Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Metab ; 4(11): 1532-1550, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36344765

RESUMO

Animals must adapt their dietary choices to meet their nutritional needs. How these needs are detected and translated into nutrient-specific appetites that drive food-choice behaviours is poorly understood. Here we show that enteroendocrine cells of the adult female Drosophila midgut sense nutrients and in response release neuropeptide F (NPF), which is an ortholog of mammalian neuropeptide Y-family gut-brain hormones. Gut-derived NPF acts on glucagon-like adipokinetic hormone (AKH) signalling to induce sugar satiety and increase consumption of protein-rich food, and on adipose tissue to promote storage of ingested nutrients. Suppression of NPF-mediated gut signalling leads to overconsumption of dietary sugar while simultaneously decreasing intake of protein-rich yeast. Furthermore, gut-derived NPF has a female-specific function in promoting consumption of protein-containing food in mated females. Together, our findings suggest that gut NPF-to-AKH signalling modulates specific appetites and regulates food choice to ensure homeostatic consumption of nutrients, providing insight into the hormonal mechanisms that underlie nutrient-specific hungers.


Assuntos
Proteínas de Drosophila , Hormônios Gastrointestinais , Feminino , Animais , Drosophila , Apetite , Açúcares , Proteínas de Drosophila/genética , Mamíferos
2.
Nat Ecol Evol ; 6(11): 1753-1765, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36192540

RESUMO

Ant colonies are higher-level organisms consisting of specialized reproductive and non-reproductive individuals that differentiate early in development, similar to germ-soma segregation in bilateral Metazoa. Analogous to diverging cell lines, developmental differentiation of individual ants has often been considered in epigenetic terms but the sets of genes that determine caste phenotypes throughout larval and pupal development remain unknown. Here, we reconstruct the individual developmental trajectories of two ant species, Monomorium pharaonis and Acromyrmex echinatior, after obtaining >1,400 whole-genome transcriptomes. Using a new backward prediction algorithm, we show that caste phenotypes can be accurately predicted by genome-wide transcriptome profiling. We find that caste differentiation is increasingly canalized from early development onwards, particularly in germline individuals (gynes/queens) and that the juvenile hormone signalling pathway plays a key role in this process by regulating body mass divergence between castes. We quantified gene-specific canalization levels and found that canalized genes with gyne/queen-biased expression were enriched for ovary and wing functions while canalized genes with worker-biased expression were enriched in brain and behavioural functions. Suppression in gyne larvae of Freja, a highly canalized gyne-biased ovary gene, disturbed pupal development by inducing non-adaptive intermediate phenotypes between gynes and workers. Our results are consistent with natural selection actively maintaining canalized caste phenotypes while securing robustness in the life cycle ontogeny of ant colonies.


Assuntos
Formigas , Animais , Feminino , Formigas/genética , Perfilação da Expressão Gênica , Larva/genética , Fenótipo , Transcriptoma
3.
Curr Biol ; 32(7): 1548-1562.e6, 2022 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-35245460

RESUMO

Nutrition is one of the most important influences on growth and the timing of maturational transitions including mammalian puberty and insect metamorphosis. Childhood obesity is associated with precocious puberty, but the assessment mechanism that links body fat to early maturation is unknown. During development, the intake of nutrients promotes signaling through insulin-like systems that govern the growth of cells and tissues and also regulates the timely production of the steroid hormones that initiate the juvenile-adult transition. We show here that the dietary lipid cholesterol, which is required as a component of cell membranes and as a substrate for steroid biosynthesis, also governs body growth and maturation in Drosophila via promoting the expression and release of insulin-like peptides. This nutritional input acts via the nutrient sensor TOR, which is regulated by the Niemann-Pick-type-C 1 (Npc1) cholesterol transporter, in the glia of the blood-brain barrier and cells of the adipose tissue to remotely drive systemic insulin signaling and body growth. Furthermore, increasing intracellular cholesterol levels in the steroid-producing prothoracic gland strongly promotes endoreduplication, leading to an accelerated attainment of a nutritional checkpoint that normally ensures that animals do not initiate maturation prematurely. These findings, therefore, show that a Npc1-TOR signaling system couples the sensing of the lipid cholesterol with cellular and systemic growth control and maturational timing, which may help explain both the link between cholesterol and cancer as well as the connection between body fat (obesity) and early puberty.


Assuntos
Proteínas de Drosophila , Obesidade Infantil , Animais , Colesterol , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Insulina/metabolismo , Larva , Mamíferos , Esteroides/metabolismo
4.
Nat Commun ; 13(1): 692, 2022 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-35121731

RESUMO

The intestine is a central regulator of metabolic homeostasis. Dietary inputs are absorbed through the gut, which senses their nutritional value and relays hormonal information to other organs to coordinate systemic energy balance. However, the gut-derived hormones affecting metabolic and behavioral responses are poorly defined. Here we show that the endocrine cells of the Drosophila gut sense nutrient stress through a mechanism that involves the TOR pathway and in response secrete the peptide hormone allatostatin C, a Drosophila somatostatin homolog. Gut-derived allatostatin C induces secretion of glucagon-like adipokinetic hormone to coordinate food intake and energy mobilization. Loss of gut Allatostatin C or its receptor in the adipokinetic-hormone-producing cells impairs lipid and sugar mobilization during fasting, leading to hypoglycemia. Our findings illustrate a nutrient-responsive endocrine mechanism that maintains energy homeostasis under nutrient-stress conditions, a function that is essential to health and whose failure can lead to metabolic disorders.


Assuntos
Proteínas de Drosophila/metabolismo , Ingestão de Alimentos/fisiologia , Metabolismo Energético/fisiologia , Hormônios Gastrointestinais/metabolismo , Homeostase , Nutrientes/metabolismo , Somatostatina/metabolismo , Animais , Animais Geneticamente Modificados , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Ingestão de Alimentos/genética , Metabolismo Energético/genética , Células Enteroendócrinas/metabolismo , Hormônios Gastrointestinais/genética , Técnicas de Inativação de Genes , Humanos , Hipoglicemia/genética , Hipoglicemia/metabolismo , Hormônios de Inseto/genética , Hormônios de Inseto/metabolismo , Oligopeptídeos/genética , Oligopeptídeos/metabolismo , Ácido Pirrolidonocarboxílico/análogos & derivados , Ácido Pirrolidonocarboxílico/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/genética , Somatostatina/genética , Análise de Sobrevida
5.
Nat Commun ; 12(1): 5178, 2021 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-34462441

RESUMO

Animals maintain metabolic homeostasis by modulating the activity of specialized organs that adjust internal metabolism to external conditions. However, the hormonal signals coordinating these functions are incompletely characterized. Here we show that six neurosecretory cells in the Drosophila central nervous system respond to circulating nutrient levels by releasing Capa hormones, homologs of mammalian neuromedin U, which activate the Capa receptor (CapaR) in peripheral tissues to control energy homeostasis. Loss of Capa/CapaR signaling causes intestinal hypomotility and impaired nutrient absorption, which gradually deplete internal nutrient stores and reduce organismal lifespan. Conversely, increased Capa/CapaR activity increases fluid and waste excretion. Furthermore, Capa/CapaR inhibits the release of glucagon-like adipokinetic hormone from the corpora cardiaca, which restricts energy mobilization from adipose tissue to avoid harmful hyperglycemia. Our results suggest that the Capa/CapaR circuit occupies a central node in a homeostatic program that facilitates the digestion and absorption of nutrients and regulates systemic energy balance.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Neuropeptídeos/metabolismo , Nutrientes/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Metabolismo Energético , Feminino , Homeostase , Hormônios de Inseto/metabolismo , Longevidade , Masculino , Neuropeptídeos/genética , Oligopeptídeos/metabolismo , Ácido Pirrolidonocarboxílico/análogos & derivados , Ácido Pirrolidonocarboxílico/metabolismo , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais
6.
Curr Opin Insect Sci ; 43: 54-62, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33214126

RESUMO

Steroid hormones control major developmental transitions such as metamorphosis in insects and puberty in mammals. The juvenile must attain a sufficient size before it begins maturation in order to give rise to a properly sized and reproductively fit adult. Studies in the insect Drosophila have begun to reveal a remarkable example of the complex interplay between different organs and the neuroendocrine system that controls the production of the steroid ecdysone, which triggers metamorphosis. This review discusses the inter-organ signals mediating this crosstalk, which allows the neuroendocrine system to assess nutrient availability and growth status of internal organs, ensuring that maturation is initiated at the appropriate time. We discuss how the neuroendocrine system integrates signals from different tissues to coordinate growth and maturation. These studies are still unraveling the organ-to-organ signaling networks that control the timing of metamorphosis, defining important principles underlying the logic of growth and maturation coordination in animals.


Assuntos
Drosophila/crescimento & desenvolvimento , Metamorfose Biológica , Animais , Ecdisona/biossíntese , Sistemas Neurossecretores/fisiologia , Transdução de Sinais
7.
Genetics ; 216(2): 269-313, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33023929

RESUMO

The control of body and organ growth is essential for the development of adults with proper size and proportions, which is important for survival and reproduction. In animals, adult body size is determined by the rate and duration of juvenile growth, which are influenced by the environment. In nutrient-scarce environments in which more time is needed for growth, the juvenile growth period can be extended by delaying maturation, whereas juvenile development is rapidly completed in nutrient-rich conditions. This flexibility requires the integration of environmental cues with developmental signals that govern internal checkpoints to ensure that maturation does not begin until sufficient tissue growth has occurred to reach a proper adult size. The Target of Rapamycin (TOR) pathway is the primary cell-autonomous nutrient sensor, while circulating hormones such as steroids and insulin-like growth factors are the main systemic regulators of growth and maturation in animals. We discuss recent findings in Drosophila melanogaster showing that cell-autonomous environment and growth-sensing mechanisms, involving TOR and other growth-regulatory pathways, that converge on insulin and steroid relay centers are responsible for adjusting systemic growth, and development, in response to external and internal conditions. In addition to this, proper organ growth is also monitored and coordinated with whole-body growth and the timing of maturation through modulation of steroid signaling. This coordination involves interorgan communication mediated by Drosophila insulin-like peptide 8 in response to tissue growth status. Together, these multiple nutritional and developmental cues feed into neuroendocrine hubs controlling insulin and steroid signaling, serving as checkpoints at which developmental progression toward maturation can be delayed. This review focuses on these mechanisms by which external and internal conditions can modulate developmental growth and ensure proper adult body size, and highlights the conserved architecture of this system, which has made Drosophila a prime model for understanding the coordination of growth and maturation in animals.


Assuntos
Tamanho Corporal , Drosophila melanogaster/genética , Regulação da Expressão Gênica no Desenvolvimento , Animais , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Estágios do Ciclo de Vida , Transdução de Sinais
8.
Development ; 147(14)2020 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-32631830

RESUMO

The activation of a neuroendocrine system that induces a surge in steroid production is a conserved initiator of the juvenile-to-adult transition in many animals. The trigger for maturation is the secretion of brain-derived neuropeptides, yet the mechanisms controlling the timely onset of this event remain ill-defined. Here, we show that a regulatory feedback circuit controlling the Drosophila neuropeptide Prothoracicotropic hormone (PTTH) triggers maturation onset. We identify the Ecdysone Receptor (EcR) in the PTTH-expressing neurons (PTTHn) as a regulator of developmental maturation onset. Loss of EcR in these PTTHn impairs PTTH signaling, which delays maturation. We find that the steroid ecdysone dose-dependently affects Ptth transcription, promoting its expression at lower concentrations and inhibiting it at higher concentrations. Our findings indicate the existence of a feedback circuit in which rising ecdysone levels trigger, via EcR activity in the PTTHn, the PTTH surge that generates the maturation-inducing ecdysone peak toward the end of larval development. Because steroid feedback is also known to control the vertebrate maturation-inducing hypothalamic-pituitary-gonadal axis, our findings suggest an overall conservation of the feedback-regulatory neuroendocrine circuitry that controls the timing of maturation initiation.


Assuntos
Proteínas de Drosophila/metabolismo , Hormônios de Inseto/metabolismo , Receptores de Esteroides/metabolismo , Animais , Tamanho Corporal , Drosophila/crescimento & desenvolvimento , Drosophila/metabolismo , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/genética , Ecdisterona/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Hormônios de Inseto/antagonistas & inibidores , Hormônios de Inseto/genética , Larva/crescimento & desenvolvimento , Larva/metabolismo , Metamorfose Biológica , Microscopia de Fluorescência , Neurônios/metabolismo , Interferência de RNA , RNA Guia de Cinetoplastídeos/metabolismo , Receptores de Esteroides/antagonistas & inibidores , Receptores de Esteroides/genética , Transdução de Sinais
9.
Cell Mol Life Sci ; 77(22): 4523-4551, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32448994

RESUMO

Organisms adapt to changing environments by adjusting their development, metabolism, and behavior to improve their chances of survival and reproduction. To achieve such flexibility, organisms must be able to sense and respond to changes in external environmental conditions and their internal state. Metabolic adaptation in response to altered nutrient availability is key to maintaining energy homeostasis and sustaining developmental growth. Furthermore, environmental variables exert major influences on growth and final adult body size in animals. This developmental plasticity depends on adaptive responses to internal state and external cues that are essential for developmental processes. Genetic studies have shown that the fruit fly Drosophila, similarly to mammals, regulates its metabolism, growth, and behavior in response to the environment through several key hormones including insulin, peptides with glucagon-like function, and steroid hormones. Here we review emerging evidence showing that various environmental cues and internal conditions are sensed in different organs that, via inter-organ communication, relay information to neuroendocrine centers that control insulin and steroid signaling. This review focuses on endocrine regulation of development, metabolism, and behavior in Drosophila, highlighting recent advances in the role of the neuroendocrine system as a signaling hub that integrates environmental inputs and drives adaptive responses.


Assuntos
Adaptação Fisiológica/fisiologia , Drosophila/metabolismo , Drosophila/fisiologia , Animais , Proteínas de Drosophila/metabolismo , Homeostase/fisiologia , Humanos , Hormônios de Inseto/metabolismo , Transdução de Sinais/fisiologia
10.
PLoS Genet ; 16(4): e1008727, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32339168

RESUMO

The human 22q11.2 chromosomal deletion is one of the strongest identified genetic risk factors for schizophrenia. Although the deletion spans a number of known genes, the contribution of each of these to the 22q11.2 deletion syndrome (DS) is not known. To investigate the effect of individual genes within this interval on the pathophysiology associated with the deletion, we analyzed their role in sleep, a behavior affected in virtually all psychiatric disorders, including the 22q11.2 DS. We identified the gene LZTR1 (night owl, nowl) as a regulator of night-time sleep in Drosophila. In humans, LZTR1 has been associated with Ras-dependent neurological diseases also caused by Neurofibromin-1 (Nf1) deficiency. We show that Nf1 loss leads to a night-time sleep phenotype nearly identical to that of nowl loss and that nowl negatively regulates Ras and interacts with Nf1 in sleep regulation. Furthermore, nowl is required for metabolic homeostasis, suggesting that LZTR1 may contribute to the genetic susceptibility to obesity associated with the 22q11.2 DS. Knockdown of nowl or Nf1 in GABA-responsive sleep-promoting neurons elicits the sleep phenotype, and this defect can be rescued by increased GABAA receptor signaling, indicating that Nowl regulates sleep through modulation of GABA signaling. Our results suggest that nowl/LZTR1 may be a conserved regulator of GABA signaling important for normal sleep that contributes to the 22q11.2 DS.


Assuntos
Síndrome da Deleção 22q11/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas de Drosophila/genética , Neurônios GABAérgicos/metabolismo , Neurofibromina 1/genética , Esquizofrenia/genética , Sono/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Drosophila , Proteínas de Drosophila/metabolismo , Neurônios GABAérgicos/fisiologia , Humanos , Neurofibromina 1/metabolismo , Receptores de GABA-A/metabolismo , Fatores de Transcrição/genética
11.
Autophagy ; 15(8): 1478-1480, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31084464

RESUMO

Steroid hormones are made from cholesterol and are essential for many developmental processes and disease conditions. The production of these hormones is nutrient dependent and tightly controlled by mechanisms that involve delivery of the precursor molecule cholesterol stored in lipid droplets (LDs). Recent studies have implicated macroautophagy/autophagy, a process regulated by nutrition, in the degradation of LDs and the mobilization of stored lipids. We recently identified an autophagy-dependent mechanism that regulates steroid production via effects on cholesterol trafficking. Through gain- and loss-of-function studies in Drosophila, we found that essential autophagy-related (Atg) genes are required in steroidogenic cells for normal steroid production. Inhibition of autophagy in these cells by knockdown of Atg genes causes strong accumulation of cholesterol in LDs and reduces steroid production, resembling effects seen in some lipid-storage disorders and steroid-dependent cancer conditions. This autophagy-dependent steroid hormone regulation (ASHR) process is regulated by the wts-yki/Warts-Yorkie tumor-suppressor pathway downstream of nutrition, coupling nutrient intake with steroid-dependent developmental growth. This mechanism potentially contributes to the development of certain cancers and lipid-storage disorders and thus may be of great therapeutic relevance.


Assuntos
Autofagia , Colesterol/metabolismo , Sistema Endócrino/citologia , Animais , Autofagossomos/metabolismo , Transporte Biológico , Drosophila melanogaster/metabolismo , Humanos , Fusão de Membrana , Modelos Biológicos
12.
Nat Commun ; 10(1): 1955, 2019 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-31028268

RESUMO

Organisms adapt their metabolism and growth to the availability of nutrients and oxygen, which are essential for development, yet the mechanisms by which this adaptation occurs are not fully understood. Here we describe an RNAi-based body-size screen in Drosophila to identify such mechanisms. Among the strongest hits is the fibroblast growth factor receptor homolog breathless necessary for proper development of the tracheal airway system. Breathless deficiency results in tissue hypoxia, sensed primarily in this context by the fat tissue through HIF-1a prolyl hydroxylase (Hph). The fat relays its hypoxic status through release of one or more HIF-1a-dependent humoral factors that inhibit insulin secretion from the brain, thereby restricting systemic growth. Independently of HIF-1a, Hph is also required for nutrient-dependent Target-of-rapamycin (Tor) activation. Our findings show that the fat tissue acts as the primary sensor of nutrient and oxygen levels, directing adaptation of organismal metabolism and growth to environmental conditions.


Assuntos
Proteínas de Drosophila/metabolismo , Animais , Proteínas de Ligação a DNA/metabolismo , Drosophila , Proteínas de Drosophila/genética , Regulação da Expressão Gênica no Desenvolvimento , Secreção de Insulina/genética , Secreção de Insulina/fisiologia , Oxigênio/metabolismo , Fatores de Transcrição/metabolismo
13.
Dev Cell ; 48(5): 659-671.e4, 2019 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-30799225

RESUMO

Steroid hormones are important signaling molecules that regulate growth and drive the development of many cancers. These factors act as long-range signals that systemically regulate the growth of the entire organism, whereas the Hippo/Warts tumor-suppressor pathway acts locally to limit organ growth. We show here that autophagy, a pathway that mediates the degradation of cellular components, also controls steroid production. This process is regulated by Warts (in mammals, LATS1/2) signaling, via its effector microRNA bantam, in response to nutrients. Specifically, autophagy-mediated mobilization and trafficking of the steroid precursor cholesterol from intracellular stores controls the production of the Drosophila steroid ecdysone. Furthermore, we also show that bantam regulates this process via the ecdysone receptor and Tor signaling, identifying pathways through which bantam regulates autophagy and growth. The Warts pathway thus promotes nutrient-dependent systemic growth during development by autophagy-dependent steroid hormone regulation (ASHR). These findings uncover an autophagic trafficking mechanism that regulates steroid production.


Assuntos
Autofagia/fisiologia , Movimento Celular/fisiologia , Colesterol/metabolismo , Ecdisona/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Animais , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , MicroRNAs/genética , Proteínas Nucleares/metabolismo , Transativadores/metabolismo
14.
PLoS Genet ; 14(12): e1007623, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30566533

RESUMO

The human 1q21.1 deletion of ten genes is associated with increased risk of schizophrenia. This deletion involves the ß-subunit of the AMP-activated protein kinase (AMPK) complex, a key energy sensor in the cell. Although neurons have a high demand for energy and low capacity to store nutrients, the role of AMPK in neuronal physiology is poorly defined. Here we show that AMPK is important in the nervous system for maintaining neuronal integrity and for stress survival and longevity in Drosophila. To understand the impact of this signaling system on behavior and its potential contribution to the 1q21.1 deletion syndrome, we focused on sleep, an important role of which is proposed to be the reestablishment of neuronal energy levels that are diminished during energy-demanding wakefulness. Sleep disturbances are one of the most common problems affecting individuals with psychiatric disorders. We show that AMPK is required for maintenance of proper sleep architecture and for sleep recovery following sleep deprivation. Neuronal AMPKß loss specifically leads to sleep fragmentation and causes dysregulation of genes believed to play a role in sleep homeostasis. Our data also suggest that AMPKß loss may contribute to the increased risk of developing mental disorders and sleep disturbances associated with the human 1q21.1 deletion.


Assuntos
Proteínas Quinases Ativadas por AMP/genética , Anormalidades Múltiplas/enzimologia , Anormalidades Múltiplas/genética , Megalencefalia/enzimologia , Megalencefalia/genética , Neurônios/enzimologia , Esquizofrenia/enzimologia , Esquizofrenia/genética , Sono/genética , Sono/fisiologia , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/deficiência , Animais , Deleção Cromossômica , Cromossomos Humanos Par 1/enzimologia , Cromossomos Humanos Par 1/genética , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/genética , Drosophila melanogaster/enzimologia , Drosophila melanogaster/genética , Feminino , Técnicas de Silenciamento de Genes , Predisposição Genética para Doença , Humanos , Aprendizagem/fisiologia , Longevidade/genética , Longevidade/fisiologia , Masculino , Modelos Animais , Neurônios/citologia , Fatores de Risco , Transdução de Sinais , Transtornos do Sono-Vigília/enzimologia , Transtornos do Sono-Vigília/genética
15.
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
16.
Curr Biol ; 27(11): 1652-1659.e4, 2017 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-28528906

RESUMO

Coordination of growth between individual organs and the whole body is essential during development to produce adults with appropriate size and proportions [1, 2]. How local organ-intrinsic signals and nutrient-dependent systemic factors are integrated to generate correctly proportioned organisms under different environmental conditions is poorly understood. In Drosophila, Hippo/Warts signaling functions intrinsically to regulate tissue growth and organ size [3, 4], whereas systemic growth is controlled via antagonistic interactions of the steroid hormone ecdysone and nutrient-dependent insulin/insulin-like growth factor (IGF) (insulin) signaling [2, 5]. The interplay between insulin and ecdysone signaling regulates systemic growth and controls organismal size. Here, we show that Warts (Wts; LATS1/2) signaling regulates systemic growth in Drosophila by activating basal ecdysone production, which negatively regulates body growth. Further, we provide evidence that Wts mediates effects of insulin and the neuropeptide prothoracicotropic hormone (PTTH) on regulation of ecdysone production through Yorkie (Yki; YAP/TAZ) and the microRNA bantam (ban). Thus, Wts couples insulin signaling with ecdysone production to adjust systemic growth in response to nutritional conditions during development. Inhibition of Wts activity in the ecdysone-producing cells non-autonomously slows the growth of the developing imaginal-disc tissues while simultaneously leading to overgrowth of the animal. This indicates that ecdysone, while restricting overall body growth, is limiting for growth of certain organs. Our data show that, in addition to its well-known intrinsic role in restricting organ growth, Wts/Yki/ban signaling also controls growth systemically by regulating ecdysone production, a mechanism that we propose controls growth between tissues and organismal size in response to nutrient availability.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiologia , Ecdisona/metabolismo , MicroRNAs/metabolismo , Proteínas Nucleares/metabolismo , Tamanho do Órgão/fisiologia , Proteínas Quinases/metabolismo , Transativadores/metabolismo , Animais , Feminino , Hormônios de Inseto/metabolismo , Insulina/metabolismo , Larva/fisiologia , Masculino , Pupa/fisiologia , Transdução de Sinais/fisiologia , Proteínas de Sinalização YAP
17.
Curr Biol ; 26(18): R855-R858, 2016 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-27676307

RESUMO

Circadian clocks are important timekeepers of physiological processes. A new report shows that silencing the circadian clock specifically in steroid-producing cells of Drosophila disrupts development and causes lethality, and is more detrimental than having no clock at all.


Assuntos
Relógios Circadianos , Ritmo Circadiano , Animais , Biologia do Desenvolvimento , Drosophila , Proteínas de Drosophila
18.
Antimicrob Agents Chemother ; 60(9): 5427-36, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27381394

RESUMO

We used the fruit fly Drosophila melanogaster as a cost-effective in vivo model to evaluate the efficacy of novel antibacterial peptides and peptoids for treatment of methicillin-resistant Staphylococcus aureus (MRSA) infections. A panel of peptides with known antibacterial activity in vitro and/or in vivo was tested in Drosophila Although most peptides and peptoids that were effective in vitro failed to rescue lethal effects of S. aureus infections in vivo, we found that two lantibiotics, nisin and NAI-107, rescued adult flies from fatal infections. Furthermore, NAI-107 rescued mortality of infection with the MRSA strain USA300 with an efficacy equivalent to that of vancomycin, a widely applied antibiotic for the treatment of serious MRSA infections. These results establish Drosophila as a useful model for in vivo drug evaluation of antibacterial peptides.


Assuntos
Antibacterianos/farmacologia , Bacteriocinas/farmacologia , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/veterinária , Sequência de Aminoácidos , Animais , Modelos Animais de Doenças , Drosophila melanogaster/efeitos dos fármacos , Drosophila melanogaster/microbiologia , Staphylococcus aureus Resistente à Meticilina/crescimento & desenvolvimento , Staphylococcus aureus Resistente à Meticilina/patogenicidade , Testes de Sensibilidade Microbiana , Nisina/farmacologia , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/mortalidade , Análise de Sobrevida
19.
Dev Cell ; 37(6): 558-70, 2016 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-27326933

RESUMO

Steroid hormones control important developmental processes and are linked to many diseases. To systematically identify genes and pathways required for steroid production, we performed a Drosophila genome-wide in vivo RNAi screen and identified 1,906 genes with potential roles in steroidogenesis and developmental timing. Here, we use our screen as a resource to identify mechanisms regulating intracellular levels of cholesterol, a substrate for steroidogenesis. We identify a conserved fatty acid elongase that underlies a mechanism that adjusts cholesterol trafficking and steroidogenesis with nutrition and developmental programs. In addition, we demonstrate the existence of an autophagosomal cholesterol mobilization mechanism and show that activation of this system rescues Niemann-Pick type C1 deficiency that causes a disorder characterized by cholesterol accumulation. These cholesterol-trafficking mechanisms are regulated by TOR and feedback signaling that couples steroidogenesis with growth and ensures proper maturation timing. These results reveal genes regulating steroidogenesis during development that likely modulate disease mechanisms.


Assuntos
Drosophila melanogaster/genética , Desenvolvimento Embrionário/genética , Testes Genéticos , Genoma de Inseto , Hormônios/biossíntese , Esteroides/biossíntese , Acetiltransferases/metabolismo , Animais , Autofagia/genética , Transporte Biológico/genética , Colesterol/metabolismo , Proteínas de Drosophila/metabolismo , Ecdisona/metabolismo , Elongases de Ácidos Graxos , Metabolismo dos Lipídeos/genética , Fenótipo , Interferência de RNA , Transdução de Sinais/genética , Esfingolipídeos/metabolismo , Fatores de Tempo
20.
Sci Rep ; 5: 11680, 2015 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-26123697

RESUMO

Coordinating metabolism and feeding is important to avoid obesity and metabolic diseases, yet the underlying mechanisms, balancing nutrient intake and metabolic expenditure, are poorly understood. Several mechanisms controlling these processes are conserved in Drosophila, where homeostasis and energy mobilization are regulated by the glucagon-related adipokinetic hormone (AKH) and the Drosophila insulin-like peptides (DILPs). Here, we provide evidence that the Drosophila neuropeptide Allatostatin A (AstA) regulates AKH and DILP signaling. The AstA receptor gene, Dar-2, is expressed in both the insulin and AKH producing cells. Silencing of Dar-2 in these cells results in changes in gene expression and physiology associated with reduced DILP and AKH signaling and animals lacking AstA accumulate high lipid levels. This suggests that AstA is regulating the balance between DILP and AKH, believed to be important for the maintenance of nutrient homeostasis in response to changing ratios of dietary sugar and protein. Furthermore, AstA and Dar-2 are regulated differentially by dietary carbohydrates and protein and AstA-neuronal activity modulates feeding choices between these types of nutrients. Our results suggest that AstA is involved in assigning value to these nutrients to coordinate metabolic and feeding decisions, responses that are important to balance food intake according to metabolic needs.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Neuropeptídeos/metabolismo , Animais , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/genética , Ingestão de Alimentos , Metabolismo Energético , Feminino , Hormônios de Inseto/antagonistas & inibidores , Hormônios de Inseto/genética , Hormônios de Inseto/metabolismo , Metabolismo dos Lipídeos , Masculino , Neurônios/metabolismo , Neuropeptídeos/antagonistas & inibidores , Neuropeptídeos/genética , Oligopeptídeos/antagonistas & inibidores , Oligopeptídeos/genética , Oligopeptídeos/metabolismo , Ácido Pirrolidonocarboxílico/análogos & derivados , Ácido Pirrolidonocarboxílico/antagonistas & inibidores , Ácido Pirrolidonocarboxílico/metabolismo , Interferência de RNA , RNA Guia de Cinetoplastídeos/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Neuropeptídeos/antagonistas & inibidores , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...