Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 257
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 116(20): 9893-9902, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31019084

ABSTRACT

The ecdysone pathway was among the first experimental systems employed to study the impact of steroid hormones on the genome. In Drosophila and other insects, ecdysone coordinates developmental transitions, including wholesale transformation of the larva into the adult during metamorphosis. Like other hormones, ecdysone controls gene expression through a nuclear receptor, which functions as a ligand-dependent transcription factor. Although it is clear that ecdysone elicits distinct transcriptional responses within its different target tissues, the role of its receptor, EcR, in regulating target gene expression is incompletely understood. In particular, EcR initiates a cascade of transcription factor expression in response to ecdysone, making it unclear which ecdysone-responsive genes are direct EcR targets. Here, we use the larval-to-prepupal transition of developing wings to examine the role of EcR in gene regulation. Genome-wide DNA binding profiles reveal that EcR exhibits widespread binding across the genome, including at many canonical ecdysone response genes. However, the majority of its binding sites reside at genes with wing-specific functions. We also find that EcR binding is temporally dynamic, with thousands of binding sites changing over time. RNA-seq reveals that EcR acts as both a temporal gate to block precocious entry to the next developmental stage as well as a temporal trigger to promote the subsequent program. Finally, transgenic reporter analysis indicates that EcR regulates not only temporal changes in target enhancer activity but also spatial patterns. Together, these studies define EcR as a multipurpose, direct regulator of gene expression, greatly expanding its role in coordinating developmental transitions.


Subject(s)
Drosophila/physiology , Ecdysone/physiology , Metamorphosis, Biological , Receptors, Steroid/metabolism , Animals , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental , Genome, Insect , Transcription Factors/metabolism , Wings, Animal/metabolism
2.
Development ; 145(6)2018 03 19.
Article in English | MEDLINE | ID: mdl-29467243

ABSTRACT

The wing imaginal disks of Lepidoptera can be grown in tissue culture, but require both insulin and ecdysone to grow normally. Here, we investigate the contributions the two hormones make to growth. Ecdysone is required to maintain mitoses, whereas in the presence of insulin alone mitoses stop. Both ecdysone and insulin stimulate protein synthesis, but only ecdysone stimulates DNA synthesis. Insulin stimulates primarily cytoplasmic growth and an increase in cell size, whereas ecdysone, by virtue of its stimulation of DNA synthesis and mitosis, stimulates growth by an increase in cell number. Although both hormones stimulate protein synthesis, they do so in different spatial patterns. Both hormones stimulate protein synthesis in the inter-vein regions, but ecdysone stimulates synthesis more strongly in the veins and in the margin of the wing disk. We propose that the balance of insulin and ecdysone signaling must be regulated to maintain normal growth, and when growth appears to be due primarily to an increase in cell number, or an increase in cell size, this may indicate growth occurred under conditions that favored a stronger role for ecdysone, or insulin, respectively.


Subject(s)
Butterflies/physiology , Ecdysone/physiology , Imaginal Discs/growth & development , Insulin/physiology , Wings, Animal/growth & development , Animals , Imaginal Discs/physiology , Larva/metabolism , Mitosis/physiology , Protein Biosynthesis/physiology , Wings, Animal/physiology
3.
Proc Natl Acad Sci U S A ; 114(5): 1057-1062, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28096379

ABSTRACT

Juvenile hormone (JH) represses precocious metamorphosis of larval to pupal and adult transitions in holometabolous insects. The early JH-inducible gene Krüppel homolog 1 (Kr-h1) plays a key role in the repression of metamorphosis as a mediator of JH action. Previous studies demonstrated that Kr-h1 inhibits precocious larval-pupal transition in immature larva via direct transcriptional repression of the pupal specifier Broad-Complex (BR-C). JH was recently reported to repress the adult specifier gene Ecdysone-induced protein 93F (E93); however, its mechanism of action remains unclear. Here, we found that JH suppressed ecdysone-inducible E93 expression in the epidermis of the silkworm Bombyx mori and in a B. mori cell line. Reporter assays in the cell line revealed that the JH-dependent suppression was mediated by Kr-h1. Genome-wide ChIP-seq analysis identified a consensus Kr-h1 binding site (KBS, 14 bp) located in the E93 promoter region, and EMSA confirmed that Kr-h1 directly binds to the KBS. Moreover, we identified a C-terminal conserved domain in Kr-h1 essential for the transcriptional repression of E93 Based on these results, we propose a mechanism in which JH-inducible Kr-h1 directly binds to the KBS site upstream of the E93 locus to repress its transcription in a cell-autonomous manner, thereby preventing larva from bypassing the pupal stage and progressing to precocious adult development. These findings help to elucidate the molecular mechanisms regulating the metamorphic genetic network, including the functional significance of Kr-h1, BR-C, and E93 in holometabolous insect metamorphosis.


Subject(s)
Bombyx/growth & development , Ecdysone/physiology , Insect Proteins/physiology , Metamorphosis, Biological/physiology , Transcription Factors/physiology , Animals , Binding Sites , Bombyx/genetics , Cell Line , Chromatin Immunoprecipitation , Consensus Sequence , Female , Gene Expression Regulation, Developmental , Genes, Reporter , Insect Proteins/genetics , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/physiology , Larva , Male , Methoprene/pharmacology , Promoter Regions, Genetic , Protein Domains , Protein Isoforms/genetics , Protein Isoforms/metabolism , Pupa , Transcription Factors/genetics , Transcription, Genetic
4.
Apoptosis ; 24(11-12): 972-989, 2019 12.
Article in English | MEDLINE | ID: mdl-31641960

ABSTRACT

Developmentally regulated programmed cell death (PCD) is one of the key cellular events for precise controlling of neuronal population during postembryonic development of the central nervous system. Previously we have shown that a group of corazonin-producing peptidergic neurons (vCrz) undergo apoptosis in response to ecdysone signaling via ecdysone receptor (EcR)-B isoforms and Ultraspiracle during early phase of metamorphosis. Further utilizing genetic, transgenic, and mosaic analyses, we have found that TGF-ß signaling mediated by a glia-produced ligand, Myoglianin, type-I receptor Baboon (particularly Babo-A isoform) and dSmad2, is also required autonomously for PCD of the vCrz neurons. Our studies show that TGF-ß signaling is not acting epistatically to EcR or vice versa. We also show that ectopic expression of a constitutively active phosphomimetic form of dSmad2 (dSmad2PM) is capable of inducing premature death of vCrz neurons in larva but not other larval neurons. Intriguingly, the dSmad2PM-mediated killing is completely suppressed by coexpression of a dominant-negative form of EcR (EcRDN), suggesting that EcR function is required for the proapoptotic dSmad2PM function. Based on these data, we suggest that TGF-ß and ecdysone signaling pathways act cooperatively to induce vCrz neuronal PCD. We propose that this type of two-factor authentication is a key developmental strategy to ensure the timely PCD of specific larval neurons during metamorphosis.


Subject(s)
Activin Receptors/metabolism , Apoptosis , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Metamorphosis, Biological/genetics , Neurons/metabolism , Receptors, Steroid/metabolism , Activin Receptors/genetics , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Animals , Animals, Genetically Modified , Apoptosis/physiology , Central Nervous System/cytology , Central Nervous System/growth & development , Central Nervous System/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Ecdysone/metabolism , Ecdysone/physiology , Gene Expression Regulation, Developmental/genetics , Insect Proteins/genetics , Insect Proteins/metabolism , Larva/cytology , Larva/metabolism , Metamorphosis, Biological/physiology , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neuropeptides/genetics , Neuropeptides/metabolism , Protein Isoforms/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Steroid/genetics , Signal Transduction/genetics , Smad Proteins, Receptor-Regulated/genetics , Smad Proteins, Receptor-Regulated/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/physiology
5.
RNA ; 21(10): 1807-17, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26289344

ABSTRACT

The accurate and efficient transfer of genetic information into amino acid sequences is carried out through codon-anticodon interactions between mRNA and tRNA, respectively. In this way, tRNAs function at the interface between gene expression and protein synthesis. Whether tRNA levels are dynamically regulated and to what degree tRNA abundance influences the cellular proteome remains largely unexplored. Here we profile tRNA, transcript and protein levels in Drosophila Kc167 cells, a plasmatocyte cell line that, upon treatment with 20-hydroxyecdysone, differentiates into macrophages. We find that high abundance tRNAs associate with codons that are overrepresented in the Kc167 cell proteome, whereas tRNAs that are in low supply associate with codons that are underrepresented. Ecdysone-induced differentiation of Kc167 cells leads to changes in mRNA codon usage in a manner consistent with the developmental progression of the cell. At both early and late time points, ecdysone treatment concomitantly increases the abundance of tRNAThr(CGU), which decodes a differentiation-associated codon that becomes enriched in the macrophage proteome. These results together suggest that tRNA levels may provide a meaningful regulatory mechanism for defining the cellular proteomic landscape.


Subject(s)
Ecdysone/physiology , Proteins/physiology , RNA, Messenger/genetics , RNA, Transfer/genetics , Signal Transduction , Animals , Cell Differentiation , Cell Line , Codon , Drosophila , Humans , Proteomics , Transcription, Genetic
6.
Article in English | MEDLINE | ID: mdl-28251296

ABSTRACT

Most brachyuran females become receptive during the intermolt period, a condition considered "derived". However, as far as we know, studies testing the existence and function of pheromones in decapods are based on species which have mating linked to molting, a condition considered as "ancestral". For the first time, we studied some physiological and morphological processes involved in Neohelice granulata intermolt female crabs becoming receptive and potentially attracting males. We found that receptive females have mobile vulvae opercula due to a softening process of the cuticle hinge which showed lower calcium levels compared to the hinge of unreceptive females. Local softening of the hinge was stimulated by a low concentration of ecdysone during the intermolt period. A putative pheromone liberated by receptive females to attract males is presumed to be released through the mobile vulvae and not through the urine.


Subject(s)
Animal Shells/physiology , Brachyura/physiology , Ecdysone/physiology , Molting/physiology , Sexual Behavior, Animal/physiology , Animals , Female , Male
7.
Biochim Biophys Acta ; 1849(2): 181-6, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24939835

ABSTRACT

Although a great deal of information is available concerning the role of ecdysone in insect oogenesis, research has tended to focus on vitellogenesis and choriogenesis. As such, the study of oogenesis in a strict sense has received much less attention. This situation changed recently when a number of observations carried out in the meroistic polytrophic ovarioles of Drosophila melanogaster started to unravel the key roles played by ecdysone in different steps of oogenesis. Thus, in larval stages, a non-autonomous role of ecdysone, first in repression and later in activation, of stem cell niche and primordial germ cell differentiation has been reported. In the adult, ecdysone stimulates the proliferation of germline stem cells, plays a role in stem cell niche maintenance and is needed non-cell-autonomously for correct differentiation of germline stem cells. Moreover, in somatic cells ecdysone is required for 16-cell cyst formation and for ovarian follicle development. In the transition from stages 8 to 9 of oogenesis, ecdysone signalling is fundamental when deciding whether or not to go ahead with vitellogenesis depending on the nutritional status, as well as to start border cell migration. This article is part of a Special Issue entitled: Nuclear receptors in animal development.


Subject(s)
Cockroaches/growth & development , Drosophila melanogaster/growth & development , Ecdysone/physiology , Ovarian Follicle/growth & development , Stem Cells/physiology , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cockroaches/genetics , Cockroaches/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Ecdysone/pharmacology , Female , Oogenesis/drug effects , Oogenesis/genetics , Signal Transduction , Vitellogenesis/drug effects , Vitellogenesis/genetics
8.
Dev Biol ; 385(2): 253-62, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24247008

ABSTRACT

Hormone-induced changes in gene expression initiate periodic molts and metamorphosis during insect development. Successful execution of these developmental steps depends upon successive phases of rising and falling 20-hydroxyecdysone (20E) levels, leading to a cascade of nuclear receptor-driven transcriptional activity that enables stage- and tissue-specific responses to the steroid. Among the cellular processes associated with declining steroids is acquisition of secretory competence in endocrine Inka cells, the source of ecdysis triggering hormones (ETHs). We show here that Inka cell secretory competence is conferred by the orphan nuclear receptor ßFTZ-F1. Selective RNA silencing of ßftz-f1 in Inka cells prevents ETH release, causing developmental arrest at all stages. Affected larvae display buttoned-up, the ETH-null phenotype characterized by double mouthparts, absence of ecdysis behaviors, and failure to shed the old cuticle. During the mid-prepupal period, individuals fail to translocate the air bubble, execute head eversion and elongate incipient wings and legs. Those that escape to the adult stage are defective in wing expansion and cuticle sclerotization. Failure to release ETH in ßftz-f1 silenced animals is indicated by persistent ETH immunoreactivity in Inka cells. Arrested larvae are rescued by precisely-timed ETH injection or Inka cell-targeted ßFTZ-F1 expression. Moreover, premature ßftz-f1 expression in these cells also results in developmental arrest. The Inka cell therefore functions as a "gateway cell", whose secretion of ETH serves as a key downstream physiological output enabling stage-specific responses to 20E that are required to advance through critical developmental steps. This secretory function depends on transient and precisely timed ßFTZ-F1 expression late in the molt as steroids decline.


Subject(s)
DNA-Binding Proteins/physiology , Drosophila melanogaster/growth & development , Ecdysone/physiology , Endocrine Glands/cytology , Receptors, Steroid/physiology , Animals , Base Sequence , DNA Primers , DNA-Binding Proteins/genetics , Drosophila melanogaster/physiology , Gene Knockdown Techniques , Polymerase Chain Reaction , RNA, Messenger/genetics , Receptors, Steroid/genetics
9.
PLoS Genet ; 8(11): e1003085, 2012.
Article in English | MEDLINE | ID: mdl-23209440

ABSTRACT

Steroid hormones act, through their respective nuclear receptors, to regulate target gene expression. Despite their critical role in development, physiology, and disease, however, it is still unclear how these systemic cues are refined into tissue-specific responses. We identified a mutation in the evolutionarily conserved DEAD box RNA helicase belle/DDX3 that disrupts a subset of responses to the steroid hormone ecdysone during Drosophila melanogaster metamorphosis. We demonstrate that belle directly regulates translation of E74A, an ets transcription factor and critical component of the ecdysone-induced transcriptional cascade. Although E74A mRNA accumulates to abnormally high levels in belle mutant tissues, no E74A protein is detectable, resulting in misregulation of E74A-dependent ecdysone response genes. The accumulation of E74A mRNA in belle mutant salivary glands is a result of auto-regulation, fulfilling a prediction made by Ashburner nearly 40 years ago. In this model, Ashburner postulates that, in addition to regulating secondary response genes, protein products of primary response genes like E74A also inhibit their own ecdysone-induced transcription. Moreover, although ecdysone-triggered transcription of E74A appears to be ubiquitous during metamorphosis, belle-dependent translation of E74A mRNA is spatially restricted. These results demonstrate that translational control plays a critical, and previously unknown, role in refining transcriptional responses to the steroid hormone ecdysone.


Subject(s)
DEAD-box RNA Helicases , DNA-Binding Proteins , Drosophila melanogaster , Ecdysone , Protein Biosynthesis , Transcription Factors , Animals , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/physiology , Ecdysone/genetics , Ecdysone/metabolism , Ecdysone/physiology , Gene Expression , Metamorphosis, Biological , Mutation , Organ Specificity , Salivary Glands/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
10.
Dev Biol ; 380(2): 344-50, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23707898

ABSTRACT

BRMS1 was first discovered as a human breast carcinoma metastasis suppressor gene. However, the mechanism of BRMS1 in tumor metastasis and its developmental role remain unclear. In this paper, we first report the identification of the Drosophila ortholog of human BRMS1, dBrms1. Through a genetic approach, the role of dBrms1 during development has been investigated. We found that dBrms1 is an essential gene and loss of dBrms1 function results in lethality at early developmental stages. dBrms1mutants displayed phenotypes such as developmental delay and failure to initiate metamorphosis. Further analysis suggests that these phenotypes are contributed by defective ecdysone signaling and expression of target genes of the ecdysone pathway. Therefore, dBrms1 is required for growth control by acting as a modulator of ecdysone signaling in Drosophila and is required for metamorphosis for normal development.


Subject(s)
Ecdysone/physiology , Gene Expression Regulation, Developmental , Genes, Tumor Suppressor , Neoplasm Proteins/genetics , Animals , Drosophila , Metamorphosis, Biological , Mutation , Repressor Proteins , Signal Transduction , Time Factors , Transgenes
11.
Dev Biol ; 374(1): 127-41, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23149076

ABSTRACT

The adult optic lobe of Drosophila develops from the primordium during metamorphosis from mid-3rd larval stage to adult. Many cells die during development of the optic lobe with a peak of the number of dying cells at 24 h after puparium formation (h APF). Dying cells were observed in spatio-temporal specific clusters. Here, we analyzed the function of a component of the insect steroid hormone receptor, EcR, in this cell death. We examined expression patterns of two EcR isoforms, EcR-A and EcR-B1, in the optic lobe. Expression of each isoform altered during development in isoform-specific manner. EcR-B1 was not expressed in optic lobe neurons from 0 to 6h APF, but was expressed between 9 and 48 h APF and then disappeared by 60 h APF. In each cortex, its expression was stronger in older glia-ensheathed neurons than in younger ones. EcR-B1 was also expressed in some types of glia. EcR-A was expressed in optic lobe neurons and many types of glia from 0 to 60 h APF in a different pattern from EcR-B1. Then, we genetically analyzed EcR function in the optic lobe cell death. At 0 h APF, the optic lobe cell death was independent of any EcR isoforms. In contrast, EcR-B1 was required for most optic lobe cell death after 24 h APF. It was suggested that cell death cell-autonomously required EcR-B1 expressed after puparium formation. ßFTZ-F1 was also involved in cell death in many dying-cell clusters, but not in some of them at 24 h APF. Altogether, the optic lobe cell death occurred in ecdysone-independent manner at prepupal stage and ecdysone-dependent manner after 24 h APF. The acquisition of ecdysone-dependence was not directly correlated with the initiation or increase of EcR-B1 expression.


Subject(s)
Apoptosis , Drosophila/metabolism , Ecdysone/metabolism , Ecdysone/physiology , Gene Expression Regulation, Developmental , Optic Lobe, Nonmammalian/embryology , Animals , Crosses, Genetic , Drosophila/embryology , Metamorphosis, Biological , Microscopy, Confocal/methods , Models, Biological , Mutation , Neurons/metabolism , Protein Isoforms , RNA, Double-Stranded/metabolism , Time Factors
12.
J Cell Biol ; 222(3)2023 03 06.
Article in English | MEDLINE | ID: mdl-36648440

ABSTRACT

Mechanical forces actively shape cells during development, but little is known about their roles during neuronal morphogenesis. Developmental neurite pruning, a critical circuit specification mechanism, often involves neurite abscission at predetermined sites by unknown mechanisms. Pruning of Drosophila sensory neuron dendrites during metamorphosis is triggered by the hormone ecdysone, which induces local disassembly of the dendritic cytoskeleton. Subsequently, dendrites are severed at positions close to the soma by an unknown mechanism. We found that ecdysone signaling causes the dendrites to become mechanically fragile. Severing occurs during periods of increased pupal morphogenetic tissue movements, which exert mechanical forces on the destabilized dendrites. Tissue movements and dendrite severing peak during pupal ecdysis, a period of strong abdominal contractions, and abolishing ecdysis causes non-cell autonomous dendrite pruning defects. Thus, our data establish mechanical tearing as a novel mechanism during neurite pruning.


Subject(s)
Dendrites , Drosophila , Neurites , Animals , Dendrites/physiology , Drosophila/growth & development , Ecdysone/physiology , Neurites/physiology , Sensory Receptor Cells/physiology , Metamorphosis, Biological , Pupa/growth & development
13.
Dev Biol ; 351(1): 146-55, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21215267

ABSTRACT

Niemann-Pick C (NPC) disease is a lethal neurodegenerative disorder affecting cellular sterol trafficking. Besides neurodegeneration, NPC patients also exhibit other pleiotropic conditions, indicating that NPC protein is required for other physiological processes. Previous studies indicated that a sterol shortage that in turn leads to a shortage of steroid hormones (for example, ecdysone in Drosophila) is likely to be the cause of NPC disease pathology. We have shown that mutations in Drosophila npc1, one of the two NPC disease-related genes, leads to larval lethal and male infertility. Here, we reported that npc1 mutants are defective in spermatogenesis and in particular in the membrane-remodeling individualization process. Interestingly, we found that ecdysone, the steroid hormone responsible for the larval lethal phenotype in npc1 mutants, is not required for individualization. However, supplying 7-dehydrocholesterol can partially rescue the male infertility of npc1 mutants, suggesting that a sterol shortage is responsible for the spermatogenesis defects. In addition, the individualization defects of npc1 mutants were enhanced at high temperature, suggesting that the sterol shortage may lead to temperature-sensitive defects in the membrane-remodeling process. Together, our study reveals a sterol-dependent, ecdysone-independent mechanism of NPC1 function in Drosophila spermatogenesis.


Subject(s)
Carrier Proteins/physiology , Cholesterol/metabolism , Drosophila Proteins/physiology , Drosophila/physiology , Spermatogenesis , Animals , Dehydrocholesterols/pharmacology , Ecdysone/physiology , Female , Infertility, Male/etiology , Male , Membrane Proteins , Microscopy, Electron, Transmission , Niemann-Pick C1 Protein , Receptors, Steroid/physiology , Temperature , Testis/ultrastructure
14.
Biochem Biophys Res Commun ; 425(1): 113-8, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22828514

ABSTRACT

Bombyx mori Cathepsin D (BmCatD) is specifically expressed in the fat body, and plays a critical role for the programmed cell death of the larval fat body and pupal gut during metamorphosis. To better understand the transcriptional control of BmCatD expression, we conducted this study to identify the ecdysone response elements (EcREs) in the BmCatD promoter and clarify their regulational functions. We inserted EcREs into a recombinant AcMNPV (Autographa californica multiple nucleopolyhedrovirus) vector and performed luciferase assay with a dual-luciferase quantitative assay system. Three putative EcREs were located at positions -109 to -99, -836 to -826 and -856 to -846 relative to the transcription start site. Overlapping deletion studies of this EcRE region showed that the three EcREs could suppress the ectopic expression of the BmCatD promoter. EcRE mutations resulted in the loss of the fat body-specific expression of the BmCatD gene. These results suggest that the EcREs are vital for activation of the promoter by 20-hydroxyecdysone (20E) in the larval fat body and further support the crucial role of ecdysone signaling to control cathepsin D gene transcription. It may suggest that the heterodimeric complex EcR/USP mediates the activation of ecdysone-dependent BmCatD transcription in the larval fat body of B. mori.


Subject(s)
Bombyx/genetics , Cathepsin D/genetics , Ecdysone/physiology , Response Elements/genetics , Transcriptional Activation , Animals , Base Sequence , Bombyx/growth & development , Ecdysone/pharmacology , Ecdysterone/pharmacology , Luciferases/genetics , Molecular Sequence Data , Mutation , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Response Elements/drug effects , Transcription Initiation Site
15.
Article in English | MEDLINE | ID: mdl-21140155

ABSTRACT

Decapod crustaceans show proliferation of the nerve cells in the olfactory lobe throughout their lives. However, the regulation of this process is still poorly understood, since it may vary with endogenous and exogenous factors. The objective of the present investigation was to quantify the proliferation of nerve cells and number of nerve cells with ecdysone receptors in the clusters of the central olfactory system in Neohelice granulata, according to moult stages and in different seasons (summer and winter). Three injections of bromodeoxyuridine (BrdU) were administered to the crabs. Brains were sectioned by microtome and fixed on slides for immunohistochemistry with anti-BrdU and anti-EcR antibodies. The proliferation of nerve cells was higher in winter than in summer, probably because in winter the crabs do not breed and the premoult and postmoult periods are longer. Crabs in postmoult exhibited more BrdU-labelled cells than crabs in premoult or intermoult in winter, because of a greater number of mitoses related to an increase in body size and addition of olfactory receptor neurons. The number of EcR-labelled cells was higher in premoult than in postmoult or intermoult in winter. The proliferation of nerve cells is regulated seasonally and according to moult stages.


Subject(s)
Brachyura , Cell Proliferation , Molting/physiology , Neurons/cytology , Receptors, Steroid/metabolism , Seasons , Animals , Cell Count , Central Nervous System/cytology , Central Nervous System/physiology , Ecdysone/metabolism , Ecdysone/physiology , Ganglia, Invertebrate/cytology , Ganglia, Invertebrate/metabolism , Neurons/metabolism , Olfactory Pathways/cytology , Olfactory Pathways/metabolism
17.
Insect Sci ; 28(6): 1582-1590, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33205532

ABSTRACT

Insect growth and development are precisely controlled by hormone homeostasis. The prothoracicotropic hormone (PTTH) receptor, Torso, is a member of the receptor tyrosine kinase family in insects. Activation of Torso by PTTH triggers biosynthesis and release of the steroid hormone in the prothoracic gland (PG). Although numbers of genes functioning in steroid hormone synthesis and metabolism have been identified in insects, the PTTH transduction pathway via its receptor Torso is poorly understood. In the current study, we describe a loss-of-function analysis of Torso in the silkworm, Bombyx mori, by targeted gene disruption using the transgenic CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/RNA-guided Cas9 nucleases) system. Depletion of B. mori Torso (BmTorso) did not eventually affect larval ecdysis and metamorphosis processes. Instead, BmTorso deficiency resulted in significant extension of developing time during larval and pupal stages with increased pupa and cocoon sizes. The ecdysteriod titers in the hemolymph of BmTorso mutants sharpy declined. Transcriptional levels of genes involved in ecdysone biosynthesis and ecdysteroid signaling pathways were significantly reduced in BmTorso-deficient animals. Additionally, RNA-Seq analysis revealed that genes involved in the longevity pathway and protein processing in the endoplasmic reticulum pathway were affected after BmTorso deletion. These results indicate that Torso is critical for maintaining steroid hormone homeostasis in insects.


Subject(s)
Bombyx , Ecdysone/physiology , Protein-Tyrosine Kinases , Animals , Bombyx/embryology , Bombyx/enzymology , Homeostasis , Larva , Pupa
18.
Sci Rep ; 11(1): 18807, 2021 09 22.
Article in English | MEDLINE | ID: mdl-34552169

ABSTRACT

Animal development relies on a sequence of specific stages that allow the formation of adult structures with a determined size. In general, juvenile stages are dedicated mainly to growth, whereas last stages are devoted predominantly to the maturation of adult structures. In holometabolous insects, metamorphosis marks the end of the growth period as the animals stops feeding and initiate the final differentiation of the tissues. This transition is controlled by the steroid hormone ecdysone produced in the prothoracic gland. In Drosophila melanogaster different signals have been shown to regulate the production of ecdysone, such as PTTH/Torso, TGFß and Egfr signaling. However, to which extent the roles of these signals are conserved remains unknown. Here, we study the role of Egfr signaling in post-embryonic development of the basal holometabolous beetle Tribolium castaneum. We show that Tc-Egfr and Tc-pointed are required to induced a proper larval-pupal transition through the control of the expression of ecdysone biosynthetic genes. Furthermore, we identified an additional Tc-Egfr ligand in the Tribolium genome, the neuregulin-like protein Tc-Vein (Tc-Vn), which contributes to induce larval-pupal transition together with Tc-Spitz (Tc-Spi). Interestingly, we found that in addition to the redundant role in the control of pupa formation, each ligand possesses different functions in organ morphogenesis. Whereas Tc-Spi acts as the main ligand in urogomphi and gin traps, Tc-Vn is required in wings and elytra. Altogether, our findings show that in Tribolium, post-embryonic Tc-Egfr signaling activation depends on the presence of two ligands and that its role in metamorphic transition is conserved in holometabolous insects.


Subject(s)
ErbB Receptors/physiology , Insect Proteins/physiology , Metamorphosis, Biological/physiology , Tribolium/growth & development , Animals , Ecdysone/physiology , ErbB Receptors/genetics , Gene Expression Regulation, Developmental , Insect Proteins/genetics , Larva/growth & development , Metamorphosis, Biological/genetics , Phylogeny , Pupa/growth & development , Real-Time Polymerase Chain Reaction , Signal Transduction , Tribolium/genetics
19.
Dev Biol ; 328(2): 541-51, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19389369

ABSTRACT

Ecdysone signaling plays key roles in Drosophila oogenesis, as its activity is required at multiple steps during egg chamber maturation. Recently, its involvement has been reported on eggshell production by controlling chorion gene transcription and amplification. Here, we present evidence that ecdysone signaling also controls the expression of the eggshell gene VM32E, whose product is a component of vitelline membrane and endochorion layers. Specifically blocking the function of the different Ecdysone receptor (EcR) isoforms we demonstrate that EcR-B1 is responsible for ecdysone-mediated VM32E transcriptional regulation. Moreover, we show that the EcR partner Ultraspiracle (Usp) is also necessary for VM32E expression. By analyzing the activity of specific VM32E regulatory regions in usp(2) clones we identify the promoter region mediating ecdysone-dependent VM32E expression. By in vitro binding assay and site-directed mutagenesis we demonstrate that this region contains a Usp binding site necessary for VM32E regulation. Our results further support the crucial role of ecdysone signaling in controlling transcription of eggshell structural genes and suggest that the heterodimeric complex EcR-B1/Usp mediates the ecdysone-dependent VM32E transcriptional activation in the main body follicle cells.


Subject(s)
DNA-Binding Proteins/physiology , Drosophila Proteins/physiology , Drosophila/physiology , Egg Proteins/physiology , Receptors, Steroid/physiology , Transcription Factors/physiology , Animals , DNA-Binding Proteins/genetics , Drosophila/genetics , Drosophila Proteins/genetics , Ecdysone/physiology , Egg Proteins/genetics , Gene Expression Regulation, Developmental , Mutagenesis, Site-Directed , Oocytes/physiology , Oogenesis , Promoter Regions, Genetic , Protein Isoforms/genetics , Protein Isoforms/physiology , Receptors, Steroid/genetics , Transcription Factors/genetics
20.
Cell Death Differ ; 27(1): 1-14, 2020 01.
Article in English | MEDLINE | ID: mdl-31745213

ABSTRACT

The removal of superfluous and unwanted cells is a critical part of animal development. In insects the steroid hormone ecdysone, the focus of this review, is an essential regulator of developmental transitions, including molting and metamorphosis. Like other steroid hormones, ecdysone works via nuclear hormone receptors to direct spatial and temporal regulation of gene transcription including genes required for cell death. During insect metamorphosis, pulses of ecdysone orchestrate the deletion of obsolete larval tissues, including the larval salivary glands and the midgut. In this review we discuss the molecular machinery and mechanisms of ecdysone-dependent cell and tissue removal, with a focus on studies in Drosophila and Lepidopteran insects.


Subject(s)
Ecdysone/physiology , Animals , Cell Death , Drosophila/growth & development , Lepidoptera/growth & development , Metamorphosis, Biological , Molting
SELECTION OF CITATIONS
SEARCH DETAIL