Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
PLoS One ; 10(7): e0133017, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26168160

RESUMO

The neuroendocrine peptides CCHamide-1 and -2, encoded by the genes ccha1 and -2, are produced by endocrine cells in the midgut and by neurons in the brain of Drosophila melanogaster. Here, we used the CRISPR/Cas9 technique to disrupt the ccha1 and -2 genes and identify mutant phenotypes with a focus on ccha-2 mutants. We found that both larval and adult ccha2 mutants showed a significantly reduced food intake as measured in adult flies by the Capillary Feeding (CAFE) assay (up to 72% reduced food intake compared to wild-type). Locomotion tests in adult flies showed that ccha2 mutants had a significantly reduced locomotor activity especially around 8 a.m. and 8 p.m., where adult Drosophila normally feeds (up to 70% reduced locomotor activity compared to wild-type). Reduced larval feeding is normally coupled to a delayed larval development, a process that is mediated by insulin. Accordingly, we found that the ccha2 mutants had a remarkably delayed development, showing pupariation 70 hours after the pupariation time point of the wild-type. In contrast, the ccha-1 mutants were not developmentally delayed. We also found that the ccha2 mutants had up to 80% reduced mRNA concentrations coding for the Drosophila insulin-like-peptides-2 and -3, while these concentrations were unchanged for the ccha1 mutants. From these experiments we conclude that CCHamide-2 is an orexigenic peptide and an important factor for controlling developmental timing in Drosophila.


Assuntos
Encéfalo/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Mucosa Intestinal/metabolismo , Neuropeptídeos/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , DNA , Drosophila/crescimento & desenvolvimento , Drosophila/fisiologia , Proteínas de Drosophila/genética , Comportamento Alimentar , Larva/metabolismo , Locomoção , Dados de Sequência Molecular , Mutação , Neuropeptídeos/genética
8.
PLoS Genet ; 10(6): e1004343, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24945799

RESUMO

Specialized endocrine cells produce and release steroid hormones that govern development, metabolism and reproduction. In order to synthesize steroids, all the genes in the biosynthetic pathway must be coordinately turned on in steroidogenic cells. In Drosophila, the steroid producing endocrine cells are located in the prothoracic gland (PG) that releases the steroid hormone ecdysone. The transcriptional regulatory network that specifies the unique PG specific expression pattern of the ecdysone biosynthetic genes remains unknown. Here, we show that two transcription factors, the POU-domain Ventral veins lacking (Vvl) and the nuclear receptor Knirps (Kni), have essential roles in the PG during larval development. Vvl is highly expressed in the PG during embryogenesis and is enriched in the gland during larval development, suggesting that Vvl might function as a master transcriptional regulator in this tissue. Vvl and Kni bind to PG specific cis-regulatory elements that are required for expression of the ecdysone biosynthetic genes. Knock down of either vvl or kni in the PG results in a larval developmental arrest due to failure in ecdysone production. Furthermore, Vvl and Kni are also required for maintenance of TOR/S6K and prothoracicotropic hormone (PTTH) signaling in the PG, two major pathways that control ecdysone biosynthesis and PG cell growth. We also show that the transcriptional regulator, Molting defective (Mld), controls early biosynthetic pathway steps. Our data show that Vvl and Kni directly regulate ecdysone biosynthesis by transcriptional control of biosynthetic gene expression and indirectly by affecting PTTH and TOR/S6K signaling. This provides new insight into the regulatory network of transcription factors involved in the coordinated regulation of steroidogenic cell specific transcription, and identifies a new function of Vvl and Knirps in endocrine cells during post-embryonic development.


Assuntos
Proteínas de Drosophila/metabolismo , Ecdisona/biossíntese , Hormônios de Inseto/biossíntese , Proteínas Nucleares/metabolismo , Fatores do Domínio POU/metabolismo , Proteínas Repressoras/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/biossíntese , Animais , Sítios de Ligação , Transporte Biológico/genética , Colesterol/metabolismo , Proteínas de Ligação a DNA , Proteínas de Drosophila/biossíntese , Proteínas de Drosophila/genética , Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Ecdisona/genética , Ecdisona/metabolismo , Regulação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Hormônios de Inseto/metabolismo , Proteínas de Membrana/biossíntese , Fatores do Domínio POU/biossíntese , Fatores do Domínio POU/genética , Interferência de RNA , RNA Interferente Pequeno , Proteínas Repressoras/biossíntese , Proteínas Repressoras/genética , Serina-Treonina Quinases TOR/biossíntese , Transcrição Gênica
9.
Development ; 140(23): 4730-9, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24173800

RESUMO

Steroid hormones trigger the onset of sexual maturation in animals by initiating genetic response programs that are determined by steroid pulse frequency, amplitude and duration. Although steroid pulses coordinate growth and timing of maturation during development, the mechanisms generating these pulses are not known. Here we show that the ecdysone steroid pulse that drives the juvenile-adult transition in Drosophila is determined by feedback circuits in the prothoracic gland (PG), the major steroid-producing tissue of insect larvae. These circuits coordinate the activation and repression of hormone synthesis, the two key parameters determining pulse shape (amplitude and duration). We show that ecdysone has a positive-feedback effect on the PG, rapidly amplifying its own synthesis to trigger pupariation as the onset of maturation. During the prepupal stage, a negative-feedback signal ensures the decline in ecdysone levels required to produce a temporal steroid pulse that drives developmental progression to adulthood. The feedback circuits rely on a developmental switch in the expression of Broad isoforms that transcriptionally activate or silence components in the ecdysone biosynthetic pathway. Remarkably, our study shows that the same well-defined genetic program that stimulates a systemic downstream response to ecdysone is also utilized upstream to set the duration and amplitude of the ecdysone pulse. Activation of this switch-like mechanism ensures a rapid, self-limiting PG response that functions in producing steroid oscillations that can guide the decision to terminate growth and promote maturation.


Assuntos
Drosophila melanogaster/embriologia , Ecdisona/metabolismo , Hormônios de Inseto/metabolismo , Maturidade Sexual/genética , Animais , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Ecdisona/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Larva/crescimento & desenvolvimento , Larva/metabolismo , Metamorfose Biológica/genética , Regiões Promotoras Genéticas , Transdução de Sinais
10.
Science ; 341(6150): 1113-6, 2013 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-24009394

RESUMO

Animal development is coupled with innate behaviors that maximize chances of survival. Here, we show that the prothoracicotropic hormone (PTTH), a neuropeptide that controls the developmental transition from juvenile stage to sexual maturation, also regulates light avoidance in Drosophila melanogaster larvae. PTTH, through its receptor Torso, acts on two light sensors--the Bolwig's organ and the peripheral class IV dendritic arborization neurons--to regulate light avoidance. We found that PTTH concomitantly promotes steroidogenesis and light avoidance at the end of larval stage, driving animals toward a darker environment to initiate the immobile maturation phase. Thus, PTTH controls the decisions of when and where animals undergo metamorphosis, optimizing conditions for adult development.


Assuntos
Drosophila melanogaster/crescimento & desenvolvimento , Reação de Fuga , Hormônios de Inseto/fisiologia , Transdução de Sinal Luminoso , Luz , Neurônios/fisiologia , Sistemas Neurossecretores/fisiologia , Animais , Escuridão , Proteínas de Drosophila/agonistas , Proteínas de Drosophila/genética , Ativação Enzimática , Hormônios de Inseto/genética , Larva/crescimento & desenvolvimento , Interferência de RNA , Receptores Proteína Tirosina Quinases/agonistas , Receptores Proteína Tirosina Quinases/genética
11.
Curr Top Dev Biol ; 105: 37-67, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23962838

RESUMO

In animals, developmental timing of sexual maturation is tightly linked to nutrition and growth. Maturation only occurs once the juvenile has acquired sufficient nutrients and completed enough growth to produce a reproductively mature adult with a genetically predefined body size. Animals therefore adjust the duration of juvenile development to the dietary conditions. When nutrients are scarce the juvenile growth phase is extended to compensate for slow growth. Conversely, development is accelerated in nutrient rich environments where animals rapidly reach their genetic target size. To achieve such flexibility, nutrient-dependent growth regulators must feed into the endocrine system that controls the timing of maturation. Work on the fruit fly Drosophila has revealed a central role of secreted signal molecules with similarity to the conserved insulin-like growth factors (IGFs) in the decision making process. These molecules are involved in checkpoints that allow the endocrine system to decide whether to release the steroid hormone, ecdysone, that triggers maturation or extent development, depending on nutrient levels and growth status. Importantly, different dietary components influence timing of maturation in Drosophila, with proteins having the greatest impact; fat and sugar play a minor role, at least within the limits of what can be considered a balanced diet. Remarkably, excess dietary sugar concentrations that mimic physiological conditions associated with diabetes, negatively affect growth and delays maturation. Altogether, this shows that the source of energy in the diet is important for timing and may provide a paradigm for understanding the emerging links between diet, obesity and diabetes, and the onset of puberty. Here, we provide an overview of the system underlying developmental timing of maturation in Drosophila and review recent success in understanding its coupling to nutrition and growth.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal/fisiologia , Proteínas Alimentares/farmacologia , Sacarose Alimentar/farmacologia , Metabolismo Energético/fisiologia , Crescimento e Desenvolvimento/fisiologia , Maturidade Sexual/fisiologia , Transdução de Sinais/fisiologia , Fenômenos Fisiológicos da Nutrição Animal/efeitos dos fármacos , Animais , Crescimento e Desenvolvimento/efeitos dos fármacos , Insulina/metabolismo , Modelos Biológicos , Maturidade Sexual/efeitos dos fármacos , Especificidade da Espécie , Serina-Treonina Quinases TOR/metabolismo , Fatores de Tempo
12.
PLoS One ; 8(2): e55131, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23383307

RESUMO

Insect steroid hormones (ecdysteroids) are important for female reproduction in many insect species and are required for the initiation and coordination of vital developmental processes. Ecdysteroids are also important for adult male physiology and behavior, but their exact function and site of synthesis remains unclear, although previous studies suggest that the reproductive system may be their source. We have examined expression profiles of the ecdysteroidogenic Halloween genes, during development and in adults of the flour beetle Tribolium castaneum. Genes required for the biosynthesis of ecdysone (E), the precursor of the molting hormone 20-hydroxyecdysone (20E), are expressed in the tubular accessory glands (TAGs) of adult males. In contrast, expression of the gene encoding the enzyme mediating 20E synthesis was detected in the ovaries of females. Further, Spookiest (Spot), an enzyme presumably required for endowing tissues with competence to produce ecdysteroids, is male specific and predominantly expressed in the TAGs. We also show that prothoracicotropic hormone (PTTH), a regulator of E synthesis during larval development, regulates ecdysteroid levels in the adult stage in Drosophila melanogaster and the gene for its receptor Torso seems to be expressed specifically in the accessory glands of males. The composite results suggest strongly that the accessory glands of adult male insects are the main source of E, but not 20E. The finding of a possible male-specific source of E raises the possibility that E and 20E have sex-specific roles analogous to the vertebrate sex steroids, where males produce primarily testosterone, the precursor of estradiol. Furthermore this study provides the first evidence that PTTH regulates ecdysteroid synthesis in the adult stage and could explain the original finding that some adult insects are a rich source of PTTH.


Assuntos
Drosophila melanogaster/metabolismo , Ecdisona/biossíntese , Glândulas Exócrinas/metabolismo , Hormônios de Inseto/metabolismo , Tribolium/metabolismo , Animais , Sistema Enzimático do Citocromo P-450/genética , Ecdisona/genética , Ecdisterona/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Hibridização In Situ , Masculino , Microscopia de Fluorescência , Ovário/metabolismo , Reação em Cadeia da Polimerase , Interferência de RNA
13.
Curr Top Dev Biol ; 103: 1-33, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23347514

RESUMO

The transition from juvenile to adult is a fundamental process that allows animals to allocate resource toward reproduction after completing a certain amount of growth. In insects, growth to a species-specific target size induces pulses of the steroid hormone ecdysone that triggers metamorphosis and reproductive maturation. The past few years have seen significant progress in understanding the interplay of mechanisms that coordinate timing of ecdysone production and release. These studies show that the neuroendocrine system monitors complex size-related and nutritional signals, as well as external cues, to time production and release of ecdysone. Based on results discussed here, we suggest that developmental progression to adulthood is controlled by checkpoints that regulate the genetic timing program enabling it to adapt to different environmental conditions. These checkpoints utilize a number of signaling pathways to modulate ecdysone production in the prothoracic gland. Release of ecdysone activates an autonomous cascade of both feedforward and feedback signals that determine the duration of the ecdysone pulse at each developmental transitions. Conservation of the genetic mechanisms that coordinate the juvenile-adult transition suggests that insights from the fruit fly Drosophila will provide a framework for future investigation of developmental timing in metazoans.


Assuntos
Retroalimentação Fisiológica , Insetos/crescimento & desenvolvimento , Metamorfose Biológica , Estruturas Animais/crescimento & desenvolvimento , Animais , Ecdisona/metabolismo , Transdução de Sinais , Fatores de Tempo
14.
Annu Rev Entomol ; 58: 497-516, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23072462

RESUMO

The steroid hormone ecdysone is the central regulator of insect developmental transitions. Recent new advances in our understanding of ecdysone action have relied heavily on the application of Drosophila melanogaster molecular genetic tools to study insect metamorphosis. In this review, we focus on three major aspects of Drosophila ecdysone biology: (a) factors that regulate the timing of ecdysone release, (b) molecular basis of stage- and tissue-specific responses to ecdysone, and (c) feedback regulation and coordination of ecdysone signaling.


Assuntos
Drosophila melanogaster/fisiologia , Ecdisona/metabolismo , Transdução de Sinais , Animais , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Metamorfose Biológica
15.
Artigo em Inglês | MEDLINE | ID: mdl-22649397

RESUMO

During development, multicellular organisms must become sexually mature in order to reproduce. The developmental timing of this transition is controlled by pulses of steroid hormones, but how these pulses are generated have remained unclear? A recent paper shows that in Drosophila larvae, nucleocytoplasmic trafficking of DHR4, a nuclear receptor, in response to prothoracicotropic hormone signaling, is critical for producing the correct temporal pulses of steroid hormones that coordinate the juvenile-adult transition.

16.
Dev Cell ; 19(6): 895-902, 2010 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-21145504

RESUMO

Steroid hormones are systemic signaling molecules that regulate juvenile-adult transitions in both insects and mammals. In insects, pulses of the steroid hormone 20-hydroxyecdysone (20E) are generated by increased biosynthesis followed by inactivation/clearance. Although mechanisms that control 20E synthesis have received considerable recent attention, the physiological significance of 20E inactivation remains largely unknown. We show that the cytochrome P450 Cyp18a1 lowers 20E titer during the Drosophila prepupal to pupal transition. Furthermore, this reduction of 20E levels is a prerequisite to induce ßFTZ-F1, a key factor in the genetic hierarchy that controls early metamorphosis. Resupplying ßFTZ-F1 rescues Cyp18a1-deficient prepupae. Because Cyp18a1 is 20E-inducible, it appears that the increased production of steroid is responsible for its eventual decline, thereby generating the regulatory pulse required for proper temporal progression of metamorphosis. The coupling of hormone clearance to ßFTZ-F1 expression suggests a general mechanism by which transient signaling drives unidirectional progression through a multistep process.


Assuntos
Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Ecdisterona/antagonistas & inibidores , Animais , Animais Geneticamente Modificados , Sequência de Bases , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Primers do DNA/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Ecdisterona/genética , Ecdisterona/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genes de Insetos , Metamorfose Biológica , Mutação , Fenótipo , Interferência de RNA , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Transdução de Sinais
17.
Science ; 326(5958): 1403-5, 2009 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-19965758

RESUMO

Holometabolous insects undergo complete metamorphosis to become sexually mature adults. Metamorphosis is initiated by brain-derived prothoracicotropic hormone (PTTH), which stimulates the production of the molting hormone ecdysone via an incompletely defined signaling pathway. Here we demonstrate that Torso, a receptor tyrosine kinase that regulates embryonic terminal cell fate in Drosophila, is the PTTH receptor. Trunk, the embryonic Torso ligand, is related to PTTH, and ectopic expression of PTTH in the embryo partially rescues trunk mutants. In larvae, torso is expressed specifically in the prothoracic gland (PG), and its loss phenocopies the removal of PTTH. The activation of Torso by PTTH stimulates extracellular signal-regulated kinase (ERK) phosphorylation, and the loss of ERK in the PG phenocopies the loss of PTTH and Torso. We conclude that PTTH initiates metamorphosis by activation of the Torso/ERK pathway.


Assuntos
Bombyx/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crescimento & desenvolvimento , Hormônios de Inseto/metabolismo , Metamorfose Biológica , Receptores Proteína Tirosina Quinases/metabolismo , Sequência de Aminoácidos , Animais , Bombyx/metabolismo , Linhagem Celular , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Embrião não Mamífero/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hormônios de Inseto/química , Larva/crescimento & desenvolvimento , Ligantes , Dados de Sequência Molecular , Neurônios/metabolismo , Fosforilação , Pupa/crescimento & desenvolvimento , Interferência de RNA , Receptores Proteína Tirosina Quinases/genética , Transdução de Sinais
18.
Insect Biochem Mol Biol ; 39(7): 475-83, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19422916

RESUMO

In insects, the neuropeptide prothoracicotropic hormone (PTTH) stimulates production of ecdysone (E) in the prothoracic glands (PGs). E is the precursor of the principal steroid hormone, 20-hydroxyecdysone (20E), that is responsible for eliciting molting and metamorphosis. In this study, we used quantitative phosphoproteomics to investigate signal transduction events initiated by PTTH. We identified Spook (CYP307A1), a suspected rate-limiting enzyme for E biosynthesis, and components of the mitogen-activated protein kinase (MAPK) pathway, as major phosphorylation targets of PTTH signaling. Further, proteins not previously linked to PTTH and ecdysone biosynthesis were identified as targets of PTTH signaling. These include proteins involved in signal transduction, endosomal trafficking, constituents of the cytoskeleton and regulators of transcription and translation. Our screen shows that PTTH likely stimulates E production by activation of Spook, an integral enzyme in the E biosynthetic pathway. This directly connects PTTH signaling to the pathway that produces E. A new mechanism for regulation of E biosynthesis in insects is proposed.


Assuntos
Hormônios de Inseto/metabolismo , Proteínas de Insetos/metabolismo , Manduca/crescimento & desenvolvimento , Muda , Fosfoproteínas/metabolismo , Proteômica/métodos , Transdução de Sinais , Sequência de Aminoácidos , Animais , Ecdisona/biossíntese , Ecdisona/genética , Hormônios de Inseto/genética , Proteínas de Insetos/química , Proteínas de Insetos/genética , Manduca/química , Manduca/genética , Manduca/metabolismo , Metamorfose Biológica , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Dados de Sequência Molecular , Fosfoproteínas/química , Fosfoproteínas/genética , Fosforilação , Alinhamento de Sequência
19.
Biochem Biophys Res Commun ; 377(4): 1135-40, 2008 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-18983823

RESUMO

The shore crab Carcinus maenas has a high capacity for metabolizing polycyclic aromatic hydrocarbons (PAHs). Cytochrome P450 (CYP) enzymes are involved in this metabolism and also have a role in development and reproduction. This investigation is a systematic gene expression analysis of six CYPs in C. maenas. Expression of CYP2 and CYP3-like genes was predominant in hepatopancreas, while expression of CYP4-like genes was predominant in gills and epidermis. Expression of all six CYP genes fluctuated over the moult cycle in the hepatopancreas and structurally related genes were regulated coordinately. The study suggests that hepatopancreas is a major site of CYP gene expression in C. maenas confirming previous biochemical studies showing that this tissue is the major compartment for CYP mediated xenobiotic metabolism in crustaceans. In addition, the data show that CYP2 and CYP3 related genes respond to ecdysteroid and xenobiotic treatment, while those related to CYP4 genes do not and likely are involved in a more general physiological function such as fatty acid metabolism. The developmental variations of CYP expression suggest a molecular mechanism for the stage specific susceptibility of crabs exposed to environmental pollutants.


Assuntos
Braquiúros/enzimologia , Sistema Enzimático do Citocromo P-450/genética , Regulação Enzimológica da Expressão Gênica , Muda/genética , Animais , Braquiúros/efeitos dos fármacos , Braquiúros/genética , Braquiúros/crescimento & desenvolvimento , Ecdisteroides/metabolismo , Ecdisteroides/farmacologia , Expressão Gênica/efeitos dos fármacos , Hidrocarbonetos Policíclicos Aromáticos/metabolismo , Análise de Sequência de Proteína , Xenobióticos/metabolismo , Xenobióticos/farmacologia
20.
BMC Evol Biol ; 8: 60, 2008 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-18298845

RESUMO

BACKGROUND: In crustaceans and insects, development and reproduction are controlled by the steroid hormone, 20-hydroxyecdysone (20E). Like other steroids, 20E, is synthesized from cholesterol through reactions involving cytochrome P450s (CYPs). In insects, the CYP enzymes mediating 20E biosynthesis have been identified, but evidence of their probable presence in crustaceans is indirect, relying solely on the ability of crustaceans to synthesize 20E. RESULTS: To investigate the presence of these genes in crustaceans, the genome of Daphnia pulex was examined for orthologs of these genes, the Halloween genes, encoding those biosynthetic CYP enzymes. Single homologs of spook-CYP307A1, phantom-CYP306A1, disembodied-CYP302A1, shadow-CYP315A1 and shade-CYP314A1 were identified in the Daphnia data base. Phylogenetic analysis indicates an orthologous relationship between the insect and Daphnia genes. Conserved intron/exon structures and microsynteny further support the conclusion that these steroidogenic CYPs have been conserved in insects and crustaceans through some 400 million years of evolution. CONCLUSION: Although these arthropod steroidogenic CYPs are related to steroidogenic CYPs in Caenorhabditis elegans and vertebrates, the data suggest that the arthropod steroidogenic CYPs became functionally specialized in a common ancestor of arthropods and are unique to these animals.


Assuntos
Sistema Enzimático do Citocromo P-450/genética , Daphnia/genética , Ecdisona/biossíntese , Evolução Molecular , Genes de Insetos , Animais , Daphnia/metabolismo , Bases de Dados de Ácidos Nucleicos , Regulação Enzimológica da Expressão Gênica , Íntrons , Filogenia , Sintenia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...