Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.952
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Immunity ; 54(8): 1683-1697.e3, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34107298

RESUMO

Microbe-derived acetate activates the Drosophila immunodeficiency (IMD) pathway in a subset of enteroendocrine cells (EECs) of the anterior midgut. In these cells, the IMD pathway co-regulates expression of antimicrobial and enteroendocrine peptides including tachykinin, a repressor of intestinal lipid synthesis. To determine whether acetate acts on a cell surface pattern recognition receptor or an intracellular target, we asked whether acetate import was essential for IMD signaling. Mutagenesis and RNA interference revealed that the putative monocarboxylic acid transporter Tarag was essential for enhancement of IMD signaling by dietary acetate. Interference with histone deacetylation in EECs augmented transcription of genes regulated by the steroid hormone ecdysone including IMD targets. Reduced expression of the histone acetyltransferase Tip60 decreased IMD signaling and blocked rescue by dietary acetate and other sources of intracellular acetyl-CoA. Thus, microbe-derived acetate induces chromatin remodeling within enteroendocrine cells, co-regulating host metabolism and intestinal innate immunity via a Tip60-steroid hormone axis that is conserved in mammals.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/imunologia , Células Enteroendócrinas/metabolismo , Microbioma Gastrointestinal/imunologia , Histona Acetiltransferases/metabolismo , Intestinos/imunologia , Acetatos/imunologia , Acetilcoenzima A/metabolismo , Animais , Montagem e Desmontagem da Cromatina/fisiologia , Drosophila melanogaster/microbiologia , Ecdisona/metabolismo , Imunidade Inata/imunologia , Intestinos/microbiologia , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Interferência de RNA , Transdução de Sinais/imunologia , Taquicininas/metabolismo
2.
Cell ; 163(4): 907-19, 2015 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-26544939

RESUMO

Steroid hormones are a large family of cholesterol derivatives regulating development and physiology in both the animal and plant kingdoms, but little is known concerning mechanisms of their secretion from steroidogenic tissues. Here, we present evidence that in Drosophila, endocrine release of the steroid hormone ecdysone is mediated through a regulated vesicular trafficking mechanism. Inhibition of calcium signaling in the steroidogenic prothoracic gland results in the accumulation of unreleased ecdysone, and the knockdown of calcium-mediated vesicle exocytosis components in the gland caused developmental defects due to deficiency of ecdysone. Accumulation of synaptotagmin-labeled vesicles in the gland is observed when calcium signaling is disrupted, and these vesicles contain an ABC transporter that functions as an ecdysone pump to fill vesicles. We propose that trafficking of steroid hormones out of endocrine cells is not always through a simple diffusion mechanism as presently thought, but instead can involve a regulated vesicle-mediated release process.


Assuntos
Drosophila melanogaster/metabolismo , Ecdisona/metabolismo , Vesículas Secretórias/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Difusão , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/crescimento & desenvolvimento , Glândulas Endócrinas/metabolismo , Exocitose , Técnicas de Silenciamento de Genes , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Larva/citologia , Larva/metabolismo , Sinaptotagminas/metabolismo
3.
Cell ; 158(4): 874-888, 2014 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-25126791

RESUMO

Stem cells are highly abundant during early development but become a rare population in most adult organs. The molecular mechanisms causing stem cells to exit proliferation at a specific time are not well understood. Here, we show that changes in energy metabolism induced by the steroid hormone ecdysone and the Mediator initiate an irreversible cascade of events leading to cell-cycle exit in Drosophila neural stem cells. We show that the timely induction of oxidative phosphorylation and the mitochondrial respiratory chain are required in neuroblasts to uncouple the cell cycle from cell growth. This results in a progressive reduction in neuroblast cell size and ultimately in terminal differentiation. Brain tumor mutant neuroblasts fail to undergo this shrinkage process and continue to proliferate until adulthood. Our findings show that cell size control can be modified by systemic hormonal signaling and reveal a unique connection between metabolism and proliferation in stem cells.


Assuntos
Proliferação de Células , Drosophila melanogaster/citologia , Ecdisona/metabolismo , Células-Tronco Neurais/citologia , Animais , Tamanho Celular , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Metabolismo Energético , Genoma de Inseto , Complexo Mediador/metabolismo , Células-Tronco Neurais/metabolismo
4.
Nature ; 603(7899): 112-118, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35197627

RESUMO

The assembly of neural circuits is dependent on precise spatiotemporal expression of cell recognition molecules1-5. Factors controlling cell type specificity have been identified6-8, but how timing is determined remains unknown. Here we describe induction of a cascade of transcription factors by a steroid hormone (ecdysone) in all fly visual system neurons spanning target recognition and synaptogenesis. We demonstrate through single-cell sequencing that the ecdysone pathway regulates the expression of a common set of targets required for synaptic maturation and cell-type-specific targets enriched for cell-surface proteins regulating wiring specificity. Transcription factors in the cascade regulate the expression of the same wiring genes in complex ways, including activation in one cell type and repression in another. We show that disruption of the ecdysone pathway generates specific defects in dendritic and axonal processes and synaptic connectivity, with the order of transcription factor expression correlating with sequential steps in wiring. We also identify shared targets of a cell-type-specific transcription factor and the ecdysone pathway that regulate specificity. We propose that neurons integrate a global temporal transcriptional module with cell-type-specific transcription factors to generate different cell-type-specific patterns of cell recognition molecules regulating wiring.


Assuntos
Proteínas de Drosophila , Ecdisona , Animais , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiologia , Ecdisona/metabolismo , Regulação da Expressão Gênica , Sinapses/metabolismo , Fatores de Transcrição/metabolismo
5.
Mol Cell ; 77(2): 205-206, 2020 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-31951544

RESUMO

Glastad et al. (2019) describe a role for the neuronal CoREST corepressor and changes in juvenile hormone (JH) and ecdysone signaling during the reprogramming of social behavioral phenotypes in ants that are reflective of a natural mechanism differentiating "Major" and "Minor" worker ants.


Assuntos
Formigas , Animais , Ecdisona , Epigênese Genética , Hormônios Juvenis , Comportamento Social
6.
Development ; 151(12)2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38885078

RESUMO

The steroid hormone ecdysone (Ec) is secreted from the prothoracic gland for growth in the developing Drosophila larva. How Ec-dependent regeneration can occur despite a drop in circulating Ec in the injured developing larvae remains unclear. In a new study in Development, Kenneth Moberg and colleagues find that injury induces local Ec synthesis at the wounded site to delay development and promote tissue repair in Drosophila. To learn more about the story behind the paper, we caught up with first author Douglas Terry and corresponding author Kenneth Moberg, Professor of Cell Biology at Emory University School of Medicine, USA.


Assuntos
Ecdisona , Animais , Ecdisona/metabolismo , Drosophila , História do Século XXI , História do Século XX , Biologia do Desenvolvimento/história , Humanos , Larva/crescimento & desenvolvimento , Drosophila melanogaster
7.
Development ; 151(12)2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38775023

RESUMO

Regenerative ability often declines as animals mature past embryonic and juvenile stages, suggesting that regeneration requires redirection of growth pathways that promote developmental growth. Intriguingly, the Drosophila larval epithelia require the hormone ecdysone (Ec) for growth but require a drop in circulating Ec levels to regenerate. Examining Ec dynamics more closely, we find that transcriptional activity of the Ec-receptor (EcR) drops in uninjured regions of wing discs, but simultaneously rises in cells around the injury-induced blastema. In parallel, blastema depletion of genes encoding Ec biosynthesis enzymes blocks EcR activity and impairs regeneration but has no effect on uninjured wings. We find that local Ec/EcR signaling is required for injury-induced pupariation delay following injury and that key regeneration regulators upd3 and Ets21c respond to Ec levels. Collectively, these data indicate that injury induces a local source of Ec within the wing blastema that sustains a transcriptional signature necessary for developmental delay and tissue repair.


Assuntos
Proteínas de Drosophila , Ecdisona , Regeneração , Asas de Animais , Animais , Ecdisona/metabolismo , Asas de Animais/metabolismo , Asas de Animais/crescimento & desenvolvimento , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Epitélio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Larva/metabolismo , Larva/crescimento & desenvolvimento , Transdução de Sinais , Drosophila , Receptores de Esteroides/metabolismo , Receptores de Esteroides/genética
8.
Development ; 151(11)2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38832826

RESUMO

Germline maintenance relies on adult stem cells to continually replenish lost gametes over a lifetime and respond to external cues altering the demands on the tissue. Mating worsens germline homeostasis over time, yet a negative impact on stem cell behavior has not been explored. Using extended live imaging of the Drosophila testis stem cell niche, we find that short periods of mating in young males disrupts cytokinesis in germline stem cells (GSCs). This defect leads to failure of abscission, preventing release of differentiating cells from the niche. We find that GSC abscission failure is caused by increased Ecdysone hormone signaling induced upon mating, which leads to disrupted somatic encystment of the germline. Abscission failure is rescued by isolating males from females, but recurs with resumption of mating. Importantly, reiterative mating also leads to increased GSC loss, requiring increased restoration of stem cells via symmetric renewal and de-differentiation. Together, these results suggest a model whereby acute mating results in hormonal changes that negatively impact GSC cytokinesis but preserves the stem cell population.


Assuntos
Citocinese , Drosophila melanogaster , Ecdisona , Células Germinativas , Testículo , Animais , Masculino , Ecdisona/metabolismo , Testículo/metabolismo , Feminino , Drosophila melanogaster/metabolismo , Células Germinativas/metabolismo , Células Germinativas/citologia , Nicho de Células-Tronco , Células-Tronco/metabolismo , Células-Tronco/citologia , Diferenciação Celular , Transdução de Sinais , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética
9.
Cell ; 149(4): 807-18, 2012 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-22579285

RESUMO

The molecular chaperone Heat shock protein 90 (Hsp90) promotes the maturation of several important proteins and plays a key role in development, cancer progression, and evolutionary diversification. By mapping chromatin-binding sites of Hsp90 at high resolution across the Drosophila genome, we uncover an unexpected mechanism by which Hsp90 orchestrates cellular physiology. It localizes near promoters of many coding and noncoding genes including microRNAs. Using computational and biochemical analyses, we find that Hsp90 maintains and optimizes RNA polymerase II pausing via stabilization of the negative elongation factor complex (NELF). Inhibition of Hsp90 leads to upregulation of target genes, and Hsp90 is required for maximal activation of paused genes in Drosophila and mammalian cells in response to environmental stimuli. Our findings add a molecular dimension to the chaperone's functionality with wide ramifications into its roles in health, disease, and evolution.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas de Choque Térmico/metabolismo , RNA Polimerase II/metabolismo , Transcrição Gênica , Animais , Drosophila melanogaster/genética , Ecdisona/metabolismo , Regulação da Expressão Gênica , Humanos
10.
PLoS Genet ; 20(3): e1011204, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38452112

RESUMO

We investigate the contribution of a candidate gene, fiz (fezzik), to complex polygenic adaptation to juvenile malnutrition in Drosophila melanogaster. Experimental populations maintained for >250 generations of experimental evolution to a nutritionally poor larval diet (Selected populations) evolved several-fold lower fiz expression compared to unselected Control populations. Here we show that this divergence in fiz expression is mediated by a cis-regulatory polymorphism. This polymorphism, originally sampled from a natural population in Switzerland, is distinct from a second cis-regulatory SNP previously identified in non-African D. melanogaster populations, implying that two independent cis-regulatory variants promoting high fiz expression segregate in non-African populations. Enzymatic analyses of Fiz protein expressed in E. coli demonstrate that it has ecdysone oxidase activity acting on both ecdysone and 20-hydroxyecdysone. Four of five fiz paralogs annotated to ecdysteroid metabolism also show reduced expression in Selected larvae, implying that malnutrition-driven selection favored general downregulation of ecdysone oxidases. Finally, as an independent test of the role of fiz in poor diet adaptation, we show that fiz knockdown by RNAi results in faster larval growth on the poor diet, but at the cost of greatly reduced survival. These results imply that downregulation of fiz in Selected populations was favored by selection on the nutritionally poor diet because of its role in suppressing growth in response to nutrient shortage. However, they suggest that fiz downregulation is only adaptive in combination with other changes evolved by Selected populations, which ensure that the organism can sustain the faster growth promoted by fiz downregulation.


Assuntos
3-Hidroxiesteroide Desidrogenases , Drosophila , Desnutrição , Animais , Drosophila/fisiologia , Drosophila melanogaster/fisiologia , Ecdisona/genética , Escherichia coli , Larva
11.
PLoS Genet ; 20(4): e1011232, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38669270

RESUMO

Animals often grow and develop in unpredictable environments where factors like food availability, temperature, and oxygen levels can fluctuate dramatically. To ensure proper sexual maturation into adulthood, juvenile animals need to adapt their growth and developmental rates to these fluctuating environmental conditions. Failure to do so can result in impaired maturation and incorrect body size. Here we describe a mechanism by which Drosophila larvae adapt their development in low oxygen (hypoxia). During normal development, larvae grow and increase in mass until they reach critical weight (CW), after which point a neuroendocrine circuit triggers the production of the steroid hormone ecdysone from the prothoracic gland (PG), which promotes maturation to the pupal stage. However, when raised in hypoxia (5% oxygen), larvae slow their growth and delay their maturation to the pupal stage. We find that, although hypoxia delays the attainment of CW, the maturation delay occurs mainly because of hypoxia acting late in development to suppress ecdysone production. This suppression operates through a distinct mechanism from nutrient deprivation, occurs independently of HIF-1 alpha and does not involve dilp8 or modulation of Ptth, the main neuropeptide that initiates ecdysone production in the PG. Instead, we find that hypoxia lowers the expression of the EGF ligand, spitz, and that the delay in maturation occurs due to reduced EGFR/ERK signaling in the PG. Our study sheds light on how animals can adjust their development rate in response to changing oxygen levels in their environment. Given that hypoxia is a feature of both normal physiology and many diseases, our findings have important implications for understanding how low oxygen levels may impact animal development in both normal and pathological situations.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster , Ecdisona , Fator de Crescimento Epidérmico , Larva , Transdução de Sinais , Animais , Ecdisona/metabolismo , Larva/crescimento & desenvolvimento , Larva/genética , Larva/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Fator de Crescimento Epidérmico/metabolismo , Fator de Crescimento Epidérmico/genética , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Hipóxia/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Receptores ErbB/metabolismo , Receptores ErbB/genética , Oxigênio/metabolismo , Pupa/crescimento & desenvolvimento , Pupa/metabolismo , Pupa/genética
12.
PLoS Genet ; 20(3): e1011196, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38466721

RESUMO

Hematophagous mosquitoes require vertebrate blood for their reproductive cycles, making them effective vectors for transmitting dangerous human diseases. Thus, high-intensity metabolism is needed to support reproductive events of female mosquitoes. However, the regulatory mechanism linking metabolism and reproduction in mosquitoes remains largely unclear. In this study, we found that the expression of estrogen-related receptor (ERR), a nuclear receptor, is activated by the direct binding of 20-hydroxyecdysone (20E) and ecdysone receptor (EcR) to the ecdysone response element (EcRE) in the ERR promoter region during the gonadotropic cycle of Aedes aegypti (named AaERR). RNA interference (RNAi) of AaERR in female mosquitoes led to delayed development of ovaries. mRNA abundance of genes encoding key enzymes involved in carbohydrate metabolism (CM)-glucose-6-phosphate isomerase (GPI) and pyruvate kinase (PYK)-was significantly decreased in AaERR knockdown mosquitoes, while the levels of metabolites, such as glycogen, glucose, and trehalose, were elevated. The expression of fatty acid synthase (FAS) was notably downregulated, and lipid accumulation was reduced in response to AaERR depletion. Dual luciferase reporter assays and electrophoretic mobility shift assays (EMSA) determined that AaERR directly activated the expression of metabolic genes, such as GPI, PYK, and FAS, by binding to the corresponding AaERR-responsive motif in the promoter region of these genes. Our results have revealed an important role of AaERR in the regulation of metabolism during mosquito reproduction and offer a novel target for mosquito control.


Assuntos
Aedes , Receptores de Esteroides , Animais , Feminino , Humanos , Aedes/genética , Aedes/metabolismo , Ecdisona/metabolismo , Mosquitos Vetores/genética , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Homeostase/genética , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo
13.
J Cell Sci ; 137(5)2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38323986

RESUMO

Migratory cells - either individually or in cohesive groups - are critical for spatiotemporally regulated processes such as embryonic development and wound healing. Their dysregulation is the underlying cause of formidable health problems such as congenital abnormalities and metastatic cancers. Border cell behavior during Drosophila oogenesis provides an effective model to study temporally regulated, collective cell migration in vivo. Developmental timing in flies is primarily controlled by the steroid hormone ecdysone, which acts through a well-conserved, nuclear hormone receptor complex. Ecdysone signaling determines the timing of border cell migration, but the molecular mechanisms governing this remain obscure. We found that border cell clusters expressing a dominant-negative form of ecdysone receptor extended ineffective protrusions. Additionally, these clusters had aberrant spatial distributions of E-cadherin (E-cad), apical domain markers and activated myosin that did not overlap. Remediating their expression or activity individually in clusters mutant for ecdysone signaling did not restore proper migration. We propose that ecdysone signaling synchronizes the functional distribution of E-cadherin, atypical protein kinase C (aPKC), Discs large (Dlg1) and activated myosin post-transcriptionally to coordinate adhesion, polarity and contractility and temporally control collective cell migration.


Assuntos
Proteínas de Drosophila , Animais , Proteínas de Drosophila/metabolismo , Ecdisona/metabolismo , Drosophila/metabolismo , Caderinas/genética , Caderinas/metabolismo , Movimento Celular/fisiologia , Miosinas/metabolismo , Drosophila melanogaster/metabolismo , Polaridade Celular/fisiologia , Adesão Celular
14.
Development ; 150(10)2023 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-37092314

RESUMO

Adipose tissue is a central organ for controlling systemic metabolism both in invertebrates and vertebrates. Here, we have investigated the developmental processes of the adult-type fat body (AFB) in Drosophila. We have established genetic tools that allow visualization and genetic manipulations of cells in the AFB lineage from early in metamorphosis. We identified precursor cells that give rise to the AFB and delineated dynamic cellular behaviors underlying AFB formation. These precursor cells displayed polarized cell shapes and oriented motility, with emigration from the thorax and subsequent dispersal to the abdomen and head. After the migration period, these cells adhered to each other, assembling into the AFB with a sheet-like architecture. Continuous cell proliferation occurred during and after the large-scale migration to achieve appropriate fat tissue mass. Homotypic cell fusion after the sheet formation contributed to the establishment of multinucleated cells in the AFB. We also examined candidate gene functions, and our results argue that ecdysone signaling and the transcription factor Serpent support adult fat body organogenesis.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster , Animais , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Fatores de Transcrição/metabolismo , Drosophila/metabolismo , Metamorfose Biológica/genética , Ecdisona/metabolismo , Tecido Adiposo/metabolismo , Larva/metabolismo , Regulação da Expressão Gênica no Desenvolvimento
15.
PLoS Biol ; 21(5): e3002115, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37205703

RESUMO

Neuron specification and maturation are essential for proper central nervous system development. However, the precise mechanisms that govern neuronal maturation, essential to shape and maintain neuronal circuitry, remain poorly understood. Here, we analyse early-born secondary neurons in the Drosophila larval brain, revealing that the early maturation of secondary neurons goes through 3 consecutive phases: (1) Immediately after birth, neurons express pan-neuronal markers but do not transcribe terminal differentiation genes; (2) Transcription of terminal differentiation genes, such as neurotransmitter-related genes VGlut, ChAT, or Gad1, starts shortly after neuron birth, but these transcripts are, however, not translated; (3) Translation of neurotransmitter-related genes only begins several hours later in mid-pupa stages in a coordinated manner with animal developmental stage, albeit in an ecdysone-independent manner. These results support a model where temporal regulation of transcription and translation of neurotransmitter-related genes is an important mechanism to coordinate neuron maturation with brain development.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila/metabolismo , Neurônios/fisiologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Neurogênese , Ecdisona , Regulação da Expressão Gênica no Desenvolvimento
16.
Nature ; 584(7821): 415-419, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32641829

RESUMO

Sexual dimorphism arises from genetic differences between male and female cells, and from systemic hormonal differences1-3. How sex hormones affect non-reproductive organs is poorly understood, yet highly relevant to health given the sex-biased incidence of many diseases4. Here we report that steroid signalling in Drosophila from the ovaries to the gut promotes growth of the intestine specifically in mated females, and enhances their reproductive output. The active ovaries of the fly produce the steroid hormone ecdysone, which stimulates the division and expansion of intestinal stem cells in two distinct proliferative phases via the steroid receptors EcR and Usp and their downstream targets Broad, Eip75B and Hr3. Although ecdysone-dependent growth of the female gut augments fecundity, the more active and more numerous intestinal stem cells also increase female susceptibility to age-dependent gut dysplasia and tumorigenesis, thus potentially reducing lifespan. This work highlights the trade-offs in fitness traits that occur when inter-organ signalling alters stem-cell behaviour to optimize organ size.


Assuntos
Drosophila melanogaster/metabolismo , Fertilidade/fisiologia , Intestinos/crescimento & desenvolvimento , Longevidade/fisiologia , Tamanho do Órgão/fisiologia , Ovário/metabolismo , Esteroides/metabolismo , Envelhecimento , Animais , Carcinogênese , Proliferação de Células , Copulação/fisiologia , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/anatomia & histologia , Drosophila melanogaster/citologia , Drosophila melanogaster/fisiologia , Ecdisona/metabolismo , Feminino , Mucosa Intestinal/anatomia & histologia , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Intestinos/anatomia & histologia , Intestinos/citologia , Intestinos/patologia , Masculino , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo
17.
Proc Natl Acad Sci U S A ; 120(49): e2313224120, 2023 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-38015844

RESUMO

The decision to stop growing and mature into an adult is a critical point in development that determines adult body size, impacting multiple aspects of an adult's biology. In many animals, growth cessation is a consequence of hormone release that appears to be tied to the attainment of a particular body size or condition. Nevertheless, the size-sensing mechanism animals use to initiate hormone synthesis is poorly understood. Here, we develop a simple mathematical model of growth cessation in Drosophila melanogaster, which is ostensibly triggered by the attainment of a critical weight (CW) early in the last instar. Attainment of CW is correlated with the synthesis of the steroid hormone ecdysone, which causes a larva to stop growing, pupate, and metamorphose into the adult form. Our model suggests that, contrary to expectation, the size-sensing mechanism that initiates metamorphosis occurs before the larva reaches CW; that is, the critical-weight phenomenon is a downstream consequence of an earlier size-dependent developmental decision, not a decision point itself. Further, this size-sensing mechanism does not require a direct assessment of body size but emerges from the interactions between body size, ecdysone, and nutritional signaling. Because many aspects of our model are evolutionarily conserved among all animals, the model may provide a general framework for understanding how animals commit to maturing from their juvenile to adult form.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila melanogaster , Ecdisona , Tamanho Corporal , Larva , Metamorfose Biológica
18.
PLoS Genet ; 19(9): e1010924, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37683015

RESUMO

TAIMAN (TAI), the only insect ortholog of mammalian Steroid Receptor Coactivators (SRCs), is a critical modulator of ecdysone and juvenile hormone (JH) signaling pathways, which govern insect development and reproduction. The modulatory effect is mediated by JH-dependent TAI's heterodimerization with JH receptor Methoprene-tolerant and association with the Ecdysone Receptor complex. Insect hormones regulate insect physiology and development in concert with abiotic cues, such as photo- and thermoperiod. Here we tested the effects of JH and ecdysone signaling on the circadian clock by a combination of microsurgical operations, application of hormones and hormone mimics, and gene knockdowns in the linden bug Pyrrhocoris apterus males. Silencing taiman by each of three non-overlapping double-strand RNA fragments dramatically slowed the free-running period (FRP) to 27-29 hours, contrasting to 24 hours in controls. To further corroborate TAIMAN's clock modulatory function in the insect circadian clock, we performed taiman knockdown in the cockroach Blattella germanica. Although Blattella and Pyrrhocoris lineages separated ~380 mya, B. germanica taiman silencing slowed the FRP by more than 2 hours, suggesting a conserved TAI clock function in (at least) some insect groups. Interestingly, the pace of the linden bug circadian clock was neither changed by blocking JH and ecdysone synthesis, by application of the hormones or their mimics nor by the knockdown of corresponding hormone receptors. Our results promote TAI as a new circadian clock modulator, a role described for the first time in insects. We speculate that TAI participation in the clock is congruent with the mammalian SRC-2 role in orchestrating metabolism and circadian rhythms, and that TAI/SRCs might be conserved components of the circadian clock in animals.


Assuntos
Relógios Circadianos , Animais , Masculino , Relógios Circadianos/genética , Ecdisona/genética , Insetos , Ritmo Circadiano/genética , Membrana Celular , Hormônios Juvenis/genética , Mamíferos
19.
PLoS Genet ; 19(6): e1010815, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37363926

RESUMO

In prostate cancer, loss of the tumour suppressor gene, Retinoblastoma (Rb), and consequent activation of transcription factor E2F1 typically occurs at a late-stage of tumour progression. It appears to regulate a switch to an androgen-independent form of cancer, castration-resistant prostate cancer (CRPC), which frequently still requires androgen receptor (AR) signalling. We have previously shown that upon mating, binucleate secondary cells (SCs) of the Drosophila melanogaster male accessory gland (AG), which share some similarities with prostate epithelial cells, switch their growth regulation from a steroid-dependent to a steroid-independent form of Ecdysone Receptor (EcR) control. This physiological change induces genome endoreplication and allows SCs to rapidly replenish their secretory compartments, even when ecdysone levels are low because the male has not previously been exposed to females. Here, we test whether the Drosophila Rb homologue, Rbf, and E2F1 regulate this switch. Surprisingly, we find that excess Rbf activity reversibly suppresses binucleation in adult SCs. We also demonstrate that Rbf, E2F1 and the cell cycle regulators, Cyclin D (CycD) and Cyclin E (CycE), are key regulators of mating-dependent SC endoreplication, as well as SC growth in both virgin and mated males. Importantly, we show that the CycD/Rbf/E2F1 axis requires the EcR, but not ecdysone, to trigger CycE-dependent endoreplication and endoreplication-associated growth in SCs, mirroring changes seen in CRPC. Furthermore, Bone Morphogenetic Protein (BMP) signalling, mediated by the BMP ligand Decapentaplegic (Dpp), intersects with CycD/Rbf/E2F1 signalling to drive endoreplication in these fly cells. Overall, our work reveals a signalling switch, which permits rapid growth of SCs and increased secretion after mating, independently of previous exposure to females. The changes observed share mechanistic parallels with the pathological switch to hormone-independent AR signalling seen in CRPC, suggesting that the latter may reflect the dysregulation of a currently unidentified physiological process.


Assuntos
Proteínas de Drosophila , Neoplasias de Próstata Resistentes à Castração , Humanos , Animais , Feminino , Masculino , Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Próstata/patologia , Neoplasias de Próstata Resistentes à Castração/metabolismo , Neoplasias de Próstata Resistentes à Castração/patologia , Endorreduplicação , Ecdisona/genética , Ecdisona/metabolismo , Fator de Transcrição E2F1/genética , Fatores de Transcrição/genética , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo
20.
Dev Biol ; 512: 35-43, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38710381

RESUMO

The larval stage of the Drosophila melanogaster life cycle is characterized by rapid growth and nutrient storage that occur over three instar stages separated by molts. In the third instar, the steroid hormone ecdysone drives key developmental processes and behaviors that occur in a temporally-controlled sequence and prepare the animal to undergo metamorphosis. Accurately staging Drosophila larvae within the final third instar is critical due to the rapid developmental progress at this stage, but it is challenging because the rate of development varies widely across a population of animals even if eggs are laid within a short period of time. Moreover, many methods to stage third instar larvae are cumbersome, and inherent variability in the rate of development confounds some of these approaches. Here we demonstrate the usefulness of the Sgs3-GFP transgene, a fusion of the Salivary gland secretion 3 (Sgs3) and GFP proteins, for staging third instar larvae. Sgs3-GFP is expressed in the salivary glands in an ecdysone-dependent manner from the midpoint of the third instar, and its expression pattern changes reproducibly as larvae progress through the third instar. We show that Sgs3-GFP can easily be incorporated into experiments, that it allows collection of developmentally-equivalent individuals from a mixed population of larvae, and that its use enables precise assessment of changing levels of hormones, metabolites, and gene expression during the second half of the third instar.


Assuntos
Drosophila melanogaster , Ecdisona , Proteínas de Fluorescência Verde , Larva , Fenótipo , Glândulas Salivares , Animais , Larva/metabolismo , Larva/genética , Glândulas Salivares/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Fluorescência Verde/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Ecdisona/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Genes Reporter , Regulação da Expressão Gênica no Desenvolvimento/genética , Animais Geneticamente Modificados , Metamorfose Biológica/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA