Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Development ; 147(14)2020 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-32631830

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila/metabolismo , Hormonas de Insectos/metabolismo , Receptores de Esteroides/metabolismo , Animales , Tamaño Corporal , Drosophila/crecimiento & desarrollo , Drosophila/metabolismo , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Ecdisterona/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hormonas de Insectos/antagonistas & inhibidores , Hormonas de Insectos/genética , Larva/crecimiento & desarrollo , Larva/metabolismo , Metamorfosis Biológica , Microscopía Fluorescente , Neuronas/metabolismo , Interferencia de ARN , ARN Guía de Kinetoplastida/metabolismo , Receptores de Esteroides/antagonistas & inhibidores , Receptores de Esteroides/genética , Transducción de Señal
2.
PLoS Genet ; 16(4): e1008727, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32339168

RESUMEN

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.


Asunto(s)
Síndrome de Deleción 22q11/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas de Drosophila/genética , Neuronas GABAérgicas/metabolismo , Neurofibromina 1/genética , Esquizofrenia/genética , Sueño/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Drosophila , Proteínas de Drosophila/metabolismo , Neuronas GABAérgicas/fisiología , Humanos , Neurofibromina 1/metabolismo , Receptores de GABA-A/metabolismo , Factores de Transcripción/genética
3.
PLoS Biol ; 16(9): e2005004, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30248087

RESUMEN

Stem cell maintenance is established by neighboring niche cells that promote stem cell self-renewal. However, it is poorly understood how stem cell activity is regulated by systemic, tissue-extrinsic signals in response to environmental cues and changes in physiological status. Here, we show that neuropeptide F (NPF) signaling plays an important role in the pathway regulating mating-induced germline stem cell (GSC) proliferation in the fruit fly Drosophila melanogaster. NPF expressed in enteroendocrine cells (EECs) of the midgut is released in response to the seminal-fluid protein sex peptide (SP) upon mating. This midgut-derived NPF controls mating-induced GSC proliferation via ovarian NPF receptor (NPFR) activity, which modulates bone morphogenetic protein (BMP) signaling levels in GSCs. Our study provides a molecular mechanism that describes how a gut-derived systemic factor couples stem cell behavior to physiological status, such as mating, through interorgan communication.


Asunto(s)
Sistema Digestivo/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Células Germinativas/citología , Neuropéptidos/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Secuencia de Bases , Proteínas Morfogenéticas Óseas/metabolismo , Recuento de Células , División Celular , Proliferación Celular , Ecdisteroides/metabolismo , Células Enteroendocrinas/metabolismo , Femenino , Células Germinativas/metabolismo , Modelos Biológicos , Ovario/metabolismo , Receptores de Neuropéptido/metabolismo , Conducta Sexual Animal , Transducción de Señal
4.
Cell Mol Life Sci ; 77(22): 4523-4551, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32448994

RESUMEN

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.


Asunto(s)
Adaptación Fisiológica/fisiología , Drosophila/metabolismo , Drosophila/fisiología , Animales , Proteínas de Drosophila/metabolismo , Homeostasis/fisiología , Humanos , Hormonas de Insectos/metabolismo , Transducción de Señal/fisiología
5.
PLoS Genet ; 14(11): e1007767, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30457986

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Neuropéptidos/genética , Neuropéptidos/fisiología , Animales , Animales Modificados Genéticamente , Conducta Animal/fisiología , Diuresis/genética , Diuresis/fisiología , Proteínas de Drosophila/deficiencia , Metabolismo Energético/genética , Metabolismo Energético/fisiología , Femenino , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Insulina/fisiología , Masculino , Actividad Motora/genética , Actividad Motora/fisiología , Mutación , Neuronas/clasificación , Neuronas/fisiología , Neuropéptidos/deficiencia , Periodo Posprandial/genética , Periodo Posprandial/fisiología , Receptores de Neuropéptido/deficiencia , Receptores de Neuropéptido/genética , Receptores de Neuropéptido/fisiología , Transducción de Señal
6.
Proc Natl Acad Sci U S A ; 113(9): E1256-65, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26884206

RESUMEN

The development of sexually dimorphic morphology and the potential for sexually dimorphic behavior in Drosophila are regulated by the Fruitless (Fru) and Doublesex (Dsx) transcription factors. Several direct targets of Dsx have been identified, but direct Fru targets have not been definitively identified. We show that Drosophila leucine-rich repeat G protein-coupled receptor 3 (Lgr3) is regulated by Fru and Dsx in separate populations of neurons. Lgr3 is a member of the relaxin-receptor family and a receptor for Dilp8, necessary for control of organ growth. Lgr3 expression in the anterior central brain of males is inhibited by the B isoform of Fru, whose DNA binding domain interacts with a short region of an Lgr3 intron. Fru A and C isoform mutants had no observed effect on Lgr3 expression. The female form of Dsx (Dsx(F)) separately up- and down-regulates Lgr3 expression in distinct neurons in the abdominal ganglion through female- and male-specific Lgr3 enhancers. Excitation of neural activity in the Dsx(F)-up-regulated abdominal ganglion neurons inhibits female receptivity, indicating the importance of these neurons for sexual behavior. Coordinated regulation of Lgr3 by Fru and Dsx marks a point of convergence of the two branches of the sex-determination hierarchy.


Asunto(s)
Proteínas de Drosophila/metabolismo , Neuronas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Factores Sexuales , Animales , Drosophila , Femenino , Masculino
7.
PLoS Genet ; 11(5): e1005209, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-26020940

RESUMEN

The coordination of growth with nutritional status is essential for proper development and physiology. Nutritional information is mostly perceived by peripheral organs before being relayed to the brain, which modulates physiological responses. Hormonal signaling ensures this organ-to-organ communication, and the failure of endocrine regulation in humans can cause diseases including obesity and diabetes. In Drosophila melanogaster, the fat body (adipose tissue) has been suggested to play an important role in coupling growth with nutritional status. Here, we show that the peripheral tissue-derived peptide hormone CCHamide-2 (CCHa2) acts as a nutrient-dependent regulator of Drosophila insulin-like peptides (Dilps). A BAC-based transgenic reporter revealed strong expression of CCHa2 receptor (CCHa2-R) in insulin-producing cells (IPCs) in the brain. Calcium imaging of brain explants and IPC-specific CCHa2-R knockdown demonstrated that peripheral-tissue derived CCHa2 directly activates IPCs. Interestingly, genetic disruption of either CCHa2 or CCHa2-R caused almost identical defects in larval growth and developmental timing. Consistent with these phenotypes, the expression of dilp5, and the release of both Dilp2 and Dilp5, were severely reduced. Furthermore, transcription of CCHa2 is altered in response to nutritional levels, particularly of glucose. These findings demonstrate that CCHa2 and CCHa2-R form a direct link between peripheral tissues and the brain, and that this pathway is essential for the coordination of systemic growth with nutritional availability. A mammalian homologue of CCHa2-R, Bombesin receptor subtype-3 (Brs3), is an orphan receptor that is expressed in the islet ß-cells; however, the role of Brs3 in insulin regulation remains elusive. Our genetic approach in Drosophila melanogaster provides the first evidence, to our knowledge, that bombesin receptor signaling with its endogenous ligand promotes insulin production.


Asunto(s)
Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/genética , Insulina/metabolismo , Insulinas/biosíntesis , Neuropéptidos/genética , Receptores de Bombesina/genética , Receptores Odorantes/genética , Animales , Animales Modificados Genéticamente , Encéfalo/metabolismo , Drosophila melanogaster , Cuerpo Adiposo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Células Secretoras de Insulina/metabolismo , Insulinas/genética , Neuropéptidos/biosíntesis , Receptores Odorantes/biosíntesis
9.
Exp Cell Res ; 317(4): 474-87, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21126519

RESUMEN

The Drosophila gene yuri gagarin is a complex locus encoding three protein isoform classes that are ubiquitously expressed in the organism. Mutations to the gene affect processes as diverse as gravitactic behavior and spermatogenesis. The larger Yuri isoforms contain extensive coiled-coil regions. Our previous studies indicate that one of the large isoform classes (Yuri-65) is required for formation of specialized F-actin-containing structures generated during spermatogenesis, including the so-called actin "cones" that mediate spermatid individualization. We used the tandem affinity purification of a tagged version of Yuri-65 (the TAP-tagging technique) to identify proteins associated with Yuri-65 in the intact organism. Tropomyosin, primarily as the 284-residue isoform derived from the ubiquitously expressed Tropomyosin 1 gene was thus identified as a major Yuri interaction partner. Co-immunoprecipitation experiments confirmed this interaction. We have established that the stable F-actin cones of spermatogenesis contain Tropomyosin 1 (Tm1) and that in mutant yuri(F64), failure of F-actin cone formation is associated with failure of Tm1 to accumulate at the cone initiation sites. In investigating possible interactions of Tm1 and Yuri in other tissues, we discovered that Tm1 and Yuri frequently colocalize with the endoplasmic reticulum. Tropomyosin has been implicated in actin-mediated membrane trafficking activity in other systems. Our findings suggest that Yuri-Tm1 complexes participate in related functions.


Asunto(s)
Proteínas de Drosophila/metabolismo , Mapeo de Interacción de Proteínas , Tropomiosina/metabolismo , Actinas , Animales , Drosophila , Proteínas de Drosophila/análisis , Retículo Endoplásmico/química , Inmunoprecipitación , Unión Proteica , Transporte de Proteínas , Tropomiosina/análisis
10.
Curr Biol ; 32(7): 1548-1562.e6, 2022 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-35245460

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila , Obesidad Infantil , Animales , Colesterol , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Insulina/metabolismo , Larva , Mamíferos , Esteroides/metabolismo
11.
Nat Metab ; 4(11): 1532-1550, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36344765

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila , Hormonas Gastrointestinales , Femenino , Animales , Drosophila , Apetito , Azúcares , Proteínas de Drosophila/genética , Mamíferos
12.
Nat Commun ; 13(1): 692, 2022 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-35121731

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila/metabolismo , Ingestión de Alimentos/fisiología , Metabolismo Energético/fisiología , Hormonas Gastrointestinales/metabolismo , Homeostasis , Nutrientes/metabolismo , Somatostatina/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Ingestión de Alimentos/genética , Metabolismo Energético/genética , Células Enteroendocrinas/metabolismo , Hormonas Gastrointestinales/genética , Técnicas de Inactivación de Genes , Humanos , Hipoglucemia/genética , Hipoglucemia/metabolismo , Hormonas de Insectos/genética , Hormonas de Insectos/metabolismo , Oligopéptidos/genética , Oligopéptidos/metabolismo , Ácido Pirrolidona Carboxílico/análogos & derivados , Ácido Pirrolidona Carboxílico/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/genética , Somatostatina/genética , Análisis de Supervivencia
13.
Genetics ; 216(2): 269-313, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33023929

RESUMEN

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.


Asunto(s)
Tamaño Corporal , Drosophila melanogaster/genética , Regulación del Desarrollo de la Expresión Génica , Animales , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Estadios del Ciclo de Vida , Transducción de Señal
14.
Autophagy ; 15(8): 1478-1480, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31084464

RESUMEN

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.


Asunto(s)
Autofagia , Colesterol/metabolismo , Sistema Endocrino/citología , Animales , Autofagosomas/metabolismo , Transporte Biológico , Drosophila melanogaster/metabolismo , Humanos , Fusión de Membrana , Modelos Biológicos
15.
Dev Cell ; 48(5): 659-671.e4, 2019 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-30799225

RESUMEN

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.


Asunto(s)
Autofagia/fisiología , Movimiento Celular/fisiología , Colesterol/metabolismo , Ecdisona/metabolismo , Regulación del Desarrollo de la Expresión Génica , Animales , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , MicroARNs/genética , Proteínas Nucleares/metabolismo , Transactivadores/metabolismo
16.
Nat Commun ; 10(1): 1955, 2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-31028268

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila/metabolismo , Animales , Proteínas de Unión al ADN/metabolismo , Drosophila , Proteínas de Drosophila/genética , Regulación del Desarrollo de la Expresión Génica , Secreción de Insulina/genética , Secreción de Insulina/fisiología , Oxígeno/metabolismo , Factores de Transcripción/metabolismo
17.
Curr Biol ; 27(11): 1652-1659.e4, 2017 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-28528906

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Ecdisona/metabolismo , MicroARNs/metabolismo , Proteínas Nucleares/metabolismo , Tamaño de los Órganos/fisiología , Proteínas Quinasas/metabolismo , Transactivadores/metabolismo , Animales , Femenino , Hormonas de Insectos/metabolismo , Insulina/metabolismo , Larva/fisiología , Masculino , Pupa/fisiología , Transducción de Señal/fisiología , Proteínas Señalizadoras YAP
18.
Elife ; 52016 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-27494275

RESUMEN

Neuronal circuits are known to integrate nutritional information, but the identity of the circuit components is not completely understood. Amino acids are a class of nutrients that are vital for the growth and function of an organism. Here, we report a neuronal circuit that allows Drosophila larvae to overcome amino acid deprivation and pupariate. We find that nutrient stress is sensed by the class IV multidendritic cholinergic neurons. Through live calcium imaging experiments, we show that these cholinergic stimuli are conveyed to glutamatergic neurons in the ventral ganglion through mAChR. We further show that IP3R-dependent calcium transients in the glutamatergic neurons convey this signal to downstream medial neurosecretory cells (mNSCs). The circuit ultimately converges at the ring gland and regulates expression of ecdysteroid biosynthetic genes. Activity in this circuit is thus likely to be an adaptation that provides a layer of regulation to help surpass nutritional stress during development.


Asunto(s)
Señalización del Calcio , Drosophila/crecimiento & desarrollo , Drosophila/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Interneuronas/fisiología , Aminoácidos/metabolismo , Animales , Neuronas Colinérgicas/fisiología , Ecdisteroides/biosíntesis , Alimentos , Larva/metabolismo , Red Nerviosa , Sistemas Neurosecretores/fisiología , Pupa/metabolismo
19.
Elife ; 52016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27845623

RESUMEN

NeuromedinU is a potent regulator of food intake and activity in mammals. In Drosophila, neurons producing the homologous neuropeptide hugin regulate feeding and locomotion in a similar manner. Here, we use EM-based reconstruction to generate the entire connectome of hugin-producing neurons in the Drosophila larval CNS. We demonstrate that hugin neurons use synaptic transmission in addition to peptidergic neuromodulation and identify acetylcholine as a key transmitter. Hugin neuropeptide and acetylcholine are both necessary for the regulatory effect on feeding. We further show that subtypes of hugin neurons connect chemosensory to endocrine system by combinations of synaptic and peptide-receptor connections. Targets include endocrine neurons producing DH44, a CRH-like peptide, and insulin-like peptides. Homologs of these peptides are likewise downstream of neuromedinU, revealing striking parallels in flies and mammals. We propose that hugin neurons are part of an ancient physiological control system that has been conserved at functional and molecular level.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/anatomía & histología , Drosophila/fisiología , Ingestión de Alimentos , Vías Nerviosas/anatomía & histología , Neuronas/metabolismo , Neuropéptidos/metabolismo , Transmisión Sináptica/efectos de los fármacos , Acetilcolina/metabolismo , Animales , Larva/anatomía & histología , Larva/fisiología , Microscopía Electrónica , Neurotransmisores/metabolismo
20.
Gravit Space Biol Bull ; 18(2): 17-29, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16038090

RESUMEN

Drosophila melanogaster has been intensely studied for almost 100 years. The sophisticated array of genetic and molecular tools that have evolved for analysis of gene function in this organism are unique. Further, Drosophila is a complex multi-cellular organism in which many aspects of development and behavior parallel those in human beings. These combined advantages have permitted research in Drosophila to make seminal contributions to the understanding of fundamental biological processes and ensure that Drosophila will continue to provide unique insights in the genomic era. An overview of the genetic methodologies available in Drosophila is given here, together with examples of outstanding recent contributions of Drosophila to our understanding of cell and organismal biology. The growing contribution of Drosophila to our knowledge of gravity-related responses is addressed.


Asunto(s)
Drosophila melanogaster/genética , Genes de Insecto , Genómica , Modelos Biológicos , Animales , Biología Evolutiva , Regulación del Desarrollo de la Expresión Génica , Gravitación , Sensación de Gravedad/fisiología , Mutagénesis , Vuelo Espacial , Ingravidez
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA