and when MS1 neurons are silenced, the normal male sleep suppression in female presence is attenuated and mating behavior is impaired. MS1 neurons do not express the sexually dimorphic FRUITLESS (FRU) transcription factor, but form male-specific contacts with FRU-expressing neurons; calcium imaging experiments reveal bidirectional functional connectivity between MS1 and FRU neurons. We propose octopaminergic MS1 neurons interact with the FRU network to mediate sleep suppression by male sex drive.


Asunto(s)
Dípteros/fisiología , Neuronas/fisiología , Octopamina/metabolismo , Conducta Sexual Animal , Sueño , Animales , Masculino
10.
Sci Rep ; 7: 40304, 2017 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-28084307

RESUMEN

Sleep is a highly conserved and essential behaviour in many species, including the fruit fly Drosophila melanogaster. In the wild, sensory signalling encoding environmental information must be integrated with sleep drive to ensure that sleep is not initiated during detrimental conditions. However, the molecular and circuit mechanisms by which sleep timing is modulated by the environment are unclear. Here we introduce a novel behavioural paradigm to study this issue. We show that in male fruit flies, onset of the daytime siesta is delayed by ambient temperatures above 29 °C. We term this effect Prolonged Morning Wakefulness (PMW). We show that signalling through the TrpA1 thermo-sensor is required for PMW, and that TrpA1 specifically impacts siesta onset, but not night sleep onset, in response to elevated temperatures. We identify two critical TrpA1-expressing circuits and show that both contact DN1p clock neurons, the output of which is also required for PMW. Finally, we identify the circadian blue-light photoreceptor CRYPTOCHROME as a molecular regulator of PMW, and propose a model in which the Drosophila nervous system integrates information encoding temperature, light, and time to dynamically control when sleep is initiated. Our results provide a platform to investigate how environmental inputs co-ordinately regulate sleep plasticity.


Asunto(s)
Ritmo Circadiano/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Sueño/genética , Canal Catiónico TRPA1/genética , Animales , Drosophila melanogaster/fisiología , Humanos , Canales Iónicos , Luz , Modelos Animales , Actividad Motora/genética , Neuronas/metabolismo , Neuronas/fisiología , Sueño/fisiología , Temperatura , Vigilia/genética , Vigilia/fisiología
11.
Curr Biol ; 25(13): 1717-26, 2015 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-26096977

RESUMEN

Sleep is an essential and conserved behavior whose regulation at the molecular and anatomical level remains to be elucidated. Here, we identify TARANIS (TARA), a Drosophila homolog of the Trip-Br (SERTAD) family of transcriptional coregulators, as a molecule that is required for normal sleep patterns. Through a forward-genetic screen, we isolated tara as a novel sleep gene associated with a marked reduction in sleep amount. Targeted knockdown of tara suggests that it functions in cholinergic neurons to promote sleep. tara encodes a conserved cell-cycle protein that contains a Cyclin A (CycA)-binding homology domain. TARA regulates CycA protein levels and genetically and physically interacts with CycA to promote sleep. Furthermore, decreased levels of Cyclin-dependent kinase 1 (Cdk1), a kinase partner of CycA, rescue the short-sleeping phenotype of tara and CycA mutants, while increased Cdk1 activity mimics the tara and CycA phenotypes, suggesting that Cdk1 mediates the role of TARA and CycA in sleep regulation. Finally, we describe a novel wake-promoting role for a cluster of ∼14 CycA-expressing neurons in the pars lateralis (PL), previously proposed to be analogous to the mammalian hypothalamus. We propose that TARANIS controls sleep amount by regulating CycA protein levels and inhibiting Cdk1 activity in a novel arousal center.


Asunto(s)
Nivel de Alerta/fisiología , Proteína Quinasa CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ciclina A/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/fisiología , Sueño/fisiología , Animales , Western Blotting , Proteínas de Ciclo Celular/genética , Proteínas de Drosophila/genética , Técnicas de Silenciamiento del Gen , Neuronas/fisiología , Porción Reticular de la Sustancia Negra/citología , Porción Reticular de la Sustancia Negra/fisiología , Interferencia de ARN
12.
Fly (Austin) ; 9(4): 165-72, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26925838

RESUMEN

Sleep is essential for health and cognition, but the molecular and neural mechanisms of sleep regulation are not well understood. We recently reported the identification of TARANIS (TARA) as a sleep-promoting factor that acts in a previously unknown arousal center in Drosophila. tara mutants exhibit a dose-dependent reduction in sleep amount of up to ∼60%. TARA and its mammalian homologs, the Trip-Br (Transcriptional Regulators Interacting with PHD zinc fingers and/or Bromodomains) family of proteins, are primarily known as transcriptional coregulators involved in cell cycle progression, and contain a conserved Cyclin-A (CycA) binding homology domain. We found that tara and CycA synergistically promote sleep, and CycA levels are reduced in tara mutants. Additional data demonstrated that Cyclin-dependent kinase 1 (Cdk1) antagonizes tara and CycA to promote wakefulness. Moreover, we identified a subset of CycA expressing neurons in the pars lateralis, a brain region proposed to be analogous to the mammalian hypothalamus, as an arousal center. In this Extra View article, we report further characterization of tara mutants and provide an extended discussion of our findings and future directions within the framework of a working model, in which a network of cell cycle genes, tara, CycA, and Cdk1, interact in an arousal center to regulate sleep.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ciclina A/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Animales , Conducta Animal , Relojes Biológicos , Femenino , Masculino , Neuronas/metabolismo , Sueño
13.
Development ; 141(23): 4548-57, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25359729

RESUMEN

Synaptic scaffold proteins control the localization of ion channels and receptors, and facilitate molecular associations between signaling components that modulate synaptic transmission and plasticity. Here, we define novel roles for a recently described scaffold protein, Dsychronic (DYSC), at the Drosophila larval neuromuscular junction. DYSC is the Drosophila homolog of whirlin/DFNB31, a PDZ domain protein linked to Usher syndrome, the most common form of human deaf-blindness. We show that DYSC is expressed presynaptically and is often localized adjacent to the active zone, the site of neurotransmitter release. Loss of DYSC results in marked alterations in synaptic morphology and cytoskeletal organization. Moreover, active zones are frequently enlarged and misshapen in dysc mutants. Electrophysiological analyses further demonstrate that dysc mutants exhibit substantial increases in both evoked and spontaneous synaptic transmission. We have previously shown that DYSC binds to and regulates the expression of the Slowpoke (SLO) BK potassium channel. Consistent with this, slo mutant larvae exhibit similar alterations in synapse morphology, active zone size and neurotransmission, and simultaneous loss of dysc and slo does not enhance these phenotypes, suggesting that dysc and slo act in a common genetic pathway to modulate synaptic development and output. Our data expand our understanding of the neuronal functions of DYSC and uncover non-canonical roles for the SLO potassium channel at Drosophila synapses.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/crecimiento & desarrollo , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Proteínas de la Membrana/metabolismo , Unión Neuromuscular/crecimiento & desarrollo , Sinapsis/fisiología , Animales , Inmunohistoquímica , Larva/crecimiento & desarrollo , Potenciales de la Membrana , Microscopía Confocal , Dominios PDZ/genética , Técnicas de Placa-Clamp , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sinapsis/metabolismo
14.
Neuron ; 82(1): 151-66, 2014 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-24631345

RESUMEN

How the circadian clock regulates the timing of sleep is poorly understood. Here, we identify a Drosophila mutant, wide awake (wake), that exhibits a marked delay in sleep onset at dusk. Loss of WAKE in a set of arousal-promoting clock neurons, the large ventrolateral neurons (l-LNvs), impairs sleep onset. WAKE levels cycle, peaking near dusk, and the expression of WAKE in l-LNvs is Clock dependent. Strikingly, Clock and cycle mutants also exhibit a profound delay in sleep onset, which can be rescued by restoring WAKE expression in LNvs. WAKE interacts with the GABAA receptor Resistant to Dieldrin (RDL), upregulating its levels and promoting its localization to the plasma membrane. In wake mutant l-LNvs, GABA sensitivity is decreased and excitability is increased at dusk. We propose that WAKE acts as a clock output molecule specifically for sleep, inhibiting LNvs at dusk to promote the transition from wake to sleep.


Asunto(s)
Ritmo Circadiano/fisiología , Neuronas/fisiología , Sueño/fisiología , Factores de Transcripción ARNTL/genética , Animales , Animales Modificados Genéticamente , Encéfalo/citología , Proteínas CLOCK/genética , Ritmo Circadiano/efectos de los fármacos , Péptidos y Proteínas de Señalización del Ritmo Circadiano/genética , Péptidos y Proteínas de Señalización del Ritmo Circadiano/metabolismo , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas Fluorescentes Verdes/genética , Antagonistas de Hormonas/farmacología , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Mifepristona/farmacología , Mutación/genética , Neuronas/efectos de los fármacos , Tiempo de Reacción/genética , Sueño/efectos de los fármacos , Sueño/genética , Factores de Tiempo
15.
J Neurosci ; 33(42): 16767-77, 2013 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-24133277

RESUMEN

SLOB (SLOWPOKE-binding protein) modulates the Drosophila SLOWPOKE calcium-activated potassium channel. We have shown previously that SLOB deletion or RNAi knockdown decreases excitability of neurosecretory pars intercerebralis (PI) neurons in the adult Drosophila brain. In contrast, we found that SLOB deletion/knockdown enhances neurotransmitter release from motor neurons at the fly larval neuromuscular junction, suggesting an increase in excitability. Because two prominent SLOB isoforms, SLOB57 and SLOB71, modulate SLOWPOKE channels in opposite directions in vitro, we investigated whether divergent expression patterns of these two isoforms might underlie the differential modulation of excitability in PI and motor neurons. By performing detailed in vitro and in vivo analysis, we found strikingly different modes of regulatory control by the slob57 and slob71 promoters. The slob71, but not slob57, promoter contains binding sites for the Hunchback and Mirror transcriptional repressors. Furthermore, several core promoter elements that are absent in the slob57 promoter coordinately drive robust expression of a luciferase vector by the slob71 promoter in vitro. In addition, we visualized the expression patterns of the slob57 and slob71 promoters in vivo and found clear spatiotemporal differences in promoter activity. SLOB57 is expressed prominently in adult PI neurons, whereas larval motor neurons exclusively express SLOB71. In contrast, at the larval neuromuscular junction, SLOB57 expression appears to be restricted mainly to a subset of glial cells. Our results illustrate how the use of alternative transcriptional start sites within an ion channel modulator locus coupled with functionally relevant alternative splicing can be used to fine-tune neuronal excitability in a cell-specific manner.


Asunto(s)
Encéfalo/metabolismo , Proteínas de Drosophila/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Canales de Potasio/metabolismo , Isoformas de Proteínas/metabolismo , Animales , Animales Modificados Genéticamente , Drosophila , Proteínas de Drosophila/genética , Larva/genética , Larva/metabolismo , Unión Neuromuscular/genética , Unión Neuromuscular/metabolismo , Canales de Potasio/genética , Regiones Promotoras Genéticas , Isoformas de Proteínas/genética , Transmisión Sináptica/fisiología
16.
PLoS Genet ; 9(9): e1003605, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24039590

RESUMEN

A robust, bistable switch regulates the fluctuations between wakefulness and natural sleep as well as those between wakefulness and anesthetic-induced unresponsiveness. We previously provided experimental evidence for the existence of a behavioral barrier to transitions between these states of arousal, which we call neural inertia. Here we show that neural inertia is controlled by processes that contribute to sleep homeostasis and requires four genes involved in electrical excitability: Sh, sss, na and unc79. Although loss of function mutations in these genes can increase or decrease sensitivity to anesthesia induction, surprisingly, they all collapse neural inertia. These effects are genetically selective: neural inertia is not perturbed by loss-of-function mutations in all genes required for the sleep/wake cycle. These effects are also anatomically selective: sss acts in different neurons to influence arousal-promoting and arousal-suppressing processes underlying neural inertia. Supporting the idea that anesthesia and sleep share some, but not all, genetic and anatomical arousal-regulating pathways, we demonstrate that increasing homeostatic sleep drive widens the neural inertial barrier. We propose that processes selectively contributing to sleep homeostasis and neural inertia may be impaired in pathophysiological conditions such as coma and persistent vegetative states.


Asunto(s)
Proteínas de Drosophila/genética , Canales Iónicos/genética , Proteínas de la Membrana/genética , Canales de Potasio de la Superfamilia Shaker/genética , Vigilia/genética , Animales , Nivel de Alerta/genética , Nivel de Alerta/fisiología , Encéfalo/metabolismo , Encéfalo/fisiología , Ritmo Circadiano/fisiología , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Homeostasis/fisiología , Humanos , Mutación , Neuronas/metabolismo , Sueño/genética
17.
PLoS Genet ; 8(4): e1002671, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22532808

RESUMEN

Many aspects of behavior and physiology are under circadian control. In Drosophila, the molecular clock that regulates rhythmic patterns of behavior has been extensively characterized. In contrast, genetic loci involved in linking the clock to alterations in motor activity have remained elusive. In a forward-genetic screen, we uncovered a new component of the circadian output pathway, which we have termed dyschronic (dysc). dysc mutants exhibit arrhythmic locomotor behavior, yet their eclosion rhythms are normal and clock protein cycling remains intact. Intriguingly, dysc is the closest Drosophila homolog of whirlin, a gene linked to type II Usher syndrome, the leading cause of deaf-blindness in humans. Whirlin and other Usher proteins are expressed in the mammalian central nervous system, yet their function in the CNS has not been investigated. We show that DYSC is expressed in major neuronal tracts and regulates expression of the calcium-activated potassium channel SLOWPOKE (SLO), an ion channel also required in the circadian output pathway. SLO and DYSC are co-localized in the brain and control each other's expression post-transcriptionally. Co-immunoprecipitation experiments demonstrate they form a complex, suggesting they regulate each other through protein-protein interaction. Furthermore, electrophysiological recordings of neurons in the adult brain show that SLO-dependent currents are greatly reduced in dysc mutants. Our work identifies a Drosophila homolog of a deaf-blindness gene as a new component of the circadian output pathway and an important regulator of ion channel expression, and suggests novel roles for Usher proteins in the mammalian nervous system.


Asunto(s)
Encéfalo , Ritmo Circadiano/genética , Drosophila melanogaster , Neuronas , Animales , Conducta Animal , Encéfalo/metabolismo , Trastornos Sordoceguera/genética , Trastornos Sordoceguera/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Regulación de la Expresión Génica , Humanos , Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Proteínas de la Membrana , Actividad Motora/genética , Neuronas/metabolismo , Neuronas/fisiología , Mapas de Interacción de Proteínas/genética
18.
J Neurosci ; 31(27): 9982-90, 2011 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-21734289

RESUMEN

The molecular circadian clock consists of a feedback loop in which canonical clock proteins negatively regulate transcription of their own genes. Timed nuclear entry of these proteins is critical, but regulation of this event is poorly understood. In Drosophila melanogaster, the idea that nuclear entry of PERIOD (PER) is controlled by its partner protein TIMELESS (TIM) has been challenged by several studies. We identify here a novel mutation in the tim gene that eliminates behavioral rhythms while allowing robust expression of TIM and PER. Mutant TIM can bind to and stabilize PER. However, neither protein is expressed cyclically, and phosphorylation of both is reduced. In addition, TIM and PER are localized in the cytoplasm at all times of day, and mutant TIM attenuates transcriptional feedback by PER in cultured cells, suggesting that it holds PER in the cytoplasm. In fact, much of the reduced phosphorylation of PER in the new tim mutant appears to result from the cytoplasmic localization of PER. Interestingly, mutating a threonine near the original mutation produces similar phenotypes, raising the possibility that defective phosphorylation is the basis of TIM dysfunction in the novel tim mutant. We also show that a stable form of PER is cytoplasmic in tim-null flies. These studies establish an essential role of TIM in the timed nuclear entry of PER.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Mutación/genética , Proteínas Circadianas Period/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Animales , Animales Modificados Genéticamente , Ritmo Circadiano/genética , Drosophila , Regulación de la Expresión Génica/genética , Inmunoprecipitación , Proteínas Luminiscentes/genética , Proteínas Circadianas Period/genética , Procesamiento Proteico-Postraduccional/genética , Fracciones Subcelulares/metabolismo
19.
Nat Neurosci ; 13(1): 69-75, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20010822

RESUMEN

Sleep is a whole-organism phenomenon accompanied by global changes in neural activity. We previously identified SLEEPLESS (SSS) as a glycosylphosphatidyl inositol-anchored protein required for sleep in Drosophila. Here we found that SSS is critical for regulating the sleep-modulating potassium channel Shaker. SSS and Shaker shared similar expression patterns in the brain and specifically affected each other's expression levels. sleepless (sss) loss-of-function mutants exhibited altered Shaker localization, reduced Shaker current density and slower Shaker current kinetics. Transgenic expression of sss in sss mutants rescued defects in Shaker expression and activity cell-autonomously and suggested that SSS functions in wake-promoting, cholinergic neurons. In heterologous cells, SSS accelerated the kinetics of Shaker currents and was co-immunoprecipitated with Shaker, suggesting that SSS modulates Shaker activity via a direct interaction. SSS is predicted to belong to the Ly-6/neurotoxin superfamily, suggesting a mechanism for regulation of neuronal excitability by endogenous toxin-like molecules.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiología , Regulación de la Expresión Génica/genética , Proteínas de la Membrana/fisiología , Canales de Potasio de la Superfamilia Shaker/metabolismo , Animales , Animales Modificados Genéticamente , Conducta Animal/fisiología , Encéfalo/citología , Encéfalo/metabolismo , Línea Celular Transformada , Drosophila , Proteínas de Drosophila/genética , Femenino , Proteínas Fluorescentes Verdes/genética , Humanos , Inmunoprecipitación/métodos , Técnicas In Vitro , Larva , Mutación/genética , Unión Neuromuscular/genética , Unión Neuromuscular/fisiología , Células Receptoras Sensoriales/metabolismo , Sueño/genética , Vigilia/genética
20.
J Neurosci ; 29(35): 10920-7, 2009 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-19726650

RESUMEN

The Drosophila PAR domain protein 1 (Pdp1) gene encodes a transcription factor with multiple functions. One isoform, PDP1epsilon, was proposed to be an essential activator of the core clock gene, Clock (Clk). However, a central clock function for PDP1epsilon was recently disputed, and genetic analysis has been difficult due to developmental lethality of Pdp1-null mutants. Here we report the discovery of a mutation that specifically disrupts the Pdp1epsilon isoform. Homozygous Pdp1epsilon mutants are viable and exhibit arrhythmic circadian behavior in constant darkness and also in the presence of light:dark cycles. Importantly, the mutants show diminished expression of CLK and PERIOD (PER) in the central clock cells. In addition, expression of PDF (pigment-dispersing factor) is reduced in a subset of the central clock cells. Loss of Pdp1epsilon also alters the phosphorylation status of the CLK protein and disrupts cyclic expression of a per-luciferase reporter in peripheral clocks under free-running conditions. Transgenic expression of PDP1epsilon in clock neurons of Pdp1epsilon mutants can restore rhythmic circadian behavior. However, transgenic expression of CLK in these mutants rescues the expression of PER in the central clock, but fails to restore behavioral rhythms, suggesting that PDP1epsilon has effects outside the core molecular clock. Together, these data support a model in which PDP1epsilon functions in the central circadian oscillator as well as in the output pathway.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/fisiología , Ritmo Circadiano/genética , Proteínas de Drosophila/fisiología , Animales , Animales Modificados Genéticamente , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Ritmo Circadiano/fisiología , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Masculino , Actividad Motora/genética , Actividad Motora/fisiología , Mutación/genética , Mutación/fisiología , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA