1
|
Zipper L, Corominas-Murtra B, Reiff T. Steroid hormone-induced wingless ligands tune female intestinal size in Drosophila. Nat Commun 2025; 16:436. [PMID: 39762218 PMCID: PMC11704138 DOI: 10.1038/s41467-024-55664-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Female reproduction comes at great expense to energy metabolism compensated by extensive organ adaptations including intestinal size. Upon mating, endocrine signals orchestrate a 30% net increase of absorptive epithelium. Mating increases production of the steroid hormone Ecdysone released by the Drosophila ovaries that stimulates intestinal stem cell (ISC) divisions. Here, we uncover the transcription factor crooked legs (crol) as an intraepithelial coordinator of Ecdysone-induced ISC mitosis. For the precise investigation of non-autonomous factors on ISC behaviour, we establish Rapport, a spatiotemporally-controlled dual expression and tracing system for the analysis of paracrine genetic manipulation while tracing ISC behaviour. Rapport tracing reveals that Ecdysone-induced Crol controls mitogenic Wnt/Wg-ligand expression from epithelial enterocytes activating ISC mitosis. Paracrine Wg stimulation is counterbalanced by Crol-repression of string/CDC25 and CyclinB autonomously in ISC. Rapport-based ISC tumours confirm paracrine stimulation through the Ecdysone-Crol-Wg axis on mitotic behaviour, whereas the autonomous anti-proliferative role of Crol in ISC is conserved in models of colorectal cancer. Finally, mathematical modelling corroborates increasing enterocyte numbers and Wnt/Wg-degradation to set a stable post-mating intestinal size. Together, our findings provide insights into the complex endocrine growth control mechanisms during mating-induced adaptations and might help untangling pleiotropic hormonal effects observed in gastrointestinal tumorigenesis.
Collapse
Affiliation(s)
- Lisa Zipper
- Department of Biology, Institute of Genetics, The Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | | | - Tobias Reiff
- Department of Biology, Institute of Genetics, The Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
2
|
Parasram K, Zuccato A, Shin M, Willms R, DeVeale B, Foley E, Karpowicz P. The emergence of circadian timekeeping in the intestine. Nat Commun 2024; 15:1788. [PMID: 38413599 PMCID: PMC10899604 DOI: 10.1038/s41467-024-45942-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 02/08/2024] [Indexed: 02/29/2024] Open
Abstract
The circadian clock is a molecular timekeeper, present from cyanobacteria to mammals, that coordinates internal physiology with the external environment. The clock has a 24-h period however development proceeds with its own timing, raising the question of how these interact. Using the intestine of Drosophila melanogaster as a model for organ development, we track how and when the circadian clock emerges in specific cell types. We find that the circadian clock begins abruptly in the adult intestine and gradually synchronizes to the environment after intestinal development is complete. This delayed start occurs because individual cells at earlier stages lack the complete circadian clock gene network. As the intestine develops, the circadian clock is first consolidated in intestinal stem cells with changes in Ecdysone and Hnf4 signalling influencing the transcriptional activity of Clk/cyc to drive the expression of tim, Pdp1, and vri. In the mature intestine, stem cell lineage commitment transiently disrupts clock activity in differentiating progeny, mirroring early developmental clock-less transitions. Our data show that clock function and differentiation are incompatible and provide a paradigm for studying circadian clocks in development and stem cell lineages.
Collapse
Affiliation(s)
- Kathyani Parasram
- Department of Biomedical Sciences, University of Windsor, Windsor, ON, N9B 3P4, Canada
| | - Amy Zuccato
- Department of Biomedical Sciences, University of Windsor, Windsor, ON, N9B 3P4, Canada
| | - Minjeong Shin
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Reegan Willms
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Brian DeVeale
- Department of Biomedical Sciences, University of Windsor, Windsor, ON, N9B 3P4, Canada
| | - Edan Foley
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Phillip Karpowicz
- Department of Biomedical Sciences, University of Windsor, Windsor, ON, N9B 3P4, Canada.
| |
Collapse
|
3
|
Monier M, Nuez I, Borne F, Courtier-Orgogozo V. Higher evolutionary dynamics of gene copy number for Drosophila glue genes located near short repeat sequences. BMC Ecol Evol 2024; 24:18. [PMID: 38308233 PMCID: PMC10835880 DOI: 10.1186/s12862-023-02178-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 11/23/2023] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND During evolution, genes can experience duplications, losses, inversions and gene conversions. Why certain genes are more dynamic than others is poorly understood. Here we examine how several Sgs genes encoding glue proteins, which make up a bioadhesive that sticks the animal during metamorphosis, have evolved in Drosophila species. RESULTS We examined high-quality genome assemblies of 24 Drosophila species to study the evolutionary dynamics of four glue genes that are present in D. melanogaster and are part of the same gene family - Sgs1, Sgs3, Sgs7 and Sgs8 - across approximately 30 millions of years. We annotated a total of 102 Sgs genes and grouped them into 4 subfamilies. We present here a new nomenclature for these Sgs genes based on protein sequence conservation, genomic location and presence/absence of internal repeats. Two types of glue genes were uncovered. The first category (Sgs1, Sgs3x, Sgs3e) showed a few gene losses but no duplication, no local inversion and no gene conversion. The second group (Sgs3b, Sgs7, Sgs8) exhibited multiple events of gene losses, gene duplications, local inversions and gene conversions. Our data suggest that the presence of short "new glue" genes near the genes of the latter group may have accelerated their dynamics. CONCLUSIONS Our comparative analysis suggests that the evolutionary dynamics of glue genes is influenced by genomic context. Our molecular, phylogenetic and comparative analysis of the four glue genes Sgs1, Sgs3, Sgs7 and Sgs8 provides the foundation for investigating the role of the various glue genes during Drosophila life.
Collapse
Affiliation(s)
- Manon Monier
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013, Paris, France
| | - Isabelle Nuez
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013, Paris, France
| | - Flora Borne
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013, Paris, France
- Department of Biological Sciences, Columbia University, New York city, New York, USA
| | | |
Collapse
|
4
|
Abstract
Endocrine signaling networks control diverse biological processes and life history traits across metazoans. In both invertebrate and vertebrate taxa, steroid hormones regulate immune system function in response to intrinsic and environmental stimuli, such as microbial infection. The mechanisms of this endocrine-immune regulation are complex and constitute an ongoing research endeavor facilitated by genetically tractable animal models. The 20-hydroxyecdysone (20E) is the major steroid hormone in arthropods, primarily studied for its essential role in mediating developmental transitions and metamorphosis; 20E also modulates innate immunity in a variety of insect taxa. This review provides an overview of our current understanding of 20E-mediated innate immune responses. The prevalence of correlations between 20E-driven developmental transitions and innate immune activation are summarized across a range of holometabolous insects. Subsequent discussion focuses on studies conducted using the extensive genetic resources available in Drosophila that have begun to reveal the mechanisms underlying 20E regulation of immunity in the contexts of both development and bacterial infection. Lastly, I propose directions for future research into 20E regulation of immunity that will advance our knowledge of how interactive endocrine networks coordinate animals' physiological responses to environmental microbes.
Collapse
Affiliation(s)
- Scott A. Keith
- Department of Entomology, Cornell University, Ithaca, New York, United States of America
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
5
|
Opportunistic binding of EcR to open chromatin drives tissue-specific developmental responses. Proc Natl Acad Sci U S A 2022; 119:e2208935119. [PMID: 36161884 DOI: 10.1073/pnas.2208935119] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Steroid hormones perform diverse biological functions in developing and adult animals. However, the mechanistic basis for their tissue specificity remains unclear. In Drosophila, the ecdysone steroid hormone is essential for coordinating developmental timing across physically separated tissues. Ecdysone directly impacts genome function through its nuclear receptor, a heterodimer of the EcR and ultraspiracle proteins. Ligand binding to EcR triggers a transcriptional cascade, including activation of a set of primary response transcription factors. The hierarchical organization of this pathway has left the direct role of EcR in mediating ecdysone responses obscured. Here, we investigate the role of EcR in controlling tissue-specific ecdysone responses, focusing on two tissues that diverge in their response to rising ecdysone titers: the larval salivary gland, which undergoes programmed destruction, and the wing imaginal disc, which initiates morphogenesis. We find that EcR functions bimodally, with both gene repressive and activating functions, even at the same developmental stage. EcR DNA binding profiles are highly tissue-specific, and transgenic reporter analyses demonstrate that EcR plays a direct role in controlling enhancer activity. Finally, despite a strong correlation between tissue-specific EcR binding and tissue-specific open chromatin, we find that EcR does not control chromatin accessibility at genomic targets. We conclude that EcR contributes extensively to tissue-specific ecdysone responses. However, control over access to its binding sites is subordinated to other transcription factors.
Collapse
|
6
|
Monier M, Courtier-Orgogozo V. Drosophila Glue: A Promising Model for Bioadhesion. INSECTS 2022; 13:734. [PMID: 36005360 PMCID: PMC9409817 DOI: 10.3390/insects13080734] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 06/15/2023]
Abstract
The glue produced by Drosophila larvae to attach themselves to a substrate for several days and resist predation until the end of metamorphosis represents an attractive model to develop new adhesives for dry environments. The adhesive properties of this interesting material have been investigated recently, and it was found that it binds as well as strongly adhesive commercial tapes to various types of substrates. This glue hardens rapidly after excretion and is made of several proteins. In D. melanogaster, eight glue proteins have been identified: four are long glycosylated mucoproteins containing repeats rich in prolines, serines and threonines, and four others are shorter proteins rich in cysteines. This protein mix is produced by the salivary glands through a complex packaging process that is starting to be elucidated. Drosophila species have adapted to stick to various substrates in diverse environmental conditions and glue genes appear to evolve rapidly in terms of gene number, number of repeats and sequence of the repeat motifs. Interestingly, besides its adhesive properties, the glue may also have antimicrobial activities. We discuss future perspectives and avenues of research for the development of new bioadhesives mimicking Drosophila fly glue.
Collapse
|
7
|
Kumar Ghosh S, Chatterjee T, Mitra S, Chakravarty A, Chakravarty S, Kumar Basak A. Benzaldehyde-induced developmental genotoxicity triggers both neural and non-neuronal cells including the cells of immunity in Drosophila melanogaster. Biol Futur 2022; 73:245-257. [DOI: 10.1007/s42977-022-00116-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 04/05/2022] [Indexed: 11/30/2022]
|
8
|
Ohde T, Mito T, Niimi T. A hemimetabolous wing development suggests the wing origin from lateral tergum of a wingless ancestor. Nat Commun 2022; 13:979. [PMID: 35190538 PMCID: PMC8861169 DOI: 10.1038/s41467-022-28624-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 01/24/2022] [Indexed: 11/26/2022] Open
Abstract
The origin and evolution of the novel insect wing remain enigmatic after a century-long discussion. The mechanism of wing development in hemimetabolous insects, in which the first functional wings evolved, is key to understand where and how insect wings evolutionarily originate. This study explored the developmental origin and the postembryonic dramatic growth of wings in the cricket Gryllus bimaculatus. We find that the lateral tergal margin, which is homologous between apterygote and pterygote insects, comprises a growth organizer to expand the body wall to form adult wing blades in Gryllus. We also find that Wnt, Fat-Dachsous, and Hippo pathways are involved in the disproportional growth of Gryllus wings. These data provide insights into where and how insect wings originate. Wings evolved from the pre-existing lateral terga of a wingless insect ancestor, and the reactivation or redeployment of Wnt/Fat-Dachsous/Hippo-mediated feed-forward circuit might have expanded the lateral terga. Here, the authors investigate wing development in cricket and find support for evolution of the novel insect wing from the pre-existing dorsal body wall of a wingless ancestor by activation of an evolutionarily conserved growth mechanism.
Collapse
|
9
|
Chang Y, Zhang B, Du M, Geng Z, Wei J, Guan R, An S, Zhao W. The vital hormone 20-hydroxyecdysone controls ATP production by upregulating binding of trehalase 1 with ATP synthase subunit α in Helicoverpa armigera. J Biol Chem 2022; 298:101565. [PMID: 34999119 PMCID: PMC8819028 DOI: 10.1016/j.jbc.2022.101565] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 12/02/2022] Open
Abstract
Trehalose is the major “blood sugar” of insects and it plays a crucial role in energy supply and as a stress protectant. The hydrolysis of trehalose occurs only under the enzymatic control of trehalase (Treh), which plays important roles in growth and development, energy supply, chitin biosynthesis, and abiotic stress responses. Previous reports have revealed that the vital hormone 20-hydroxyecdysone (20E) regulates Treh, but the detailed mechanism underlying 20E regulating Treh remains unclear. In this study, we investigated the function of HaTreh1 in Helicoverpa armigera larvae. The results showed that the transcript levels and enzymatic activity of HaTreh1 were elevated during molting and metamorphosis stages in the epidermis, midgut, and fat body, and that 20E upregulated the transcript levels of HaTreh1 through the classical nuclear receptor complex EcR-B1/USP1. HaTreh1 is a mitochondria protein. We also found that knockdown of HaTreh1 in the fifth- or sixth-instar larvae resulted in weight loss and increased mortality. Yeast two-hybrid, coimmunoprecipitation, and glutathione-S-transferase (GST) pull-down experiments demonstrated that HaTreh1 bound with ATP synthase subunit alpha (HaATPs-α) and that this binding increased under 20E treatment. In addition, 20E enhanced the transcript level of HaATPs-α and ATP content. Finally, the knockdown of HaTreh1 or HaATPs-α decreased the induction effect of 20E on ATP content. Altogether, these findings demonstrate that 20E controls ATP production by up-regulating the binding of HaTreh1 to HaATPs-α in H. armigera.
Collapse
Affiliation(s)
- Yanpeng Chang
- State key Laboratory of Wheat and Maize Crop Science/College of Plant Protection, Henan Agricultural University, Zhengzhou, China
| | - Bo Zhang
- State key Laboratory of Wheat and Maize Crop Science/College of Plant Protection, Henan Agricultural University, Zhengzhou, China
| | - Mengfang Du
- State key Laboratory of Wheat and Maize Crop Science/College of Plant Protection, Henan Agricultural University, Zhengzhou, China
| | - Zichen Geng
- State key Laboratory of Wheat and Maize Crop Science/College of Plant Protection, Henan Agricultural University, Zhengzhou, China
| | - Jizhen Wei
- State key Laboratory of Wheat and Maize Crop Science/College of Plant Protection, Henan Agricultural University, Zhengzhou, China
| | - Ruobing Guan
- State key Laboratory of Wheat and Maize Crop Science/College of Plant Protection, Henan Agricultural University, Zhengzhou, China
| | - Shiheng An
- State key Laboratory of Wheat and Maize Crop Science/College of Plant Protection, Henan Agricultural University, Zhengzhou, China
| | - Wenli Zhao
- State key Laboratory of Wheat and Maize Crop Science/College of Plant Protection, Henan Agricultural University, Zhengzhou, China.
| |
Collapse
|
10
|
Borne F, Kulathinal RJ, Courtier-Orgogozo V. Glue Genes Are Subjected to Diverse Selective Forces during Drosophila Development. Genome Biol Evol 2021; 13:6426080. [PMID: 34788814 PMCID: PMC8643421 DOI: 10.1093/gbe/evab248] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2021] [Indexed: 12/18/2022] Open
Abstract
Molecular evolutionary studies usually focus on genes with clear roles in adult fitness or on developmental genes expressed at multiple time points during the life of the organism. Here, we examine the evolutionary dynamics of Drosophila glue genes, a set of eight genes tasked with a singular primary function during a specific developmental stage: the production of glue that allows animal pupa to attach to a substrate for several days during metamorphosis. Using phenotypic assays and available data from transcriptomics, PacBio genomes, and sequence variation from global populations, we explore the selective forces acting on glue genes within the cosmopolitan Drosophila melanogaster species and its five closely related species, D. simulans, D. sechellia, D. mauritiana, D. yakuba, and D. teissieri. We observe a three-fold difference in glue adhesion between the least and the most adhesive D. melanogaster strain, indicating a strong genetic component to phenotypic variation. These eight glue genes are among the most highly expressed genes in salivary glands yet they display no notable codon bias. New copies of Sgs3 and Sgs7 are found in D. yakuba and D. teissieri with the Sgs3 coding sequence evolving rapidly after duplication in the D. yakuba branch. Multiple sites along the various glue genes appear to be constrained. Our population genetics analysis in D. melanogaster suggests signals of local adaptive evolution for Sgs3, Sgs5, and Sgs5bis and traces of selective sweeps for Sgs1, Sgs3, Sgs7, and Sgs8. Our work shows that stage-specific genes can be subjected to various dynamic evolutionary forces.
Collapse
Affiliation(s)
- Flora Borne
- CNRS, Institut Jacques Monod, Université de Paris, France
| | - Rob J Kulathinal
- CNRS, Institut Jacques Monod, Université de Paris, France.,Department of Biology, Temple University, USA
| | | |
Collapse
|
11
|
Suzuki Y, Toh L. Constraints and Opportunities for the Evolution of Metamorphic Organisms in a Changing Climate. Front Ecol Evol 2021. [DOI: 10.3389/fevo.2021.734031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We argue that developmental hormones facilitate the evolution of novel phenotypic innovations and timing of life history events by genetic accommodation. Within an individual’s life cycle, metamorphic hormones respond readily to environmental conditions and alter adult phenotypes. Across generations, the many effects of hormones can bias and at times constrain the evolution of traits during metamorphosis; yet, hormonal systems can overcome constraints through shifts in timing of, and acquisition of tissue specific responses to, endocrine regulation. Because of these actions of hormones, metamorphic hormones can shape the evolution of metamorphic organisms. We present a model called a developmental goblet, which provides a visual representation of how metamorphic organisms might evolve. In addition, because developmental hormones often respond to environmental changes, we discuss how endocrine regulation of postembryonic development may impact how organisms evolve in response to climate change. Thus, we propose that developmental hormones may provide a mechanistic link between climate change and organismal adaptation.
Collapse
|
12
|
Dib A, Zanet J, Mancheno-Ferris A, Gallois M, Markus D, Valenti P, Marques-Prieto S, Plaza S, Kageyama Y, Chanut-Delalande H, Payre F. Pri smORF Peptides Are Wide Mediators of Ecdysone Signaling, Contributing to Shape Spatiotemporal Responses. Front Genet 2021; 12:714152. [PMID: 34527021 DOI: 10.3389/fgene.2021.714152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/28/2021] [Indexed: 11/13/2022] Open
Abstract
There is growing evidence that peptides encoded by small open-reading frames (sORF or smORF) can fulfill various cellular functions and define a novel class regulatory molecules. To which extend transcripts encoding only smORF peptides compare with canonical protein-coding genes, yet remain poorly understood. In particular, little is known on whether and how smORF-encoding RNAs might need tightly regulated expression within a given tissue, at a given time during development. We addressed these questions through the analysis of Drosophila polished rice (pri, a.k.a. tarsal less or mille pattes), which encodes four smORF peptides (11-32 amino acids in length) required at several stages of development. Previous work has shown that the expression of pri during epidermal development is regulated in the response to ecdysone, the major steroid hormone in insects. Here, we show that pri transcription is strongly upregulated by ecdysone across a large panel of cell types, suggesting that pri is a core component of ecdysone response. Although pri is produced as an intron-less short transcript (1.5 kb), genetic assays reveal that the developmental functions of pri require an unexpectedly large array of enhancers (spanning over 50 kb), driving a variety of spatiotemporal patterns of pri expression across developing tissues. Furthermore, we found that separate pri enhancers are directly activated by the ecdysone nuclear receptor (EcR) and display distinct regulatory modes between developmental tissues and/or stages. Alike major developmental genes, the expression of pri in a given tissue often involves several enhancers driving apparently redundant (or shadow) expression, while individual pri enhancers can harbor pleiotropic functions across tissues. Taken together, these data reveal the broad role of Pri smORF peptides in ecdysone signaling and show that the cis-regulatory architecture of the pri gene contributes to shape distinct spatial and temporal patterns of ecdysone response throughout development.
Collapse
Affiliation(s)
- Azza Dib
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), CNRS, UPS, University of Toulouse, Toulouse, France
| | - Jennifer Zanet
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), CNRS, UPS, University of Toulouse, Toulouse, France
| | - Alexandra Mancheno-Ferris
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), CNRS, UPS, University of Toulouse, Toulouse, France
| | - Maylis Gallois
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), CNRS, UPS, University of Toulouse, Toulouse, France
| | - Damien Markus
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), CNRS, UPS, University of Toulouse, Toulouse, France
| | - Philippe Valenti
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), CNRS, UPS, University of Toulouse, Toulouse, France
| | - Simon Marques-Prieto
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), CNRS, UPS, University of Toulouse, Toulouse, France
| | - Serge Plaza
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), CNRS, UPS, University of Toulouse, Toulouse, France
| | - Yuji Kageyama
- Department of Biology, Graduate School of Science, Kobe University, Kobe, Japan.,Biosignal Research Center, Kobe University, Kobe, Japan
| | - Hélène Chanut-Delalande
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), CNRS, UPS, University of Toulouse, Toulouse, France
| | - François Payre
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), CNRS, UPS, University of Toulouse, Toulouse, France
| |
Collapse
|
13
|
Comparative transcriptome analysis reveals a potential mechanism for host nutritional manipulation after parasitization by Leptopilina boulardi. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2021; 39:100862. [PMID: 34120097 DOI: 10.1016/j.cbd.2021.100862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 05/13/2021] [Accepted: 06/04/2021] [Indexed: 01/18/2023]
Abstract
Parasitoids have been extensively found to manipulate nutrient amounts of their hosts to benefit their own development and survival, but the underlying mechanisms are largely unknown. Leptopilina boulardi (Hymenoptera: Figitidae) is a larval-pupal endoparasitoid wasp of Drosophila melanogaster whose survival relies on the nutrients provided by its Drosophila host. Here, we used RNA-seq to compare the gene expression levels of the host midgut at 24 h and 48 h post L. boulardi parasitization. We obtained 95 and 191 differentially expressed genes (DEGs) in the parasitized host midgut at 24 h and 48 h post L. boulardi parasitization, respectively. A KEGG analysis revealed that several metabolic pathways were significantly enriched in the upregulated DEGs, and these pathways included "starch and sucrose metabolism" and "galactose metabolism". A functional annotation analysis showed that four classes of genes involved in carbohydrate digestion process had increased expression levels in the midgut post L.boulardi parasitization than nonparasitized groups: glucosidase, mannosidase, chitinase and amylase. Genes involved in protein digestion process were also found among the DEGs, and most of these genes, which belonged to the metallopeptidase and serine-type endopeptidase families, were found at higher expression levels in the parasitized host midgut comparing with nonparasitized hosts. Moreover, some immune genes, particularly those involved in the Toll and Imd pathways, also exhibited high expression levels after L.boulardi parasitization. Our study provides large-scale transcriptome data and identifies sets of DEGs between parasitized and nonparasitized host midgut tissues at 24 h and 48 h post L. boulardi parasitization. These resources help improve our understanding of how parasitoid infection affects the nutrient components in the hosts.
Collapse
|
14
|
Heredia F, Volonté Y, Pereirinha J, Fernandez-Acosta M, Casimiro AP, Belém CG, Viegas F, Tanaka K, Menezes J, Arana M, Cardoso GA, Macedo A, Kotowicz M, Prado Spalm FH, Dibo MJ, Monfardini RD, Torres TT, Mendes CS, Garelli A, Gontijo AM. The steroid-hormone ecdysone coordinates parallel pupariation neuromotor and morphogenetic subprograms via epidermis-to-neuron Dilp8-Lgr3 signal induction. Nat Commun 2021; 12:3328. [PMID: 34099654 PMCID: PMC8184853 DOI: 10.1038/s41467-021-23218-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 03/16/2021] [Indexed: 02/07/2023] Open
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.
Collapse
Affiliation(s)
- Fabiana Heredia
- CEDOC, Chronic Diseases Research Center, NOVA Medical School | Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Yanel Volonté
- CEDOC, Chronic Diseases Research Center, NOVA Medical School | Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
- INIBIBB, Instituto de Investigaciones Bioquímicas de Bahia Blanca, Universidad Nacional del Sur - CONICET, Bahía Blanca, Argentina
| | - Joana Pereirinha
- CEDOC, Chronic Diseases Research Center, NOVA Medical School | Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
- Institute of Molecular Biology, Mainz, Germany
| | - Magdalena Fernandez-Acosta
- CEDOC, Chronic Diseases Research Center, NOVA Medical School | Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Andreia P Casimiro
- CEDOC, Chronic Diseases Research Center, NOVA Medical School | Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Cláudia G Belém
- CEDOC, Chronic Diseases Research Center, NOVA Medical School | Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
- The Francis Crick Institute, London, UK
| | - Filipe Viegas
- CEDOC, Chronic Diseases Research Center, NOVA Medical School | Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Kohtaro Tanaka
- Instituto Gulbenkian de Ciências, Oeiras, Portugal
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Juliane Menezes
- CEDOC, Chronic Diseases Research Center, NOVA Medical School | Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Maite Arana
- INIBIBB, Instituto de Investigaciones Bioquímicas de Bahia Blanca, Universidad Nacional del Sur - CONICET, Bahía Blanca, Argentina
| | - Gisele A Cardoso
- CEDOC, Chronic Diseases Research Center, NOVA Medical School | Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
- Laboratório de Genômica e Evolução de Artrópodes, Departamento de Genética e Biologia Evolutiva, Universidade de São Paulo, São Paulo, Brazil
- CBMEG, Universidade Estadual de Campinas, Campinas, Brazil
| | - André Macedo
- CEDOC, Chronic Diseases Research Center, NOVA Medical School | Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Malwina Kotowicz
- CEDOC, Chronic Diseases Research Center, NOVA Medical School | Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
- DZNE, Helmholtz Association, Bonn, Germany
| | - Facundo H Prado Spalm
- INIBIBB, Instituto de Investigaciones Bioquímicas de Bahia Blanca, Universidad Nacional del Sur - CONICET, Bahía Blanca, Argentina
| | - Marcos J Dibo
- INIBIBB, Instituto de Investigaciones Bioquímicas de Bahia Blanca, Universidad Nacional del Sur - CONICET, Bahía Blanca, Argentina
| | - Raquel D Monfardini
- CEDOC, Chronic Diseases Research Center, NOVA Medical School | Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
- Laboratório de Genômica e Evolução de Artrópodes, Departamento de Genética e Biologia Evolutiva, Universidade de São Paulo, São Paulo, Brazil
| | - Tatiana T Torres
- Laboratório de Genômica e Evolução de Artrópodes, Departamento de Genética e Biologia Evolutiva, Universidade de São Paulo, São Paulo, Brazil
| | - César S Mendes
- CEDOC, Chronic Diseases Research Center, NOVA Medical School | Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Andres Garelli
- CEDOC, Chronic Diseases Research Center, NOVA Medical School | Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal.
- INIBIBB, Instituto de Investigaciones Bioquímicas de Bahia Blanca, Universidad Nacional del Sur - CONICET, Bahía Blanca, Argentina.
| | - Alisson M Gontijo
- CEDOC, Chronic Diseases Research Center, NOVA Medical School | Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal.
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Rua do Instituto Bacteriológico 5, 1150-190, Lisbon, Portugal.
| |
Collapse
|
15
|
Rork AM, Xu S, Attygalle A, Renner T. Primary Metabolism co-Opted for Defensive Chemical Production in the Carabid Beetle, Harpalus pensylvanicus. J Chem Ecol 2021; 47:334-349. [PMID: 33689113 DOI: 10.1007/s10886-021-01253-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/18/2020] [Accepted: 02/02/2021] [Indexed: 11/25/2022]
Abstract
Of the approximately one million described insect species, ground beetles (Coleoptera: Carabidae) have long captivated the attention of evolutionary biologists due to the diversity of defensive compounds they synthesize. Produced using defensive glands in the abdomen, ground beetle chemicals represent over 250 compounds including predator-deterring formic acid, which has evolved as a defensive strategy at least three times across Insecta. Despite being a widespread method of defense, formic acid biosynthesis is poorly understood in insects. Previous studies have suggested that the folate cycle of one-carbon (C1) metabolism, a pathway involved in nucleotide biosynthesis, may play a key role in defensive-grade formic acid production in ants. Here, we report on the defensive gland transcriptome of the formic acid-producing ground beetle Harpalus pensylvanicus. The full suite of genes involved in the folate cycle of C1 metabolism are significantly differentially expressed in the defensive glands of H. pensylvanicus when compared to gene expression profiles in the rest of the body. We also find support for two additional pathways potentially involved in the biosynthesis of defensive-grade formic acid, the kynurenine pathway and the methionine salvage cycle. Additionally, we have found an array of differentially expressed genes in the secretory lobes involved in the biosynthesis and transport of cofactors necessary for formic acid biosynthesis, as well as genes presumably involved in the detoxification of secondary metabolites including formic acid. We also provide insight into the evolution of the predominant gene family involved in the folate cycle (MTHFD) and suggest that high expression of folate cycle genes rather than gene duplication and/or neofunctionalization may be more important for defensive-grade formic acid biosynthesis in H. pensylvanicus. This provides the first evidence in Coleoptera and one of a few examples in Insecta of a primary metabolic process being co-opted for defensive chemical biosynthesis. Our results shed light on potential mechanisms of formic acid biosynthesis in the defensive glands of a ground beetle and provide a foundation for further studies into the evolution of formic acid-based chemical defense strategies in insects.
Collapse
Affiliation(s)
- Adam M Rork
- Department of Entomology, The Pennsylvania State University, 501 ASI Building, University Park, PA, 16802, USA.
| | - Sihang Xu
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Athula Attygalle
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Tanya Renner
- Department of Entomology, The Pennsylvania State University, 501 ASI Building, University Park, PA, 16802, USA
| |
Collapse
|
16
|
Guo S, Tian Z, Wu QW, King-Jones K, Liu W, Zhu F, Wang XP. Steroid hormone ecdysone deficiency stimulates preparation for photoperiodic reproductive diapause. PLoS Genet 2021; 17:e1009352. [PMID: 33529191 PMCID: PMC7880476 DOI: 10.1371/journal.pgen.1009352] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 02/12/2021] [Accepted: 01/09/2021] [Indexed: 01/21/2023] Open
Abstract
Diapause, a programmed developmental arrest primarily induced by seasonal environmental changes, is very common in the animal kingdom, and found in vertebrates and invertebrates alike. Diapause provides an adaptive advantage to animals, as it increases the odds of surviving adverse conditions. In insects, individuals perceive photoperiodic cues and modify endocrine signaling to direct reproductive diapause traits, such as ovary arrest and increased fat accumulation. However, it remains unclear as to which endocrine factors are involved in this process and how they regulate the onset of reproductive diapause. Here, we found that the long day-mediated drop in the concentration of the steroid hormone ecdysone is essential for the preparation of photoperiodic reproductive diapause in Colaphellus bowringi, an economically important cabbage beetle. The diapause-inducing long-day condition reduced the expression of ecdysone biosynthetic genes, explaining the drop in the titer of 20-hydroxyecdysone (20E, the active form of ecdysone) in female adults. Application of exogenous 20E induced vitellogenesis and ovarian development but reduced fat accumulation in the diapause-destined females. Knocking down the ecdysone receptor (EcR) in females destined for reproduction blocked reproductive development and induced diapause traits. RNA-seq and hormone measurements indicated that 20E stimulates the production of juvenile hormone (JH), a key endocrine factor in reproductive diapause. To verify this, we depleted three ecdysone biosynthetic enzymes via RNAi, which confirmed that 20E is critical for JH biosynthesis and reproductive diapause. Importantly, impairing Met function, a component of the JH intracellular receptor, partially blocked the 20E-regulated reproductive diapause preparation, indicating that 20E regulates reproductive diapause in both JH-dependent and -independent manners. Finally, we found that 20E deficiency decreased ecdysis-triggering hormone signaling and reduced JH production, thereby inducing diapause. Together, these results suggest that 20E signaling is a pivotal regulator that coordinates reproductive plasticity in response to environmental inputs. Developmental arrest pervades organismal development and physiology where it facilitates an enormous range of adaptive responses to stressful conditions. Many animals exhibit various forms of developmental arrest that ensures survival under the most adverse environments. Reproductive diapause occurs when adults temporarily suspend reproduction in response to environmental stress and has been documented for a variety of invertebrates, particularly insects. Endocrine signals play a central role in translating environmental cues such as photoperiod into reproductive diapause-related physiology and behavior. However, it has been an unresolved issue as to which endocrine factors can respond to photoperiodic inputs and regulate diapause outputs. In this study, we found that a decrease in ecdysone levels is critical for reproductive diapause to occur. Also, ecdysone could interact with juvenile hormone to regulate the occurrence of reproductive diapause in response to photoperiodic cues. Our findings provide new insight into endocrine mechanisms of photoperiodic reproductive diapause and an example of phenotypic plasticity in animals.
Collapse
Affiliation(s)
- Shuang Guo
- Hubei Key Laboratory of Insect Resources Utilization and Sustainable Pest Management, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| | - Zhong Tian
- Hubei Key Laboratory of Insect Resources Utilization and Sustainable Pest Management, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| | - Qing-Wen Wu
- Hubei Key Laboratory of Insect Resources Utilization and Sustainable Pest Management, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| | - Kirst King-Jones
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Wen Liu
- Hubei Key Laboratory of Insect Resources Utilization and Sustainable Pest Management, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, PR China
- * E-mail: (WL); (X-PW)
| | - Fen Zhu
- Hubei Key Laboratory of Insect Resources Utilization and Sustainable Pest Management, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| | - Xiao-Ping Wang
- Hubei Key Laboratory of Insect Resources Utilization and Sustainable Pest Management, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, PR China
- * E-mail: (WL); (X-PW)
| |
Collapse
|
17
|
Niederhuber MJ, McKay DJ. Mechanisms underlying the control of dynamic regulatory element activity and chromatin accessibility during metamorphosis. CURRENT OPINION IN INSECT SCIENCE 2021; 43:21-28. [PMID: 32979530 PMCID: PMC7985040 DOI: 10.1016/j.cois.2020.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 08/25/2020] [Indexed: 05/10/2023]
Abstract
Cis-regulatory modules of metazoan genomes determine the when and where of gene expression during development. Here we discuss insights into the genetic and molecular mechanisms behind cis-regulatory module usage that have come from recent application of genomics assays to insect metamorphosis. Assays including FAIRE-seq, ATAC-seq, and CUT&RUN indicate that sequential changes in chromatin accessibility play a key role in mediating stage-specific cis-regulatory module activity and gene expression. We review the current understanding of what controls precisely coordinated changes in chromatin accessibility during metamorphosis and describe evidence that points to systemic hormone signaling as a primary signal to trigger genome-wide shifts in accessibility patterns and cis-regulatory module usage.
Collapse
Affiliation(s)
- Matthew J Niederhuber
- Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States; Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States; Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States; Integrative Program for Biological and Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Daniel J McKay
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States; Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States; Integrative Program for Biological and Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States.
| |
Collapse
|
18
|
Hanna L, Abouheif E. The origin of wing polyphenism in ants: An eco-evo-devo perspective. Curr Top Dev Biol 2021; 141:279-336. [PMID: 33602491 DOI: 10.1016/bs.ctdb.2020.12.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The evolution of eusociality, where solitary individuals integrate into a single colony, is a major transition in individuality. In ants, the origin of eusociality coincided with the origin of a wing polyphenism approximately 160 million years ago, giving rise to colonies with winged queens and wingless workers. As a consequence, both eusociality and wing polyphenism are nearly universal features of all ants. Here, we synthesize fossil, ecological, developmental, and evolutionary data in an attempt to understand the factors that contributed to the origin of wing polyphenism in ants. We propose multiple models and hypotheses to explain how wing polyphenism is orchestrated at multiple levels, from environmental cues to gene networks. Furthermore, we argue that the origin of wing polyphenism enabled the subsequent evolution of morphological diversity across the ants. We finally conclude by outlining several outstanding questions for future work.
Collapse
Affiliation(s)
- Lisa Hanna
- Department of Biology, McGill University, Montreal, QC, Canada
| | - Ehab Abouheif
- Department of Biology, McGill University, Montreal, QC, Canada.
| |
Collapse
|
19
|
Finger DS, Whitehead KM, Phipps DN, Ables ET. Nuclear receptors linking physiology and germline stem cells in Drosophila. VITAMINS AND HORMONES 2021; 116:327-362. [PMID: 33752824 PMCID: PMC8063499 DOI: 10.1016/bs.vh.2020.12.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Maternal nutrition and physiology are intimately associated with reproductive success in diverse organisms. Despite decades of study, the molecular mechanisms linking maternal diet to the production and quality of oocytes remain poorly defined. Nuclear receptors (NRs) link nutritional signals to cellular responses and are essential for oocyte development. The fruit fly, Drosophila melanogaster, is an excellent genetically tractable model to study the relationship between NR signaling and oocyte production. In this review, we explore how NRs in Drosophila regulate the earliest stages of oocyte development. Long-recognized as an essential mediator of developmental transitions, we focus on the intrinsic roles of the Ecdysone Receptor and its ligand, ecdysone, in oogenesis. We also review recent studies suggesting broader roles for NRs as regulators of maternal physiology and their impact specifically on oocyte production. We propose that NRs form the molecular basis of a broad physiological surveillance network linking maternal diet with oocyte production. Given the functional conservation between Drosophila and humans, continued experimental investigation into the molecular mechanisms by which NRs promote oogenesis will likely aid our understanding of human fertility.
Collapse
Affiliation(s)
- Danielle S Finger
- Department of Biology, East Carolina University, Greenville, NC, United States
| | - Kaitlin M Whitehead
- Department of Biology, East Carolina University, Greenville, NC, United States
| | - Daniel N Phipps
- Department of Biology, East Carolina University, Greenville, NC, United States
| | - Elizabeth T Ables
- Department of Biology, East Carolina University, Greenville, NC, United States.
| |
Collapse
|
20
|
Wu Z, He Q, Zeng B, Zhou H, Zhou S. Juvenile hormone acts through FoxO to promote Cdc2 and Orc5 transcription for polyploidy-dependent vitellogenesis. Development 2020; 147:dev.188813. [PMID: 32907849 DOI: 10.1242/dev.188813] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 07/20/2020] [Indexed: 12/21/2022]
Abstract
Vitellogenin (Vg) is a prerequisite for egg production and embryonic development after ovipositioning in oviparous animals. In many insects, juvenile hormone (JH) promotes fat body cell polyploidization for the massive Vg synthesis required for the maturation of multiple oocytes, but the underlying mechanisms remain poorly understood. Using the migratory locust Locusta migratoria as a model system, we report here that JH induces the dephosphorylation of Forkhead box O transcription factor (FoxO) through a signaling cascade including leucine carboxyl methyltransferase 1 (LCMT1) and protein phosphatase 2A (PP2A). JH promotes PP2A activity via LCMT1-mediated methylation, consequently triggering FoxO dephosphorylation. Dephosphorylated FoxO binds to the upstream region of two endocycle-related genes, cell-division-cycle 2 (Cdc2) and origin-recognition-complex subunit 5 (Orc5), and activates their transcription. Depletion of FoxO, Cdc2 or Orc5 results in blocked polyploidization of fat body cells, accompanied by markedly reduced Vg expression, impaired oocyte maturation and arrested ovarian development. The results suggest that JH acts via LCMT1-PP2A-FoxO to regulate Cdc2 and Orc5 expression, and to enhance ploidy of fat body cells in preparation for the large-scale Vg synthesis required for synchronous maturation of multiple eggs.
Collapse
Affiliation(s)
- Zhongxia Wu
- Key Laboratory of Plant Stress Biology, State Key Laboratory of Cotton Biology, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Qiongjie He
- Key Laboratory of Plant Stress Biology, State Key Laboratory of Cotton Biology, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Baojuan Zeng
- Key Laboratory of Plant Stress Biology, State Key Laboratory of Cotton Biology, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Haodan Zhou
- Key Laboratory of Plant Stress Biology, State Key Laboratory of Cotton Biology, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Shutang Zhou
- Key Laboratory of Plant Stress Biology, State Key Laboratory of Cotton Biology, School of Life Sciences, Henan University, Kaifeng 475004, China
| |
Collapse
|
21
|
Temporal Coordination of Collective Migration and Lumen Formation by Antagonism between Two Nuclear Receptors. iScience 2020; 23:101335. [PMID: 32682323 PMCID: PMC7366032 DOI: 10.1016/j.isci.2020.101335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/13/2020] [Accepted: 06/26/2020] [Indexed: 02/01/2023] Open
Abstract
During development, cells undergo multiple, distinct morphogenetic processes to form a tissue or organ, but how their temporal order and time interval are determined remain poorly understood. Here we show that the nuclear receptors E75 and DHR3 regulate the temporal order and time interval between the collective migration and lumen formation of a coherent group of cells named border cells during Drosophila oogenesis. We show that E75, in response to ecdysone signaling, antagonizes the activity of DHR3 during border cell migration, and DHR3 is necessary and sufficient for the subsequent lumen formation that is critical for micropyle morphogenesis. DHR3's lumen-inducing function is mainly mediated through βFtz-f1, another nuclear receptor and transcription factor. Furthermore, both DHR3 and βFtz-f1 are required for chitin secretion into the lumen, whereas DHR3 is sufficient for chitin secretion. Lastly, DHR3 and βFtz-f1 suppress JNK signaling in the border cells to downregulate cell adhesion during lumen formation. E75 antagonizes DHR3's function in inducing lumen formation of border cells (BCs) E75 and DHR3 temporally coordinate collective migration and lumen formation of BCs DHR3 is required and sufficient for chitin secretion into the lumen DHR3 and βFtz-f1 downregulate JNK signaling and cell adhesion in the BCs
Collapse
|
22
|
Cruz J, Martín D, Franch-Marro X. Egfr Signaling Is a Major Regulator of Ecdysone Biosynthesis in the Drosophila Prothoracic Gland. Curr Biol 2020; 30:1547-1554.e4. [DOI: 10.1016/j.cub.2020.01.092] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 12/13/2019] [Accepted: 01/30/2020] [Indexed: 12/21/2022]
|
23
|
McDonald SI, Beachum AN, Hinnant TD, Blake AJ, Bynum T, Hickman EP, Barnes J, Churchill KL, Roberts TS, Zangwill DE, Ables ET. Novel cis-regulatory regions in ecdysone responsive genes are sufficient to promote gene expression in Drosophila ovarian cells. Gene Expr Patterns 2019; 34:119074. [PMID: 31563631 PMCID: PMC6996244 DOI: 10.1016/j.gep.2019.119074] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 06/18/2019] [Accepted: 09/26/2019] [Indexed: 12/14/2022]
Abstract
The insect steroid hormone ecdysone is a key regulator of oogenesis in Drosophila melanogaster and many other species. Despite the diversity of cellular functions of ecdysone in oogenesis, the molecular regulation of most ecdysone-responsive genes in ovarian cells remains largely unexplored. We performed a functional screen using the UAS/Gal4 system to identify non-coding cis-regulatory elements within well-characterized ecdysone-response genes capable of driving transcription of an indelible reporter in ovarian cells. Using two publicly available transgenic collections (the FlyLight and Vienna Tiles resources), we tested 62 Gal4 drivers corresponding to ecdysone-response genes EcR, usp, E75, br, ftz-f1 and Hr3. We observed 31 lines that were sufficient to drive a UAS-lacZ reporter in discrete cell populations in the ovary. Reporter expression was reproducibly observed in both somatic and germ cells at distinct stages of oogenesis, including those previously characterized as critical points of ecdysone regulation. Our studies identified several useful new reagents, adding to the UAS/Gal4 toolkit available for genetic analysis of oogenesis in Drosophila. Further, our study provides novel insight into the molecular regulation of ecdysone signaling in oogenesis.
Collapse
Affiliation(s)
| | - Allison N Beachum
- Department of Biology, East Carolina University, Greenville, NC, 27858, USA
| | - Taylor D Hinnant
- Department of Biology, East Carolina University, Greenville, NC, 27858, USA
| | - Amelia J Blake
- Department of Biology, East Carolina University, Greenville, NC, 27858, USA
| | - Tierra Bynum
- Department of Biology, East Carolina University, Greenville, NC, 27858, USA
| | - E Parris Hickman
- Department of Biology, East Carolina University, Greenville, NC, 27858, USA
| | - Joseph Barnes
- Department of Biology, East Carolina University, Greenville, NC, 27858, USA
| | - Kaely L Churchill
- Department of Biology, East Carolina University, Greenville, NC, 27858, USA
| | - Tamesia S Roberts
- Department of Biology, East Carolina University, Greenville, NC, 27858, USA
| | - Denise E Zangwill
- Department of Biology, East Carolina University, Greenville, NC, 27858, USA
| | - Elizabeth T Ables
- Department of Biology, East Carolina University, Greenville, NC, 27858, USA.
| |
Collapse
|
24
|
Ma Y, McKay DJ, Buttitta L. Changes in chromatin accessibility ensure robust cell cycle exit in terminally differentiated cells. PLoS Biol 2019; 17:e3000378. [PMID: 31479438 PMCID: PMC6743789 DOI: 10.1371/journal.pbio.3000378] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 09/13/2019] [Accepted: 08/13/2019] [Indexed: 12/12/2022] Open
Abstract
During terminal differentiation, most cells exit the cell cycle and enter into a prolonged or permanent G0 in which they are refractory to mitogenic signals. Entry into G0 is usually initiated through the repression of cell cycle gene expression by formation of a transcriptional repressor complex called dimerization partner (DP), retinoblastoma (RB)-like, E2F and MuvB (DREAM). However, when DREAM repressive function is compromised during terminal differentiation, additional unknown mechanisms act to stably repress cycling and ensure robust cell cycle exit. Here, we provide evidence that developmentally programmed, temporal changes in chromatin accessibility at a small subset of critical cell cycle genes act to enforce cell cycle exit during terminal differentiation in the Drosophila melanogaster wing. We show that during terminal differentiation, chromatin closes at a set of pupal wing enhancers for the key rate-limiting cell cycle regulators Cyclin E (cycE), E2F transcription factor 1 (e2f1), and string (stg). This closing coincides with wing cells entering a robust postmitotic state that is strongly refractory to cell cycle reactivation, and the regions that close contain known binding sites for effectors of mitogenic signaling pathways such as Yorkie and Notch. When cell cycle exit is genetically disrupted, chromatin accessibility at cell cycle genes remains unaffected, and the closing of distal enhancers at cycE, e2f1, and stg proceeds independent of the cell cycling status. Instead, disruption of cell cycle exit leads to changes in accessibility and expression of a subset of hormone-induced transcription factors involved in the progression of terminal differentiation. Our results uncover a mechanism that acts as a cell cycle–independent timer to limit the response to mitogenic signaling and aberrant cycling in terminally differentiating tissues. In addition, we provide a new molecular description of the cross talk between cell cycle exit and terminal differentiation during metamorphosis. The longer a cell remains in G0, the more refractory it becomes to re-entering the cell cycle. This study shows that in terminally differentiated cells in vivo, regulatory elements at genes encoding just three key cell cycle regulators (cycE, e2f1 and stg) become inaccessible, limiting their aberrant activation and maintaining a prolonged, robust G0.
Collapse
Affiliation(s)
- Yiqin Ma
- Department of Molecular Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Daniel J McKay
- Department of Biology, Department of Genetics, Integrative Program for Biological and Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Laura Buttitta
- Department of Molecular Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
25
|
Basak AK, Chatterjee T, Chakravarty A, Ghosh SK. Silver nanoparticle-induced developmental inhibition of Drosophila melanogaster accompanies disruption of genetic material of larval neural stem cells and non-neuronal cells. ENVIRONMENTAL MONITORING AND ASSESSMENT 2019; 191:497. [PMID: 31312907 DOI: 10.1007/s10661-019-7630-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 07/02/2019] [Indexed: 06/10/2023]
Abstract
A few studies had determined the effects of silver nanoparticles on the development of Drosophila melanogaster. However, none had addressed its genotoxic effects on specific larval cells of the fly in details. This study was conducted to determine the effects of silver nanoparticle on the development of D. melanogaster with simultaneous evaluation of its genotoxic potential on specific larval cell types that play important roles in immunological defenses as well as growth and development. Five male and five female flies were maintained in standard Drosophila melanogaster culture medium containing varying concentrations of silver nanoparticles, i.e., 25, 50, 100, 200, and 300 mg/l with control culture medium containing no nanoparticle. Total time needed for stage-specific development, population yield, and genotoxic effects on third instar larval polytene chromosomes, hemocytes, and neuroblasts was determined. Body pigmentation of pupae and young adults was examined visually. In comparison with control, silver nanoparticles dose dependently inhibited the metamororphosis and population yields of pupae and young adults of Drosophila melanogaster. Every concentration of the nanoparticles inhibited pupa to adult conversion, with huge reduction under the influence of nanoparticle concentration of 100 mg/ml and above. Developmental inhibition was accompanied by dose-dependent and significant structural aberrations of larval polytene chromosomes and deformities of hemocytes and neuroblasts. Pupae and young adults also exhibited gradual discoloration of body with the increase in exposure to nanoparticle concentration.
Collapse
Affiliation(s)
- Ashim Kumar Basak
- Department of Molecular Biology, Institute of Genetic Engineering, 30, Thakurhat Road, Kolkata, West Bengal, 700128, India
| | - Tridip Chatterjee
- Department of Molecular Biology, Institute of Genetic Engineering, 30, Thakurhat Road, Kolkata, West Bengal, 700128, India
| | - Amit Chakravarty
- Institute of Genetic Engineering; Institute of Genetic Medicine and Genomic Sciences, 30, Thakurhat Road, Kolkata, West Bengal, 700128, India
| | - Swapan Kumar Ghosh
- Molecular Mycopathology Lab, Cancer Research Unit, PG Department of Botany, Ramakrishna Mission Vivekananda Centenary College (Autonomous), Rahara, Kolkata, 700118, India.
| |
Collapse
|
26
|
Liu X, Zhang S, Shen ZJ, Liu Y, Li Z, Liu X. Vrille is required for larval moulting and metamorphosis of Helicoverpa armigera (Lepidoptera: Noctuidae). INSECT MOLECULAR BIOLOGY 2019; 28:355-371. [PMID: 30485565 DOI: 10.1111/imb.12557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Vrille (Vri), a basic leucine zipper transcription factor, plays important roles in insect circadian clock regulation, tracheal development, proliferation, flight and metamorphosis. Here, Helicoverpa armigera was used as a model to investigate the role of Vri in larval moulting and metamorphosis. Sequence analysis results revealed that H. armigera Vri (HaVri) shares a high amino acid identity with other Lepidoptera Vri homologues. Spatial-temporal expression pattern data showed that HaVri expression was highly abundant in larval moulting and metamorphosis stages and was mainly expressed in the midgut and Malpighian tubule during metamorphosis. HaVri knockdown by RNA interference in the fourth-instar larvae prevented larval moulting, and HaVri knockdown in the fifth-instar larvae suppressed midgut remodelling and delayed or blocked metamorphosis. Further studies confirmed that 20-hydroxyecdysone (20E) activated HaVri expression via its heterodimer receptors, ecdysone receptor (EcRB1) and ultraspiracle protein (USP1), whereas methoprene [juvenile hormone analogue (JHA)] promoted HaVri expression via its intracellular receptor methoprene-tolerant (Met1). However, 20E and JHA can counteract each other in the activation of HaVri expression. Together, the present results suggested that HaVri was involved in larval moulting and metamorphosis and was regulated by 20E and JHA in H. armigera.
Collapse
Affiliation(s)
- X Liu
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, 100193, Beijing, China
| | - S Zhang
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, 100193, Beijing, China
| | - Z J Shen
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, 100193, Beijing, China
| | - Y Liu
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, 100193, Beijing, China
| | - Z Li
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, 100193, Beijing, China
| | - X Liu
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, 100193, Beijing, China
| |
Collapse
|
27
|
Tran NM, Mykles DL, Elizur A, Ventura T. Characterization of G-protein coupled receptors from the blackback land crab Gecarcinus lateralis Y organ transcriptome over the molt cycle. BMC Genomics 2019; 20:74. [PMID: 30669976 PMCID: PMC6341585 DOI: 10.1186/s12864-018-5363-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 12/11/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND G-protein coupled receptors (GPCRs) are ancient, ubiquitous, constitute the largest family of transducing cell surface proteins, and are integral to cell communication via an array of ligands/neuropeptides. Molt inhibiting hormone (MIH) is a key neuropeptide that controls growth and reproduction in crustaceans by regulating the molt cycle. It inhibits ecdysone biosynthesis by a pair of endocrine glands (Y-organs; YOs) through binding a yet uncharacterized GPCR, which triggers a signalling cascade, leading to inhibition of the ecdysis sequence. When MIH release stops, ecdysone is synthesized and released to the hemolymph. A peak in ecdysone titer is followed by a molting event. A transcriptome of the blackback land crab Gecarcinus lateralis YOs across molt was utilized in this study to curate the list of GPCRs and their expression in order to better assess which GPCRs are involved in the molt process. RESULTS Ninety-nine G. lateralis putative GPCRs were obtained by screening the YO transcriptome against the Pfam database. Phylogenetic analysis classified 49 as class A (Rhodopsin-like receptor), 35 as class B (Secretin receptor), and 9 as class C (metabotropic glutamate). Further phylogenetic analysis of class A GPCRs identified neuropeptide GPCRs, including those for Allatostatin A, Allatostatin B, Bursicon, CCHamide, FMRFamide, Proctolin, Corazonin, Relaxin, and the biogenic amine Serotonin. Three GPCRs clustered with recently identified putative CHH receptors (CHHRs), and differential expression over the molt cycle suggests that they are associated with ecdysteroidogenesis regulation. Two putative Corazonin receptors showed much higher expression in the YOs compared with all other GPCRs, suggesting an important role in molt regulation. CONCLUSIONS Molting requires an orchestrated regulation of YO ecdysteroid synthesis by multiple neuropeptides. In this study, we curated a comprehensive list of GPCRs expressed in the YO and followed their expression across the molt cycle. Three putative CHH receptors were identified and could include an MIH receptor whose activation negatively regulates molting. Orthologs of receptors that were found to be involved in molt regulation in insects were also identified, including LGR3 and Corazonin receptor, the latter of which was expressed at much higher level than all other receptors, suggesting a key role in YO regulation.
Collapse
Affiliation(s)
- Nhut M Tran
- GeneCology Research Centre, School of Science and Engineering University of the Sunshine Coast, Queensland, 4556, Australia
| | - Donald L Mykles
- Department of Biology, Colorado State University, Fort Collins, CO, 80523, USA
| | - Abigail Elizur
- GeneCology Research Centre, School of Science and Engineering University of the Sunshine Coast, Queensland, 4556, Australia
| | - Tomer Ventura
- GeneCology Research Centre, School of Science and Engineering University of the Sunshine Coast, Queensland, 4556, Australia.
| |
Collapse
|
28
|
Andoh M, Ueno T, Kawasaki K. Tissue-dependent induction of antimicrobial peptide genes after body wall injury in house fly ( Musca domestica) larvae. Drug Discov Ther 2018; 12:355-362. [DOI: 10.5582/ddt.2018.01063] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Minako Andoh
- Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts
| | - Takayuki Ueno
- Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts
| | - Kiyoshi Kawasaki
- Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts
| |
Collapse
|
29
|
Okamoto N, Viswanatha R, Bittar R, Li Z, Haga-Yamanaka S, Perrimon N, Yamanaka N. A Membrane Transporter Is Required for Steroid Hormone Uptake in Drosophila. Dev Cell 2018; 47:294-305.e7. [PMID: 30293839 PMCID: PMC6219898 DOI: 10.1016/j.devcel.2018.09.012] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/06/2018] [Accepted: 09/09/2018] [Indexed: 02/08/2023]
Abstract
Steroid hormones are a group of lipophilic hormones that are believed to enter cells by simple diffusion to regulate diverse physiological processes through intracellular nuclear receptors. Here, we challenge this model in Drosophila by demonstrating that Ecdysone Importer (EcI), a membrane transporter identified from two independent genetic screens, is involved in cellular uptake of the steroid hormone ecdysone. EcI encodes an organic anion transporting polypeptide of the evolutionarily conserved solute carrier organic anion superfamily. In vivo, EcI loss of function causes phenotypes indistinguishable from ecdysone- or ecdysone receptor (EcR)-deficient animals, and EcI knockdown inhibits cellular uptake of ecdysone. Furthermore, EcI regulates ecdysone signaling in a cell-autonomous manner and is both necessary and sufficient for inducing ecdysone-dependent gene expression in culture cells expressing EcR. Altogether, our results challenge the simple diffusion model for cellular uptake of ecdysone and may have wide implications for basic and medical aspects of steroid hormone studies.
Collapse
Affiliation(s)
- Naoki Okamoto
- Department of Entomology, Institute for Integrative Genome Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Raghuvir Viswanatha
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Riyan Bittar
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Zhongchi Li
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Sachiko Haga-Yamanaka
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA; Howard Hughes Medical Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Naoki Yamanaka
- Department of Entomology, Institute for Integrative Genome Biology, University of California, Riverside, Riverside, CA 92521, USA.
| |
Collapse
|
30
|
Wu Z, Guo W, Yang L, He Q, Zhou S. Juvenile hormone promotes locust fat body cell polyploidization and vitellogenesis by activating the transcription of Cdk6 and E2f1. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2018; 102:1-10. [PMID: 30205150 DOI: 10.1016/j.ibmb.2018.09.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 09/07/2018] [Accepted: 09/07/2018] [Indexed: 06/08/2023]
Abstract
Juvenile hormone (JH) is known to promote cell polyploidization for insect vitellogenesis and egg production, but the underlying mechanisms remain poorly understood. Using the migratory locust Locusta migratoria as a model system, we report here that the expression of cyclin-dependent kinase 6 (Cdk6) and adenovirus E2 factor-1 (E2f1), the core mediators in cell cycle progression is regulated by JH and its receptor Methoprene-tolerant (Met). JH acts through its receptor complex comprised of Met and Taiman to directly activate the transcription of Cdk6 and E2f1. Depletion of Cdk6 or E2f1 results in significantly decreased ploidy, precocious mitotic entry and increased cell numbers in the fat body, accompanied by substantial reduction of Vitellogenin gene expression, blocked ovarian growth and arrested oocyte maturation. These findings indicate a crucial role of Cdk6 and E2f1 in JH-regulated polyploidization and vitellogenesis as well as a novel regulatory machinery for endocycling in insects.
Collapse
Affiliation(s)
- Zhongxia Wu
- Key Laboratory of Plant Stress Biology, State Key Laboratory of Cotton Biology, School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Wei Guo
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Libin Yang
- Key Laboratory of Plant Stress Biology, State Key Laboratory of Cotton Biology, School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Qiongjie He
- Key Laboratory of Plant Stress Biology, State Key Laboratory of Cotton Biology, School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Shutang Zhou
- Key Laboratory of Plant Stress Biology, State Key Laboratory of Cotton Biology, School of Life Sciences, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
31
|
Xu T, Denton D, Kumar S. Hedgehog and Wingless signaling are not essential for autophagy-dependent cell death. Biochem Pharmacol 2018; 162:3-13. [PMID: 30879494 DOI: 10.1016/j.bcp.2018.10.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 10/24/2018] [Indexed: 01/05/2023]
Abstract
Autophagy-dependent cell death is a distinct mode of regulated cell death required in a context specific manner. One of the best validated genetic models of autophagy-dependent cell death is the removal of the Drosophila larval midgut during larval-pupal transition. We have previously shown that down-regulation of growth signaling is essential for autophagy induction and larval midgut degradation. Sustained growth signaling through Ras and PI3K blocks autophagy and consequently inhibits midgut degradation. In addition, the morphogen Dpp plays an important role in regulating the correct timing of midgut degradation. Here we explore the potential roles of Hh and Wg signaling in autophagy-dependent midgut cell death. We demonstrate that Hh and Wg signaling are not involved in the regulation of autophagy-dependent cell death. However, surprisingly we found that one key component of these pathways, the Drosophila Glycogen Synthase Kinase 3, Shaggy (Sgg), may regulate midgut cell size independent of Hh and Wg signaling.
Collapse
Affiliation(s)
- Tianqi Xu
- Centre for Cancer Biology, University of South Australia & SA Pathology, GPO Box 2471, Adelaide, SA 5001, Australia
| | - Donna Denton
- Centre for Cancer Biology, University of South Australia & SA Pathology, GPO Box 2471, Adelaide, SA 5001, Australia.
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia & SA Pathology, GPO Box 2471, Adelaide, SA 5001, Australia.
| |
Collapse
|
32
|
Schumann I, Kenny N, Hui J, Hering L, Mayer G. Halloween genes in panarthropods and the evolution of the early moulting pathway in Ecdysozoa. ROYAL SOCIETY OPEN SCIENCE 2018; 5:180888. [PMID: 30839709 PMCID: PMC6170570 DOI: 10.1098/rsos.180888] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/17/2018] [Indexed: 05/15/2023]
Abstract
Moulting is a characteristic feature of Ecdysozoa-the clade of moulting animals that includes the hyperdiverse arthropods and less speciose groups, such as onychophorans, tardigrades and nematodes. Moulting has been best analysed in arthropods, specifically in insects and crustaceans, in which a complex neuroendocrine system acts at the genomic level and initiates the transcription of genes responsible for moulting. The key moulting hormones, ecdysone and 20-hydroxyecdysone, are subsequently synthesized from cholesterol ingested with food. Their biosynthesis is regulated by the Rieske-domain protein Neverland and cytochrome P450 enzymes encoded by the so-called 'Halloween' genes. Ecdysone is then released into the haemolymph and modified into 20-hydroxyecdysone, which binds to the nuclear receptor EcR/USP and initiates transcription of the Early genes. As little is known about the moulting pathway of other ecdysozoans, we examined the occurrence of genes involved in ecdysteroid biosynthesis and the early moulting cascade across ecdysozoan subgroups. Genomic and transcriptomic searches revealed no Halloween genes in cycloneuralians, whereas only shadow (CYP315A1) is present in onychophorans and tardigrades, suggesting that the Halloween genes evolved stepwise in panarthropods. These findings imply that the genes which were responsible for the ecdysteroid biosynthesis in the last common ancestor of Ecdysozoa are currently unknown.
Collapse
Affiliation(s)
- Isabell Schumann
- Department of Zoology, Institute of Biology, University of Kassel, Kassel, Germany
- Molecular Evolution and Animal Systematics, Institute of Biology, University of Leipzig, Leipzig, Germany
| | - Nathan Kenny
- School of Life Sciences, Simon F.S. Li Marine Science Laboratory, Center of Soybean Research, State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong, People's Republic of China
| | - Jerome Hui
- School of Life Sciences, Simon F.S. Li Marine Science Laboratory, Center of Soybean Research, State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong, People's Republic of China
| | - Lars Hering
- Department of Zoology, Institute of Biology, University of Kassel, Kassel, Germany
| | - Georg Mayer
- Department of Zoology, Institute of Biology, University of Kassel, Kassel, Germany
| |
Collapse
|
33
|
Dynamic changes in ORC localization and replication fork progression during tissue differentiation. BMC Genomics 2018; 19:623. [PMID: 30134926 PMCID: PMC6103881 DOI: 10.1186/s12864-018-4992-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/02/2018] [Indexed: 12/23/2022] Open
Abstract
Background Genomic regions repressed for DNA replication, resulting in either delayed replication in S phase or underreplication in polyploid cells, are thought to be controlled by inhibition of replication origin activation. Studies in Drosophila polytene cells, however, raised the possibility that impeding replication fork progression also plays a major role. Results We exploited genomic regions underreplicated (URs) with tissue specificity in Drosophila polytene cells to analyze mechanisms of replication repression. By localizing the Origin Recognition Complex (ORC) in the genome of the larval fat body and comparing this to ORC binding in the salivary gland, we found that sites of ORC binding show extensive tissue specificity. In contrast, there are common domains nearly devoid of ORC in the salivary gland and fat body that also have reduced density of ORC binding sites in diploid cells. Strikingly, domains lacking ORC can still be replicated in some polytene tissues, showing absence of ORC and origins is insufficient to repress replication. Analysis of the width and location of the URs with respect to ORC position indicates that whether or not a genomic region lacking ORC is replicated is controlled by whether replication forks formed outside the region are inhibited. Conclusions These studies demonstrate that inhibition of replication fork progression can block replication across genomic regions that constitutively lack ORC. Replication fork progression can be inhibited in both tissue-specific and genome region-specific ways. Consequently, when evaluating sources of genome instability it is important to consider altered control of replication forks in response to differentiation. Electronic supplementary material The online version of this article (10.1186/s12864-018-4992-3) contains supplementary material, which is available to authorized users.
Collapse
|
34
|
The biology and evolution of the Dilp8-Lgr3 pathway: A relaxin-like pathway coupling tissue growth and developmental timing control. Mech Dev 2018; 154:44-50. [PMID: 29715504 DOI: 10.1016/j.mod.2018.04.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 04/25/2018] [Accepted: 04/26/2018] [Indexed: 11/21/2022]
Abstract
Many insects, like cockroaches, moths, and flies, can regenerate tissues by extending the growth-competent phases of their life cycle. The molecular and cellular players mediating this coordination between tissue growth and developmental timing have been recently discovered in Drosophila. The insulin/relaxin-like peptide, Dilp8, was identified as a factor communicating abnormal growth status of Drosophila larval imaginal discs to the neuroendocrine centers that control the timing of the onset of metamorphosis. Dilp8 requires a neuronal relaxin receptor for this function, the Leucine rich repeat containing G protein coupled receptor, Lgr3. A review of current data supports a model where imaginal disc-derived Dilp8 acts on four central nervous system Lgr3-positive neurons to activate cyclic-AMP signaling in an Lgr3-dependent manner. This causes a reduction in ecdysone hormone production by the larval endocrine prothoracic gland, which leads to a delay in the onset of metamorphosis and a simultaneous slowing down in the growth rates of healthy imaginal tissues, promoting the generation of proportionate individuals. We discuss reports indicating that the Dilp8-Lgr3 pathway might have other functions at different life history stages, which remain to be elucidated, and review molecular evolution data on invertebrate genes related to the relaxin-pathway. The strong conservation of the relaxin pathway throughout animal evolution contrasts with instances of its complete loss in some clades, such as lepidopterans, which must coordinate growth and developmental timing using another mechanism. Research into these areas should generate exciting new insights into the biology of growth coordination, the evolution of the relaxin signaling pathway, and likely reveal unforeseen functions in other developmental stages.
Collapse
|
35
|
Abstract
The larval stage of the stingless bee Scaptotrigona depilis must consume a specific brood cell fungus in order to continue development. Here we show that this fungus is a member of the genus Zygosaccharomyces and provides essential steroid precursors to the developing bee. Insect pupation requires ecdysteroid hormones, and as insects cannot synthesize sterols de novo, they must obtain steroids in their diet. Larval in vitro culturing assays demonstrated that consuming ergosterol recapitulates the developmental effects on S. depilis as ingestion of Zygosaccharomyces sp. cells. Thus, we determined the molecular underpinning of this intimate mutualistic symbiosis. Phylogenetic analyses showed that similar cases of bee-Zygosaccharomyces symbiosis may exist. This unprecedented case of bee-fungus symbiosis driven by steroid requirement brings new perspectives regarding pollinator-microbiota interaction and preservation.
Collapse
|
36
|
Chen D, Dale RK, Lei EP. Shep regulates Drosophila neuronal remodeling by controlling transcription of its chromatin targets. Development 2018; 145:dev.154047. [PMID: 29158441 DOI: 10.1242/dev.154047] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 11/07/2017] [Indexed: 11/20/2022]
Abstract
Neuronal remodeling is crucial for formation of the mature nervous system and disruption of this process can lead to neuropsychiatric diseases. Global gene expression changes in neurons during remodeling as well as the factors that regulate these changes remain poorly defined. To elucidate this process, we performed RNA-seq on isolated Drosophila larval and pupal neurons and found upregulated synaptic signaling and downregulated gene expression regulators as a result of normal neuronal metamorphosis. We further tested the role of alan shepard (shep), which encodes an evolutionarily conserved RNA-binding protein required for proper neuronal remodeling. Depletion of shep in neurons prevents the execution of metamorphic gene expression patterns, and shep-regulated genes correspond to Shep chromatin and/or RNA-binding targets. Reduced expression of a Shep-inhibited target gene that we identified, brat, is sufficient to rescue neuronal remodeling defects of shep knockdown flies. Our results reveal direct regulation of transcriptional programs by Shep to regulate neuronal remodeling during metamorphosis.
Collapse
Affiliation(s)
- Dahong Chen
- Nuclear Organization and Gene Expression Section, Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ryan K Dale
- Nuclear Organization and Gene Expression Section, Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elissa P Lei
- Nuclear Organization and Gene Expression Section, Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
37
|
Analysis of temporal transcription expression profiles reveal links between protein function and developmental stages of Drosophila melanogaster. PLoS Comput Biol 2017; 13:e1005791. [PMID: 29045400 PMCID: PMC5662236 DOI: 10.1371/journal.pcbi.1005791] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 10/30/2017] [Accepted: 09/25/2017] [Indexed: 11/19/2022] Open
Abstract
Accurate gene or protein function prediction is a key challenge in the post-genome era. Most current methods perform well on molecular function prediction, but struggle to provide useful annotations relating to biological process functions due to the limited power of sequence-based features in that functional domain. In this work, we systematically evaluate the predictive power of temporal transcription expression profiles for protein function prediction in Drosophila melanogaster. Our results show significantly better performance on predicting protein function when transcription expression profile-based features are integrated with sequence-derived features, compared with the sequence-derived features alone. We also observe that the combination of expression-based and sequence-based features leads to further improvement of accuracy on predicting all three domains of gene function. Based on the optimal feature combinations, we then propose a novel multi-classifier-based function prediction method for Drosophila melanogaster proteins, FFPred-fly+. Interpreting our machine learning models also allows us to identify some of the underlying links between biological processes and developmental stages of Drosophila melanogaster. Despite painstaking experimental efforts and the extensive sequence similarity based annotation transfers, less than a half of the fruit fly protein sequences in UniProtKB have some functional annotation. To help fill in this gap, we test the usefulness of publicly available temporal gene expression profiles and their combination with many biophysical attributes that can be effectively derived from the corresponding protein sequence. We find that such an integrative function prediction method provides more accurate predictions than using sequence data alone and we expect these predictions to help narrow down the number of experimental assays required to characterise fly protein function. We demonstrate by highlighting correlations between predicted biological process functions and known facts about fly developmental stages.
Collapse
|
38
|
Liu HW, Wang LL, Meng Z, Tang X, Li YS, Xia QY, Zhao P. A clip domain serine protease involved in moulting in the silkworm, Bombyx mori: cloning, characterization, expression patterns and functional analysis. INSECT MOLECULAR BIOLOGY 2017; 26:507-521. [PMID: 28597953 DOI: 10.1111/imb.12312] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Clip domain serine proteases (CLIPs), characterized by one or more conserved clip domains, are essential components of extracellular signalling cascades in various biological processes, especially in innate immunity and the embryonic development of insects. Additionally, CLIPs may have additional non-immune functions in insect development. In the present study, the clip domain serine protease gene Bombyx mori serine protease 95 (BmSP95), which encodes a 527-residue protein, was cloned from the integument of B. mori. Bioinformatics analysis indicated that BmSP95 is a typical CLIP of the subfamily D and possesses a clip domain at the N terminus, a trypsin-like serine protease (tryp_spc) domain at the C terminus and a conserved proline-rich motif between these two domains. At the transcriptional level, BmSP95 is expressed in the integument during moulting and metamorphosis, and the expression pattern is consistent with the fluctuating 20-hydroxyecdysone (20E) titre in B. mori. At the translational level, BmSP95 protein is synthesized in the epidermal cells, secreted as a zymogen and activated in the moulting fluid. Immunofluorescence revealed that BmSP95 is distributed into the old endocuticle in the moulting stage. The expression of BmSP95 was upregulated by 20E. Moreover, expression of BmSP95 was downregulated by pathogen infection. RNA interference-mediated silencing of BmSP95 led to delayed moulting from pupa to moth. These results suggest that BmSP95 is involved in integument remodelling during moulting and metamorphosis.
Collapse
Affiliation(s)
- H-W Liu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - L-L Wang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Z Meng
- College of Biotechnology, Southwest University, Chongqing, China
| | - X Tang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Y-S Li
- Vitamin D Research Institute, Shaanxi University of Technology, Hanzhong, Shaanxi, China
| | - Q-Y Xia
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - P Zhao
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| |
Collapse
|
39
|
Yuan S, Huang W, Geng L, Beerntsen BT, Song H, Ling E. Differentiation of lepidoptera scale cells from epidermal stem cells followed by ecdysone-regulated DNA duplication and scale secreting. Cell Cycle 2017; 16:2156-2167. [PMID: 28933984 DOI: 10.1080/15384101.2017.1376148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Integuments are the first line to protect insects from physical damage and pathogenic infection. In lepidopteran insects, they undergo distinct morphology changes such as scale formation during metamorphosis. However, we know little about integument development and scale formation during this stage. Here, we use the silkworm, Bombyx mori, as a model and show that stem cells in the integument of each segment, but not intersegmental membrane, divide into two scale precursor cells during the spinning stage. In young pupae, the scale precursor cell divides again. One of the daughter cells becomes a mature scale-secreting cell that undergoes several rounds of DNA duplication and the other daughter cell undergoes apoptosis later on. This scale precursor cell division is crucial to the development and differentiation of scale-secreting cells because scale production can be blocked after treatment with the cell division inhibitor paclitaxel. Subsequently, the growth of scale-secreting cells is under the control of 20-hydroxyecdysone but not juvenile hormone since injection of 20-hydroxyecdysone inhibited scale formation. Further work demonstrated that 20-hydroxyecdysone injection inhibits DNA duplication in scale-secreting cells while the expression of scale-forming gene ASH1 was down-regulated by BR-C Z2. Therefore, this research demonstrates that the scale cells of the silkworm develops through stem cell division prior to pupation and then another wave of cell division differentiates these cells into scale secreting cells soon after entrance into the pupal stage. Additionally, DNA duplication and scale production in the scale-secreting cells were found to be under the regulation of 20-hydroxyecdysone.
Collapse
Affiliation(s)
- Shenglei Yuan
- a Key Laboratory of Insect Developmental and Evolutionary Biology , Institute of Plant Physiology and Ecology, Chinese Academy of Sciences , Shanghai , China.,b Department of Neurosciences , College of Life Sciences, Shanghai University , Shanghai , China
| | - Wuren Huang
- a Key Laboratory of Insect Developmental and Evolutionary Biology , Institute of Plant Physiology and Ecology, Chinese Academy of Sciences , Shanghai , China
| | - Lei Geng
- a Key Laboratory of Insect Developmental and Evolutionary Biology , Institute of Plant Physiology and Ecology, Chinese Academy of Sciences , Shanghai , China
| | - Brenda T Beerntsen
- c Department of Veterinary Pathobiology , University of Missouri , Columbia , MO , USA
| | - Hongsheng Song
- b Department of Neurosciences , College of Life Sciences, Shanghai University , Shanghai , China
| | - Erjun Ling
- a Key Laboratory of Insect Developmental and Evolutionary Biology , Institute of Plant Physiology and Ecology, Chinese Academy of Sciences , Shanghai , China
| |
Collapse
|
40
|
Kristó I, Bajusz C, Borsos BN, Pankotai T, Dopie J, Jankovics F, Vartiainen MK, Erdélyi M, Vilmos P. The actin binding cytoskeletal protein Moesin is involved in nuclear mRNA export. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1589-1604. [PMID: 28554770 DOI: 10.1016/j.bbamcr.2017.05.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 05/22/2017] [Accepted: 05/25/2017] [Indexed: 12/29/2022]
Abstract
Current models imply that the evolutionarily conserved, actin-binding Ezrin-Radixin-Moesin (ERM) proteins perform their activities at the plasma membrane by anchoring membrane proteins to the cortical actin network. Here we show that beside its cytoplasmic functions, the single ERM protein of Drosophila, Moesin, has a novel role in the nucleus. The activation of transcription by heat shock or hormonal treatment increases the amount of nuclear Moesin, indicating biological function for the protein in the nucleus. The distribution of Moesin in the nucleus suggests a function in transcription and the depletion of mRNA export factors Nup98 or its interacting partner, Rae1, leads to the nuclear accumulation of Moesin, suggesting that the nuclear function of the protein is linked to mRNA export. Moesin localizes to mRNP particles through the interaction with the mRNA export factor PCID2 and knock down of Moesin leads to the accumulation of mRNA in the nucleus. Based on our results we propose that, beyond its well-known, manifold functions in the cytoplasm, the ERM protein of Drosophila is a new, functional component of the nucleus where it participates in mRNA export.
Collapse
Affiliation(s)
- Ildikó Kristó
- Biological Research Center of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Csaba Bajusz
- Biological Research Center of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Barbara N Borsos
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
| | - Tibor Pankotai
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
| | - Joseph Dopie
- University of Helsinki, Institute of Biotechnology, Helsinki, Finland
| | - Ferenc Jankovics
- Biological Research Center of the Hungarian Academy of Sciences, Szeged, Hungary
| | | | - Miklós Erdélyi
- Biological Research Center of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Péter Vilmos
- Biological Research Center of the Hungarian Academy of Sciences, Szeged, Hungary.
| |
Collapse
|
41
|
Ables ET, Drummond-Barbosa D. Steroid Hormones and the Physiological Regulation of Tissue-Resident Stem Cells: Lessons from the Drosophila Ovary. CURRENT STEM CELL REPORTS 2017; 3:9-18. [PMID: 28458991 PMCID: PMC5407287 DOI: 10.1007/s40778-017-0070-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Stem cells respond to local paracrine signals; more recently, however, systemic hormones have also emerged as key regulators of stem cells. This review explores the role of steroid hormones in stem cells, using the Drosophila germline stem cell as a centerpiece for discussion. RECENT FINDINGS Stem cells sense and respond directly and indirectly to steroid hormones, which regulate diverse sets of target genes via interactions with nuclear hormone receptors. Hormone-regulated networks likely integrate the actions of multiple systemic signals to adjust the activity of stem cell lineages in response to changes in physiological status. SUMMARY Hormones are inextricably linked to animal physiology, and can control stem cells and their local niches. Elucidating the molecular mechanisms of hormone signaling in stem cells is essential for our understanding of the fundamental underpinnings of stem cell biology, and for informing new therapeutic interventions against cancers or for regenerative medicine.
Collapse
Affiliation(s)
- Elizabeth T. Ables
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| | - Daniela Drummond-Barbosa
- Department of Biochemistry and Molecular Biology, Division of Reproductive Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
42
|
Vellichirammal NN, Madayiputhiya N, Brisson JA. The genomewide transcriptional response underlying the pea aphid wing polyphenism. Mol Ecol 2016; 25:4146-60. [PMID: 27393739 PMCID: PMC5021599 DOI: 10.1111/mec.13749] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 06/07/2016] [Accepted: 07/05/2016] [Indexed: 01/17/2023]
Abstract
Phenotypic plasticity is a key life history strategy used by many plants and animals living in heterogeneous environments. A multitude of studies have investigated the costs and limits of plasticity, as well as the conditions under which it evolves. Much less well understood are the molecular genetic mechanisms that enable an organism to sense its environment and respond in a plastic manner. The pea aphid wing polyphenism is a compelling laboratory model to study these mechanisms. In this polyphenism, environmental stressors like high density cause asexual, viviparous adult female aphids to change the development of their embryos from wingless to winged morphs. The life history trade-offs between the two morphs have been intensively studied, but the molecular mechanisms underlying this process remain largely unknown. We therefore performed a genomewide study of the maternal transcriptome at two time points with and without a crowding stress to discover the maternal molecular changes that lead to the development of winged vs. wingless offspring. We observed significant transcriptional changes in genes associated with odorant binding, neurotransmitter transport, hormonal activity and chromatin remodelling in the maternal transcriptome. We also found that titres of serotonin, dopamine and octopamine were higher in solitary compared to crowded aphids. We use these results to posit a model for how maternal signals inform a developing embryo to be winged or wingless. Our findings add significant insights into the identity of the molecular mechanisms that underlie environmentally induced morph determination and suggest a possible role for biogenic amine regulation in polyphenisms generally.
Collapse
Affiliation(s)
| | | | - Jennifer A. Brisson
- School of Biological Sciences, University of Nebraska-Lincoln, Nebraska, USA
| |
Collapse
|
43
|
A Genetic Mosaic Screen Reveals Ecdysone-Responsive Genes Regulating Drosophila Oogenesis. G3-GENES GENOMES GENETICS 2016; 6:2629-42. [PMID: 27226164 PMCID: PMC4978916 DOI: 10.1534/g3.116.028951] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Multiple aspects of Drosophila oogenesis, including germline stem cell activity, germ cell differentiation, and follicle survival, are regulated by the steroid hormone ecdysone. While the transcriptional targets of ecdysone signaling during development have been studied extensively, targets in the ovary remain largely unknown. Early studies of salivary gland polytene chromosomes led to a model in which ecdysone stimulates a hierarchical transcriptional cascade, wherein a core group of ecdysone-sensitive transcription factors induce tissue-specific responses by activating secondary branches of transcriptional targets. More recently, genome-wide approaches have identified hundreds of putative ecdysone-responsive targets. Determining whether these putative targets represent bona fide targets in vivo, however, requires that they be tested via traditional mutant analysis in a cell-type specific fashion. To investigate the molecular mechanisms whereby ecdysone signaling regulates oogenesis, we used genetic mosaic analysis to screen putative ecdysone-responsive genes for novel roles in the control of the earliest steps of oogenesis. We identified a cohort of genes required for stem cell maintenance, stem and progenitor cell proliferation, and follicle encapsulation, growth, and survival. These genes encode transcription factors, chromatin modulators, and factors required for RNA transport, stability, and ribosome biogenesis, suggesting that ecdysone might control a wide range of molecular processes during oogenesis. Our results suggest that, although ecdysone target genes are known to have cell type-specific roles, many ecdysone response genes that control larval or pupal cell types at developmental transitions are used reiteratively in the adult ovary. These results provide novel insights into the molecular mechanisms by which ecdysone signaling controls oogenesis, laying new ground for future studies.
Collapse
|
44
|
Van Bortle K, Peterson AJ, Takenaka N, O'Connor MB, Corces VG. CTCF-dependent co-localization of canonical Smad signaling factors at architectural protein binding sites in D. melanogaster. Cell Cycle 2016; 14:2677-87. [PMID: 26125535 DOI: 10.1080/15384101.2015.1053670] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
The transforming growth factor β (TGF-β) and bone morphogenetic protein (BMP) pathways transduce extracellular signals into tissue-specific transcriptional responses. During this process, signaling effector Smad proteins translocate into the nucleus to direct changes in transcription, but how and where they localize to DNA remain important questions. We have mapped Drosophila TGF-β signaling factors Mad, dSmad2, Medea, and Schnurri genome-wide in Kc cells and find that numerous sites for these factors overlap with the architectural protein CTCF. Depletion of CTCF by RNAi results in the disappearance of a subset of Smad sites, suggesting Smad proteins localize to CTCF binding sites in a CTCF-dependent manner. Sensitive Smad binding sites are enriched at low occupancy CTCF peaks within topological domains, rather than at the physical domain boundaries where CTCF may function as an insulator. In response to Decapentaplegic, CTCF binding is not significantly altered, whereas Mad, Medea, and Schnurri are redirected from CTCF to non-CTCF binding sites. These results suggest that CTCF participates in the recruitment of Smad proteins to a subset of genomic sites and in the redistribution of these proteins in response to BMP signaling.
Collapse
|
45
|
Li Z, You L, Zeng B, Ling L, Xu J, Chen X, Zhang Z, Palli SR, Huang Y, Tan A. Ectopic expression of ecdysone oxidase impairs tissue degeneration in Bombyx mori. Proc Biol Sci 2016; 282:20150513. [PMID: 26041352 DOI: 10.1098/rspb.2015.0513] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Metamorphosis in insects includes a series of programmed tissue histolysis and remolding processes that are controlled by two major classes of hormones, juvenile hormones and ecdysteroids. Precise pulses of ecdysteroids (the most active ecdysteroid is 20-hydroxyecdysone, 20E), are regulated by both biosynthesis and metabolism. In this study, we show that ecdysone oxidase (EO), a 20E inactivation enzyme, expresses predominantly in the midgut during the early pupal stage in the lepidopteran model insect, Bombyx mori. Depletion of BmEO using the transgenic CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/RNA-guided Cas9 nucleases) system extended the duration of the final instar larval stage. Ubiquitous transgenic overexpression of BmEO using the Gal4/UAS system induced lethality during the larval-pupal transition. When BmEO was specifically overexpressed in the middle silk gland (MSG), degeneration of MSG at the onset of metamorphosis was blocked. Transmission electron microscope and LysoTracker analyses showed that the autophagy pathway in MSG is inhibited by BmEO ectopic expression. Furthermore, RNA-seq analysis revealed that the genes involved in autophagic cell death and the mTOR signal pathway are affected by overexpression of BmEO. Taken together, BmEO functional studies reported here provide insights into ecdysone regulation of tissue degeneration during metamorphosis.
Collapse
Affiliation(s)
- Zhiqian Li
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, People's Republic of China
| | - Lang You
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, People's Republic of China
| | - Baosheng Zeng
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, People's Republic of China
| | - Lin Ling
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, People's Republic of China
| | - Jun Xu
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, People's Republic of China
| | - Xu Chen
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, People's Republic of China
| | - Zhongjie Zhang
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, People's Republic of China
| | - Subba Reddy Palli
- Department of Entomology, College of Agriculture, University of Kentucky, S-225 Agriculture Science Center North, Lexington, KY 40546, USA
| | - Yongping Huang
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, People's Republic of China
| | - Anjiang Tan
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, People's Republic of China
| |
Collapse
|
46
|
Subbarayan S, Marimuthu SK, Nachimuthu SK, Zhang W, Subramanian S. Characterization and cytotoxic activity of apoptosis-inducing pierisin-5 protein from white cabbage butterfly. Int J Biol Macromol 2016; 87:16-27. [PMID: 26812112 DOI: 10.1016/j.ijbiomac.2016.01.072] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 01/19/2016] [Accepted: 01/21/2016] [Indexed: 10/22/2022]
Abstract
In this study, caspase-dependent apoptosis-inducing pierisin-5 gene was identified and characterized from cabbage white butterfly, Pieris canidia. A thousand-fold increase in expression of pierisin-5 gene was observed from second to third instar larvae, gradually decreasing before pupation. Pierisin-5 was purified from the fifth-instar larvae and was found to exhibit cytotoxicity against HeLa and HepG2 human cancer cell lines. Pierisin-5 showed growth inhibition and several morphological changes such as cell shrinkage, chromatin condensation and apoptotic body formation with programmed cell death in HeLa and HepG2 cells. Moreover, DNA fragmentation was observed after gel electrophoresis analysis. Caspase substrate assay showed further cleavage of Ac-DEVD-pNA, suggesting the activation of Caspase-3. Flow cytometry analysis revealed the cell cycle arrest at G1 phase and increased the percentage of apoptotic cells in cancer cell lines treated with pierisin-5. These findings suggest that pierisin-5 could significantly induce apoptosis in cancer cell lines and is mediated by activation of caspase-3 in the mitochondrial pathway. Phylogenetic analysis using pierisin proteins from Pierid butterflies, ADP-ribosylating toxins from bacteria, human, rat, and mouse indicated the possibility of horizontal transfer of pierisin genes from bacteria to butterflies. The single copy of pierisin gene unlike other insect toxin genes also supports lateral transfer.
Collapse
Affiliation(s)
| | | | | | - Wenqing Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Selvi Subramanian
- Department of Biotechnology, PSG College of Technology, Coimbatore, 641004 TN, India.
| |
Collapse
|
47
|
Abstract
Steroid hormones induce cascades of gene activation and repression with transformative effects on cell fate . Steroid transduction plays a major role in the development and physiology of nearly all metazoan species, and in the progression of the most common forms of cancer. Despite the paramount importance of steroids in developmental and translational biology, a complete map of transcriptional response has not been developed for any hormone . In the case of 20-hydroxyecdysone (ecdysone) in Drosophila melanogaster, these trajectories range from apoptosis to immortalization. We mapped the ecdysone transduction network in a cohort of 41 cell lines, the largest such atlas yet assembled. We found that the early transcriptional response mirrors the distinctiveness of physiological origins: genes respond in restricted patterns, conditional on the expression levels of dozens of transcription factors. Only a small cohort of genes is constitutively modulated independent of initial cell state. Ecdysone-responsive genes tend to organize into directional same-stranded units, with consecutive genes induced from the same strand. Here, we identify half of the ecdysone receptor heterodimer as the primary rate-limiting step in the response, and find that initial receptor isoform levels modulate the activated cohort of target transcription factors. This atlas of steroid response reveals organizing principles of gene regulation by a model type II nuclear receptor and lays the foundation for comprehensive and predictive understanding of the ecdysone transduction network in the fruit fly.
Collapse
|
48
|
Van Bortle K, Nichols MH, Ramos E, Corces VG. Integrated tRNA, transcript, and protein profiles in response to steroid hormone signaling. RNA (NEW YORK, N.Y.) 2015; 21:1807-17. [PMID: 26289344 PMCID: PMC4574756 DOI: 10.1261/rna.052126.115] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 07/16/2015] [Indexed: 06/04/2023]
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.
Collapse
Affiliation(s)
- Kevin Van Bortle
- Department of Biology, Emory University, Atlanta, Georgia 30322, USA
| | - Michael H Nichols
- Department of Biology, Emory University, Atlanta, Georgia 30322, USA
| | - Edward Ramos
- Department of Biology, Emory University, Atlanta, Georgia 30322, USA
| | - Victor G Corces
- Department of Biology, Emory University, Atlanta, Georgia 30322, USA
| |
Collapse
|
49
|
Roy S, Saha TT, Johnson L, Zhao B, Ha J, White KP, Girke T, Zou Z, Raikhel AS. Regulation of Gene Expression Patterns in Mosquito Reproduction. PLoS Genet 2015; 11:e1005450. [PMID: 26274815 PMCID: PMC4537244 DOI: 10.1371/journal.pgen.1005450] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 07/17/2015] [Indexed: 12/14/2022] Open
Abstract
In multicellular organisms, development, growth and reproduction require coordinated expression of numerous functional and regulatory genes. Insects, in addition to being the most speciose animal group with enormous biological and economical significance, represent outstanding model organisms for studying regulation of synchronized gene expression due to their rapid development and reproduction. Disease-transmitting female mosquitoes have adapted uniquely for ingestion and utilization of the huge blood meal required for swift reproductive events to complete egg development within a 72-h period. We investigated the network of regulatory factors mediating sequential gene expression in the fat body, a multifunctional organ analogous to the vertebrate liver and adipose tissue, of the female Aedes aegypti mosquito. Transcriptomic and bioinformatics analyses revealed that ~7500 transcripts are differentially expressed in four sequential waves during the 72-h reproductive period. A combination of RNA-interference gene-silencing and in-vitro organ culture identified the major regulators for each of these waves. Amino acids (AAs) regulate the first wave of gene activation between 3 h and 12 h post-blood meal (PBM). During the second wave, between 12 h and 36 h, most genes are highly upregulated by a synergistic action of AAs, 20-hydroxyecdysone (20E) and the Ecdysone-Receptor (EcR). Between 36 h and 48 h, the third wave of gene activation—regulated mainly by HR3—occurs. Juvenile Hormone (JH) and its receptor Methoprene-Tolerant (Met) are major regulators for the final wave between 48 h and 72 h. Each of these key regulators also has repressive effects on one or more gene sets. Our study provides a better understanding of the complexity of the regulatory mechanisms related to temporal coordination of gene expression during reproduction. We have detected the novel function of 20E/EcR responsible for transcriptional repression. This study also reveals the previously unidentified large-scale effects of HR3 and JH/Met on transcriptional regulation during the termination of vitellogenesis and remodeling of the fat body. In addition to being vectors of devastating human diseases, mosquitoes represent outstanding model organisms for studying regulatory mechanisms of differential gene expression due to their rapid reproductive cycles. About 7500 transcripts are differentially expressed in four sequential waves during the 72-h reproductive period in the fat body, a critical reproductive organ. The major regulators for these waves of gene expression are the two very important insect hormones, 20-hydroxyecdysone (20E) and Juvenile hormone (JH), their respective receptors Ecdysone Receptor (EcR) and Methoprene-Tolerant (Met), amino acids and the orphan nuclear receptor HR3. These key regulators are responsible for activation and repression of co-regulated gene sets, at different time points, within the 72-h reproductive period. Importantly, this study, apart from providing an insight into the regulatory complexity involved in the temporal coordination of gene expression, also reveals the previously unidentified roles of 20E/EcR, JH/Met and HR3 during the 72-h period post blood meal.
Collapse
Affiliation(s)
- Sourav Roy
- Department of Entomology, University of California, Riverside, Riverside, California, United States of America
- Institute of Integrative Genome Biology, University of California, Riverside, Riverside, California, United States of America
| | - Tusar T. Saha
- Department of Entomology, University of California, Riverside, Riverside, California, United States of America
- Institute of Integrative Genome Biology, University of California, Riverside, Riverside, California, United States of America
| | - Lisa Johnson
- Department of Entomology, University of California, Riverside, Riverside, California, United States of America
- Institute of Integrative Genome Biology, University of California, Riverside, Riverside, California, United States of America
- Graduate Program in Cell, Molecular and Developmental Biology, University of California, Riverside, Riverside, California, United States of America
| | - Bo Zhao
- Department of Entomology, University of California, Riverside, Riverside, California, United States of America
- Institute of Integrative Genome Biology, University of California, Riverside, Riverside, California, United States of America
| | - Jisu Ha
- Department of Entomology, University of California, Riverside, Riverside, California, United States of America
- Institute of Integrative Genome Biology, University of California, Riverside, Riverside, California, United States of America
- Graduate Program in Genetics, Genomics and Bioinformatics, University of California, Riverside, Riverside, California, United States of America
| | - Kevin P. White
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, Illinois, United States of America
| | - Thomas Girke
- Institute of Integrative Genome Biology, University of California, Riverside, Riverside, California, United States of America
- Department of Botany and Plant Sciences, University of California, Riverside, Riverside, California, United States of America
| | - Zhen Zou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- * E-mail: (ZZ); (ASR)
| | - Alexander S. Raikhel
- Department of Entomology, University of California, Riverside, Riverside, California, United States of America
- Institute of Integrative Genome Biology, University of California, Riverside, Riverside, California, United States of America
- * E-mail: (ZZ); (ASR)
| |
Collapse
|
50
|
Yadav P, Choudhury D, Sadanandappa MK, Sharma VK. Extent of mismatch between the period of circadian clocks and light/dark cycles determines time-to-emergence in fruit flies. INSECT SCIENCE 2015; 22:569-577. [PMID: 24668961 DOI: 10.1111/1744-7917.12126] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/03/2014] [Indexed: 06/03/2023]
Abstract
Circadian clocks time developmental stages of fruit flies Drosophila melanogaster, while light/dark (LD) cycles delimit emergence of adults, conceding only during the "allowed gate." Previous studies have revealed that time-to-emergence can be altered by mutations in the core clock gene period (per), or by altering the length of LD cycles. Since this evidence came from studies on genetically manipulated flies, or on flies maintained under LD cycles with limited range of periods, inferences that can be drawn are limited. Moreover, the extent of shortening or lengthening of time-to-emergence remains yet unknown. In order to pursue this further, we assayed time-to-emergence of D. melanogaster under 12 different LD cycles as well as in constant light (LL) and constant dark conditions (DD). Time-to-emergence in flies occurred earlier under LL than in LD cycles and DD. Among the LD cycles, time-to-emergence occurred earlier under T4-T8, followed by T36-T48, and then T12-T32, suggesting that egg-to-emergence duration in flies becomes shorter when the length of LD cycles deviates from 24 h, bearing a strong positive and a marginally negative correlation with day length, for values shorter and longer than 24 h, respectively. These results suggest that the extent of mismatch between the period of circadian clocks and environmental cycles determines the time-to-emergence in Drosophila.
Collapse
Affiliation(s)
- Pankaj Yadav
- Chronobiology Laboratory, Evolutionary and Organismal Biology Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore 560064, Karnataka, India
| | - Deepak Choudhury
- Chronobiology Laboratory, Evolutionary and Organismal Biology Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore 560064, Karnataka, India
| | - Madhumala K Sadanandappa
- Chronobiology Laboratory, Evolutionary and Organismal Biology Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore 560064, Karnataka, India
| | - Vijay Kumar Sharma
- Chronobiology Laboratory, Evolutionary and Organismal Biology Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore 560064, Karnataka, India
| |
Collapse
|