Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Cell ; 154(4): 904-13, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23932121

RESUMEN

Nervous systems process information by integrating the electrical activity of neurons in complex networks. This motivates the long-standing interest in using optical methods to simultaneously monitor the membrane potential of multiple genetically targeted neurons via expression of genetically encoded fluorescent voltage indicators (GEVIs) in intact neural circuits. No currently available GEVIs have demonstrated robust signals in intact brain tissue that enable reliable recording of individual electrical events simultaneously in multiple neurons. Here, we show that the recently developed "ArcLight" GEVI robustly reports both subthreshold events and action potentials in genetically targeted neurons in the intact Drosophila fruit fly brain and reveals electrical signals in neurite branches. In the same way that genetically encoded fluorescent sensors have revolutionized the study of intracellular Ca(2+) signals, ArcLight now enables optical measurement in intact neural circuits of membrane potential, the key cellular parameter that underlies neuronal information processing.


Asunto(s)
Drosophila melanogaster/fisiología , Fenómenos Electrofisiológicos , Red Nerviosa , Optogenética/métodos , Animales , Encéfalo/fisiología , Relojes Circadianos , Drosophila melanogaster/citología , Proteínas Fluorescentes Verdes/genética , Neuronas/fisiología
2.
J Neurogenet ; 32(3): 209-220, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30175644

RESUMEN

Presynaptic Ca2+ appears to play multiple roles in synaptic development and physiology. We examined the effect of buffering presynaptic Ca2+ by expressing parvalbumin (PV) in Drosophila neurons, which do not normally express PV. The studies were performed on the identified Ib terminal that innervates muscle fiber 5. The volume-averaged, residual Ca2+ resulting from single action potentials (APs) and AP trains was measured using the fluorescent Ca2+ indicator, OGB-1. PV reduced the amplitude and decay time constant (τ) for single-AP Ca2+ transients. For AP trains, there was a reduction in the rate of rise and decay of [Ca2+]i but the plateau [Ca2+]i was not affected. Electrophysiological recordings from muscle fiber 5 showed a reduction in paired-pulse facilitation, particularly the F1 component; this was likely due to the reduction in residual Ca2+. These synapses also showed reduced synaptic enhancement during AP trains, presumably due to less buildup of synaptic facilitation. The transmitter release for single APs was increased for the PV-expressing terminals and this may have been a homeostatic response to the decrease in facilitation. Confocal microscopy was used to examine the structure of the motor terminals and PV expression resulted in smaller motor terminals with fewer synaptic boutons and active zones. This result supports earlier proposals that increased AP activity promotes motor terminal growth through increases in presynaptic [Ca2+]i.


Asunto(s)
Potenciales de Acción/fisiología , Señalización del Calcio/fisiología , Neurogénesis/fisiología , Unión Neuromuscular/fisiología , Parvalbúminas/metabolismo , Animales , Calcio/metabolismo , Drosophila , Larva
3.
Proc Natl Acad Sci U S A ; 112(2): E220-9, 2015 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-25550513

RESUMEN

Complex animal behaviors are built from dynamical relationships between sensory inputs, neuronal activity, and motor outputs in patterns with strategic value. Connecting these patterns illuminates how nervous systems compute behavior. Here, we study Drosophila larva navigation up temperature gradients toward preferred temperatures (positive thermotaxis). By tracking the movements of animals responding to fixed spatial temperature gradients or random temperature fluctuations, we calculate the sensitivity and dynamics of the conversion of thermosensory inputs into motor responses. We discover three thermosensory neurons in each dorsal organ ganglion (DOG) that are required for positive thermotaxis. Random optogenetic stimulation of the DOG thermosensory neurons evokes behavioral patterns that mimic the response to temperature variations. In vivo calcium and voltage imaging reveals that the DOG thermosensory neurons exhibit activity patterns with sensitivity and dynamics matched to the behavioral response. Temporal processing of temperature variations carried out by the DOG thermosensory neurons emerges in distinct motor responses during thermotaxis.


Asunto(s)
Conducta Animal/fisiología , Drosophila melanogaster/fisiología , Termorreceptores/fisiología , Animales , Animales Modificados Genéticamente , Señalización del Calcio , Ganglios/fisiología , Larva/fisiología , Locomoción/fisiología , Optogenética , Sensación Térmica/fisiología
4.
Crit Rev Biochem Mol Biol ; 50(1): 18-30, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25410535

RESUMEN

Members of the class B1 family of G-protein coupled receptors (GPCRs) whose ligands are neuropeptides have been implicated in regulation of circadian rhythms and sleep in diverse metazoan clades. This review discusses the cellular and molecular mechanisms by which class B1 GPCRs, especially the mammalian VPAC2 receptor and its functional homologue PDFR in Drosophila and C. elegans, regulate arousal and daily rhythms of sleep and wake. There are remarkable parallels in the cellular and molecular roles played by class B1 intercellular signaling pathways in coordinating arousal and circadian timekeeping across multiple cells and tissues in these very different genetic model organisms.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Ritmo Circadiano/fisiología , Proteínas de Drosophila/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sueño/fisiología , Animales , Caenorhabditis elegans/fisiología , Drosophila/fisiología , Humanos , Filogenia , Receptores Acoplados a Proteínas G/genética
5.
J Neurosci ; 36(46): 11739-11754, 2016 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-27852781

RESUMEN

Body temperature exhibits rhythmic fluctuations over a 24 h period (Refinetti and Menaker, 1992) and decreases during the night, which is associated with sleep initiation (Gilbert et al., 2004; Kräuchi, 2007a,b). However, the underlying mechanism of this temperature decrease is largely unknown. We have previously shown that Drosophila exhibit a daily temperature preference rhythm (TPR), in which their preferred temperatures increase during the daytime and then decrease at the transition from day to night (night-onset) (Kaneko et al., 2012). Because Drosophila are small ectotherms, their body temperature is very close to that of the ambient temperature (Stevenson, 1985), suggesting that their TPR generates their body temperature rhythm. Here, we demonstrate that the neuropeptide diuretic hormone 31 (DH31) and pigment-dispersing factor receptor (PDFR) contribute to regulate the preferred temperature decrease at night-onset. We show that PDFR and tethered-DH31 expression in dorsal neurons 2 (DN2s) restore the preferred temperature decrease at night-onset, suggesting that DH31 acts on PDFR in DN2s. Notably, we previously showed that the molecular clock in DN2s is important for TPR. Although PDF (another ligand of PDFR) is a critical factor for locomotor activity rhythms, Pdf mutants exhibit normal preferred temperature decreases at night-onset. This suggests that DH31-PDFR signaling specifically regulates a preferred temperature decrease at night-onset. Thus, we propose that night-onset TPR and locomotor activity rhythms are differentially controlled not only by clock neurons but also by neuropeptide signaling in the brain. SIGNIFICANCE STATEMENT: Body temperature rhythm (BTR) is fundamental for the maintenance of functions essential for homeostasis, such as generating metabolic energy and sleep. One major unsolved question is how body temperature decreases dramatically during the night. Previously, we demonstrated that a BTR-like mechanism, referred to as temperature preference rhythm (TPR), exists in Drosophila Here, we demonstrate that the diuretic hormone 31 (DH31) neuropeptide and pigment-dispersing factor receptor (PDFR) regulate preferred temperature decreases at night-onset via dorsal neurons 2. This is the first in vivo evidence that DH31 could function as a ligand of PDFR. Although both DH31 and PDF are ligands of PDFR, we show that DH31 regulates night-onset TPR, but PDF does not, suggesting that night-onset TPR and locomotor activity rhythms are controlled by different neuropeptides via different clock cells.


Asunto(s)
Regulación de la Temperatura Corporal/fisiología , Ritmo Circadiano/fisiología , Proteínas de Drosophila/metabolismo , Drosophila/fisiología , Hormonas de Insectos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células Receptoras Sensoriales/fisiología , Animales , Relojes Biológicos/fisiología , Regulación de la Expresión Génica
6.
Mol Pharmacol ; 88(3): 534-5, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26162863

RESUMEN

In August 2014, an international group of researchers gathered for 5 days at the Lorentz Center in Leiden, The Netherlands, to explore the technical and conceptual issues associated with the analysis of G protein-coupled receptor functions utilizing information from crystal structure models to the use of model organisms. This collection of review articles evolved from the 5-day meeting, with brief presentations and structured discussion periods that were designed to identify key questions remaining in understanding G protein-coupled receptor function and to propose novel strategies by integrating scientific disciplines to guide future research.


Asunto(s)
Congresos como Asunto , Receptores Acoplados a Proteínas G/metabolismo , Animales , Humanos , Receptores Acoplados a Proteínas G/química
7.
Genome Res ; 22(7): 1266-81, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22472103

RESUMEN

Eukaryotic circadian clocks include transcriptional/translational feedback loops that drive 24-h rhythms of transcription. These transcriptional rhythms underlie oscillations of protein abundance, thereby mediating circadian rhythms of behavior, physiology, and metabolism. Numerous studies over the last decade have used microarrays to profile circadian transcriptional rhythms in various organisms and tissues. Here we use RNA sequencing (RNA-seq) to profile the circadian transcriptome of Drosophila melanogaster brain from wild-type and period-null clock-defective animals. We identify several hundred transcripts whose abundance oscillates with 24-h periods in either constant darkness or 12 h light/dark diurnal cycles, including several noncoding RNAs (ncRNAs) that were not identified in previous microarray studies. Of particular interest are U snoRNA host genes (Uhgs), a family of diurnal cycling noncoding RNAs that encode the precursors of more than 50 box-C/D small nucleolar RNAs, key regulators of ribosomal biogenesis. Transcriptional profiling at the level of individual exons reveals alternative splice isoforms for many genes whose relative abundances are regulated by either period or circadian time, although the effect of circadian time is muted in comparison to that of period. Interestingly, period loss of function significantly alters the frequency of RNA editing at several editing sites, suggesting an unexpected link between a key circadian gene and RNA editing. We also identify tens of thousands of novel splicing events beyond those previously annotated by the modENCODE Consortium, including several that affect key circadian genes. These studies demonstrate extensive circadian control of ncRNA expression, reveal the extent of clock control of alternative splicing and RNA editing, and provide a novel, genome-wide map of splicing in Drosophila brain.


Asunto(s)
Empalme Alternativo , Encéfalo/fisiología , Relojes Circadianos , Drosophila melanogaster/genética , Perfilación de la Expresión Génica/métodos , Animales , Secuencia de Bases , Encéfalo/citología , Ritmo Circadiano , Drosophila melanogaster/metabolismo , Exones , Anotación de Secuencia Molecular , Fotoperiodo , Edición de ARN , Isoformas de ARN/genética , Isoformas de ARN/metabolismo , Sitios de Empalme de ARN , ARN no Traducido/genética , ARN no Traducido/metabolismo , Alineación de Secuencia , Análisis de Secuencia de ARN/métodos , Transcripción Genética , Transcriptoma
8.
Physiology (Bethesda) ; 28(3): 164-71, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23636262

RESUMEN

Detection of secreted signaling molecules by cognate cell surface receptors is a major intercellular communication pathway in cellular circuits that control biological processes. Understanding the biological significance of these connections would allow us to understand how cellular circuits operate as a whole. Membrane-tethered ligands are recombinant transgenes with structural modules that allow them to act on cell-surface receptors and ion channel subtypes with pharmacological specificity in a cell-autonomous manner. Membrane-tethered ligands have been successful in the specific manipulation of ion channels as well as G-protein-coupled receptors, and, in combination with cell-specific promoters, such manipulations have been restricted to genetically defined subpopulations within cellular circuits in vivo to induce specific phenotypes controlled by those circuits. These studies establish the membrane-tethering approach as a generally applicable method for dissecting neural and physiological circuits.


Asunto(s)
Canales Iónicos/metabolismo , Receptores de Superficie Celular/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Humanos , Ligandos , Neurotoxinas/genética , Neurotoxinas/metabolismo , Unión Proteica , Receptores Nicotínicos/metabolismo , Proteínas Recombinantes de Fusión/genética , Transgenes
9.
J Neurosci ; 31(22): 8181-93, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21632940

RESUMEN

Neuropeptide PDF (pigment-dispersing factor)-secreting large ventrolateral neurons (lLN(v)s) in the Drosophila brain regulate daily patterns of rest and arousal. These bilateral wake-promoting neurons are light responsive and integrate information from the circadian system, sleep circuits, and light environment. To begin to dissect the synaptic circuitry of the circadian neural network, we performed simultaneous dual whole-cell patch-clamp recordings of pairs of lLN(v)s. Both ipsilateral and contralateral pairs of lLN(v)s exhibit synchronous rhythmic membrane activity with a periodicity of ∼ 5-10 s. This rhythmic lLN(v) activity is blocked by TTX, voltage-gated sodium blocker, or α-bungarotoxin, nicotinic acetylcholine receptor antagonist, indicating that action potential-dependent cholinergic synaptic connections are required for rhythmic lLN(v) activity. Since injecting current into one neuron of the pair had no effect on the membrane activity of the other neuron of the pair, this suggests that the synchrony is attributable to bilateral inputs and not coupling between the pairs of lLN(v)s. To further elucidate the nature of these synaptic inputs to lLN(v)s, we blocked or activated a variety of neurotransmitter receptors and measured effects on network activity and ionic conductances. These measurements indicate the lLN(v)s possess excitatory nicotinic ACh receptors, inhibitory ionotropic GABA(A) receptors, and inhibitory ionotropic GluCl (glutamate-gated chloride) receptors. We demonstrate that cholinergic input, but not GABAergic input, is required for synchronous membrane activity, whereas GABA can modulate firing patterns. We conclude that neuropeptidergic lLN(v)s that control rest and arousal receive synchronous synaptic inputs mediated by ACh.


Asunto(s)
Encéfalo/fisiología , Ritmo Circadiano/fisiología , Drosophila melanogaster , Neuronas/fisiología , Receptores de GABA/fisiología , Receptores Nicotínicos/fisiología , Transmisión Sináptica/fisiología , Animales , Encéfalo/efectos de los fármacos , Canales de Cloruro/fisiología , Ritmo Circadiano/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Técnicas de Placa-Clamp/métodos , Receptores de GABA-A/fisiología , Transmisión Sináptica/efectos de los fármacos
10.
Brain Behav Immun ; 26(3): 407-13, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22019350

RESUMEN

In mammals, circadian and daily rhythms influence nearly all aspects of physiology, ranging from behavior to gene expression. Functional molecular clocks have been described in the murine spleen and splenic NK cells. The aim of our study was to investigate the existence of molecular clock mechanisms in other immune cells. Therefore, we measured the circadian changes in gene expression of clock genes (Per1, Per2, Bmal1, and Clock) and clock-controlled transcription factors (Rev-erbα and Dbp) in splenic enriched macrophages, dendritic cells, and B cells in both mice entrained to a light-dark cycle and under constant environmental conditions. Our study reveals the existence of functional molecular clock mechanisms in splenic macrophages, dendritic cells, and B cells.


Asunto(s)
Linfocitos B/metabolismo , Péptidos y Proteínas de Señalización del Ritmo Circadiano/genética , Ritmo Circadiano/genética , Células Dendríticas/metabolismo , Macrófagos/metabolismo , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Animales , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Expresión Génica , Ratones , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Fotoperiodo , Bazo/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
11.
Proc Natl Acad Sci U S A ; 106(19): 8049-54, 2009 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-19416829

RESUMEN

Class B1 (secretin family) G protein-coupled receptors (GPCRs) modulate a wide range of physiological functions, including glucose homeostasis, feeding behavior, fat deposition, bone remodeling, and vascular contractility. Endogenous peptide ligands for these GPCRs are of intermediate length (27-44 aa) and include receptor affinity (C-terminal) as well as receptor activation (N-terminal) domains. We have developed a technology in which a peptide ligand tethered to the cell membrane selectively modulates corresponding class B1 GPCR-mediated signaling. The engineered cDNA constructs encode a single protein composed of (i) a transmembrane domain (TMD) with an intracellular C terminus, (ii) a poly(asparagine-glycine) linker extending from the TMD into the extracellular space, and (iii) a class B1 receptor ligand positioned at the N terminus. We demonstrate that membrane-tethered peptides, like corresponding soluble ligands, trigger dose-dependent receptor activation. The broad applicability of this approach is illustrated by experiments using tethered versions of 7 mammalian endogenous class B1 GPCR agonists. In parallel, we carried out mutational studies focused primarily on incretin ligands of the glucagon-like peptide-1 receptor. These experiments suggest that tethered ligand activity is conferred in large part by the N-terminal domain of the peptide hormone. Follow-up studies revealed that interconversion of tethered agonists and antagonists can be achieved with the introduction of selected point mutations. Such complementary receptor modulators provide important new tools for probing receptor structure-function relationships as well as for future studies aimed at dissecting the tissue-specific biological role of a GPCR in vivo (e.g., in the brain vs. in the periphery).


Asunto(s)
Membrana Celular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Bioquímica/métodos , Línea Celular , Eliminación de Gen , Humanos , Incretinas/metabolismo , Ligandos , Modelos Biológicos , Hormonas Peptídicas/metabolismo , Péptidos/química , Mutación Puntual , Estructura Terciaria de Proteína , Receptores Acoplados a Proteínas G/genética
12.
Elife ; 112022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35363138

RESUMEN

Insects adapt their response to stimuli, such as odours, according to their pairing with positive or negative reinforcements, such as sugar or shock. Recent electrophysiological and imaging findings in Drosophila melanogaster allow detailed examination of the neural mechanisms supporting the acquisition, forgetting, and assimilation of memories. We propose that this data can be explained by the combination of a dopaminergic plasticity rule that supports a variety of synaptic strength change phenomena, and a circuit structure (derived from neuroanatomy) between dopaminergic and output neurons that creates different roles for specific neurons. Computational modelling shows that this circuit allows for rapid memory acquisition, transfer from short term to long term, and exploration/exploitation trade-off. The model can reproduce the observed changes in the activity of each of the identified neurons in conditioning paradigms and can be used for flexible behavioural control.


Asunto(s)
Drosophila melanogaster , Cuerpos Pedunculados , Animales , Drosophila melanogaster/fisiología , Memoria/fisiología , Motivación , Cuerpos Pedunculados/fisiología , Odorantes
13.
Curr Biol ; 18(2): R84-93, 2008 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-18211849

RESUMEN

Molecular genetics has revealed the identities of several components of the fundamental circadian molecular oscillator - an evolutionarily conserved molecular mechanism of transcription and translation that can operate in a cell-autonomous manner. Therefore, it was surprising when studies of circadian rhythmic behavior in the fruit fly Drosophila suggested that the normal operations of circadian clock cells, which house the molecular oscillator, in fact depend on non-cell-autonomous effects - interactions between the clock cells themselves. Here we review several genetic analyses that broadly extend that viewpoint. They support a model whereby the approximately 150 circadian clock cells in the brain of the fly are sub-divided into functionally discrete rhythmic centers. These centers alternatively cooperate or compete to control the different episodes of rhythmic behavior that define the fly's daily activity profile.


Asunto(s)
Relojes Biológicos/fisiología , Ritmo Circadiano/fisiología , Drosophila melanogaster/fisiología , Animales , Encéfalo/citología , Encéfalo/metabolismo , Encéfalo/fisiología , Proteínas de Drosophila/metabolismo , Ambiente , Ácido Glutámico/metabolismo , Luz , Locomoción/fisiología , Neuropéptidos/metabolismo , Temperatura
14.
PLoS Biol ; 6(11): e273, 2008 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-18986214

RESUMEN

Drosophila clock neurons are self-sustaining cellular oscillators that rely on negative transcriptional feedback to keep circadian time. Proper regulation of organismal rhythms of physiology and behavior requires coordination of the oscillations of individual clock neurons within the circadian control network. Over the last decade, it has become clear that a key mechanism for intercellular communication in the circadian network is signaling between a subset of clock neurons that secrete the neuropeptide pigment dispersing factor (PDF) and clock neurons that possess its G protein-coupled receptor (PDFR). Furthermore, the specific hypothesis has been proposed that PDF-secreting clock neurons entrain the phase of organismal rhythms, and the cellular oscillations of other clock neurons, via the temporal patterning of secreted PDF signals. In order to test this hypothesis, we have devised a novel technique for altering the phase relationship between circadian transcriptional feedback oscillation and PDF secretion by using an ion channel-directed spider toxin to modify voltage-gated Na(+) channel inactivation in vivo. This technique relies on the previously reported "tethered-toxin" technology for cell-autonomous modulation of ionic conductances via heterologous expression of subtype-specific peptide ion channel toxins as chimeric fusion proteins tethered to the plasma membrane with a glycosylphosphatidylinositol (GPI) anchor. We demonstrate for the first time, to our knowledge, the utility of the tethered-toxin technology in a transgenic animal, validating four different tethered spider toxin ion channel modifiers for use in Drosophila. Focusing on one of these toxins, we show that GPI-tethered Australian funnel-web spider toxin delta-ACTX-Hv1a inhibits Drosophila para voltage-gated Na(+) channel inactivation when coexpressed in Xenopus oocytes. Transgenic expression of membrane-tethered delta-ACTX-Hv1a in vivo in the PDF-secreting subset of clock neurons induces rhythmic action potential bursts and depolarized plateau potentials. These in vitro and in vivo electrophysiological effects of membrane-tethered delta-ACTX-Hv1a are consistent with the effects of soluble delta-ACTX-Hv1a purified from venom on Na(+) channel physiological and biophysical properties in cockroach neurons. Membrane-tethered delta-ACTX-Hv1a expression in the PDF-secreting subset of clock neurons induces an approximately 4-h phase advance of the rhythm of PDF accumulation in their terminals relative to both the phase of the day:night cycle and the phase of the circadian transcriptional feedback loops. As a consequence, the morning anticipatory peak of locomotor activity preceding dawn, which has been shown to be driven by the clocks of the PDF-secreting subset of clock neurons, phase advances coordinately with the phase of the PDF rhythm of the PDF-secreting clock neurons, rather than maintaining its phase relationship with the day:night cycle and circadian transcriptional feedback loops. These results (1) validate the tethered-toxin technology for cell-autonomous modulation of ion channel biophysical properties in vivo in transgenic Drosophila, (2) demonstrate that the kinetics of para Na(+) channel inactivation is a key parameter for determining the phase relationship between circadian transcriptional feedback oscillation and PDF secretion, and (3) provide experimental support for the hypothesis that PDF-secreting clock neurons entrain the phase of organismal rhythms via the temporal patterning of secreted PDF signals.


Asunto(s)
Relojes Biológicos/fisiología , Ritmo Circadiano/fisiología , Proteínas de Drosophila/metabolismo , Drosophila/fisiología , Neuronas/fisiología , Neuropéptidos/metabolismo , Canales de Sodio/metabolismo , Animales , Animales Modificados Genéticamente , Drosophila/genética , Drosophila/metabolismo , Regulación de la Expresión Génica , Actividad Motora/fisiología , Oocitos/metabolismo , Venenos de Araña/farmacología , Xenopus
15.
Nat Commun ; 12(1): 4131, 2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34226544

RESUMEN

Feeding decisions are fundamental to survival, and decision making is often disrupted in disease. Here, we show that neural activity in a small population of neurons projecting to the fan-shaped body higher-order central brain region of Drosophila represents food choice during sensory conflict. We found that food deprived flies made tradeoffs between appetitive and aversive values of food. We identified an upstream neuropeptidergic and dopaminergic network that relays internal state and other decision-relevant information to a specific subset of fan-shaped body neurons. These neurons were strongly inhibited by the taste of the rejected food choice, suggesting that they encode behavioral food choice. Our findings reveal that fan-shaped body taste responses to food choices are determined not only by taste quality, but also by previous experience (including choice outcome) and hunger state, which are integrated in the fan-shaped body to encode the decision before relay to downstream motor circuits for behavioral implementation.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Drosophila/metabolismo , Neuronas/metabolismo , Animales , Encéfalo/metabolismo , Toma de Decisiones , Conducta Alimentaria/fisiología , Preferencias Alimentarias , Hambre/fisiología , Gusto/fisiología , Percepción del Gusto
16.
Nat Commun ; 12(1): 1115, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33602917

RESUMEN

Animals form and update learned associations between otherwise neutral sensory cues and aversive outcomes (i.e., punishment) to predict and avoid danger in changing environments. When a cue later occurs without punishment, this unexpected omission of aversive outcome is encoded as reward via activation of reward-encoding dopaminergic neurons. How such activation occurs remains unknown. Using real-time in vivo functional imaging, optogenetics, behavioral analysis and synaptic reconstruction from electron microscopy data, we identify the neural circuit mechanism through which Drosophila reward-encoding dopaminergic neurons are activated when an olfactory cue is unexpectedly no longer paired with electric shock punishment. Reduced activation of punishment-encoding dopaminergic neurons relieves depression of olfactory synaptic inputs to cholinergic neurons. Synaptic excitation by these cholinergic neurons of reward-encoding dopaminergic neurons increases their odor response, thus decreasing aversiveness of the odor. These studies reveal how an excitatory cholinergic relay from punishment- to reward-encoding dopaminergic neurons encodes the absence of punishment as reward, revealing a general circuit motif for updating aversive memories that could be present in mammals.


Asunto(s)
Dopamina/metabolismo , Drosophila melanogaster/fisiología , Castigo , Recompensa , Animales , Reacción de Prevención/fisiología , Condicionamiento Clásico , Neuronas Dopaminérgicas/fisiología , Memoria/fisiología , Aprendizaje Inverso , Olfato/fisiología , Sinapsis/fisiología
17.
Science ; 371(6533): 1059-1063, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33674494

RESUMEN

Color detection is used by animals of diverse phyla to navigate colorful natural environments and is thought to require evolutionarily conserved opsin photoreceptor genes. We report that Caenorhabditis elegans roundworms can discriminate between colors despite the fact that they lack eyes and opsins. Specifically, we found that white light guides C. elegans foraging decisions away from a blue-pigment toxin secreted by harmful bacteria. These foraging decisions are guided by specific blue-to-amber ratios of light. The color specificity of color-dependent foraging varies notably among wild C. elegans strains, which indicates that color discrimination is ecologically important. We identified two evolutionarily conserved cellular stress response genes required for opsin-independent, color-dependent foraging by C. elegans, and we speculate that cellular stress response pathways can mediate spectral discrimination by photosensitive cells and organisms-even by those lacking opsins.


Asunto(s)
Caenorhabditis elegans/fisiología , Caenorhabditis elegans/efectos de la radiación , Visión de Colores , Conducta Alimentaria , Animales , Reacción de Prevención , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/fisiología , Secuencia Conservada , Escherichia coli , Luz , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Proteínas Quinasas/genética , Proteínas Quinasas/fisiología , Pseudomonas aeruginosa/metabolismo , Piocianina/metabolismo , Piocianina/toxicidad
18.
J Neurosci ; 28(25): 6493-501, 2008 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-18562620

RESUMEN

Drosophila circadian rhythms are controlled by a neural circuit containing approximately 150 clock neurons. Although much is known about mechanisms of autonomous cellular oscillation, the connection between cellular oscillation and functional outputs that control physiological and behavioral rhythms is poorly understood. To address this issue, we performed whole-cell patch-clamp recordings on lateral ventral clock neurons (LN(v)s), including large (lLN(v)s) and small LN(v)s (sLN(v)s), in situ in adult fly whole-brain explants. We found two distinct sizes of action potentials (APs) in >50% of lLN(v)s that fire APs spontaneously, and determined that large APs originate in the ipsilateral optic lobe and small APs in the contralateral. lLN(v) resting membrane potential (RMP), spontaneous AP firing rate, and membrane resistance are cyclically regulated as a function of time of day in 12 h light/dark conditions (LD). lLN(v) RMP becomes more hyperpolarized as time progresses from dawn to dusk with a concomitant decrease in spontaneous AP firing rate and membrane resistance. From dusk to dawn, lLN(v) RMP becomes more depolarized, with spontaneous AP firing rate and membrane resistance remaining stable. In contrast, circadian defective per(0) null mutant lLN(v) membrane excitability is nearly constant in LD. Over 24 h in constant darkness (DD), wild-type lLN(v) membrane excitability is not cyclically regulated, although RMP gradually becomes slightly more depolarized. sLN(v) RMP is most depolarized around lights-on, with substantial variability centered around lights-off in LD. Our results indicate that LN(v) membrane excitability encodes time of day via a circadian clock-dependent mechanism, and likely plays a critical role in regulating Drosophila circadian behavior.


Asunto(s)
Membrana Celular/fisiología , Ritmo Circadiano/fisiología , Proteínas de Drosophila/fisiología , Drosophila melanogaster/fisiología , Neuronas/fisiología , Factores de Transcripción/fisiología , Potenciales de Acción/fisiología , Animales , Relojes Biológicos/fisiología , Proteínas CLOCK , Drosophila melanogaster/citología
19.
J Biol Rhythms ; 23(2): 117-28, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18375861

RESUMEN

Drosophila clock neurons exhibit self-sustaining cellular oscillations that rely in part on rhythmic transcriptional feedback loops. We have previously determined that electrical silencing of the pigment dispersing factor (PDF)-expressing lateral-ventral (LN(V)) pacemaker subset of fly clock neurons via expression of an inward-rectifier K(+) channel (Kir2.1) severely disrupts free-running rhythms of locomotor activity-most flies are arrhythmic and those that are not exhibit weak short-period rhythms-and abolishes LN(V) molecular oscillation in constant darkness. PDF is known to be an important LN(V) output signal. Here we examine the effects of electrical silencing of the LN(V) pacemakers on molecular rhythms in other, nonsilenced, subsets of clock neurons. In contrast to previously described cell-autonomous abolition of free-running molecular rhythms, we find that electrical silencing of the LN(V) pacemakers via Kir2.1 expression does not impair molecular rhythms in LN(D), DN1, and DN2 subsets of clock neurons. However, free-running molecular rhythms in these non-LN(V) clock neurons occur with advanced phase. Electrical silencing of LN(V)s phenocopies PDF null mutation (pdf (01) ) at both behavioral and molecular levels except for the complete abolition of free-running cellular oscillation in the LN(V)s themselves. LN(V) electrically silenced or pdf 01 flies exhibit weak free-running behavioral rhythms with short period, and the molecular oscillation in non-LN(V) neurons phase advances in constant darkness. That LN( V) electrical silencing leads to the same behavioral and non-LN( V) molecular phenotypes as pdf 01 suggests that persistence of LN(V) molecular oscillation in pdf 01 flies has no functional effect, either on behavioral rhythms or on non-LN(V) molecular rhythms. We thus conclude that functionally relevant signals from LN(V)s to non-LN(V) clock neurons and other downstream targets rely both on PDF signaling and LN(V) electrical activity, and that LN( V)s do not ordinarily send functionally relevant signals via PDF-independent mechanisms.


Asunto(s)
Relojes Biológicos/fisiología , Ritmo Circadiano/fisiología , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Potenciales de la Membrana/fisiología , Neuronas/metabolismo , Neuropéptidos/metabolismo , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Encéfalo/citología , Proteínas de Drosophila/genética , Masculino , Actividad Motora/fisiología , Neuropéptidos/genética , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo
20.
J Neurosci ; 27(46): 12489-99, 2007 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-18003827

RESUMEN

Although circadian oscillation in dynamics of intracellular Ca2+ signals has been observed in both plant and animal cells, it has remained unknown whether Ca2+ signals play an in vivo role in cellular oscillation itself. To address this question, we modified the dynamics of intracellular Ca2+ signals in circadian pacemaker neurons in vivo by targeted expression of varying doses of a Ca2+ buffer protein in transgenic Drosophila melanogaster. Intracellular Ca2+ buffering in pacemaker neurons results in dose-dependent slowing of free-running behavioral rhythms, with average period >3 h longer than control at the highest dose. The rhythmic nuclear accumulation of a transcription factor known to be essential for cellular circadian oscillation is also slowed. We also determined that Ca2+ buffering interacts synergistically with genetic manipulations that interfere with either calmodulin or calmodulin-dependent protein kinase II function. These results suggest a role for intracellular Ca2+ signaling in regulating intrinsic cellular oscillation in vivo.


Asunto(s)
Relojes Biológicos/genética , Señalización del Calcio/genética , Calcio/metabolismo , Ritmo Circadiano/genética , Drosophila melanogaster/metabolismo , Parvalbúminas/genética , Animales , Animales Modificados Genéticamente , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Calmodulina/metabolismo , Células Cultivadas , Proteínas de Drosophila/genética , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Regulación de la Expresión Génica/genética , Líquido Intracelular/metabolismo , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Sistema Nervioso/citología , Sistema Nervioso/metabolismo , Vías Nerviosas/citología , Vías Nerviosas/metabolismo , Neuronas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA