Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Elife ; 122023 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-37750673

RESUMEN

Tastes typically evoke innate behavioral responses that can be broadly categorized as acceptance or rejection. However, research in Drosophila melanogaster indicates that taste responses also exhibit plasticity through experience-dependent changes in mushroom body circuits. In this study, we develop a novel taste learning paradigm using closed-loop optogenetics. We find that appetitive and aversive taste memories can be formed by pairing gustatory stimuli with optogenetic activation of sensory neurons or dopaminergic neurons encoding reward or punishment. As with olfactory memories, distinct dopaminergic subpopulations drive the parallel formation of short- and long-term appetitive memories. Long-term memories are protein synthesis-dependent and have energetic requirements that are satisfied by a variety of caloric food sources or by direct stimulation of MB-MP1 dopaminergic neurons. Our paradigm affords new opportunities to probe plasticity mechanisms within the taste system and understand the extent to which taste responses depend on experience.

2.
Annu Rev Physiol ; 85: 25-45, 2023 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-36332657

RESUMEN

Salt taste, the taste of sodium chloride (NaCl), is mechanistically one of the most complex and puzzling among basic tastes. Sodium has essential functions in the body but causes harm in excess. Thus, animals use salt taste to ingest the right amount of salt, which fluctuates by physiological needs: typically, attraction to low salt concentrations and rejection of high salt. This concentration-valence relationship is universally observed in terrestrial animals, and research has revealed complex peripheral codes for NaCl involving multiple taste pathways of opposing valence. Sodium-dependent and -independent pathways mediate attraction and aversion to NaCl, respectively. Gustatory sensors and cells that transduce NaCl have been uncovered, along with downstream signal transduction and neurotransmission mechanisms. However, much remains unknown. This article reviews classical and recent advances in our understanding of the molecular and cellular mechanisms underlying salt taste in mammals and insects and discusses perspectives on human salt taste.


Asunto(s)
Papilas Gustativas , Gusto , Animales , Humanos , Gusto/fisiología , Cloruro de Sodio/metabolismo , Papilas Gustativas/metabolismo , Sodio/metabolismo , Transducción de Señal , Mamíferos/metabolismo
3.
Curr Biol ; 32(14): 3070-3081.e5, 2022 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-35772408

RESUMEN

Dietary salt detection and consumption are crucial to maintaining fluid and ionic homeostasis. To optimize salt intake, animals employ salt-dependent activation of multiple taste pathways. Generally, sodium activates attractive taste cells, but attraction is overridden at high salt concentrations by cation non-selective activation of aversive taste cells. In flies, high salt avoidance is driven by both "bitter" taste neurons and a class of glutamatergic "high salt" neurons expressing pickpocket23 (ppk23). Although the cellular basis of salt taste has been described, many of the molecular mechanisms remain elusive. Here, we show that ionotropic receptor 7c (IR7c) is expressed in glutamatergic high salt neurons, where it functions with co-receptors IR76b and IR25a to detect high salt and is essential for monovalent salt taste. Misexpression of IR7c in sweet neurons, which endogenously express IR76b and IR25a, confers responsiveness to non-sodium salts, indicating that IR7c is sufficient to convert a sodium-selective gustatory receptor neuron to a cation non-selective one. Furthermore, the resultant transformation of taste neuron tuning switches potassium chloride from an aversive to an attractive tastant. This research provides insight into the molecular basis of monovalent and divalent salt-taste coding.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Cloruro de Sodio/farmacología , Gusto/fisiología , Percepción del Gusto/fisiología
4.
Sci Adv ; 7(49): eabj0186, 2021 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-34851668

RESUMEN

In flies, neuronal sensors detect prandial changes in circulating fructose levels and either sustain or terminate feeding, depending on internal state. Here, we describe a three-part neural circuit that imparts satiety-dependent modulation of fructose sensing. We show that dorsal fan-shaped body neurons display oscillatory calcium activity when hemolymph glucose is high and that these oscillations require glutamatergic input from SLP-AB or "Janus" neurons projecting from the protocerebrum to the asymmetric body. Suppression of activity in this circuit, either by starvation or by genetic silencing, promotes specific drive for fructose ingestion. This is achieved through neuropeptidergic signaling by tachykinin, which is released from the fan-shaped body when glycemia is high. Tachykinin, in turn, signals to Gr43a-positive fructose sensors to modulate their response to fructose. Together, our results demonstrate how a three-layer neural circuit links the detection of two sugars to produce precise satiety-dependent control of feeding behavior.

5.
Curr Biol ; 31(16): 3525-3537.e6, 2021 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-34197729

RESUMEN

Sour has been studied almost exclusively as an aversive taste modality. Yet recent work in Drosophila demonstrates that specific carboxylic acids are attractive at ecologically relevant concentrations. Here, we demonstrate that lactic acid is an appetitive and energetic tastant, which stimulates feeding through activation of sweet gustatory receptor neurons (GRNs). This activation displays distinct, mechanistically separable stimulus onset and removal phases. Ionotropic receptor 25a (IR25a) primarily mediates the onset response, which shows specificity for the lactate anion and drives feeding initiation through proboscis extension. Conversely, sweet gustatory receptors (Gr64a-f) mediate a non-specific removal response to low pH that primarily impacts ingestion. While mutations in either receptor family have marginal impacts on feeding, lactic acid attraction is completely abolished in combined mutants. Thus, specific components of lactic acid are detected through two classes of receptors to activate a single set of sensory neurons in physiologically distinct ways, ultimately leading to robust behavioral attraction.


Asunto(s)
Drosophila melanogaster , Ácido Láctico , Receptores de Superficie Celular , Células Receptoras Sensoriales , Gusto , Animales , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Receptores de Superficie Celular/genética , Células Receptoras Sensoriales/fisiología
6.
G3 (Bethesda) ; 11(5)2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-33714999

RESUMEN

Feeding is an essential part of animal life that is greatly impacted by the sense of taste. Although the characterization of taste-detection at the periphery has been extensive, higher order taste and feeding circuits are still being elucidated. Here, we use an automated closed-loop optogenetic activation screen to detect novel taste and feeding neurons in Drosophila melanogaster. Out of 122 Janelia FlyLight Project GAL4 lines preselected based on expression pattern, we identify six lines that acutely promote feeding and 35 lines that inhibit it. As proof of principle, we follow up on R70C07-GAL4, which labels neurons that strongly inhibit feeding. Using split-GAL4 lines to isolate subsets of the R70C07-GAL4 population, we find both appetitive and aversive neurons. Furthermore, we show that R70C07-GAL4 labels putative second-order taste interneurons that contact both sweet and bitter sensory neurons. These results serve as a resource for further functional dissection of fly feeding circuits.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Optogenética , Gusto
7.
Curr Biol ; 31(10): 2065-2074.e5, 2021 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-33740428

RESUMEN

How social interactions influence cognition is a fundamental question, yet rarely addressed at the neurobiological level. It is well established that the presence of conspecifics affects learning and memory performance, but the neural basis of this process has only recently begun to be investigated. In the fruit fly Drosophila melanogaster, the presence of other flies improves retrieval of a long-lasting olfactory memory. Here, we demonstrate that this is a composite memory composed of two distinct elements. One is an individual memory that depends on outputs from the α'ß' Kenyon cells (KCs) of the mushroom bodies (MBs), the memory center in the insect brain. The other is a group memory requiring output from the αß KCs, a distinct sub-part of the MBs. We show that social facilitation of memory increases with group size and is triggered by CO2 released by group members. Among the different known neurons carrying CO2 information in the brain, we establish that the bilateral ventral projection neuron (biVPN), which projects onto the MBs, is necessary for social facilitation. Moreover, we demonstrate that CO2-evoked memory engages a serotoninergic pathway involving the dorsal-paired medial (DPM) neurons, revealing a new role for this pair of serotonergic neurons. Overall, we identified both the sensorial cue and the neural circuit (biVPN>αß>DPM>αß) governing social facilitation of memory in flies. This study provides demonstration that being in a group recruits the expression of a cryptic memory and that variations in CO2 concentration can affect cognitive processes in insects.


Asunto(s)
Dióxido de Carbono/metabolismo , Drosophila melanogaster/metabolismo , Memoria a Largo Plazo/fisiología , Facilitación Social , Animales , Femenino , Masculino , Cuerpos Pedunculados/citología , Cuerpos Pedunculados/fisiología , Neuronas
8.
Cell Calcium ; 91: 102259, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32739609

RESUMEN

In their recent paper, Li and colleagues discover that cold food tastes less sweet to flies, in part by activating bitter sensory neurons through a rhodopsin-dependent mechanism [1]. This work establishes temperature as an important variable in understanding fly taste processing and adds diversity to the sensory roles for rhodopsin receptors.


Asunto(s)
Drosophila , Gusto , Animales , Drosophila melanogaster , Rodopsina , Temperatura
9.
Cell Rep ; 32(4): 107972, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32726635

RESUMEN

Drosophila development is governed by distinct ecdysone steroid pulses that initiate spatially and temporally defined gene expression programs. The translation of these signals into tissue-specific responses is crucial for metamorphosis, but the mechanisms that confer specificity to systemic ecdysone pulses are far from understood. Here, we identify Bric-à-brac 2 (Bab2) as an ecdysone-responsive transcriptional repressor that controls temporal gene expression during larval to pupal transition. Bab2 is necessary to terminate Salivary gland secretion (Sgs) gene expression, while premature Bab2 expression blocks Sgs genes and causes precocious salivary gland histolysis. The timely expression of bab2 is controlled by the ecdysone-responsive transcription factor Broad, and manipulation of EcR/USP/Broad signaling induces inappropriate Bab2 expression and termination of Sgs gene expression. Bab2 directly binds to Sgs loci in vitro and represses all Sgs genes in vivo. Our work characterizes Bab2 as a temporal regulator of somatic gene expression in response to systemic ecdysone signaling.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Factores de Transcripción/genética , Animales , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Ecdisona/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Larva/metabolismo , Metamorfosis Biológica/genética , Factores de Transcripción/metabolismo , Transcripción Genética/genética
10.
Elife ; 82019 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-31322499

RESUMEN

Manipulating feeding circuits in freely moving animals is challenging, in part because the timing of sensory inputs is affected by the animal's behavior. To address this challenge in Drosophila, we developed the Sip-Triggered Optogenetic Behavior Enclosure ('STROBE'). The STROBE is a closed-looped system for real-time optogenetic activation of feeding flies, designed to evoke neural excitation coincident with food contact. We previously demonstrated the STROBE's utility in probing the valence of fly sensory neurons (Jaeger et al., 2018). Here we provide a thorough characterization of the STROBE system, demonstrate that STROBE-driven behavior is modified by hunger and the presence of taste ligands, and find that mushroom body dopaminergic input neurons and their respective post-synaptic partners drive opposing feeding behaviors following activation. Together, these results establish the STROBE as a new tool for dissecting fly feeding circuits and suggest a role for mushroom body circuits in processing naïve taste responses.


Asunto(s)
Drosophila/fisiología , Entomología/métodos , Conducta Alimentaria , Red Nerviosa/fisiología , Optogenética/métodos , Animales
11.
Elife ; 72018 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-30307393

RESUMEN

Each taste modality is generally encoded by a single, molecularly defined, population of sensory cells. However, salt stimulates multiple taste pathways in mammals and insects, suggesting a more complex code for salt taste. Here, we examine salt coding in Drosophila. After creating a comprehensive molecular map comprised of five discrete sensory neuron classes across the fly labellum, we find that four are activated by salt: two exhibiting characteristics of 'low salt' cells, and two 'high salt' classes. Behaviorally, low salt attraction depends primarily on 'sweet' neurons, with additional input from neurons expressing the ionotropic receptor IR94e. High salt avoidance is mediated by 'bitter' neurons and a population of glutamatergic neurons expressing Ppk23. Interestingly, the impact of these glutamatergic neurons depends on prior salt consumption. These results support a complex model for salt coding in flies that combinatorially integrates inputs from across cell types to afford robust and flexible salt behaviors.


Asunto(s)
Drosophila melanogaster/fisiología , Cloruro de Sodio/farmacología , Gusto/fisiología , Animales , Reacción de Prevención/efectos de los fármacos , Calcio/metabolismo , Drosophila melanogaster/anatomía & histología , Modelos Biológicos , Feromonas/farmacología , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/fisiología , Toxina Tetánica/farmacología
12.
Cell ; 174(6): 1450-1464.e23, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30100184

RESUMEN

Synapses are fundamental units of communication in the brain. The prototypical synapse-organizing complex neurexin-neuroligin mediates synapse development and function and is central to a shared genetic risk pathway in autism and schizophrenia. Neurexin's role in synapse development is thought to be mediated purely by its protein domains, but we reveal a requirement for a rare glycan modification. Mice lacking heparan sulfate (HS) on neurexin-1 show reduced survival, as well as structural and functional deficits at central synapses. HS directly binds postsynaptic partners neuroligins and LRRTMs, revealing a dual binding mode involving intrinsic glycan and protein domains for canonical synapse-organizing complexes. Neurexin HS chains also bind novel ligands, potentially expanding the neurexin interactome to hundreds of HS-binding proteins. Because HS structure is heterogeneous, our findings indicate an additional dimension to neurexin diversity, provide a molecular basis for fine-tuning synaptic function, and open therapeutic directions targeting glycan-binding motifs critical for brain development.


Asunto(s)
Heparitina Sulfato/metabolismo , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Sinapsis/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de Unión al Calcio , Moléculas de Adhesión Celular Neuronal/antagonistas & inhibidores , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Drosophila , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Femenino , Glicopéptidos/análisis , Heparitina Sulfato/química , Humanos , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso , Moléculas de Adhesión de Célula Nerviosa/antagonistas & inhibidores , Moléculas de Adhesión de Célula Nerviosa/genética , Neuronas/citología , Neuronas/metabolismo , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Alineación de Secuencia
13.
Curr Biol ; 26(21): 2854-2861, 2016 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-27720624

RESUMEN

Nutrient deprivation can lead to dramatic changes in feeding behavior, including acceptance of foods that are normally rejected. In flies, this behavioral shift depends in part on reciprocal sensitization and desensitization of sweet and bitter taste, respectively. However, the mechanisms for bitter taste modulation remain unclear. Here, we identify a set of octopaminergic/tyraminergic neurons, named OA-VLs, that directly modulate bitter sensory neuron output in response to starvation. OA-VLs are in close proximity to bitter sensory neuron axon terminals and show reduced tonic firing following starvation. We find that octopamine and tyramine potentiate bitter sensory neuron responses, suggesting that starvation-induced reduction in OA-VL activity depotentiates bitter taste. Consistent with this model, artificial silencing of OA-VL activity induces a starvation-like reduction in bitter sensory neuron output. These results demonstrate that OA-VLs mediate a critical step in starvation-dependent bitter taste modulation, allowing flies to dynamically balance the risks associated with bitter food consumption against the threat of severe starvation.


Asunto(s)
Drosophila melanogaster/fisiología , Privación de Alimentos , Depresión Sináptica a Largo Plazo , Percepción del Gusto , Animales , Femenino , Células Receptoras Sensoriales/fisiología
14.
Nat Commun ; 6: 6667, 2015 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-25807033

RESUMEN

The fly pharyngeal sense organs lie at the transition between external and internal nutrient-sensing mechanisms. Here we investigate the function of pharyngeal sweet gustatory receptor neurons, demonstrating that they express a subset of the nine previously identified sweet receptors and respond to stimulation with a panel of sweet compounds. We show that pox-neuro (poxn) mutants lacking taste function in the legs and labial palps have intact pharyngeal sweet taste, which is both necessary and sufficient to drive preferred consumption of sweet compounds by prolonging ingestion. Moreover, flies putatively lacking all sweet taste show little preference for nutritive or non-nutritive sugars in a short-term feeding assay. Together, our data demonstrate that pharyngeal sense organs play an important role in directing sustained consumption of sweet compounds, and suggest that post-ingestive sugar sensing does not effectively drive food choice in a simple short-term feeding paradigm.


Asunto(s)
Preferencias Alimentarias/fisiología , Edulcorantes no Nutritivos , Edulcorantes Nutritivos , Faringe , Papilas Gustativas/fisiología , Gusto/fisiología , Animales , Drosophila , Proteínas de Drosophila/genética , Conducta Alimentaria/fisiología , Mutación , Proteínas del Tejido Nervioso/genética , Neuronas , Factores de Transcripción Paired Box/genética
15.
Curr Biol ; 24(17): 1978-84, 2014 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-25131672

RESUMEN

The sense of taste is critical in determining the nutritional suitability of foods. Sweet and bitter are primary taste modalities in mammals, and their behavioral relevance is similar in flies. Sweet taste drives the appetitive response to energy sources, whereas bitter taste drives avoidance of potential toxins and also suppresses the sweet response [1, 2]. Despite their importance to survival, little is known about the neural circuit mechanisms underlying integration of sweet and bitter taste. Here, we describe a presynaptic gain control mechanism in Drosophila that differentially affects sweet and bitter taste channels and mediates integration of these opposing stimuli. Gain control is known to play an important role in fly olfaction, where GABAB receptor (GABABR) mediates intra- and interglomerular presynaptic inhibition of sensory neuron output [3-5]. In the taste system, we find that gustatory receptor neurons (GRNs) responding to sweet compounds express GABABR, whereas those that respond to bitter do not. GABABR mediates presynaptic inhibition of calcium responses in sweet GRNs, and both sweet and bitter stimuli evoke GABAergic neuron activity in the vicinity of GRN axon terminals. Pharmacological blockade and genetic reduction of GABABR both lead to increased sugar responses and decreased suppression of the sweet response by bitter compounds. We propose a model in which GABA acts via GABABR to expand the dynamic range of sweet GRNs through presynaptic gain control and suppress the output of sweet GRNs in the presence of opposing bitter stimuli.


Asunto(s)
Drosophila melanogaster/fisiología , Percepción del Gusto , Animales , Femenino , Células Receptoras Sensoriales/fisiología , Sacarosa/química
16.
Neuron ; 83(1): 164-77, 2014 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-24991960

RESUMEN

Feeding is dynamically regulated by the palatability of the food source and the physiological needs of the animal. How consumption is controlled by external sensory cues and internal metabolic state remains under intense investigation. Here, we identify four GABAergic interneurons in the Drosophila brain that establish a central feeding threshold which is required to inhibit consumption. Inactivation of these cells results in indiscriminate and excessive intake of all compounds, independent of taste quality or nutritional state. Conversely, acute activation of these neurons suppresses consumption of water and nutrients. The output from these neurons is required to gate activity in motor neurons that control meal initiation and consumption. Thus, our study reveals a layer of inhibitory control in feeding circuits that is required to suppress a latent state of unrestricted and nonselective consumption.


Asunto(s)
Conducta Alimentaria/fisiología , Neuronas GABAérgicas/fisiología , Interneuronas/fisiología , Animales , Animales Modificados Genéticamente , Drosophila , Femenino , Tracto Gastrointestinal/inervación , Tracto Gastrointestinal/fisiología
17.
Neuron ; 79(4): 754-65, 2013 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-23972600

RESUMEN

The decision to engage in one behavior often precludes the selection of others, suggesting cross-inhibition between incompatible behaviors. For example, the likelihood to initiate feeding might be influenced by an animal's commitment to other behaviors. Here, we examine the modulation of feeding behavior in the fruit fly, Drosophila melanogaster, and identify a pair of interneurons in the ventral nerve cord that is activated by stimulation of mechanosensory neurons and inhibits feeding initiation, suggesting that these neurons suppress feeding while the fly is walking. Conversely, inhibiting activity in these neurons promotes feeding initiation and inhibits locomotion. These studies demonstrate the mutual exclusivity between locomotion and feeding initiation in the fly, isolate interneurons that influence this behavioral choice, and provide a framework for studying the neural basis for behavioral exclusivity in Drosophila.


Asunto(s)
Conducta de Elección/fisiología , Conducta Alimentaria/fisiología , Interneuronas/fisiología , Locomoción/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Análisis de Varianza , Animales , Animales Modificados Genéticamente , Encéfalo/citología , Calmodulina/genética , Calmodulina/metabolismo , Relación Dosis-Respuesta a Droga , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Conducta Alimentaria/efectos de los fármacos , Privación de Alimentos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Interneuronas/clasificación , Locomoción/efectos de los fármacos , Quinasa de Cadena Ligera de Miosina/genética , Quinasa de Cadena Ligera de Miosina/metabolismo , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Rodopsina/genética , Sacarosa/administración & dosificación , Edulcorantes/administración & dosificación , Temperatura , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/metabolismo
18.
J Neurosci ; 32(42): 14767-74, 2012 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-23077061

RESUMEN

Animals use gustatory information to assess the suitability of potential food sources and make critical decisions on what to consume. For example, the taste of sugar generally signals a potent dietary source of carbohydrates. However, the intensity of the sensory response to a particular sugar, or "sweetness," is not always a faithful reporter of its nutritional value, and recent evidence suggests that animals can sense the caloric content of food independently of taste. Here, we demonstrate that the vinegar fly Drosophila melanogaster uses both taste and calorie sensing to determine feeding choices, and that the relative contribution of each changes over time. Using the capillary feeder assay, we allowed flies to choose between sources of sugars that varied in their ratio of sweetness to caloric value. We found that flies initially consume sugars according to taste. However, over several hours their preference shifts toward the food source with higher caloric content. This behavioral shift occurs more rapidly following food deprivation and is modulated by cAMP and insulin signaling within neurons. Our results are consistent with the existence of a taste-independent calorie sensor in flies, and suggest that calorie-based reward modifies long-term feeding preferences.


Asunto(s)
Ingestión de Energía/fisiología , Preferencias Alimentarias/fisiología , Gusto/fisiología , Animales , Drosophila melanogaster , Femenino , Masculino
19.
PLoS One ; 6(11): e26993, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22069480

RESUMEN

Wnt proteins comprise a large family of secreted ligands implicated in a wide variety of biological roles. WntD has previously been shown to inhibit the nuclear accumulation of Dorsal/NF-κB protein during embryonic dorsal/ventral patterning and the adult innate immune response, independent of the well-studied Armadillo/ß-catenin pathway. In this paper, we present a novel phenotype for WntD mutant embryos, suggesting that this gene is involved in migration of primordial germ cells (PGC) to the embryonic gonad. Additionally, we describe a genetic suppressor/enhancer screen aimed at identifying genes required for WntD signal transduction, based on the previous observation that maternal overexpression of WntD results in lethally dorsalized embryos. Using an algorithm to narrow down our hits from the screen, we found two novel WntD signaling components: Fz4, a member of the Frizzled family, and the Drosophila Ceramide Kinase homolog, Dcerk. We show here that Dcerk and Dmulk (Drosophila Multi-substrate lipid kinase) redundantly mediate PGC migration. Our data are consistent with a model in which the activity of lipid phosphate phosphatases shapes a concentration gradient of ceramide-1-phosphate (C1P), the product of Dcerk, allowing proper PGC migration.


Asunto(s)
Movimiento Celular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila/genética , Pruebas Genéticas , Células Germinativas/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Receptores Acoplados a Proteínas G/genética , Animales , Animales Modificados Genéticamente , Southern Blotting , Western Blotting , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Ceramidas/metabolismo , Drosophila/crecimiento & desarrollo , Drosophila/metabolismo , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Elementos de Facilitación Genéticos , Femenino , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular/genética , Metabolismo de los Lípidos , Masculino , Filogenia , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Acoplados a Proteínas G/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Supresión Genética , beta Catenina/metabolismo
20.
Neuron ; 61(4): 519-26, 2009 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-19249273

RESUMEN

Mating induces changes in the receptivity and egg-laying behavior in Drosophila females, primarily due to a peptide pheromone called sex peptide which is transferred with the sperm into the female reproductive tract during copulation. Whereas sex peptide is generally believed to modulate fruitless-GAL4-expressing neurons in the central nervous system to produce behavioral changes, we found that six to eight sensory neurons on the reproductive tract labeled by both ppk-GAL4 and fruitless-GAL4 can sense sex peptide to control the induction of postmating behaviors. In these sensory neurons, sex peptide appears to act through Pertussis toxin-sensitive G proteins and suppression of protein kinase A activity to reduce synaptic output. Our results uncover a neuronal mechanism by which sex peptide exerts its control over reproductive behaviors in Drosophila females.


Asunto(s)
Drosophila/fisiología , Conducta Sexual Animal/fisiología , Animales , Clonación Molecular , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Activación Enzimática/fisiología , Femenino , Proteínas de Unión al GTP Heterotriméricas/genética , Proteínas de Unión al GTP Heterotriméricas/fisiología , Inmunohistoquímica , Masculino , Fenotipo , Canales de Potasio de Rectificación Interna/biosíntesis , Canales de Potasio de Rectificación Interna/genética , Transducción de Señal/fisiología , Canales de Sodio/genética , Canales de Sodio/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...