Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 135
Filter
1.
PLoS Genet ; 20(3): e1011204, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38452112

ABSTRACT

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.


Subject(s)
3-Hydroxysteroid Dehydrogenases , Drosophila , Malnutrition , Animals , Drosophila/physiology , Drosophila melanogaster/physiology , Ecdysone/genetics , Escherichia coli , Larva
2.
Curr Opin Genet Dev ; 84: 102148, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38271845

ABSTRACT

Specifically timed pulses of the moulting hormone ecdysone are necessary for developmental progression in insects, guiding development through important milestones such as larval moults, pupation and metamorphosis. It also coordinates the acquisition of cell identities, known as cell patterning, and growth in a tissue-specific manner. In the absence of ecdysone, the ecdysone receptor heterodimer Ecdysone Receptor and Ultraspiracle represses expression of target primary response genes, which become de-repressed as the ecdysone titre rises. However, ecdysone signalling elicits both repressive and activating responses in a temporal and tissue-specific manner. To understand how ecdysone achieves such specificity, this review explores the layers of gene regulation involved in stage-appropriate ecdysone responses in Drosophila fruit flies.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Drosophila/genetics , Drosophila/metabolism , Ecdysone/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Steroids , Gene Expression Regulation , Larva , Gene Expression Regulation, Developmental/genetics , Drosophila melanogaster
3.
PLoS Genet ; 19(9): e1010924, 2023 09.
Article in English | MEDLINE | ID: mdl-37683015

ABSTRACT

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.


Subject(s)
Circadian Clocks , Animals , Male , Circadian Clocks/genetics , Ecdysone/genetics , Insecta , Circadian Rhythm/genetics , Cell Membrane , Juvenile Hormones/genetics , Mammals
4.
J Exp Biol ; 226(18)2023 09 15.
Article in English | MEDLINE | ID: mdl-37671530

ABSTRACT

Previous studies have shown that selection for starvation resistance in Drosophila melanogaster results in delayed eclosion and increased adult fat stores. It is assumed that these traits are caused by the starvation selection pressure, but its mechanism is unknown. We found that our starvation-selected (SS) population stores more fat during larval development and has extended larval development and pupal development time. Developmental checkpoints in the third instar associated with ecdysteroid hormone pulses are increasingly delayed. The delay in the late larval period seen in the SS population is indicative of reduced and delayed ecdysone signaling. An enzyme immunoassay for ecdysteroids (with greatest affinity to the metabolically active 20-hydroxyecdysone and the α-ecdysone precursor) confirmed that the SS population had reduced and delayed hormone production compared with that of fed control (FC) flies. Feeding third instar larvae on food supplemented with α-ecdysone partially rescued the developmental delay and reduced subsequent adult starvation resistance. This work suggests that starvation selection causes reduced and delayed production of ecdysteroids in the larval stage and affects the developmental delay phenotype that contributes to subsequent adult fat storage and starvation resistance.


Subject(s)
Ecdysone , Ecdysteroids , Animals , Ecdysone/genetics , Drosophila melanogaster/genetics , Larva , Phenotype
5.
PLoS Genet ; 19(6): e1010815, 2023 06.
Article in English | MEDLINE | ID: mdl-37363926

ABSTRACT

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.


Subject(s)
Drosophila Proteins , Prostatic Neoplasms, Castration-Resistant , Humans , Animals , Female , Male , Drosophila/metabolism , Drosophila melanogaster/metabolism , Prostate/pathology , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Endoreduplication , Ecdysone/genetics , Ecdysone/metabolism , E2F1 Transcription Factor/genetics , Transcription Factors/genetics , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism
6.
Dev Biol ; 498: 87-96, 2023 06.
Article in English | MEDLINE | ID: mdl-36967076

ABSTRACT

Signaling networks are redeployed across different developmental times and places to generate phenotypic diversity from a limited genetic toolkit. Hormone signaling networks in particular have well-studied roles in multiple developmental processes. In insects, the ecdysone pathway controls critical events in late embryogenesis and throughout post-embryonic development. While this pathway has not been shown to function in the earliest stage of embryonic development in the model insect Drosophila melanogaster, one component of the network, the nuclear receptor E75A, is necessary for proper segment generation in the milkweed bug Oncopeltus fasciatus. Published expression data from several other species suggests possible conservation of this role across hundreds of millions of years of insect evolution. Previous work also demonstrates a second nuclear receptor in the ecdysone pathway, Ftz-F1, plays a role in segmentation in multiple insect species. Here we report tightly linked expression patterns of ftz-F1 and E75A in two hemimetabolous insect species, the German cockroach Blattella germanica and the two-spotted cricket Gryllus bimaculatus. In both species, the genes are expressed segmentally in adjacent cells, but they are never co-expressed. Using parental RNAi, we show the two genes have distinct roles in early embryogenesis. E75A appears necessary for abdominal segmentation in B. germanica, while ftz-F1 is essential for proper germband formation. Our results suggest that the ecdysone network is critical for early embryogenesis in hemimetabolous insects.


Subject(s)
Ecdysone , Heteroptera , Animals , Ecdysone/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Embryonic Development/genetics , Insecta/genetics , Insecta/metabolism , Insect Proteins/genetics , Insect Proteins/metabolism , Gene Expression Regulation, Developmental/genetics
7.
Mol Biol (Mosk) ; 56(4): 557-563, 2022.
Article in Russian | MEDLINE | ID: mdl-35964312

ABSTRACT

Previously we showed that the CG9890 protein, which has zinc finger domains, interacts with ENY2-containing complexes and is localized mainly on the promoters of active genes. The CG9890 protein is involved in the regulation of the expression of some of the genes on the promoters of which it is located, and among these genes there are genes for the ecdysone cascade. In this work, the role of the CG9890 protein in the regulation of ecdysone-dependent inducible transcription was studied. For this, 12 ecdysone-dependent genes on the promoters of which the CG9890 protein is localized were identified. Their activation was studied after the addition of 20-hydroxyecdysone to cells, both in normal conditions and after RNA interference of CG9890. The expression of ecdysone-dependent genes is significantly increased in response to the treatment of cells with ecdysone, in contrast to the control genes. Moreover, in the cell line after RNA interference CG9890, the transcription of 8 out of 12 genes was significantly higher than in the control line. Thus, the CG9890 protein is involved in the regulation of transcription of ecdysone-dependent genes, and, in most cases, acts as a repressor.


Subject(s)
Drosophila Proteins , Ecdysone , Animals , Drosophila/genetics , Drosophila Proteins/metabolism , Ecdysone/genetics , Ecdysone/metabolism , Promoter Regions, Genetic , Zinc Fingers/genetics
8.
Gene ; 809: 146002, 2022 Jan 30.
Article in English | MEDLINE | ID: mdl-34648919

ABSTRACT

We aimed to explain the reason and function of the successive expression of ecdysone-responsive transcription factors (ERTFs) and related cuticular protein (CP) genes during transformation from larva to pupa. The regulation of the expression of CP genes by ERTFs was examined by in vitro wing disc culture and reporter assay using a gene gun transduction system. Two CP genes that showed expression peaks at different stages-BmorCPG12 at W3L and BmorCPH2 at P0 stage-were selected and examined. Reporter constructs conveying putative BHR3, ßFTZ-F1, BHR39, and E74A binding sites of BmorCPG12 and BmorCPH2 showed promoter activity when introduced into wing discs. In the present study, we showed the functioning of the putative BHR3 and E74A binding sites, together with putative ßFTZ-F1 binding sites, on the activation of CP genes, and different ERTF binding sites functioned in one CP gene. From these, we conclude that BHR3, ßFTZ-F1, and E74A that are successively expressed bring about the successive expression of CP genes, resulting in insect metamorphosis. In addition to this, reporter constructs conveying putative BHR39 binding sites of BmorCPG12 and BmorCPH2 showed negative regulation.


Subject(s)
Bombyx/genetics , Ecdysone/metabolism , Insect Proteins/genetics , Metamorphosis, Biological/genetics , Transcription Factors/genetics , Animals , Binding Sites , Bombyx/physiology , Ecdysone/genetics , Gene Expression Regulation, Developmental , Genes, Reporter , Insect Proteins/metabolism , Larva/genetics , Mutagenesis, Site-Directed , Pupa/genetics , Transcription Factors/metabolism , Wings, Animal/growth & development
9.
PLoS Genet ; 17(11): e1009916, 2021 11.
Article in English | MEDLINE | ID: mdl-34843450

ABSTRACT

Insect metamorphosis is triggered by the production, secretion and degradation of 20-hydroxyecdysone (ecdysone). In addition to its role in developmental regulation, increasing evidence suggests that ecdysone is involved in innate immunity processes, such as phagocytosis and the induction of antimicrobial peptide (AMP) production. AMP regulation includes systemic responses as well as local responses at surface epithelia that contact with the external environment. At pupariation, Drosophila melanogaster increases dramatically the expression of three AMP genes, drosomycin (drs), drosomycin-like 2 (drsl2) and drosomycin-like 5 (drsl5). We show that the systemic action of drs at pupariation is dependent on ecdysone signalling in the fat body and operates via the ecdysone downstream target, Broad. In parallel, ecdysone also regulates local responses, specifically through the activation of drsl2 expression in the gut. Finally, we confirm the relevance of this ecdysone dependent AMP expression for the control of bacterial load by showing that flies lacking drs expression in the fat body have higher bacterial persistence over metamorphosis. In contrast, local responses may be redundant with the systemic effect of drs since reduction of ecdysone signalling or of drsl2 expression has no measurable negative effect on bacterial load control in the pupa. Together, our data emphasize the importance of the association between ecdysone signalling and immunity using in vivo studies and establish a new role for ecdysone at pupariation, which impacts developmental success by regulating the immune system in a stage-dependent manner. We speculate that this co-option of immune effectors by the hormonal system may constitute an anticipatory mechanism to control bacterial numbers in the pupa, at the core of metamorphosis evolution.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Intercellular Signaling Peptides and Proteins/genetics , Metamorphosis, Biological/genetics , Animals , Antimicrobial Peptides/genetics , Drosophila melanogaster/growth & development , Ecdysone/genetics , Ecdysterone/genetics , Gene Expression Regulation, Developmental/genetics , Larva/genetics , Larva/growth & development , Pupa/genetics , Pupa/growth & development , Signal Transduction/genetics
10.
Int J Mol Sci ; 22(16)2021 Aug 23.
Article in English | MEDLINE | ID: mdl-34445782

ABSTRACT

Spodoptera exigua is a worldwide pest afflicting edible vegetables and has developed varying levels of resistance to insecticides. Methoxyfenozide (MET), an ecdysteroid agonist, is effective against lepidopteran pests such as S. exigua. However, the mechanism of MET to S. exigua remains unclear. In this study, we analyzed the expression patterns of genes related to the ecdysone signaling pathway in transcriptome data treated with sublethal doses of MET and analyzed how expression levels of key genes affect the toxicity of MET on S. exigua. Our results demonstrated that 2639 genes were up-regulated and 2512 genes were down-regulated in S. exigua treated with LC30 of MET. Of these, 15 genes were involved in the ecdysone signaling pathway. qPCR results demonstrated that ecdysone receptor A (EcRA) expression levels significantly increased in S. exigua when treated with different doses of MET, and that the RNAi-mediated silencing of EcRA significantly increased mortality to 55.43% at 72 h when L3 S. exigua larvae were exposed to MET at the LC30 dose. Additionally, knocking down EcRA suppressed the most genes expressed in the ecdysone signaling pathway. The combination of MET and dsEcRA affected the expression of E74 and enhanced the expression of TREA. These results demonstrate that the adverse effects of sublethal MET disturb the ecdysone signaling pathway in S. exigua, and EcRA is closely related to MET toxic effect. This study increases our collective understanding of the mechanisms of MET in insect pests.


Subject(s)
Ecdysone/genetics , Hydrazines/pharmacology , Juvenile Hormones/pharmacology , RNA Interference/physiology , Signal Transduction/drug effects , Spodoptera/drug effects , Transcriptome/genetics , Animals , Gene Expression Profiling/methods , Insecticides/pharmacology , Larva/drug effects , Larva/genetics , Receptors, Steroid/genetics , Spodoptera/genetics
11.
Nat Commun ; 12(1): 3328, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099654

ABSTRACT

Innate behaviors consist of a succession of genetically-hardwired motor and physiological subprograms that can be coupled to drastic morphogenetic changes. How these integrative responses are orchestrated is not completely understood. Here, we provide insight into these mechanisms by studying pupariation, a multi-step innate behavior of Drosophila larvae that is critical for survival during metamorphosis. We find that the steroid-hormone ecdysone triggers parallel pupariation neuromotor and morphogenetic subprograms, which include the induction of the relaxin-peptide hormone, Dilp8, in the epidermis. Dilp8 acts on six Lgr3-positive thoracic interneurons to couple both subprograms in time and to instruct neuromotor subprogram switching during behavior. Our work reveals that interorgan feedback gates progression between subunits of an innate behavior and points to an ancestral neuromodulatory function of relaxin signaling.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Ecdysone/pharmacology , Epidermis/metabolism , Morphogenesis/drug effects , Neurons/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Ecdysone/genetics , Epidermal Cells/metabolism , Intercellular Signaling Peptides and Proteins , Larva/metabolism , Metamorphosis, Biological , Morphogenesis/genetics , Receptors, G-Protein-Coupled/genetics , Relaxin/metabolism
12.
Dev Cell ; 56(12): 1742-1755.e4, 2021 06 21.
Article in English | MEDLINE | ID: mdl-34081907

ABSTRACT

Organogenesis requires exquisite spatiotemporal coordination of cell morphogenesis, migration, proliferation, and differentiation of multiple cell types. For gonads, this involves complex interactions between somatic and germline tissues. During Drosophila ovary morphogenesis, primordial germ cells (PGCs) either are sequestered in stem cell niches and are maintained in an undifferentiated germline stem cell state or transition directly toward differentiation. Here, we identify a mechanism that links hormonal triggers of somatic tissue morphogenesis with PGC differentiation. An early ecdysone pulse initiates somatic swarm cell (SwC) migration, positioning these cells close to PGCs. A second hormone peak activates Torso-like signal in SwCs, which stimulates the Torso receptor tyrosine kinase (RTK) signaling pathway in PGCs promoting their differentiation by de-repression of the differentiation gene, bag of marbles. Thus, systemic temporal cues generate a transitory signaling center that coordinates ovarian morphogenesis with stem cell self-renewal and differentiation programs, highlighting a more general role for such centers in reproductive and developmental biology.


Subject(s)
Cell Differentiation/genetics , Drosophila Proteins/genetics , Germ Cells/growth & development , Morphogenesis/genetics , Ovary/growth & development , Receptor Protein-Tyrosine Kinases/genetics , Animals , Cell Movement/genetics , Cell Proliferation/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Ecdysone/genetics , Female , Gene Expression Regulation, Developmental/genetics , Larva/genetics , Larva/growth & development , Ovary/metabolism
13.
Development ; 148(6)2021 03 19.
Article in English | MEDLINE | ID: mdl-33658221

ABSTRACT

Regeneration of Drosophila imaginal discs, larval precursors to adult tissues, activates a regeneration checkpoint that coordinates regenerative growth with developmental progression. This regeneration checkpoint results from the release of the relaxin-family peptide Dilp8 from regenerating imaginal tissues. Secreted Dilp8 protein is detected within the imaginal disc lumen, in which it is separated from its receptor target Lgr3, which is expressed in the brain and prothoracic gland, by the disc epithelial barrier. Here, we demonstrate that following damage the imaginal disc epithelial barrier limits Dilp8 signaling and the duration of regeneration checkpoint delay. We also find that the barrier becomes increasingly impermeable to the transepithelial diffusion of labeled dextran during the second half of the third instar. This change in barrier permeability is driven by the steroid hormone ecdysone and correlates with changes in localization of Coracle, a component of the septate junctions that is required for the late-larval impermeable epithelial barrier. Based on these observations, we propose that the imaginal disc epithelial barrier regulates the duration of the regenerative checkpoint, providing a mechanism by which tissue function can signal the completion of regeneration.


Subject(s)
Drosophila Proteins/genetics , Imaginal Discs/growth & development , Intercellular Signaling Peptides and Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Regeneration/genetics , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Ecdysone/genetics , Gene Expression Regulation, Developmental , Imaginal Discs/metabolism , Larva/genetics , Larva/growth & development , Neurons/metabolism , Signal Transduction/genetics
14.
Curr Opin Genet Dev ; 69: 82-87, 2021 08.
Article in English | MEDLINE | ID: mdl-33740694

ABSTRACT

Phenotypic plasticity in response to environmental cues is common in butterflies, and is a major driver of butterfly wing pattern diversity. The endocrine signal ecdysone has been revealed as a major modulator of plasticity in butterflies. External cues such as day length or temperature are translated internally into variation in ecdysone titers, which in turn lead to alternate phenotypes such as seasonal wing patterns. Here we review the evidence showing that ecdysone-mediated plasticity of different wing pattern features such as wing color and eyespot size can evolve independently. Recent studies show that ecdysone regulates gene expression in Drosophila melanogaster via a chromatin remodeling mechanism. We thus propose that environmentally responsive ecdysone titers in butterflies may also function via chromatin regulation to promote different seasonal phenotypes. We present a model of ecdysone response evolution that integrates both gene regulatory architecture and organismal development, and propose a set of testable mechanistic hypotheses for how plastic response profiles of specific genes can evolve.


Subject(s)
Biological Evolution , Butterflies/genetics , Pigmentation/genetics , Wings, Animal/anatomy & histology , Adaptation, Physiological/genetics , Animals , Butterflies/anatomy & histology , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/genetics , Ecdysone/genetics , Gene Expression Regulation, Developmental/genetics , Phenotype , Seasons , Wings, Animal/growth & development
15.
Open Biol ; 11(2): 200373, 2021 02.
Article in English | MEDLINE | ID: mdl-33593157

ABSTRACT

In both mammals and insects, steroid hormones play a major role in directing the animal's progression through developmental stages. To maximize fitness outcomes, steroid hormone production is regulated by the environmental conditions experienced by the animal. In insects, the steroid hormone ecdysone mediates transitions between developmental stages and is regulated in response to environmental factors such as nutrition. These environmental signals are communicated to the ecdysone-producing gland via the action of neuropeptide and peptide hormone signalling pathways. While some of these pathways have been well characterized, there is evidence to suggest more signalling pathways than has previously been thought function to control ecdysone production, potentially in response to a greater range of environmental conditions. Here, we review the neuropeptide and peptide hormone signalling pathways known to regulate the production of ecdysone in the model genetic insect Drosophila melanogaster, as well as what is known regarding the environmental signals that trigger these pathways. Areas for future research are highlighted that can further contribute to our overall understanding of the complex orchestration of environmental, physiological and developmental cues that together produce a functioning adult organism.


Subject(s)
Drosophila Proteins/metabolism , Ecdysone/biosynthesis , Neuropeptides/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Ecdysone/genetics , Gene Expression Regulation, Developmental , Neuropeptides/genetics
16.
PLoS Genet ; 17(1): e1009300, 2021 01.
Article in English | MEDLINE | ID: mdl-33507966

ABSTRACT

Highly reproducible tissue development is achieved by robust, time-dependent coordination of cell proliferation and cell death. To study the mechanisms underlying robust tissue growth, we analyzed the developmental process of wing imaginal discs in Drosophila Minute mutants, a series of heterozygous mutants for a ribosomal protein gene. Minute animals show significant developmental delay during the larval period but develop into essentially normal flies, suggesting there exists a mechanism ensuring robust tissue growth during abnormally prolonged developmental time. Surprisingly, we found that both cell death and compensatory cell proliferation were dramatically increased in developing wing pouches of Minute animals. Blocking the cell-turnover by inhibiting cell death resulted in morphological defects, indicating the essential role of cell-turnover in Minute wing morphogenesis. Our analyses showed that Minute wing discs elevate Wg expression and JNK-mediated Dilp8 expression that causes developmental delay, both of which are necessary for the induction of cell-turnover. Furthermore, forced increase in Wg expression together with developmental delay caused by ecdysone depletion induced cell-turnover in the wing pouches of non-Minute animals. Our findings suggest a novel paradigm for robust coordination of tissue growth by cell-turnover, which is induced when developmental time axis is distorted.


Subject(s)
Drosophila Proteins/genetics , Imaginal Discs/growth & development , Intercellular Signaling Peptides and Proteins/genetics , Ribosomal Proteins/genetics , Wnt1 Protein/genetics , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Ecdysone/genetics , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental/genetics , Imaginal Discs/metabolism , Larva/genetics , Larva/growth & development , Metamorphosis, Biological/genetics , Organogenesis/genetics , Signal Transduction/genetics , Transcription Factors/genetics , Wings, Animal/growth & development , Wings, Animal/metabolism
17.
Mech Dev ; 163: 103626, 2020 09.
Article in English | MEDLINE | ID: mdl-32526278

ABSTRACT

'Developmental robustness' is the ability of biological systems to maintain a stable phenotype despite genetic, environmental or physiological perturbations. In holometabolous insects, accurate patterning and development is guaranteed by alignment of final gene expression patterns in tissues at specific developmental stage such as molting and pupariation, irrespective of individual rate of development. In the present study, we used faster developing Drosophila melanogaster populations that show reduction of ~22% in egg to adult development time. Flies from the faster developing population exhibit phenotype constancy, although significantly small in size. The reduction in development time in faster developing flies is possibly due to coordination between higher ecdysteroid release and higher expression of developmental genes. The two together might be ensuring appropriate pattern formation and early exit at each development stage in the populations selected for faster pre-adult development compared to their ancestral controls. We report that apart from plasticity in the rate of pattern progression, alteration in the level of gene expression may be responsible for pattern integrity even under reduced development time.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Ecdysone/genetics , Wings, Animal/growth & development , Wnt1 Protein/genetics , Animals , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental/genetics , Larva/genetics , Larva/growth & development , Signal Transduction
18.
Biochim Biophys Acta Gene Regul Mech ; 1863(8): 194576, 2020 08.
Article in English | MEDLINE | ID: mdl-32389826

ABSTRACT

Juvenile hormones (JH) and ecdysone coordinately regulate metamorphosis in Aedes aegypti. We studied the function of an epigenetic regulator and multifunctional transactivator, CREB binding protein (CBP) in A. aegypti. RNAi-mediated knockdown of CBP in Ae. aegypti larvae resulted in suppression of JH primary response gene, Krüppel-homolog 1 (Kr-h1), and induction of primary ecdysone response gene, E93, resulting in multiple effects including early metamorphosis, larval-pupal intermediate formation, mortality and inhibition of compound eye development. RNA sequencing identified hundreds of genes, including JH and ecdysone response genes regulated by CBP. In the presence of JH, CBP upregulates Kr-h1 by acetylating core histones at the Kr-h1 promoter and facilitating the recruitment of JH receptor and other proteins. CBP suppresses metamorphosis regulators, EcR-A, USP-A, BR-C, and E93 through the upregulation of Kr-h1 and E75A. CBP regulates the expression of core eye specification genes including those involved in TGF-ß and EGFR signaling. These studies demonstrate that CBP is an essential player in JH and 20E action and regulates metamorphosis and compound eye development in Ae. aegypti.


Subject(s)
Aedes/metabolism , CREB-Binding Protein/metabolism , Eye/growth & development , Metamorphosis, Biological/physiology , Organogenesis/physiology , Aedes/genetics , Animals , CREB-Binding Protein/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins , Ecdysone/genetics , Ecdysone/metabolism , Ecdysone/pharmacology , Female , Gene Expression Regulation, Developmental , Histones/metabolism , Insect Proteins/genetics , Insect Proteins/metabolism , Juvenile Hormones/metabolism , Juvenile Hormones/pharmacology , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Larva , Organogenesis/drug effects , Organogenesis/genetics , Promoter Regions, Genetic , Pupa/growth & development , Signal Transduction , Transcription Factors/metabolism , Yellow Fever/genetics
19.
PLoS One ; 15(4): e0231451, 2020.
Article in English | MEDLINE | ID: mdl-32282855

ABSTRACT

Insect molting hormone (ecdysteroids) and juvenile hormone regulate molting and metamorphic events in a variety of insect species. Mealybugs undergo sexually dimorphic metamorphosis: males develop into winged adults through non-feeding, pupa-like stages called prepupa and pupa, while females emerge as neotenic wingless adults. We previously demonstrated, in the Japanese mealybug Planococcus kraunhiae (Kuwana), that the juvenile hormone titer is higher in males than in females at the end of the juvenile stage, which suggests that juvenile hormone may regulate male-specific adult morphogenesis. Here, we examined the involvement of ecdysteroids in sexually dimorphic metamorphosis. To estimate ecdysteroid titers, quantitative RT-PCR analyses of four Halloween genes encoding for cytochrome P450 monooxygenases in ecdysteroid biosynthesis, i.e., spook, disembodied, shadow and shade, were performed. Overall, their expression levels peaked before each nymphal molt. Transcript levels of spook, disembodied and shadow, genes that catalyze the steps in ecdysteroid biosynthesis in the prothoracic gland, were higher in males from the middle of the second nymphal instar to adult emergence. In contrast, the expression of shade, which was reported to be involved in the conversion of ecdysone into 20-hydroxyecdysone in peripheral tissues, was similar between males and females. These results suggest that ecdysteroid biosynthesis in the prothoracic gland is more active in males than in females, although the final conversion into 20-hydroxyecdysone occurs at similar levels in both sexes. Moreover, expression profiles of ecdysone response genes, ecdysone receptor and ecdysone-induced protein 75B, were also analyzed. Based on these expression profiles, we propose that the changes in ecdysteroid titer differ between males and females, and that high ecdysteroid titer is essential for directing male adult development.


Subject(s)
Ecdysone/genetics , Ecdysteroids/genetics , Insect Proteins/genetics , Insecta/genetics , Animals , Cytochrome P-450 Enzyme System/genetics , Ecdysterone/genetics , Female , Gene Expression Regulation, Developmental/genetics , Insecta/growth & development , Juvenile Hormones/genetics , Larva/genetics , Larva/growth & development , Male , Metamorphosis, Biological/genetics , Morphogenesis/genetics , Pupa/genetics , Pupa/growth & development , Sex Characteristics , Wings, Animal/growth & development
20.
Int J Biol Macromol ; 150: 965-973, 2020 May 01.
Article in English | MEDLINE | ID: mdl-31758989

ABSTRACT

The development of insect wings is a complex process controlled by a series of genes, whereas the mechanism of wing development of orthoptera insects is less frequently reported. In the present study, a BTB domain-containing protein 6 (LmBTBD6) gene was identified from Locusta migratoria. Its encoded protein belongs to the BTB-BACK-PHR subfamily, and is highly conserved among insect species. LmBTBD6 was mainly expressed in the wing pads and showed high expression on day 7 of fifth-instar nymphs. LmBTBD6 responded to induction by 20-Hydroxyecdysone (20E) in vivo, and its expression was significantly suppressed after knocking down the ecdysone receptor gene LmEcR and nuclear receptor gene LmHR39. Deficiency of LmBTBD6 did not show visible phenotype in the wing pads transition from nymph to nymph of L. migratoria, but caused wing defects in the transition from nymph to adult. After silencing of LmBTBD6, the transcription of wing development-related genes (LmSal411, LmSal468, and LmHth) and the wing-specific cuticle protein genes (LmACP7 and LmACP8) of L. migratoria were significantly suppressed. Thus, LmBTBD6 that regulated by the LmEcR-LmHR39-mediated 20E signaling pathway is involved in wing development during the nymph to adult transition by regulating the expression of wing development-related genes and wing-specific cuticle protein genes.


Subject(s)
BTB-POZ Domain/physiology , Locusta migratoria/metabolism , Nymph/genetics , Nymph/metabolism , Wings, Animal/growth & development , Wings, Animal/metabolism , Animals , Ecdysone/genetics , Ecdysone/metabolism , Ecdysterone , Gene Expression Regulation, Developmental , Insect Proteins/genetics , Insect Proteins/metabolism , Locusta migratoria/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA Interference , Sequence Alignment , Signal Transduction , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...