Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.994
Filter
1.
Proc Biol Sci ; 291(2023): 20240518, 2024 May.
Article in English | MEDLINE | ID: mdl-38747703

ABSTRACT

Drosophila remains a pre-eminent insect model system for host-virus interaction, but the host range and fitness consequences of the drosophilid virome are poorly understood. Metagenomic studies have reported approximately 200 viruses associated with Drosophilidae, but few isolates are available to characterize the Drosophila immune response, and most characterization has relied on injection and systemic infection. Here, we use a more natural infection route to characterize the fitness effects of infection and to study a wider range of viruses. We exposed laboratory Drosophila melanogaster to 23 naturally occurring viruses from wild-collected drosophilids. We recorded transmission rates along with two components of female fitness: survival and the lifetime number of adult offspring produced. Nine different viruses transmitted during contact with laboratory D. melanogaster, although for the majority, rates of transmission were less than 20%. Five virus infections led to a significant decrease in lifespan (D. melanogaster Nora virus, D. immigrans Nora virus, Muthill virus, galbut virus and Prestney Burn virus), and three led to a reduction in the total number of offspring. Our findings demonstrate the utility of the Drosophila model for community-level studies of host-virus interactions, and suggest that viral infection could be a substantial fitness burden on wild flies.


Subject(s)
Drosophila melanogaster , Longevity , Animals , Drosophila melanogaster/virology , Drosophila melanogaster/physiology , Female , Insect Viruses/physiology , Host-Pathogen Interactions
2.
Curr Biol ; 34(10): 2186-2199.e3, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38723636

ABSTRACT

Animals exhibit rhythmic patterns of behavior that are shaped by an internal circadian clock and the external environment. Although light intensity varies across the day, there are particularly robust differences at twilight (dawn/dusk). These periods are also associated with major changes in behavioral states, such as the transition from arousal to sleep. However, the neural mechanisms by which time and environmental conditions promote these behavioral transitions are poorly defined. Here, we show that the E1 subclass of Drosophila evening clock neurons promotes the transition from arousal to sleep at dusk. We first demonstrate that the cell-autonomous clocks of E2 neurons primarily drive and adjust the phase of evening anticipation, the canonical behavior associated with "evening" clock neurons. We next show that conditionally silencing E1 neurons causes a significant delay in sleep onset after dusk. However, rather than simply promoting sleep, activating E1 neurons produces time- and light-dependent effects on behavior. Activation of E1 neurons has no effect early in the day but then triggers arousal before dusk and induces sleep after dusk. Strikingly, these activation-induced phenotypes depend on the presence of light during the day. Despite their influence on behavior around dusk, in vivo voltage imaging of E1 neurons reveals that their spiking rate and pattern do not significantly change throughout the day. Moreover, E1-specific clock ablation has no effect on arousal or sleep. Thus, we suggest that, rather than specifying "evening" time, E1 neurons act, in concert with other rhythmic neurons, to promote behavioral transitions at dusk.


Subject(s)
Arousal , Circadian Clocks , Circadian Rhythm , Drosophila melanogaster , Neurons , Sleep , Animals , Sleep/physiology , Arousal/physiology , Neurons/physiology , Drosophila melanogaster/physiology , Circadian Clocks/physiology , Circadian Rhythm/physiology , Drosophila Proteins/metabolism , Drosophila Proteins/genetics
3.
Proc Natl Acad Sci U S A ; 121(21): e2316799121, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38753511

ABSTRACT

The mammalian brain implements sophisticated sensory processing algorithms along multilayered ("deep") neural networks. Strategies that insects use to meet similar computational demands, while relying on smaller nervous systems with shallow architectures, remain elusive. Using Drosophila as a model, we uncover the algorithmic role of odor preprocessing by a shallow network of compartmentalized olfactory receptor neurons. Each compartment operates as a ratiometric unit for specific odor-mixtures. This computation arises from a simple mechanism: electrical coupling between two differently sized neurons. We demonstrate that downstream synaptic connectivity is shaped to optimally leverage amplification of a hedonic value signal in the periphery. Furthermore, peripheral preprocessing is shown to markedly improve novel odor classification in a higher brain center. Together, our work highlights a far-reaching functional role of the sensory periphery for downstream processing. By elucidating the implementation of powerful computations by a shallow network, we provide insights into general principles of efficient sensory processing algorithms.


Subject(s)
Odorants , Olfactory Receptor Neurons , Smell , Animals , Odorants/analysis , Olfactory Receptor Neurons/physiology , Smell/physiology , Drosophila melanogaster/physiology , Algorithms , Drosophila/physiology , Olfactory Pathways/physiology , Models, Neurological , Nerve Net/physiology
4.
Sci Rep ; 14(1): 11285, 2024 05 17.
Article in English | MEDLINE | ID: mdl-38760389

ABSTRACT

Feeding behaviors are determined by two main factors. One is the internal state, such as hunger or previous experiences; the other is external factors, such as sensory stimulation. During starvation, animals must balance food-seeking behavior with energy conservation. The fruit fly, Drosophila melanogaster, serves as a useful model for studying food selectivity and various behaviors related to food intake. However, few studies have directly connected food selectivity with other behaviors, such as locomotor activity and sleep. In this study, we report that flies exhibited a preference for specific positions and spent more time in the proximity of sweet sugars, such as sucrose and sucralose, but not non-sweet and nutritious sugars like xylitol and sorbitol. On the other hand, prolonged exposure to sorbitol increased the staying time of flies in the proximity of sorbitol. Additionally, after starvation, flies immediately exhibited a position preference in the proximity of sorbitol. These findings suggest that flies prefer the proximity of sweet food, and starvation alters their preference for nutritious food, which may be beneficial for their survival.


Subject(s)
Drosophila melanogaster , Feeding Behavior , Sugars , Animals , Drosophila melanogaster/physiology , Feeding Behavior/physiology , Starvation , Food Preferences/physiology , Sorbitol/pharmacology , Sucrose/metabolism
6.
Elife ; 122024 May 03.
Article in English | MEDLINE | ID: mdl-38700995

ABSTRACT

Adenine phosphoribosyltransferase (APRT) and hypoxanthine-guanine phosphoribosyltransferase (HGPRT) are two structurally related enzymes involved in purine recycling in humans. Inherited mutations that suppress HGPRT activity are associated with Lesch-Nyhan disease (LND), a rare X-linked metabolic and neurological disorder in children, characterized by hyperuricemia, dystonia, and compulsive self-injury. To date, no treatment is available for these neurological defects and no animal model recapitulates all symptoms of LND patients. Here, we studied LND-related mechanisms in the fruit fly. By combining enzymatic assays and phylogenetic analysis, we confirm that no HGPRT activity is expressed in Drosophila melanogaster, making the APRT homolog (Aprt) the only purine-recycling enzyme in this organism. Whereas APRT deficiency does not trigger neurological defects in humans, we observed that Drosophila Aprt mutants show both metabolic and neurobehavioral disturbances, including increased uric acid levels, locomotor impairments, sleep alterations, seizure-like behavior, reduced lifespan, and reduction of adenosine signaling and content. Locomotor defects could be rescued by Aprt re-expression in neurons and reproduced by knocking down Aprt selectively in the protocerebral anterior medial (PAM) dopaminergic neurons, the mushroom bodies, or glia subsets. Ingestion of allopurinol rescued uric acid levels in Aprt-deficient mutants but not neurological defects, as is the case in LND patients, while feeding adenosine or N6-methyladenosine (m6A) during development fully rescued the epileptic behavior. Intriguingly, pan-neuronal expression of an LND-associated mutant form of human HGPRT (I42T), but not the wild-type enzyme, resulted in early locomotor defects and seizure in flies, similar to Aprt deficiency. Overall, our results suggest that Drosophila could be used in different ways to better understand LND and seek a cure for this dramatic disease.


Subject(s)
Drosophila melanogaster , Lesch-Nyhan Syndrome , Animals , Drosophila melanogaster/physiology , Drosophila melanogaster/genetics , Lesch-Nyhan Syndrome/genetics , Lesch-Nyhan Syndrome/metabolism , Purines/metabolism , Disease Models, Animal , Behavior, Animal , Hypoxanthine Phosphoribosyltransferase/genetics , Hypoxanthine Phosphoribosyltransferase/metabolism , Hypoxanthine Phosphoribosyltransferase/deficiency , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Locomotion
7.
Nat Commun ; 15(1): 3685, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38693116

ABSTRACT

Sleep, locomotor and social activities are essential animal behaviors, but their reciprocal relationships and underlying mechanisms remain poorly understood. Here, we elicit information from a cutting-edge large-language model (LLM), generative pre-trained transformer (GPT) 3.5, which interprets 10.2-13.8% of Drosophila genes known to regulate the 3 behaviors. We develop an instrument for simultaneous video tracking of multiple moving objects, and conduct a genome-wide screen. We have identified 758 fly genes that regulate sleep and activities, including mre11 which regulates sleep only in the presence of conspecifics, and NELF-B which regulates sleep regardless of whether conspecifics are present. Based on LLM-reasoning, an educated signal web is modeled for understanding of potential relationships between its components, presenting comprehensive molecular signatures that control sleep, locomotor and social activities. This LLM-aided strategy may also be helpful for addressing other complex scientific questions.


Subject(s)
Behavior, Animal , Drosophila melanogaster , Locomotion , Sleep , Animals , Sleep/physiology , Sleep/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Locomotion/physiology , Locomotion/genetics , Behavior, Animal/physiology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Social Behavior , Male
9.
Arch Insect Biochem Physiol ; 116(1): e22116, 2024 May.
Article in English | MEDLINE | ID: mdl-38739777

ABSTRACT

Studies on antiaging remedies in insect models sometimes show discrepancies in results. These discrepancies could be explained by different responses of short- and long-lived strains on the antiaging remedies. The purpose of the study was to test whether life-prolonging effects of alpha-ketoglutarate (AKG), observed in nematodes and fruit flies, would be reproduced in long-lived Drosophila melanogaster flies. Lifespan was assayed in flies kept in demographic cages. Fecundity, proportion of flies capable of negative geotaxis, starvation resistance, time of heat coma onset, levels of triacyglycerols, body glucose, glycogen, activities of glutamate dehydrogenase, catalase, glutathione-S-transferase, hexokinase, phosphofructokinase, pyruvate kinase, lactate, and glutamate dehydrogenases were assessed. Dietary AKG did not affect fly lifespan on the diet with 5% yeast and 5% sucrose (5Y:5S) and on the diet with 9% yeast and 1% sucrose (9Y:1S), but increased lifespan on the low-protein diet (1Y:9S). Twenty-five-day-old female flies fed a 5Y:5S diet with 10 mM AKG for 3 weeks, did not differ from the control group (without AKG) in climbing activity, resistance to heat stress, and starvation. The levels of glucose and glycogen were unaffected but the levels of triacylglycerols were lower in AKG-fed female flies. No differences in activities of glycolytic enzymes, NADPH-producing enzymes, glutamate dehydrogenase, oxygen consumption, and levels of oxidative stress markers were observed between the control and AKG-fed flies. However, AKG-fed flies had lower activities of catalase and glutathione-S-transferase. These results suggest that potential antiaging remedies, such as AKG, may not extend lifespan in long-living organisms despite influencing several metabolic parameters.


Subject(s)
Drosophila melanogaster , Ketoglutaric Acids , Longevity , Animals , Drosophila melanogaster/physiology , Drosophila melanogaster/drug effects , Drosophila melanogaster/metabolism , Longevity/drug effects , Ketoglutaric Acids/pharmacology , Ketoglutaric Acids/metabolism , Female , Male , Dietary Supplements
10.
PLoS Comput Biol ; 20(4): e1012029, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38648221

ABSTRACT

The circadian clock is an evolutionarily-conserved molecular oscillator that enables species to anticipate rhythmic changes in their environment. At a molecular level, the core clock genes induce circadian oscillations in thousands of genes in a tissue-specific manner, orchestrating myriad biological processes. While previous studies have investigated how the core clock circuit responds to environmental perturbations such as temperature, the downstream effects of such perturbations on circadian regulation remain poorly understood. By analyzing bulk-RNA sequencing of Drosophila fat bodies harvested from flies subjected to different environmental conditions, we demonstrate a highly condition-specific circadian transcriptome: genes are cycling in a temperature-specific manner, and the distributions of their phases also differ between the two conditions. Further employing a reference-based gene regulatory network (Reactome), we find evidence of increased gene-gene coordination at low temperatures and synchronization of rhythmic genes that are network neighbors. We report that the phase differences between cycling genes increase as a function of geodesic distance in the low temperature condition, suggesting increased coordination of cycling on the gene regulatory network. Our results suggest a potential mechanism whereby the circadian clock mediates the fly's response to seasonal changes in temperature.


Subject(s)
Circadian Clocks , Circadian Rhythm , Gene Expression Regulation , Gene Regulatory Networks , Temperature , Animals , Circadian Rhythm/genetics , Circadian Rhythm/physiology , Gene Regulatory Networks/genetics , Circadian Clocks/genetics , Circadian Clocks/physiology , Gene Expression Regulation/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Drosophila/genetics , Drosophila/physiology , Transcriptome/genetics , Computational Biology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Transcription, Genetic/genetics
11.
Curr Biol ; 34(10): 2066-2076.e3, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38657610

ABSTRACT

Flies groom in response to competing mechanosensory cues in an anterior-to-posterior order using specific legs. From behavior screens, we identified a pair of cholinergic command-like neurons, Mago-no-Te (MGT), whose optogenetic activation elicits thoracic grooming by the back legs. Thoracic grooming is typically composed of body sweeps and leg rubs in alternation, but clonal analysis coupled with amputation experiments revealed that MGT activation only commands the body sweeps: initiation of leg rubbing requires contact between the leg and thorax. With new electron microscopy (EM) connectome data for the ventral nerve cord (VNC), we uncovered a circuit-based explanation for why stimulation of posterior thoracic mechanosensory bristles initiates cleaning by the back legs. Our previous work showed that flies weigh mechanosensory inputs across the body to select which part to groom, but we did not know why the thorax was always cleaned last. Here, the connectome for the VNC enabled us to identify a pair of GABAergic inhibitory neurons, UMGT1, that receives diverse sensory inputs and synapses onto both MGT and components of its downstream circuits. Optogenetic activation of UMGT1 suppresses thoracic cleaning, representing a mechanism by which mechanosensory stimuli on other body parts could take precedence in the grooming hierarchy. We also anatomically mapped the pre-motor circuit downstream of MGT, including inhibitory feedback connections that may enable rhythmicity and coordination of limb movement during thoracic grooming. The combination of behavioral screens and connectome analysis allowed us to identify a neural circuit connecting sensory-to-motor neurons that contributes to thoracic grooming.


Subject(s)
Drosophila melanogaster , Grooming , Animals , Grooming/physiology , Drosophila melanogaster/physiology , Extremities/physiology , Connectome , Optogenetics , Mechanoreceptors/physiology , Mechanotransduction, Cellular
12.
J Exp Biol ; 227(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38680096

ABSTRACT

The metabolic responses of insects to high temperatures have been linked to their mitochondrial substrate oxidation capacity. However, the mechanism behind this mitochondrial flexibility is not well understood. Here, we used three insect species with different thermal tolerances (the honey bee, Apis mellifera; the fruit fly, Drosophila melanogaster; and the potato beetle, Leptinotarsa decemlineata) to characterize the thermal sensitivity of different metabolic enzymes. Specifically, we measured activity of enzymes involved in glycolysis (hexokinase, HK; pyruvate kinase, PK; and lactate dehydrogenase, LDH), pyruvate oxidation and the tricarboxylic acid cycle (pyruvate dehydrogenase, PDH; citrate synthase, CS; malate dehydrogenase, MDH; and aspartate aminotransferase, AAT), and the electron transport system (Complex I, CI; Complex II, CII; mitochondrial glycerol-3-phosphate dehydrogenase, mG3PDH; proline dehydrogenase, ProDH; and Complex IV, CIV), as well as that of ATP synthase (CV) at 18, 24, 30, 36, 42 and 45°C. Our results show that at high temperature, all three species have significantly increased activity of enzymes linked to FADH2 oxidation, specifically CII and mG3PDH. In fruit flies and honey bees, this coincides with a significant decrease of PDH and CS activity, respectively, that would limit NADH production. This is in line with the switch from NADH-linked substrates to FADH2-linked substrates previously observed with mitochondrial oxygen consumption. Thus, we demonstrate that even though the three insect species have a different metabolic regulation, a similar response to high temperature involving CII and mG3PDH is observed, denoting the importance of these proteins for thermal tolerance in insects.


Subject(s)
Coleoptera , Drosophila melanogaster , Energy Metabolism , Animals , Bees/enzymology , Bees/metabolism , Bees/physiology , Drosophila melanogaster/enzymology , Drosophila melanogaster/metabolism , Drosophila melanogaster/physiology , Coleoptera/enzymology , Coleoptera/metabolism , Coleoptera/physiology , Hot Temperature
13.
PLoS One ; 19(4): e0301999, 2024.
Article in English | MEDLINE | ID: mdl-38635686

ABSTRACT

To study how the nervous system processes visual information, experimenters must record neural activity while delivering visual stimuli in a controlled fashion. In animals with a nearly panoramic field of view, such as flies, precise stimulation of the entire visual field is challenging. We describe a projector-based device for stimulation of the insect visual system under a microscope. The device is based on a bowl-shaped screen that provides a wide and nearly distortion-free field of view. It is compact, cheap, easy to assemble, and easy to operate using the included open-source software for stimulus generation. We validate the virtual reality system technically and demonstrate its capabilities in a series of experiments at two levels: the cellular, by measuring the membrane potential responses of visual interneurons; and the organismal, by recording optomotor and fixation behavior of Drosophila melanogaster in tethered flight. Our experiments reveal the importance of stimulating the visual system of an insect with a wide field of view, and we provide a simple solution to do so.


Subject(s)
Drosophila melanogaster , Visual Fields , Animals , Drosophila melanogaster/physiology , Photic Stimulation , Software , Interneurons , Flight, Animal/physiology , Visual Perception/physiology
14.
Elife ; 122024 Apr 24.
Article in English | MEDLINE | ID: mdl-38655926

ABSTRACT

The brain regulates food intake in response to internal energy demands and food availability. However, can internal energy storage influence the type of memory that is formed? We show that the duration of starvation determines whether Drosophila melanogaster forms appetitive short-term or longer-lasting intermediate memories. The internal glycogen storage in the muscles and adipose tissue influences how intensely sucrose-associated information is stored. Insulin-like signaling in octopaminergic reward neurons integrates internal energy storage into memory formation. Octopamine, in turn, suppresses the formation of long-term memory. Octopamine is not required for short-term memory because octopamine-deficient mutants can form appetitive short-term memory for sucrose and to other nutrients depending on the internal energy status. The reduced positive reinforcing effect of sucrose at high internal glycogen levels, combined with the increased stability of food-related memories due to prolonged periods of starvation, could lead to increased food intake.


Deciding what and how much to eat is a complex biological process which involves balancing many types of information such as the levels of internal energy storage, the amount of food previously available in the environment, the perceived value of certain food items, and how these are remembered. At the molecular level, food contains carbohydrates that are broken down to produce glucose, which is then delivered to cells under the control of a hormone called insulin. There, glucose molecules are either immediately used or stored as glycogen until needed. Insulin signalling is also known to interact with the brain's decision-making systems that control eating behaviors; however, how our brains balance food intake with energy storage is poorly understood. Berger et al. set out to investigate this question using fruit flies as an experimental model. These insects also produce insulin-like molecules which help to relay information about glycogen levels to the brain's decision-making system. In particular, these signals reach a population of neurons that produce a messenger known as octopamine similar to the human noradrenaline, which helps regulate how much the flies find consuming certain types of foods rewarding. Berger et al. were able to investigate the role of octopamine in helping to integrate information about internal and external resource levels, memory formation and the evaluation of different food types. When the insects were fed normally, increased glycogen levels led to foods rich in carbohydrates being rated as less rewarding by the decision-making cells, and therefore being consumed less. Memories related to food intake were also short-lived ­ in other words, long-term 'food memory' was suppressed, re-setting the whole system after every meal. In contrast, long periods of starvation in insects with high carbohydrates resources produced a stable, long-term memory of food and hunger which persisted even after the flies had fed again. This experience also changed their food rating system, with highly nutritious foods no longer being perceived as sufficiently rewarding. As a result, the flies overate. This study sheds new light on the mechanisms our bodies may use to maintain energy reserves when food is limited. The persistence of 'food memory' after long periods of starvation may also explain why losing weight is difficult, especially during restrictive diets. In the future, Berger et al. hope that this knowledge will contribute to better strategies for weight management.


Subject(s)
Drosophila melanogaster , Energy Metabolism , Octopamine , Animals , Drosophila melanogaster/physiology , Octopamine/metabolism , Memory/physiology , Glycogen/metabolism , Starvation , Sucrose/metabolism , Memory, Long-Term/physiology , Eating/physiology
15.
BMC Biol ; 22(1): 89, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38644510

ABSTRACT

BACKGROUND: Innate immune responses can be activated by pathogen-associated molecular patterns (PAMPs), danger signals released by damaged tissues, or the absence of self-molecules that inhibit immunity. As PAMPs are typically conserved across broad groups of pathogens but absent from the host, it is unclear whether they allow hosts to recognize parasites that are phylogenetically similar to themselves, such as parasitoid wasps infecting insects. RESULTS: Parasitoids must penetrate the cuticle of Drosophila larvae to inject their eggs. In line with previous results, we found that the danger signal of wounding triggers the differentiation of specialized immune cells called lamellocytes. However, using oil droplets to mimic infection by a parasitoid wasp egg, we found that this does not activate the melanization response. This aspect of the immune response also requires exposure to parasite molecules. The unidentified factor enhances the transcriptional response in hemocytes and induces a specific response in the fat body. CONCLUSIONS: We conclude that a combination of danger signals and the recognition of nonself molecules is required to activate Drosophila's immune response against parasitic insects.


Subject(s)
Hemocytes , Host-Parasite Interactions , Immunity, Innate , Wasps , Animals , Wasps/physiology , Host-Parasite Interactions/immunology , Hemocytes/immunology , Drosophila melanogaster/parasitology , Drosophila melanogaster/immunology , Drosophila melanogaster/physiology , Larva/immunology , Larva/parasitology , Drosophila/parasitology , Drosophila/immunology
16.
Nat Commun ; 15(1): 2834, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38565846

ABSTRACT

The circadian clock regulates animal physiological activities. How temperature reorganizes circadian-dependent physiological activities remains elusive. Here, using in-vivo two-photon imaging with the temperature control device, we investigated the response of the Drosophila central circadian circuit to temperature variation and identified that DN1as serves as the most sensitive temperature-sensing neurons. The circadian clock gate DN1a's diurnal temperature response. Trans-synaptic tracing, connectome analysis, and functional imaging data reveal that DN1as bidirectionally targets two circadian neuronal subsets: activity-related E cells and sleep-promoting DN3s. Specifically, behavioral data demonstrate that the DN1a-E cell circuit modulates the evening locomotion peak in response to cold temperature, while the DN1a-DN3 circuit controls the warm temperature-induced nocturnal sleep reduction. Our findings systematically and comprehensively illustrate how the central circadian circuit dynamically integrates temperature and light signals to effectively coordinate wakefulness and sleep at different times of the day, shedding light on the conserved neural mechanisms underlying temperature-regulated circadian physiology in animals.


Subject(s)
Circadian Clocks , Drosophila Proteins , Animals , Circadian Rhythm/physiology , Temperature , Sleep/physiology , Drosophila , Circadian Clocks/physiology , Drosophila Proteins/genetics , Drosophila melanogaster/physiology
17.
Curr Biol ; 34(7): R288-R291, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38593775

ABSTRACT

The development of sex-specific neural circuitry is critical for reproductive behaviors. A new study traces the developmental origin of female-specific neurons that underlie an adult mating behavior to larval neurons common to both sexes in Drosophila.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Male , Female , Larva , Drosophila/physiology , Neurons/physiology , Sexual Behavior, Animal/physiology , Drosophila melanogaster/physiology
18.
BMC Biol ; 22(1): 98, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38679694

ABSTRACT

BACKGROUND: The ability of animals to regenerate damaged tissue is a complex process that involves various cellular mechanisms. As animals age, they lose their regenerative abilities, making it essential to understand the underlying mechanisms that limit regenerative ability during aging. Drosophila melanogaster wing imaginal discs are epithelial structures that can regenerate after tissue injury. While significant research has focused on investigating regenerative responses during larval stages our comprehension of the regenerative potential of pupal wings and the underlying mechanisms contributing to the decline of regenerative responses remains limited. RESULTS: Here, we explore the temporal dynamics during pupal development of the proliferative response triggered by the induction of cell death, a typical regenerative response. Our results indicate that the apoptosis-induced proliferative response can continue until 34 h after puparium formation (APF), beyond this point cell death alone is not sufficient to induce a regenerative response. Under normal circumstances, cell proliferation ceases around 24 h APF. Interestingly, the failure of reinitiating the cell cycle beyond this time point is not attributed to an incapacity to activate the JNK pathway. Instead, our results suggest that the function of the ecdysone-responsive transcription factor E93 is involved in limiting the apoptosis-induced proliferative response during pupal development. CONCLUSIONS: Our study shows that apoptosis can prolong the proliferative period of cells in the wing during pupal development as late as 34 h APF, at least 10 h longer than during normal development. After this time point, the regenerative response is diminished, a process mediated in part by the ecdysone-responsive transcription factor E93.


Subject(s)
Apoptosis , Cell Proliferation , Drosophila Proteins , Drosophila melanogaster , Pupa , Regeneration , Transcription Factors , Wings, Animal , Animals , Wings, Animal/growth & development , Wings, Animal/physiology , Drosophila melanogaster/physiology , Drosophila melanogaster/growth & development , Pupa/growth & development , Pupa/physiology , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Regeneration/physiology
19.
Int J Mol Sci ; 25(8)2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38674068

ABSTRACT

Lifespan is a complex quantitative trait involving genetic and non-genetic factors as well as the peculiarities of ontogenesis. As with all quantitative traits, lifespan shows considerable variation within populations and between individuals. Drosophila, a favourite object of geneticists, has greatly advanced our understanding of how different forms of variability affect lifespan. This review considers the role of heritable genetic variability, phenotypic plasticity and stochastic variability in controlling lifespan in Drosophila melanogaster. We discuss the major historical milestones in the development of the genetic approach to study lifespan, the breeding of long-lived lines, advances in lifespan QTL mapping, the environmental factors that have the greatest influence on lifespan in laboratory maintained flies, and the mechanisms, by which individual development affects longevity. The interplay between approaches to study ageing and lifespan limitation will also be discussed. Particular attention will be paid to the interaction of different types of variability in the control of lifespan.


Subject(s)
Drosophila melanogaster , Longevity , Animals , Longevity/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Quantitative Trait Loci , Stochastic Processes , Genetic Variation , Gene-Environment Interaction , Aging/genetics , Aging/physiology , Environment , Phenotype
20.
Curr Biol ; 34(9): 1904-1917.e6, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38642548

ABSTRACT

Neurons have differential and fluctuating energy needs across distinct cellular compartments, shaped by brain electrochemical activity associated with cognition. In vitro studies show that mitochondria transport from soma to axons is key to maintaining neuronal energy homeostasis. Nevertheless, whether the spatial distribution of neuronal mitochondria is dynamically adjusted in vivo in an experience-dependent manner remains unknown. In Drosophila, associative long-term memory (LTM) formation is initiated by an early and persistent upregulation of mitochondrial pyruvate flux in the axonal compartment of neurons in the mushroom body (MB). Through behavior experiments, super-resolution analysis of mitochondria morphology in the neuronal soma and in vivo mitochondrial fluorescence recovery after photobleaching (FRAP) measurements in the axons, we show that LTM induction, contrary to shorter-lived memories, is sustained by the departure of some mitochondria from MB neuronal soma and increased mitochondrial dynamics in the axonal compartment. Accordingly, impairing mitochondrial dynamics abolished the increased pyruvate consumption, specifically after spaced training and in the MB axonal compartment, thereby preventing LTM formation. Our results thus promote reorganization of the mitochondrial network in neurons as an integral step in elaborating high-order cognitive processes.


Subject(s)
Axons , Drosophila Proteins , Drosophila melanogaster , Memory, Long-Term , Mitochondria , Mitochondrial Dynamics , Mushroom Bodies , Animals , Memory, Long-Term/physiology , Mitochondrial Dynamics/physiology , Axons/metabolism , Axons/physiology , Mushroom Bodies/physiology , Mushroom Bodies/metabolism , Drosophila melanogaster/physiology , Mitochondria/metabolism , Mitochondria/physiology , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Neurons/metabolism , Neurons/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...