Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(11): e2308067121, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38442160

RESUMO

Circadian clocks impose daily periodicities to behavior, physiology, and metabolism. This control is mediated by a central clock and by peripheral clocks, which are synchronized to provide the organism with a unified time through mechanisms that are not fully understood. Here, we characterized in Drosophila the cellular and molecular mechanisms involved in coupling the central clock and the peripheral clock located in the prothoracic gland (PG), which together control the circadian rhythm of emergence of adult flies. The time signal from central clock neurons is transmitted via small neuropeptide F (sNPF) to neurons that produce the neuropeptide Prothoracicotropic Hormone (PTTH), which is then translated into daily oscillations of Ca2+ concentration and PTTH levels. PTTH signaling is required at the end of metamorphosis and transmits time information to the PG through changes in the expression of the PTTH receptor tyrosine kinase (RTK), TORSO, and of ERK phosphorylation, a key component of PTTH transduction. In addition to PTTH, we demonstrate that signaling mediated by other RTKs contributes to the rhythmicity of emergence. Interestingly, the ligand to one of these receptors (Pvf2) plays an autocrine role in the PG, which may explain why both central brain and PG clocks are required for the circadian gating of emergence. Our findings show that the coupling between the central and the PG clock is unexpectedly complex and involves several RTKs that act in concert and could serve as a paradigm to understand how circadian clocks are coordinated.


Assuntos
Antígenos de Grupos Sanguíneos , Relógios Circadianos , Animais , Relógios Circadianos/genética , Drosophila , Transdução de Sinais , Receptores Proteína Tirosina Quinases/genética , Fosforilação , Fatores de Crescimento do Endotélio Vascular
2.
PLoS Genet ; 20(1): e1011054, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38236837

RESUMO

Living in dynamic environments such as the social domain, where interaction with others determines the reproductive success of individuals, requires the ability to recognize opportunities to obtain natural rewards and cope with challenges that are associated with achieving them. As such, actions that promote survival and reproduction are reinforced by the brain reward system, whereas coping with the challenges associated with obtaining these rewards is mediated by stress-response pathways, the activation of which can impair health and shorten lifespan. While much research has been devoted to understanding mechanisms underlying the way by which natural rewards are processed by the reward system, less attention has been given to the consequences of failure to obtain a desirable reward. As a model system to study the impact of failure to obtain a natural reward, we used the well-established courtship suppression paradigm in Drosophila melanogaster as means to induce repeated failures to obtain sexual reward in male flies. We discovered that beyond the known reduction in courtship actions caused by interaction with non-receptive females, repeated failures to mate induce a stress response characterized by persistent motivation to obtain the sexual reward, reduced male-male social interaction, and enhanced aggression. This frustrative-like state caused by the conflict between high motivation to obtain sexual reward and the inability to fulfill their mating drive impairs the capacity of rejected males to tolerate stressors such as starvation and oxidative stress. We further show that sensitivity to starvation and enhanced social arousal is mediated by the disinhibition of a small population of neurons that express receptors for the fly homologue of neuropeptide Y. Our findings demonstrate for the first time the existence of social stress in flies and offers a framework to study mechanisms underlying the crosstalk between reward, stress, and reproduction in a simple nervous system that is highly amenable to genetic manipulation.


Assuntos
Drosophila melanogaster , Neuropeptídeos , Comportamento Sexual Animal , Humanos , Animais , Feminino , Masculino , Drosophila melanogaster/genética , Comportamento Sexual Animal/fisiologia , Reprodução/genética , Recompensa , Neurônios/metabolismo
3.
PLoS Genet ; 17(3): e1009425, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33684132

RESUMO

Environmental factors challenge the physiological homeostasis in animals, thereby evoking stress responses. Various mechanisms have evolved to counter stress at the organism level, including regulation by neuropeptides. In recent years, much progress has been made on the mechanisms and neuropeptides that regulate responses to metabolic/nutritional stress, as well as those involved in countering osmotic and ionic stresses. Here, we identified a peptidergic pathway that links these types of regulatory functions. We uncover the neuropeptide Corazonin (Crz), previously implicated in responses to metabolic stress, as a neuroendocrine factor that inhibits the release of a diuretic hormone, CAPA, and thereby modulates the tolerance to osmotic and ionic stress. Both knockdown of Crz and acute injections of Crz peptide impact desiccation tolerance and recovery from chill-coma. Mapping of the Crz receptor (CrzR) expression identified three pairs of Capa-expressing neurons (Va neurons) in the ventral nerve cord that mediate these effects of Crz. We show that Crz acts to restore water/ion homeostasis by inhibiting release of CAPA neuropeptides via inhibition of cAMP production in Va neurons. Knockdown of CrzR in Va neurons affects CAPA signaling, and consequently increases tolerance for desiccation, ionic stress and starvation, but delays chill-coma recovery. Optogenetic activation of Va neurons stimulates excretion and simultaneous activation of Crz and CAPA-expressing neurons reduces this response, supporting the inhibitory action of Crz. Thus, Crz inhibits Va neurons to maintain osmotic and ionic homeostasis, which in turn affects stress tolerance. Earlier work demonstrated that systemic Crz signaling restores nutrient levels by promoting food search and feeding. Here we additionally propose that Crz signaling also ensures osmotic homeostasis by inhibiting release of CAPA neuropeptides and suppressing diuresis. Thus, Crz ameliorates stress-associated physiology through systemic modulation of both peptidergic neurosecretory cells and the fat body in Drosophila.


Assuntos
Drosophila/fisiologia , Redes e Vias Metabólicas , Sistemas Neurossecretores/metabolismo , Pressão Osmótica , Animais , AMP Cíclico/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Imunofluorescência , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Imuno-Histoquímica , Modelos Biológicos , Neurônios/metabolismo , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Transdução de Sinais , Estresse Fisiológico
4.
PLoS Genet ; 17(8): e1009724, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34398892

RESUMO

Feeding is essential for animal survival and reproduction and is regulated by both internal states and external stimuli. However, little is known about how internal states influence the perception of external sensory cues that regulate feeding behavior. Here, we investigated the neuronal and molecular mechanisms behind nutritional state-mediated regulation of gustatory perception in control of feeding behavior in the brown planthopper and Drosophila. We found that feeding increases the expression of the cholecystokinin-like peptide, sulfakinin (SK), and the activity of a set of SK-expressing neurons. Starvation elevates the transcription of the sugar receptor Gr64f and SK negatively regulates the expression of Gr64f in both insects. Interestingly, we found that one of the two known SK receptors, CCKLR-17D3, is expressed by some of Gr64f-expressing neurons in the proboscis and proleg tarsi. Thus, we have identified SK as a neuropeptide signal in a neuronal circuitry that responds to food intake, and regulates feeding behavior by diminishing gustatory receptor gene expression and activity of sweet sensing GRNs. Our findings demonstrate one nutritional state-dependent pathway that modulates sweet perception and thereby feeding behavior, but our experiments cannot exclude further parallel pathways. Importantly, we show that the underlying mechanisms are conserved in the two distantly related insect species.


Assuntos
Comportamento Alimentar/fisiologia , Percepção Gustatória/genética , Animais , Encéfalo/metabolismo , Metabolismo dos Carboidratos/fisiologia , Carboidratos/fisiologia , Colecistocinina/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/fisiologia , Comportamento Alimentar/psicologia , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Hemípteros/genética , Hemípteros/fisiologia , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Receptores de Superfície Celular/genética , Inanição/metabolismo , Açúcares/metabolismo , Paladar/fisiologia , Percepção Gustatória/fisiologia
5.
Cell Mol Life Sci ; 79(3): 188, 2022 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-35286508

RESUMO

Neuropeptides are the most diverse messenger molecules in metazoans and are involved in regulation of daily physiology and a wide array of behaviors. Some neuropeptides and their cognate receptors are structurally and functionally well conserved over evolution in bilaterian animals. Among these are peptides related to gastrin and cholecystokinin (CCK). In mammals, CCK is produced by intestinal endocrine cells and brain neurons, and regulates gall bladder contractions, pancreatic enzyme secretion, gut functions, satiety and food intake. Additionally, CCK plays important roles in neuromodulation in several brain circuits that regulate reward, anxiety, aggression and sexual behavior. In invertebrates, CCK-type peptides (sulfakinins, SKs) are, with a few exceptions, produced by brain neurons only. Common among invertebrates is that SKs mediate satiety and regulate food ingestion by a variety of mechanisms. Also regulation of secretion of digestive enzymes has been reported. Studies of the genetically tractable fly Drosophila have advanced our understanding of SK signaling mechanisms in regulation of satiety and feeding, but also in gustatory sensitivity, locomotor activity, aggression and reproductive behavior. A set of eight SK-expressing brain neurons plays important roles in regulation of these competing behaviors. In males, they integrate internal state and external stimuli to diminish sex drive and increase aggression. The same neurons also diminish sugar gustation, induce satiety and reduce feeding. Although several functional roles of CCK/SK signaling appear conserved between Drosophila and mammals, available data suggest that the underlying mechanisms differ.


Assuntos
Agressão/fisiologia , Colecistocinina/metabolismo , Neuropeptídeos/metabolismo , Receptores de Neuropeptídeos/metabolismo , Comportamento Sexual Animal/fisiologia , Animais , Encéfalo/metabolismo , Drosophila/fisiologia , Humanos , Invertebrados/fisiologia , Mamíferos , Neuropeptídeos/química , Receptores de Neuropeptídeos/química , Transdução de Sinais/fisiologia , Paladar
6.
PLoS Biol ; 17(2): e2006409, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30759083

RESUMO

Dysregulation of sleep and feeding has widespread health consequences. Despite extensive epidemiological evidence for interactions between sleep and metabolic function, little is known about the neural or molecular basis underlying the integration of these processes. D. melanogaster potently suppress sleep in response to starvation, and powerful genetic tools allow for mechanistic investigation of sleep-metabolism interactions. We have previously identified neurons expressing the neuropeptide leucokinin (Lk) as being required for starvation-mediated changes in sleep. Here, we demonstrate an essential role for Lk neuropeptide in metabolic regulation of sleep. The activity of Lk neurons is modulated by feeding, with reduced activity in response to glucose and increased activity under starvation conditions. Both genetic silencing and laser-mediated microablation localize Lk-dependent sleep regulation to a single pair of Lk neurons within the Lateral Horn (LHLK neurons). A targeted screen identified a role for 5' adenosine monophosphate-activated protein kinase (AMPK) in starvation-modulated changes in sleep. Knockdown of AMPK in Lk neurons suppresses sleep and increases LHLK neuron activity in fed flies, phenocopying the starvation state. Further, we find a requirement for the Lk receptor in the insulin-producing cells (IPCs), suggesting LHLK-IPC connectivity is critical for sleep regulation under starved conditions. Taken together, these findings localize feeding-state-dependent regulation of sleep to a single pair of neurons within the fruit fly brain and provide a system for investigating the cellular basis of sleep-metabolism interactions.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Sono/fisiologia , Adenilato Quinase/metabolismo , Animais , Terapia a Laser , Inanição , Vigília
7.
PLoS Genet ; 14(8): e1007618, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30138334

RESUMO

Animals need to continuously adjust their water metabolism to the internal and external conditions. Homeostasis of body fluids thus requires tight regulation of water intake and excretion, and a balance between ingestion of water and solid food. Here, we investigated how these processes are coordinated in Drosophila melanogaster. We identified the first thirst-promoting and anti-diuretic hormone of Drosophila, encoded by the gene Ion transport peptide (ITP). This endocrine regulator belongs to the CHH (crustacean hyperglycemic hormone) family of peptide hormones. Using genetic gain- and loss-of-function experiments, we show that ITP signaling acts analogous to the human vasopressin and renin-angiotensin systems; expression of ITP is elevated by dehydration of the fly, and the peptide increases thirst while repressing excretion, promoting thus conservation of water resources. ITP responds to both osmotic and desiccation stress, and dysregulation of ITP signaling compromises the fly's ability to cope with these stressors. In addition to the regulation of thirst and excretion, ITP also suppresses food intake. Altogether, our work identifies ITP as an important endocrine regulator of thirst and excretion, which integrates water homeostasis with feeding of Drosophila.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster/genética , Homeostase , Neuropeptídeos/fisiologia , Sede/fisiologia , Animais , Proteínas de Drosophila/genética , Feminino , Trato Gastrointestinal/fisiologia , Regulação da Expressão Gênica , Fome , Masculino , Neuropeptídeos/genética , Transdução de Sinais , Estresse Fisiológico , Água
8.
PLoS Genet ; 14(11): e1007767, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30457986

RESUMO

Behavior and physiology are orchestrated by neuropeptides acting as central neuromodulators and circulating hormones. An outstanding question is how these neuropeptides function to coordinate complex and competing behaviors. In Drosophila, the neuropeptide leucokinin (LK) modulates diverse functions, but mechanisms underlying these complex interactions remain poorly understood. As a first step towards understanding these mechanisms, we delineated LK circuitry that governs various aspects of post-feeding physiology and behavior. We found that impaired LK signaling in Lk and Lk receptor (Lkr) mutants affects diverse but coordinated processes, including regulation of stress, water homeostasis, feeding, locomotor activity, and metabolic rate. Next, we sought to define the populations of LK neurons that contribute to the different aspects of this physiology. We find that the calcium activity in abdominal ganglia LK neurons (ABLKs), but not in the two sets of brain neurons, increases specifically following water consumption, suggesting that ABLKs regulate water homeostasis and its associated physiology. To identify targets of LK peptide, we mapped the distribution of Lkr expression, mined a brain single-cell transcriptome dataset for genes coexpressed with Lkr, and identified synaptic partners of LK neurons. Lkr expression in the brain insulin-producing cells (IPCs), gut, renal tubules and chemosensory cells, correlates well with regulatory roles detected in the Lk and Lkr mutants. Furthermore, these mutants and flies with targeted knockdown of Lkr in IPCs displayed altered expression of insulin-like peptides (DILPs) and transcripts in IPCs and increased starvation resistance. Thus, some effects of LK signaling appear to occur via DILP action. Collectively, our data suggest that the three sets of LK neurons have different targets, but modulate the establishment of post-prandial homeostasis by regulating distinct physiological processes and behaviors such as diuresis, metabolism, organismal activity and insulin signaling. These findings provide a platform for investigating feeding-related neuroendocrine regulation of vital behavior and physiology.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/genética , Drosophila melanogaster/fisiologia , Neuropeptídeos/genética , Neuropeptídeos/fisiologia , Animais , Animais Geneticamente Modificados , Comportamento Animal/fisiologia , Diurese/genética , Diurese/fisiologia , Proteínas de Drosophila/deficiência , Metabolismo Energético/genética , Metabolismo Energético/fisiologia , Feminino , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Insulina/fisiologia , Masculino , Atividade Motora/genética , Atividade Motora/fisiologia , Mutação , Neurônios/classificação , Neurônios/fisiologia , Neuropeptídeos/deficiência , Período Pós-Prandial/genética , Período Pós-Prandial/fisiologia , Receptores de Neuropeptídeos/deficiência , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/fisiologia , Transdução de Sinais
9.
Int J Mol Sci ; 22(4)2021 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-33669286

RESUMO

Leucokinins (LKs) constitute a family of neuropeptides identified in numerous insects and many other invertebrates. LKs act on G-protein-coupled receptors that display only distant relations to other known receptors. In adult Drosophila, 26 neurons/neurosecretory cells of three main types express LK. The four brain interneurons are of two types, and these are implicated in several important functions in the fly's behavior and physiology, including feeding, sleep-metabolism interactions, state-dependent memory formation, as well as modulation of gustatory sensitivity and nociception. The 22 neurosecretory cells (abdominal LK neurons, ABLKs) of the abdominal neuromeres co-express LK and a diuretic hormone (DH44), and together, these regulate water and ion homeostasis and associated stress as well as food intake. In Drosophila larvae, LK neurons modulate locomotion, escape responses and aspects of ecdysis behavior. A set of lateral neurosecretory cells, ALKs (anterior LK neurons), in the brain express LK in larvae, but inconsistently so in adults. These ALKs co-express three other neuropeptides and regulate water and ion homeostasis, feeding, and drinking, but the specific role of LK is not yet known. This review summarizes Drosophila data on embryonic lineages of LK neurons, functional roles of individual LK neuron types, interactions with other peptidergic systems, and orchestrating functions of LK.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Larva/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Animais , Ingestão de Alimentos/fisiologia , Comportamento Alimentar/fisiologia , Homeostase/fisiologia , Hormônios de Inseto/metabolismo , Transdução de Sinais/fisiologia , Sono/fisiologia , Inanição/metabolismo
10.
Int J Mol Sci ; 22(4)2021 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-33546414

RESUMO

Leucokinins (LKs) constitute a neuropeptide family first discovered in a cockroach and later identified in numerous insects and several other invertebrates. The LK receptors are only distantly related to other known receptors. Among insects, there are many examples of species where genes encoding LKs and their receptors are absent. Furthermore, genomics has revealed that LK signaling is lacking in several of the invertebrate phyla and in vertebrates. In insects, the number and complexity of LK-expressing neurons vary, from the simple pattern in the Drosophila larva where the entire CNS has 20 neurons of 3 main types, to cockroaches with about 250 neurons of many different types. Common to all studied insects is the presence or 1-3 pairs of LK-expressing neurosecretory cells in each abdominal neuromere of the ventral nerve cord, that, at least in some insects, regulate secretion in Malpighian tubules. This review summarizes the diverse functional roles of LK signaling in insects, as well as other arthropods and mollusks. These functions include regulation of ion and water homeostasis, feeding, sleep-metabolism interactions, state-dependent memory formation, as well as modulation of gustatory sensitivity and nociception. Other functions are implied by the neuronal distribution of LK, but remain to be investigated.


Assuntos
Hormônios/genética , Hormônios/metabolismo , Insetos , Invertebrados , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Animais , Regulação da Expressão Gênica , Hormônios/química , Controle de Insetos , Neurônios/metabolismo , Neuropeptídeos/química , Especificidade de Órgãos/genética , Controle de Pragas , Filogenia , Ligação Proteica , Receptores de Neuropeptídeos/metabolismo , Transdução de Sinais , Especificidade da Espécie
11.
Cell Tissue Res ; 382(2): 233-266, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32827072

RESUMO

Hormones regulate development, as well as many vital processes in the daily life of an animal. Many of these hormones are peptides that act at a higher hierarchical level in the animal with roles as organizers that globally orchestrate metabolism, physiology and behavior. Peptide hormones can act on multiple peripheral targets and simultaneously convey basal states, such as metabolic status and sleep-awake or arousal across many central neuronal circuits. Thereby, they coordinate responses to changing internal and external environments. The activity of neurosecretory cells is controlled either by (1) cell autonomous sensors, or (2) by other neurons that relay signals from sensors in peripheral tissues and (3) by feedback from target cells. Thus, a hormonal signaling axis commonly comprises several components. In mammals and other vertebrates, several hormonal axes are known, such as the hypothalamic-pituitary-gonad axis or the hypothalamic-pituitary-thyroid axis that regulate reproduction and metabolism, respectively. It has been proposed that the basic organization of such hormonal axes is evolutionarily old and that cellular homologs of the hypothalamic-pituitary system can be found for instance in insects. To obtain an appreciation of the similarities between insect and vertebrate neurosecretory axes, we review the organization of neurosecretory cell systems in Drosophila. Our review outlines the major peptidergic hormonal pathways known in Drosophila and presents a set of schemes of hormonal axes and orchestrating peptidergic systems. The detailed organization of the larval and adult Drosophila neurosecretory systems displays only very basic similarities to those in other arthropods and vertebrates.


Assuntos
Hormônios/metabolismo , Neuropeptídeos/metabolismo , Animais , Drosophila
12.
Cell Mol Life Sci ; 75(6): 1099-1115, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29043393

RESUMO

Multiple neuropeptides are known to regulate water and ion balance in Drosophila melanogaster. Several of these peptides also have other functions in physiology and behavior. Examples are corticotropin-releasing factor-like diuretic hormone (diuretic hormone 44; DH44) and leucokinin (LK), both of which induce fluid secretion by Malpighian tubules (MTs), but also regulate stress responses, feeding, circadian activity and other behaviors. Here, we investigated the functional relations between the LK and DH44 signaling systems. DH44 and LK peptides are only colocalized in a set of abdominal neurosecretory cells (ABLKs). Targeted knockdown of each of these peptides in ABLKs leads to increased resistance to desiccation, starvation and ionic stress. Food ingestion is diminished by knockdown of DH44, but not LK, and water retention is increased by LK knockdown only. Thus, the two colocalized peptides display similar systemic actions, but differ with respect to regulation of feeding and body water retention. We also demonstrated that DH44 and LK have additive effects on fluid secretion by MTs. It is likely that the colocalized peptides are coreleased from ABLKs into the circulation and act on the tubules where they target different cell types and signaling systems to regulate diuresis and stress tolerance. Additional targets seem to be specific for each of the two peptides and subserve regulation of feeding and water retention. Our data suggest that the ABLKs and hormonal actions are sufficient for many of the known DH44 and LK functions, and that the remaining neurons in the CNS play other functional roles.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Hormônios de Inseto/genética , Túbulos de Malpighi/metabolismo , Células Neuroendócrinas/metabolismo , Neuropeptídeos/genética , Equilíbrio Hidroeletrolítico/genética , Animais , Dessecação , Diurese/fisiologia , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Ingestão de Alimentos/fisiologia , Regulação da Expressão Gênica , Hormônios de Inseto/antagonistas & inibidores , Hormônios de Inseto/metabolismo , Túbulos de Malpighi/citologia , Células Neuroendócrinas/citologia , Neuropeptídeos/antagonistas & inibidores , Neuropeptídeos/metabolismo , Pressão Osmótica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Inanição/genética , Inanição/metabolismo , Estresse Fisiológico/genética
13.
Cell Mol Life Sci ; 73(2): 271-90, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26472340

RESUMO

Insulin, insulin-like growth factors (IGFs) and insulin-like peptides (ILPs) are important regulators of metabolism, growth, reproduction and lifespan, and mechanisms of insulin/IGF signaling (IIS) have been well conserved over evolution. In insects, between one and 38 ILPs have been identified in each species. Relatively few insect species have been investigated in depth with respect to ILP functions, and therefore we focus mainly on the well-studied fruitfly Drosophila melanogaster. In Drosophila eight ILPs (DILP1-8), but only two receptors (dInR and Lgr3) are known. DILP2, 3 and 5 are produced by a set of neurosecretory cells (IPCs) in the brain and their biosynthesis and release are controlled by a number of mechanisms differing between larvae and adults. Adult IPCs display cell-autonomous sensing of circulating glucose, coupled to evolutionarily conserved mechanisms for DILP release. The glucose-mediated DILP secretion is modulated by neurotransmitters and neuropeptides, as well as by factors released from the intestine and adipocytes. Larval IPCs, however, are indirectly regulated by glucose-sensing endocrine cells producing adipokinetic hormone, or by circulating factors from the intestine and fat body. Furthermore, IIS is situated within a complex physiological regulatory network that also encompasses the lipophilic hormones, 20-hydroxyecdysone and juvenile hormone. After release from IPCs, the ILP action can be modulated by circulating proteins that act either as protective carriers (binding proteins), or competitive inhibitors. Some of these proteins appear to have additional functions that are independent of ILPs. Taken together, the signaling with multiple ILPs is under complex control, ensuring tightly regulated IIS in the organism.


Assuntos
Drosophila/metabolismo , Proteínas de Insetos/metabolismo , Insetos/metabolismo , Insulina/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neuropeptídeos/metabolismo , Transdução de Sinais , Somatomedinas/metabolismo , Animais , Drosophila/genética , Drosophila/fisiologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Hormônios , Proteínas de Insetos/genética , Insetos/genética , Insetos/fisiologia , Insulina/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Neurônios/metabolismo , Neuropeptídeos/genética , Reprodução , Somatomedinas/genética
14.
BMC Genomics ; 17: 50, 2016 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-26758761

RESUMO

BACKGROUND: In models extensively used in studies of aging and extended lifespan, such as C. elegans and Drosophila, adult senescence is regulated by gene networks that are likely to be similar to ones that underlie lifespan extension during dormancy. These include the evolutionarily conserved insulin/IGF, TOR and germ line-signaling pathways. Dormancy, also known as dauer stage in the larval worm or adult diapause in the fly, is triggered by adverse environmental conditions, and results in drastically extended lifespan with negligible senescence. It is furthermore characterized by increased stress resistance and somatic maintenance, developmental arrest and reallocated energy resources. In the fly Drosophila melanogaster adult reproductive diapause is additionally manifested in arrested ovary development, improved immune defense and altered metabolism. However, the molecular mechanisms behind this adaptive lifespan extension are not well understood. RESULTS: A genome wide analysis of transcript changes in diapausing D. melanogaster revealed a differential regulation of more than 4600 genes. Gene ontology (GO) and KEGG pathway analysis reveal that many of these genes are part of signaling pathways that regulate metabolism, stress responses, detoxification, immunity, protein synthesis and processes during aging. More specifically, gene readouts and detailed mapping of the pathways indicate downregulation of insulin-IGF (IIS), target of rapamycin (TOR) and MAP kinase signaling, whereas Toll-dependent immune signaling, Jun-N-terminal kinase (JNK) and Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathways are upregulated during diapause. Furthermore, we detected transcriptional regulation of a large number of genes specifically associated with aging and longevity. CONCLUSIONS: We find that many affected genes and signal pathways are shared between dormancy, aging and lifespan extension, including IIS, TOR, JAK/STAT and JNK. A substantial fraction of the genes affected by diapause have also been found to alter their expression in response to starvation and cold exposure in D. melanogaster, and the pathways overlap those reported in GO analysis of other invertebrates in dormancy or even hibernating mammals. Our study, thus, shows that D. melanogaster is a genetically tractable model for dormancy in other organisms and effects of dormancy on aging and lifespan.


Assuntos
Envelhecimento/genética , Drosophila melanogaster/genética , Regulação da Expressão Gênica , Transcriptoma/genética , Envelhecimento/fisiologia , Animais , Drosophila melanogaster/fisiologia , Ontologia Genética , Genoma de Inseto , Células Germinativas/metabolismo , Insulina/genética , Longevidade/genética , Reprodução/genética , Transdução de Sinais
15.
Cell Mol Life Sci ; 72(16): 3143-55, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25782410

RESUMO

Food odors stimulate appetite and innate food-seeking behavior in hungry animals. The smell of food also induces salivation and release of gastric acid and insulin. Conversely, sustained odor exposure may induce satiation. We demonstrate novel effects of food odors on food ingestion, metabolism and endocrine signaling in Drosophila melanogaster. Acute exposure to attractive vinegar odor triggers a rapid and transient increase in circulating glucose, and a rapid upregulation of genes encoding the glucagon-like hormone adipokinetic hormone (AKH), four insulin-like peptides (DILPs) and some target genes in peripheral tissues. Sustained exposure to food odors, however, decreases food intake. Hunger-induced strengthening of synaptic signaling from olfactory sensory neurons (OSNs) to brain neurons increases food-seeking behavior, and conversely fed flies display reduced food odor sensitivity and feeding. We show that increasing the strength of OSN signaling chronically by genetic manipulation of local peptide neuromodulation reduces feeding, elevates carbohydrates and diminishes lipids. Furthermore, constitutively strengthened odor sensitivity altered gene transcripts for AKH, DILPs and some of their targets. Thus, we show that food odor can induce a transient anticipatory endocrine response, and that boosted sensitivity to this odor affects food intake, as well as metabolism and hormonal signaling.


Assuntos
Drosophila melanogaster/fisiologia , Sistema Endócrino/fisiologia , Comportamento Alimentar/fisiologia , Alimentos , Redes e Vias Metabólicas/fisiologia , Odorantes/análise , Transdução de Sinais/fisiologia , Ácido Acético/química , Ácido Acético/farmacologia , Análise de Variância , Animais , Animais Geneticamente Modificados , Glicemia/metabolismo , Drosophila melanogaster/genética , Imunofluorescência , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Imuno-Histoquímica , Hormônios de Inseto/metabolismo , Oligopeptídeos/metabolismo , Ácido Pirrolidonocarboxílico/análogos & derivados , Ácido Pirrolidonocarboxílico/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
16.
PLoS Genet ; 9(12): e1004052, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24385933

RESUMO

Neurons and other cells display a large variation in size in an organism. Thus, a fundamental question is how growth of individual cells and their organelles is regulated. Is size scaling of individual neurons regulated post-mitotically, independent of growth of the entire CNS? Although the role of insulin/IGF-signaling (IIS) in growth of tissues and whole organisms is well established, it is not known whether it regulates the size of individual neurons. We therefore studied the role of IIS in the size scaling of neurons in the Drosophila CNS. By targeted genetic manipulations of insulin receptor (dInR) expression in a variety of neuron types we demonstrate that the cell size is affected only in neuroendocrine cells specified by the bHLH transcription factor DIMMED (DIMM). Several populations of DIMM-positive neurons tested displayed enlarged cell bodies after overexpression of the dInR, as well as PI3 kinase and Akt1 (protein kinase B), whereas DIMM-negative neurons did not respond to dInR manipulations. Knockdown of these components produce the opposite phenotype. Increased growth can also be induced by targeted overexpression of nutrient-dependent TOR (target of rapamycin) signaling components, such as Rheb (small GTPase), TOR and S6K (S6 kinase). After Dimm-knockdown in neuroendocrine cells manipulations of dInR expression have significantly less effects on cell size. We also show that dInR expression in neuroendocrine cells can be altered by up or down-regulation of Dimm. This novel dInR-regulated size scaling is seen during postembryonic development, continues in the aging adult and is diet dependent. The increase in cell size includes cell body, axon terminations, nucleus and Golgi apparatus. We suggest that the dInR-mediated scaling of neuroendocrine cells is part of a plasticity that adapts the secretory capacity to changing physiological conditions and nutrient-dependent organismal growth.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Sistema Nervoso Central/metabolismo , Proteínas de Drosophila/genética , Drosophila/genética , Insulina/genética , Neurônios/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Linhagem da Célula , Sistema Nervoso Central/citologia , Drosophila/crescimento & desenvolvimento , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Complexo de Golgi/genética , Complexo de Golgi/metabolismo , Insulina/metabolismo , Células Neuroendócrinas/citologia , Células Neuroendócrinas/metabolismo , Neurônios/citologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Proteínas Quinases S6 Ribossômicas/genética , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais
17.
Gen Comp Endocrinol ; 221: 255-66, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25616197

RESUMO

Taking advantage of Drosophila as a genetically tractable experimental animal much progress has been made in our understanding of how the insulin/IGF signaling (IIS) pathway regulates development, growth, metabolism, stress responses and lifespan. The role of IIS in regulation of neuronal activity and behavior has also become apparent from experiments in Drosophila. This review briefly summarizes these functional roles of IIS, and also how the insulin producing cells (IPCs) are regulated in the fly. Furthermore, we discuss functional aspects of the spatio-temporal production of eight different insulin-like peptides (DILP1-8) that are thought to act on one known receptor (dInR) in Drosophila.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fator de Crescimento Insulin-Like I/metabolismo , Insulina/metabolismo , Animais , Drosophila/crescimento & desenvolvimento , Células Secretoras de Insulina/metabolismo , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais
18.
J Neurosci ; 33(12): 5340-5, 2013 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-23516298

RESUMO

In insects, many complex behaviors, including olfactory memory, are controlled by a paired brain structure, the so-called mushroom bodies (MB). In Drosophila, the development, neuroanatomy, and function of intrinsic neurons of the MB, the Kenyon cells, have been well characterized. Until now, several potential neurotransmitters or neuromodulators of Kenyon cells have been anatomically identified. However, whether these neuroactive substances of the Kenyon cells are functional has not been clarified yet. Here we show that a neuropeptide precursor gene encoding four types of short neuropeptide F (sNPF) is required in the Kenyon cells for appetitive olfactory memory. We found that activation of Kenyon cells by expressing a thermosensitive cation channel (dTrpA1) leads to a decrease in sNPF immunoreactivity in the MB lobes. Targeted expression of RNA interference against the sNPF precursor in Kenyon cells results in a highly significant knockdown of sNPF levels. This knockdown of sNPF in the Kenyon cells impairs sugar-rewarded olfactory memory. This impairment is not due to a defect in the reflexive sugar preference or odor response. Consistently, knockdown of sNPF receptors outside the MB causes deficits in appetitive memory. Altogether, these results suggest that sNPF is a functional neuromodulator released by Kenyon cells.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster/fisiologia , Memória/fisiologia , Corpos Pedunculados/citologia , Corpos Pedunculados/fisiologia , Neuropeptídeos/fisiologia , Olfato/fisiologia , Animais , Animais Geneticamente Modificados , Apetite/fisiologia , Comportamento Animal/fisiologia , Condicionamento Psicológico/fisiologia , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Feminino , Masculino , Neuropeptídeos/genética , Neurotransmissores/genética , Neurotransmissores/fisiologia , Odorantes
19.
Elife ; 122024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38904987

RESUMO

Numerous roles for the Alk receptor tyrosine kinase have been described in Drosophila, including functions in the central nervous system (CNS), however the molecular details are poorly understood. To gain mechanistic insight, we employed Targeted DamID (TaDa) transcriptional profiling to identify targets of Alk signaling in the larval CNS. TaDa was employed in larval CNS tissues, while genetically manipulating Alk signaling output. The resulting TaDa data were analyzed together with larval CNS scRNA-seq datasets performed under similar conditions, identifying a role for Alk in the transcriptional regulation of neuroendocrine gene expression. Further integration with bulk and scRNA-seq datasets from larval brains in which Alk signaling was manipulated identified a previously uncharacterized Drosophila neuropeptide precursor encoded by CG4577 as an Alk signaling transcriptional target. CG4577, which we named Sparkly (Spar), is expressed in a subset of Alk-positive neuroendocrine cells in the developing larval CNS, including circadian clock neurons. In agreement with our TaDa analysis, overexpression of the Drosophila Alk ligand Jeb resulted in increased levels of Spar protein in the larval CNS. We show that Spar protein is expressed in circadian (clock) neurons, and flies lacking Spar exhibit defects in sleep and circadian activity control. In summary, we report a novel activity regulating neuropeptide precursor gene that is regulated by Alk signaling in the Drosophila CNS.


Assuntos
Quinase do Linfoma Anaplásico , Sistema Nervoso Central , Proteínas de Drosophila , Animais , Sistema Nervoso Central/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Quinase do Linfoma Anaplásico/metabolismo , Quinase do Linfoma Anaplásico/genética , Larva/metabolismo , Larva/genética , Larva/crescimento & desenvolvimento , Neuropeptídeos/metabolismo , Neuropeptídeos/genética , Transdução de Sinais , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptores Proteína Tirosina Quinases/genética , Drosophila/genética , Drosophila/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica
20.
Cell Tissue Res ; 353(3): 511-23, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23760890

RESUMO

Four forms of short neuropeptide F (sNPF1-4), derived from the gene snpf, have been identified in Drosophila and are known to act on a single G-protein-coupled receptor (sNPFR). Several functions have been suggested for sNPFs in Drosophila, including the regulation of feeding and growth in larvae, the control of insulin signalling and the modulation of neuronal circuits in adult flies. Furthermore, sNPF has been shown to act as a nutritional state-dependent neuromodulator in the olfactory system. The role of sNPF in the larval nervous system is less well known. To analyse sites of action of sNPF in the larva, we mapped the distribution of sNPF- and sNPFR-expressing neurons. In particular, we studied circuits associated with chemosensory inputs and systems involved in the regulation of feeding, including neurosecretory cell systems and the hypocerebral ganglion. We employed a combination of immunocytochemistry and enhancer trap and promoter Gal4 lines to drive green fluorescent protein. We found a good match between the distribution of the receptor and its ligand. However, several differences between the larval and adult systems were observed. Thus, neither sNPF nor its receptor was found in the olfactory (or other sensory) systems in the larva and cells producing insulin-like peptides did not co-express sNPFR, as opposed to results from adults. Moreover, sNPF was expressed in a subpopulation of Hugin cells (second-order gustatory neurons) only in adult flies. We propose that the differences in sNPF signalling between the developmental stages is explained by differences in their feeding behaviour.


Assuntos
Sistema Nervoso Central/metabolismo , Proteínas de Drosophila/metabolismo , Gânglios dos Invertebrados/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Animais , Sistema Nervoso Central/citologia , Proteínas de Drosophila/genética , Drosophila melanogaster , Comportamento Alimentar/fisiologia , Gânglios dos Invertebrados/citologia , Larva/citologia , Larva/genética , Larva/metabolismo , Neurônios/citologia , Neuropeptídeos/genética , Receptores de Neuropeptídeos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA