Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
Nat Struct Mol Biol ; 30(9): 1260-1264, 2023 09.
Article En | MEDLINE | ID: mdl-37488356

Control of insulin mRNA translation is crucial for energy homeostasis, but the mechanisms remain largely unknown. We discovered that insulin mRNAs across invertebrates, vertebrates and mammals feature the modified base N6-methyladenosine (m6A). In flies, this RNA modification enhances insulin mRNA translation by promoting the association of the transcript with polysomes. Depleting m6A in Drosophila melanogaster insulin 2 mRNA (dilp2) directly through specific 3' untranslated region (UTR) mutations, or indirectly by mutating the m6A writer Mettl3, decreases dilp2 protein production, leading to aberrant energy homeostasis and diabetic-like phenotypes. Together, our findings reveal adenosine mRNA methylation as a key regulator of insulin protein synthesis with notable implications for energy balance and metabolic disease.


Drosophila melanogaster , Insulin , Animals , Methylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Insulin/genetics , Insulin/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , Adenosine/genetics , Adenosine/metabolism , Mammals/genetics
2.
Elife ; 122023 03 23.
Article En | MEDLINE | ID: mdl-36951889

Diet profoundly influences brain physiology, but how metabolic information is transmuted into neural activity and behavior changes remains elusive. Here, we show that the metabolic enzyme O-GlcNAc Transferase (OGT) moonlights on the chromatin of the D. melanogaster gustatory neurons to instruct changes in chromatin accessibility and transcription that underlie sensory adaptations to a high-sugar diet. OGT works synergistically with the Mitogen Activated Kinase/Extracellular signal Regulated Kinase (MAPK/ERK) rolled and its effector stripe (also known as EGR2 or Krox20) to integrate activity information. OGT also cooperates with the epigenetic silencer Polycomb Repressive Complex 2.1 (PRC2.1) to decrease chromatin accessibility and repress transcription in the high-sugar diet. This integration of nutritional and activity information changes the taste neurons' responses to sugar and the flies' ability to sense sweetness. Our findings reveal how nutrigenomic signaling generates neural activity and behavior in response to dietary changes in the sensory neurons.


Drosophila melanogaster , Nutrigenomics , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Chromatin , Chromosomes/metabolism , Sugars , N-Acetylglucosaminyltransferases/genetics
3.
Curr Biol ; 33(2): 215-227.e3, 2023 01 23.
Article En | MEDLINE | ID: mdl-36528025

In mammals, learning circuits play an essential role in energy balance by creating associations between sensory cues and the rewarding qualities of food. This process is altered by diet-induced obesity, but the causes and mechanisms are poorly understood. Here, we exploited the relative simplicity and wealth of knowledge about the D. melanogaster reinforcement learning network, the mushroom body, in order to study the relationship between the dietary environment, dopamine-induced plasticity, and food associations. We show flies that are fed a high-sugar diet cannot make associations between sensory cues and the rewarding properties of sugar. This deficit was caused by diet exposure, not fat accumulation, and specifically by lower dopamine-induced plasticity onto mushroom body output neurons (MBONs) during learning. Importantly, food memories dynamically tune the output of MBONs during eating, which instead remains fixed in sugar-diet animals. Interestingly, manipulating the activity of MBONs influenced eating and fat mass, depending on the diet. Altogether, this work advances our fundamental understanding of the mechanisms, causes, and consequences of the dietary environment on reinforcement learning and ingestive behavior.


Dopamine , Drosophila melanogaster , Animals , Drosophila melanogaster/physiology , Feeding Behavior/physiology , Learning/physiology , Sugars , Mushroom Bodies/physiology , Eating , Mammals
4.
Curr Biol ; 32(19): 4103-4113.e4, 2022 10 10.
Article En | MEDLINE | ID: mdl-35977546

Elevated sugar consumption is associated with an increased risk for metabolic diseases. Whereas evidence from humans, rodents, and insects suggests that dietary sucrose modifies sweet taste sensation, understanding of peripheral nerve or taste bud alterations is sparse. To address this, male rats were given access to 30% liquid sucrose for 4 weeks (sucrose rats). Neurophysiological responses of the chorda tympani (CT) nerve to lingual stimulation with sugars, other taste qualities, touch, and cold were then compared with controls (access to water only). Morphological and immunohistochemical analyses of fungiform papillae and taste buds were also conducted. Sucrose rats had substantially decreased CT responses to 0.15-2.0 M sucrose compared with controls. In contrast, effects were not observed for glucose, fructose, maltose, Na saccharin, NaCl, organic acid, or umami, touch, or cold stimuli. Whereas taste bud number, size, and innervation volume were unaffected, the number of PLCß2+ taste bud cells in the fungiform papilla was reduced in sucrose rats. Notably, the replacement of sucrose with water resulted in a complete recovery of all phenotypes over 4 weeks. The work reveals the selective and modality-specific effects of sucrose consumption on peripheral taste nerve responses and taste bud cells, with implications for nutrition and metabolic disease risk. VIDEO ABSTRACT.


Saccharin , Taste , Animals , Diet , Dietary Sucrose , Fructose , Glucose , Humans , Male , Maltose , Rats , Sodium Chloride , Taste/physiology , Water
5.
Front Behav Neurosci ; 15: 746299, 2021.
Article En | MEDLINE | ID: mdl-34658807

In humans, alterations in cognitive, motivated, and affective behaviors have been described with consumption of processed diets high in refined sugars and saturated fats and with high body mass index, but the causes, mechanisms, and consequences of these changes remain poorly understood. Animal models have provided an opportunity to answer these questions and illuminate the ways in which diet composition, especially high-levels of added sugar and saturated fats, contribute to brain physiology, plasticity, and behavior. Here we review findings from invertebrate (flies) and vertebrate models (rodents, zebrafish) that implicate these diets with changes in multiple behaviors, including eating, learning and memory, and motivation, and discuss limitations, open questions, and future opportunities.

6.
Neurochem Int ; 149: 105099, 2021 10.
Article En | MEDLINE | ID: mdl-34133954

Humans have known for millennia that nutrition has a profound influence on health and disease, but it is only recently that we have begun mapping the mechanisms via which the dietary environment impacts brain physiology and behavior. Here we review recent evidence on the effects of energy-dense and methionine diets on neural epigenetic marks, gene expression, and behavior in invertebrate and vertebrate model organisms. We also discuss limitations, open questions, and future directions in the emerging field of the neuroepigenetics of nutrition.


Brain/metabolism , Diet, Western/adverse effects , Energy Intake/physiology , Epigenesis, Genetic/physiology , Methionine/administration & dosage , Nutritional Status/physiology , Brain/drug effects , Energy Intake/drug effects , Feeding Behavior/drug effects , Feeding Behavior/physiology , Feeding Behavior/psychology , Food/adverse effects , Humans , Nutritional Status/drug effects
7.
Trends Endocrinol Metab ; 32(2): 95-105, 2021 02.
Article En | MEDLINE | ID: mdl-33384209

Although genetics shapes our sense of taste to prefer some foods over others, taste sensation is plastic and changes with age, disease state, and nutrition. We have known for decades that diet composition can influence the way we perceive foods, but many questions remain unanswered, particularly regarding the effects of chemosensory plasticity on feeding behavior. Here, we review recent evidence on the effects of high-nutrient diets, especially high dietary sugar, on sweet taste in vinegar flies, rodents, and humans, and discuss open questions about molecular and neural mechanisms and research priorities. We also consider ways in which diet-dependent chemosensory plasticity may influence food intake and play a role in the etiology of obesity and metabolic disease. Understanding the interplay between nutrition, taste sensation, and feeding will help us define the role of the food environment in mediating chronic disease and design better public health strategies to combat it.


Diet , Obesity/physiopathology , Feeding Behavior/physiology , Humans , Taste/physiology
8.
Am J Clin Nutr ; 113(1): 232-245, 2021 Jan 04.
Article En | MEDLINE | ID: mdl-33300030

In November 2019, the NIH held the "Sensory Nutrition and Disease" workshop to challenge multidisciplinary researchers working at the interface of sensory science, food science, psychology, neuroscience, nutrition, and health sciences to explore how chemosensation influences dietary choice and health. This report summarizes deliberations of the workshop, as well as follow-up discussion in the wake of the current pandemic. Three topics were addressed: A) the need to optimize human chemosensory testing and assessment, B) the plasticity of chemosensory systems, and C) the interplay of chemosensory signals, cognitive signals, dietary intake, and metabolism. Several ways to advance sensory nutrition research emerged from the workshop: 1) refining methods to measure chemosensation in large cohort studies and validating measures that reflect perception of complex chemosensations relevant to dietary choice; 2) characterizing interindividual differences in chemosensory function and how they affect ingestive behaviors, health, and disease risk; 3) defining circuit-level organization and function that link and interact with gustatory, olfactory, homeostatic, visceral, and cognitive systems; and 4) discovering new ligands for chemosensory receptors (e.g., those produced by the microbiome) and cataloging cell types expressing these receptors. Several of these priorities were made more urgent by the current pandemic because infection with sudden acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the ensuing coronavirus disease of 2019 has direct short- and perhaps long-term effects on flavor perception. There is increasing evidence of functional interactions between the chemosensory and nutritional sciences. Better characterization of this interface is expected to yield insights to promote health, mitigate disease risk, and guide nutrition policy.

9.
Sci Adv ; 6(46)2020 11.
Article En | MEDLINE | ID: mdl-33177090

Diets rich in sugar, salt, and fat alter taste perception and food preference, contributing to obesity and metabolic disorders, but the molecular mechanisms through which this occurs are unknown. Here, we show that in response to a high sugar diet, the epigenetic regulator Polycomb Repressive Complex 2.1 (PRC2.1) persistently reprograms the sensory neurons of Drosophila melanogaster flies to reduce sweet sensation and promote obesity. In animals fed high sugar, the binding of PRC2.1 to the chromatin of the sweet gustatory neurons is redistributed to repress a developmental transcriptional network that modulates the responsiveness of these cells to sweet stimuli, reducing sweet sensation. Half of these transcriptional changes persist despite returning the animals to a control diet, causing a permanent decrease in sweet taste. Our results uncover a new epigenetic mechanism that, in response to the dietary environment, regulates neural plasticity and feeding behavior to promote obesity.


Drosophila Proteins , Drosophila melanogaster , Animals , Diet , Drosophila Proteins/genetics , Drosophila melanogaster/physiology , Epigenesis, Genetic , Obesity/genetics , Sensory Receptor Cells/metabolism , Sugars , Taste/physiology
10.
Elife ; 92020 06 16.
Article En | MEDLINE | ID: mdl-32539934

From humans to vinegar flies, exposure to diets rich in sugar and fat lowers taste sensation, changes food choices, and promotes feeding. However, how these peripheral alterations influence eating is unknown. Here we used the genetically tractable organism D. melanogaster to define the neural mechanisms through which this occurs. We characterized a population of protocerebral anterior medial dopaminergic neurons (PAM DANs) that innervates the ß'2 compartment of the mushroom body and responds to sweet taste. In animals fed a high sugar diet, the response of PAM-ß'2 to sweet stimuli was reduced and delayed, and sensitive to the strength of the signal transmission out of the sensory neurons. We found that PAM-ß'2 DANs activity controls feeding rate and satiation: closed-loop optogenetic activation of ß'2 DANs restored normal eating in animals fed high sucrose. These data argue that diet-dependent alterations in taste weaken satiation by impairing the central processing of sensory signals.


Obesity is a major health problem affecting over 650 million adults worldwide. It is typically caused by overeating high-energy foods, which often contain a lot of sugar. Consuming sugary foods triggers the production of a reward signal called dopamine in the brains of insects and mammals, which reinforces sugar-consuming behavior. The brain balances this with a process called 'sensory-enhanced satiety', which makes foods that provide a stronger sensation of sweetness better at reducing hunger and further eating. High-energy food was scarce for most of human evolution, but over the past century sugar has become readily available in our diet leading to an increase in obesity. Last year, a study in fruit flies reported that a sugary diet reduces the sensitivity to sweet flavors, which leads to overeating and weight gain. It appears that this sensitivity is linked to the effectiveness of sensory-enhanced satiety. However, the mechanism linking diets high in sugar and overeating is still poorly understood. One hypothesis is that fruit flies estimate the energy content of food based on the degree of dopamine released in response to the sugar. May et al. compared the responses of neurons in fruit flies fed a normal diet to those in flies fed a diet high in sugar. As expected, both groups activated the neurons involved in the dopamine reward response when they tasted sugar. However, when the flies were on a sugar-heavy diet, these neurons were less active. This was because the neurons responsible for tasting sweetness were activated less in flies fed a high-sugar diet, leading to a lowered response by the neurons that produce dopamine. The flies in these experiments were genetically engineered so that the dopamine-producing neurons could be artificially activated in response to light, a technique called optogenetics. When May et al. applied this technique to the flies on a sugar-heavy diet, they were able to stop these flies from overeating. These findings provide further evidence to support the idea that a sugary diet reduces the brain's sensitivity to overeating. Given the significant healthcare cost of obesity to society, this improved understanding could help public health initiatives focusing on manufacturing food that is lower in sugar.


Dietary Sugars/administration & dosage , Dopaminergic Neurons , Drosophila melanogaster/physiology , Sucrose/metabolism , Taste Perception , Animals , Animals, Genetically Modified/physiology , Male
11.
Nat Commun ; 10(1): 4052, 2019 09 06.
Article En | MEDLINE | ID: mdl-31492856

Metabolites are active controllers of cellular physiology, but their role in complex behaviors is less clear. Here we report metabolic changes that occur during the transition between hunger and satiety in Drosophila melanogaster. To analyze these data in the context of fruit fly metabolic networks, we developed Flyscape, an open-access tool. We show that in response to eating, metabolic profiles change in quick, but distinct ways in the heads and bodies. Consumption of a high sugar diet dulls the metabolic and behavioral differences between the fasted and fed state, and reshapes the way nutrients are utilized upon eating. Specifically, we found that high dietary sugar increases TCA cycle activity, alters neurochemicals, and depletes 1-carbon metabolism and brain health metabolites N-acetyl-aspartate and kynurenine. Together, our work identifies the metabolic transitions that occur during hunger and satiation, and provides a platform to study the role of metabolites and diet in complex behavior.


Drosophila melanogaster/physiology , Hunger/physiology , Metabolic Networks and Pathways/physiology , Metabolome/physiology , Satiation/physiology , Animals , Brain/metabolism , Brain/physiology , Diet , Drosophila melanogaster/metabolism , Eating/physiology , Fasting/physiology , Humans , Metabolomics/methods
12.
Cell Rep ; 27(6): 1675-1685.e7, 2019 05 07.
Article En | MEDLINE | ID: mdl-31067455

Recent studies find that sugar tastes less intense to humans with obesity, but whether this sensory change is a cause or a consequence of obesity is unclear. To tackle this question, we study the effects of a high sugar diet on sweet taste sensation and feeding behavior in Drosophila melanogaster. On this diet, fruit flies have lower taste responses to sweet stimuli, overconsume food, and develop obesity. Excess dietary sugar, but not obesity or dietary sweetness alone, caused taste deficits and overeating via the cell-autonomous action of the sugar sensor O-linked N-Acetylglucosamine (O-GlcNAc) transferase (OGT) in the sweet-sensing neurons. Correcting taste deficits by manipulating the excitability of the sweet gustatory neurons or the levels of OGT protected animals from diet-induced obesity. Our work demonstrates that the reshaping of sweet taste sensation by excess dietary sugar drives obesity and highlights the role of glucose metabolism in neural activity and behavior.


Dietary Sugars/pharmacology , Drosophila melanogaster/physiology , Feeding Behavior/drug effects , Taste/drug effects , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/drug effects , Neurons/drug effects , Obesity/pathology , Synapses/drug effects , Synapses/physiology
14.
Curr Biol ; 26(15): 1965-1974, 2016 08 08.
Article En | MEDLINE | ID: mdl-27397890

Hunger is a powerful drive that stimulates food intake. Yet, the mechanism that determines how the energy deficits that result in hunger are represented in the brain and promote feeding is not well understood. We previously described SLC5A11-a sodium/solute co-transporter-like-(or cupcake) in Drosophila melanogaster, which is required for the fly to select a nutritive sugar over a sweeter nonnutritive sugar after periods of food deprivation. SLC5A11 acts on approximately 12 pairs of ellipsoid body (EB) R4 neurons to trigger the selection of nutritive sugars, but the underlying mechanism is not understood. Here, we report that the excitability of SLC5A11-expressing EB R4 neurons increases dramatically during starvation and that this increase is abolished in the SLC5A11 mutation. Artificial activation of SLC5A11-expresssing neurons is sufficient to promote feeding and hunger-driven behaviors; silencing these neurons has the opposite effect. Notably, SLC5A11 transcript levels in the brain increase significantly when flies are starved and decrease shortly after starved flies are refed. Furthermore, expression of SLC5A11 is sufficient for promoting hunger-driven behaviors and enhancing the excitability of SLC5A11-expressing neurons. SLC5A11 inhibits the function of the Drosophila KCNQ potassium channel in a heterologous expression system. Accordingly, a knockdown of dKCNQ expression in SLC5A11-expressing neurons produces hunger-driven behaviors even in fed flies, mimicking the overexpression of SLC5A11. We propose that starvation increases SLC5A11 expression, which enhances the excitability of SLC5A11-expressing neurons by suppressing dKCNQ channels, thereby conferring the hunger state.


Drosophila Proteins/genetics , Drosophila melanogaster/physiology , Food Deprivation , Hunger , Potassium Channels/genetics , Sodium-Glucose Transport Proteins/genetics , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Male , Neurons/metabolism , Potassium Channels/metabolism , Sodium-Glucose Transport Proteins/metabolism
15.
J Chromatogr A ; 1446: 78-90, 2016 May 13.
Article En | MEDLINE | ID: mdl-27083258

Widely targeted metabolomic assays are useful because they provide quantitative data on large groups of related compounds. We report a high performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) method that utilizes benzoyl chloride labeling for 70 neurologically relevant compounds, including catecholamines, indoleamines, amino acids, polyamines, trace amines, antioxidants, energy compounds, and their metabolites. The method includes neurotransmitters and metabolites found in both vertebrates and insects. This method was applied to analyze microdialysate from rats, human cerebrospinal fluid, human serum, fly tissue homogenate, and fly hemolymph, demonstrating its broad versatility for multiple physiological contexts and model systems. Limits of detection for most assayed compounds were below 10nM, relative standard deviations were below 10%, and carryover was less than 5% for 70 compounds separated in 20min, with a total analysis time of 33min. This broadly applicable method provides robust monitoring of multiple analytes, utilizes small sample sizes, and can be applied to diverse matrices. The assay will be of value for evaluating normal physiological changes in metabolism in neurochemical systems. The results demonstrate the utility of benzoyl chloride labeling with HPLC-MS/MS for widely targeted metabolomics assays.


Benzoates/chemistry , Metabolome , Neurotransmitter Agents/analysis , Amino Acids/analysis , Amino Acids/cerebrospinal fluid , Animals , Catecholamines/analysis , Catecholamines/blood , Catecholamines/cerebrospinal fluid , Chromatography, High Pressure Liquid/methods , Drosophila , Hemolymph/chemistry , Humans , Metabolomics , Neurotransmitter Agents/blood , Neurotransmitter Agents/cerebrospinal fluid , Rats , Rats, Sprague-Dawley , Species Specificity , Tandem Mass Spectrometry/methods
16.
Neuron ; 87(1): 139-51, 2015 Jul 01.
Article En | MEDLINE | ID: mdl-26074004

Animals can detect and consume nutritive sugars without the influence of taste. However, the identity of the taste-independent nutrient sensor and the mechanism by which animals respond to the nutritional value of sugar are unclear. Here, we report that six neurosecretory cells in the Drosophila brain that produce Diuretic hormone 44 (Dh44), a homolog of the mammalian corticotropin-releasing hormone (CRH), were specifically activated by nutritive sugars. Flies in which the activity of these neurons or the expression of Dh44 was disrupted failed to select nutritive sugars. Manipulation of the function of Dh44 receptors had a similar effect. Notably, artificial activation of Dh44 receptor-1 neurons resulted in proboscis extensions and frequent episodes of excretion. Conversely, reduced Dh44 activity led to decreased excretion. Together, these actions facilitate ingestion and digestion of nutritive foods. We propose that the Dh44 system directs the detection and consumption of nutritive sugars through a positive feedback loop.


Brain/metabolism , Drosophila Proteins/metabolism , Feeding Behavior/physiology , Insect Hormones/metabolism , Neurons/metabolism , Nutritive Sweeteners/metabolism , Animals , Drosophila , Drosophila Proteins/drug effects , Feedback, Sensory , Fructose/pharmacology , Glucose/pharmacology , Neurosecretion/drug effects , Nutritive Sweeteners/pharmacology , Receptors, Cell Surface/drug effects , Receptors, Cell Surface/metabolism , Trehalose/pharmacology
17.
Nat Neurosci ; 16(5): 526-8, 2013 May.
Article En | MEDLINE | ID: mdl-23542692

Animals can determine the nutritional value of sugar without the influence of taste. We examined a Drosophila mutant that is insensitive to the nutritional value of sugars, responding only to the concentration (that is, sweetness). The affected gene encodes a sodium/solute co-transporter-like protein, designated SLC5A11 (or cupcake), which is structurally similar to mammalian sodium/glucose co-transporters that transport sugar across the intestinal and renal lumen. However, SLC5A11 was prominently expressed in 10-13 pairs of R4 neurons of the ellipsoid body in the brain and functioned in these neurons for selecting appropriate foods.


Brain/metabolism , Choice Behavior/physiology , Drosophila Proteins/metabolism , Drosophila/physiology , Food Preferences/physiology , Sodium-Glucose Transport Proteins/metabolism , Taste/genetics , Analysis of Variance , Animals , Animals, Genetically Modified , Base Sequence , Brain/cytology , CD8 Antigens/genetics , Dose-Response Relationship, Drug , Drosophila Proteins/genetics , Food Deprivation/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glucose/administration & dosage , Glycogen/metabolism , Green Fluorescent Proteins/genetics , Hemolymph/metabolism , Humans , Mutation/genetics , Neurons/metabolism , RNA Interference , Receptors, Cell Surface/genetics , Sodium-Glucose Transport Proteins/genetics , Transcription Factors/genetics
18.
Proc Natl Acad Sci U S A ; 108(28): 11644-9, 2011 Jul 12.
Article En | MEDLINE | ID: mdl-21709242

Feeding behavior is influenced primarily by two factors: nutritional needs and food palatability. However, the role of food deprivation and metabolic needs in the selection of appropriate food is poorly understood. Here, we show that the fruit fly, Drosophila melanogaster, selects calorie-rich foods following prolonged food deprivation in the absence of taste-receptor signaling. Flies mutant for the sugar receptors Gr5a and Gr64a cannot detect the taste of sugar, but still consumed sugar over plain agar after 15 h of starvation. Similarly, pox-neuro mutants that are insensitive to the taste of sugar preferentially consumed sugar over plain agar upon starvation. Moreover, when given a choice between metabolizable sugar (sucrose or D-glucose) and nonmetabolizable (zero-calorie) sugar (sucralose or L-glucose), starved Gr5a; Gr64a double mutants preferred metabolizable sugars. These findings suggest the existence of a taste-independent metabolic sensor that functions in food selection. The preference for calorie-rich food correlates with a decrease in the two main hemolymph sugars, trehalose and glucose, and in glycogen stores, indicating that this sensor is triggered when the internal energy sources are depleted. Thus, the need to replenish depleted energy stores during periods of starvation may be met through the activity of a taste-independent metabolic sensing pathway.


Drosophila melanogaster/physiology , Animals , Carbohydrate Metabolism , Dietary Sucrose , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Drosophila melanogaster/genetics , Energy Intake , Feeding Behavior/physiology , Food Deprivation/physiology , Food Preferences/physiology , Genes, Insect , Hemolymph/metabolism , Male , Mutation , Receptors, Cell Surface/genetics , Receptors, Cell Surface/physiology , Starvation/genetics , Starvation/physiopathology , Sucrose , Taste/physiology
19.
Curr Biol ; 20(13): 1209-15, 2010 Jul 13.
Article En | MEDLINE | ID: mdl-20541409

Neural systems controlling the vital functions of sleep and feeding in mammals are tightly interconnected: sleep deprivation promotes feeding, whereas starvation suppresses sleep. Here we show that starvation in Drosophila potently suppresses sleep, suggesting that these two homeostatically regulated behaviors are also integrated in flies. The sleep-suppressing effect of starvation is independent of the mushroom bodies, a previously identified sleep locus in the fly brain, and therefore is regulated by distinct neural circuitry. The circadian clock genes Clock (Clk) and cycle (cyc) are critical for proper sleep suppression during starvation. However, the sleep suppression is independent of light cues and of circadian rhythms as shown by the fact that starved period mutants sleep like wild-type flies. By selectively targeting subpopulations of Clk-expressing neurons, we localize the observed sleep phenotype to the dorsally located circadian neurons. These findings show that Clk and cyc act during starvation to modulate the conflict of whether flies sleep or search for food.


ARNTL Transcription Factors/physiology , CLOCK Proteins/physiology , Drosophila Proteins/physiology , Drosophila/physiology , Sleep Deprivation , Animals , Feeding Behavior , Female , Male , Sleep
20.
PLoS One ; 4(7): e6182, 2009 Jul 08.
Article En | MEDLINE | ID: mdl-19584931

Heterochromatin formation plays an important role in gene regulation and the maintenance of genome integrity. Here we present results from a study of the D. melanogaster gene vig, encoding an RNAi complex component and its homolog vig2 (CG11844) that support their involvement in heterochromatin formation and/or maintenance. Protein null mutations vig(EP812) and vig2(PL470) act as modifiers of Position Effect Variegation (PEV). VIG and Vig2 are present in polytene chromosomes and partially overlap with HP1. Quantitative immunoblots show depletion of HP1 and HP2 (large isoform) in isolated nuclei from the vig(EP812) mutant. The vig2(PL470) mutant strain demonstrates a decreased level of H3K9me2. Pull-down experiments using antibodies specific to HP1 recovered both VIG and Vig2. The association between HP1 and both VIG and Vig2 proteins depends on an RNA component. The above data and the developmental profiles of the two genes suggest that Vig2 may be involved in heterochromatin targeting and establishment early in development, while VIG may have a role in stabilizing HP1/HP2 chromatin binding during later stages.


Drosophila Proteins/physiology , Drosophila melanogaster/physiology , Heterochromatin/metabolism , RNA-Induced Silencing Complex/physiology , Animals , Base Sequence , Blotting, Western , DNA Primers , Gene Expression Profiling , Gene Expression Regulation, Developmental , Immunoprecipitation , Reverse Transcriptase Polymerase Chain Reaction
...