1
|
Fujinaga D, Nolan C, Yamanaka N. Functional characterization of eicosanoid signaling in Drosophila development. PLoS Genet 2025; 21:e1011705. [PMID: 40344083 DOI: 10.1371/journal.pgen.1011705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 04/29/2025] [Indexed: 05/11/2025] Open
Abstract
20-carbon fatty acid-derived eicosanoids are versatile signaling oxylipins in mammals. In particular, a group of eicosanoids termed prostanoids are involved in multiple physiological processes, such as reproduction and immune responses. Although some eicosanoids such as prostaglandin E2 (PGE2) have been detected in some insect species, molecular mechanisms of eicosanoid synthesis and signal transduction in insects have not been thoroughly investigated. Our phylogenetic analysis indicated that, in clear contrast to the presence of numerous receptors for oxylipins and other lipid mediators in humans, the Drosophila genome only possesses a single ortholog of such receptors, which is homologous to human prostanoid receptors. This G protein-coupled receptor, named Prostaglandin Receptor or PGR, is activated by PGE2 and its isomer PGD2 in Drosophila S2 cells. PGR mutant flies die as pharate adults with insufficient tracheal development, which can be rescued by supplying high oxygen. Consistent with this, through a comprehensive mutagenesis approach, we identified a Drosophila PGE synthase whose mutants show similar pharate adult lethality with hypoxia responses. Drosophila thus has a highly simplified eicosanoid signaling pathway as compared to humans, and it may provide an ideal model system for investigating evolutionarily conserved aspects of eicosanoid signaling.
Collapse
Affiliation(s)
- Daiki Fujinaga
- Department of Entomology, Institute for Integrative Genome Biology, University of California, Riverside, Riverside, California, United States of America
| | - Cebrina Nolan
- Department of Entomology, Institute for Integrative Genome Biology, University of California, Riverside, Riverside, California, United States of America
| | - Naoki Yamanaka
- Department of Entomology, Institute for Integrative Genome Biology, University of California, Riverside, Riverside, California, United States of America
| |
Collapse
|
2
|
Jiao Y, Sengodan K, Chen J, Palli SR. Role of histone methylation in insect development: KMT5A regulates ecdysteroid biosynthesis during metamorphosis of Tribolium castaneum. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2025; 180:104316. [PMID: 40287070 PMCID: PMC12066215 DOI: 10.1016/j.ibmb.2025.104316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 04/09/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
Methylation levels of core histones play important roles in the regulation of gene expression and impact animal development. However, the methyltransferases and demethylases that determine histone methylation levels remain largely unexplored in insects. Most of our current understanding of histone methylation comes from mammalian studies. In this study, we first identified potential histone methyltransferases and demethylases encoded in the genome of the red flour beetle Tribolium castaneum. The function of these histone methylation enzymes in the metamorphosis was investigated by knocking down genes coding for these enzymes using RNA interference (RNAi). Our results showed that a lysine methyltransferase, KMT5A, plays a critical role in T. castaneum metamorphosis by regulating the biosynthesis of ecdysteroids. Treating KMT5A-knockdown larvae with 20 hydroxyecdysone can partially rescue T. castaneum pupation. Western blot analysis showed that KMT5A catalyzes H4K20 mono-methylation. However, further studies suggest that KMT5A may regulate T. castaneum pupation through mechanisms independent of H4K20 methylation. These data uncovered the roles of histone methylation enzymes in T. castaneum metamorphosis and KMT5A as a critical regulator of ecdysteroid biosynthesis.
Collapse
Affiliation(s)
- Yaoyu Jiao
- Department of Entomology, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY, 40546, USA.
| | - Karthi Sengodan
- Department of Entomology, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY, 40546, USA
| | - Jiasheng Chen
- Department of Entomology, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY, 40546, USA
| | - Subba Reddy Palli
- Department of Entomology, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY, 40546, USA.
| |
Collapse
|
3
|
Hu ZL, Wei H, Sun L, Russinova E. Plant steroids on the move: mechanisms of brassinosteroid export. Trends Biochem Sci 2025:S0968-0004(25)00052-0. [PMID: 40251078 DOI: 10.1016/j.tibs.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/02/2025] [Accepted: 03/06/2025] [Indexed: 04/20/2025]
Abstract
Brassinosteroids (BRs) are essential plant steroidal hormones that regulate growth and development. The recent discoveries of ATP-binding cassette subfamily B (ABCB) members, ABCB19 and ABCB1, as BR transporters highlight the significance of active export to the apoplast in maintaining BR homeostasis and enabling effective signaling. This review focuses on the latest progress in understanding ABCB-mediated BR transport, with particular attention to the structural and functional characterization of arabidopsis ABCB19 and ABCB1. These findings reveal both conserved and distinct features in substrate recognition and transport mechanisms, providing valuable insights into their roles in hormonal regulation. Additionally, the evolutionary conservation of ABC transporters in mediating steroid-based signaling across biological kingdoms underscores their fundamental biological significance.
Collapse
Affiliation(s)
- Zi-Liang Hu
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium; Center for Plant Systems Biology, VIB, 9052 Ghent, Belgium
| | - Hong Wei
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027 Hefei, China
| | - Linfeng Sun
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027 Hefei, China
| | - Eugenia Russinova
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium; Center for Plant Systems Biology, VIB, 9052 Ghent, Belgium.
| |
Collapse
|
4
|
Bradic I, Rewitz K. Steroid Signaling in Autophagy. J Mol Biol 2025:169134. [PMID: 40210154 DOI: 10.1016/j.jmb.2025.169134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/19/2025] [Accepted: 04/04/2025] [Indexed: 04/12/2025]
Abstract
Autophagy is a conserved cellular process essential for homeostasis and development that plays a central role in the degradation and recycling of cellular components. Recent studies reveal bidirectional interactions between autophagy and steroid-hormone signaling. Steroids are signaling molecules synthesized from cholesterol that regulate key physiological and developmental processes - including autophagic activity. Conversely, other work demonstrates that autophagy regulates steroid production by controlling the availability of precursor sterol substrate. Insights from Drosophila and mammalian models provide compelling evidence for the conservation of these mechanisms across species. In this review we explore how steroid hormones modulate autophagy in diverse tissues and contexts, such as metabolism and disease, and discuss advances in our understanding of autophagy's regulatory role in steroid hormone production. We examine the implications of these interactions for health and disease and offer perspectives on the potential for harnessing this functionality for addressing cholesterol-related disorders.
Collapse
Affiliation(s)
- Ivan Bradic
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Kim Rewitz
- Department of Biology, University of Copenhagen, 2100 Copenhagen O, Denmark.
| |
Collapse
|
5
|
Schwarzlmueller P, Triebig A, Assié G, Jouinot A, Theurich S, Maier T, Beuschlein F, Kobold S, Kroiss M. Steroid hormones as modulators of anti-tumoural immunity. Nat Rev Endocrinol 2025:10.1038/s41574-025-01102-2. [PMID: 40128599 DOI: 10.1038/s41574-025-01102-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/27/2025] [Indexed: 03/26/2025]
Abstract
Immune evasion is a hallmark of cancer progression but the role of steroid hormones in this evasion has long been underrated. This oversight is particularly notable for glucocorticoids given that exogenous glucocorticoids remain a cornerstone therapy in various oncological treatment regimens, supportive care and treatment of immune-related adverse events caused by immune-checkpoint inhibitors. Cortisol, the main endogenous glucocorticoid in humans, is secreted by the adrenal cortex in response to stress. Additionally, cortisol and its inactive metabolite cortisone can be interconverted to further modulate tissue-dependent glucocorticoid action. In the past 5 years, intratumoural production of glucocorticoids, by both immune and tumour cells, has been shown to support tumour immune evasion. Here, we summarize current progress at the crossroads of endocrinology and immuno-oncology. We outline the known effects of steroid hormones on different immune cell types with a focus on glucocorticoids and androgens. We conclude with options for pharmaceutical intervention, including the engineering of cell-based therapies that resist the immunosuppressive action of steroid hormones. Overall, local steroid production and metabolism are emerging elements of tumour immune suppression that are potentially amenable to therapeutic intervention. Targeting steroid hormones to enhance anticancer therapies could increase their efficacy but will require expertise in endocrine care.
Collapse
Affiliation(s)
| | - Alexandra Triebig
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Guillaume Assié
- Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
- Department of Endocrinology and National Reference Center for Rare Adrenal Disorders, Hôpital Cochin, Paris, France
| | - Anne Jouinot
- Department of Endocrinology and National Reference Center for Rare Adrenal Disorders, Hôpital Cochin, Paris, France
- Université Paris Cité, Institut Cochin, Paris, France
| | - Sebastian Theurich
- Department of Medicine III and Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital, Munich, Germany
- Cancer- and Immunometabolism Research Group, Gene Center, Ludwig Maximilian University (LMU), Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- German Cancer Consortium (DKTK), Munich Site, Heidelberg, Germany
| | - Tanja Maier
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Felix Beuschlein
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ), Zurich, Switzerland
- The LOOP Zurich - Medical Research Center, Zurich, Switzerland
| | - Sebastian Kobold
- German Cancer Consortium (DKTK), Munich Site, Heidelberg, Germany
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Matthias Kroiss
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany.
- Bavarian Cancer Research Center (BZKF), Munich, Germany.
- Kroiss Endokrinologie & Diabetologie, Schweinfurt, Germany.
| |
Collapse
|
6
|
Fujinaga D, Nolan C, Yamanaka N. Functional characterization of eicosanoid signaling in Drosophila development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632770. [PMID: 39868285 PMCID: PMC11761813 DOI: 10.1101/2025.01.13.632770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
20-carbon fatty acid-derived eicosanoids are versatile signaling oxylipins in mammals. In particular, a group of eicosanoids termed prostanoids are involved in multiple physiological processes, such as reproduction and immune responses. Although some eicosanoids such as prostaglandin E2 (PGE2) have been detected in some insect species, molecular mechanisms of eicosanoid synthesis and signal transduction in insects have not been thoroughly investigated. Our phylogenetic analysis indicated that, in clear contrast to the presence of numerous receptors for oxylipins and other lipid mediators in humans, the Drosophila genome only possesses a single ortholog of such receptors, which is homologous to human prostanoid receptors. This G protein-coupled receptor, named Prostaglandin Receptor or PGR, is activated by PGE2 and its isomer PGD2 in Drosophila S2 cells. PGR mutant flies die as pharate adults with insufficient tracheal development, which can be rescued by supplying high oxygen. Consistent with this, through a comprehensive mutagenesis approach, we identified a Drosophila PGE synthase whose mutants show similar pharate adult lethality with hypoxia responses. Drosophila thus has a highly simplified eicosanoid signaling pathway as compared to humans, and it may provide an ideal model system for investigating evolutionarily conserved aspects of eicosanoid signaling.
Collapse
Affiliation(s)
- Daiki Fujinaga
- Department of Entomology, Institute for Integrative Genome Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Cebrina Nolan
- Department of Entomology, Institute for Integrative Genome Biology, University of California, Riverside, Riverside, CA 92521, USA
- Current address: Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Naoki Yamanaka
- Department of Entomology, Institute for Integrative Genome Biology, University of California, Riverside, Riverside, CA 92521, USA
| |
Collapse
|
7
|
Hagenbuch B, Stieger B, Locher KP. Organic anion transporting polypeptides: Pharmacology, toxicology, structure, and transport mechanisms. Pharmacol Rev 2025; 77:100023. [PMID: 40148036 DOI: 10.1016/j.pharmr.2024.100023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/12/2024] [Indexed: 03/29/2025] Open
Abstract
Organic anion transporting polypeptides (OATPs) are membrane proteins that mediate the uptake of a wide range of substrates across the plasma membrane of various cells and tissues. They are classified into 6 subfamilies, OATP1 through OATP6. Humans contain 12 OATPs encoded by 11 solute carrier of organic anion transporting polypeptide (SLCO) genes: OATP1A2, OATP1B1, OATP1B3, the splice variant OATP1B3-1B7, OATP1C1, OATP2A1, OATP2B1, OATP3A1, OATP4A1, OATP4C1, OATP5A1, and OATP6A1. Most of these proteins are expressed in epithelial cells, where they mediate the uptake of structurally unrelated organic anions, cations, and even neutral compounds into the cytoplasm. The best-characterized members are OATP1B1 and OATP1B3, which have an important role in drug metabolism by mediating drug uptake into the liver and are involved in drug-drug interactions. In this review, we aimed to (1) provide a historical perspective on the identification of OATPs and their nomenclature and discuss their phylogenic relationships and molecular characteristics; (2) review the current knowledge of the broad substrate specificity and their role in drug disposition and drug-drug interactions, with a special emphasis on human hepatic OATPs; (3) summarize the different experimental systems that are used to study the function of OATPs and discuss their advantages and disadvantages; (4) review the available experimental 3-dimensional structures and examine how they can help elucidate the transport mechanisms of OATPs; and (5) finally, summarize the current knowledge of the regulation of OATP expression, discuss clinically important single-nucleotide polymorphisms, and outline challenges of physiologically based pharmacokinetic modeling and in vitro to in vivo extrapolation. SIGNIFICANCE STATEMENT: Organic anion transporting polypeptides (OATPs) are a family of 12 uptake transporters in the solute carrier superfamily. Several members, particularly the liver-expressed OATP1B1 and OATP1B3, are important drug transporters. They mediate the uptake of several endobiotics and xenobiotics, including statins and numerous other drugs, into hepatocytes, and their inhibition by other drugs or reduced expression due to single-nucleotide polymorphisms can lead to adverse drug effects. Their recently solved 3-dimensional structures should help to elucidate their transport mechanisms and broad substrate specificities.
Collapse
Affiliation(s)
- Bruno Hagenbuch
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas.
| | - Bruno Stieger
- Institute of Molecular Biology and Biophysics, ETH Zürich, Zürich, Switzerland
| | - Kaspar P Locher
- Institute of Molecular Biology and Biophysics, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
8
|
Natsios P, Golini L, Park BH, Steffen F. Effect of local epidural application of methylprednisolone acetate on time to ambulation in non-ambulatory dogs with thoracolumbar intervertebral disc disease: A prospective randomised, blinded control trial. Vet Rec 2025; 196:e4962. [PMID: 39727220 PMCID: PMC11869356 DOI: 10.1002/vetr.4962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/10/2024] [Accepted: 11/04/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND The objective of this study was to analyse the potential benefit of the epidural application of steroids on time to ambulation in non-ambulatory dogs affected by intervertebral disc disease (IVDD) treated with decompressive surgery. METHODS This prospective, randomised, blinded control trial involved 41 dogs with thoracolumbar disc extrusion, which were randomly allocated into two groups. In the control group, saline was locally applied after surgical decompression of the spinal cord (n = 23). In the treatment group (n = 18), local epidural application of methylprednisolone acetate (1 mg/kg) was used. Ambulation time was the primary outcome measure, defined as the ability to take 10 independent steps. RESULTS The median number of days to ambulation was 7 days (range: 1‒17 days) for the control group and 3 days (range: 1‒8 days) for the treatment group. One dog from the treatment group developed discospondylitis and abscess formation. LIMITATIONS The study's heterogeneity in dog breeds, ages and pre-existing health conditions could affect the generalisability of the findings. CONCLUSION Epidural methylprednisolone acetate applied locally at the time of surgery may accelerate recovery in dogs following IVDD surgery.
Collapse
Affiliation(s)
- Pavlos Natsios
- Small Animal Surgery ClinicVetsuisse FacultyUniversity of ZurichZurichSwitzerland
| | - Lorenzo Golini
- Division of NeurologyVetsuisse FacultyUniversity of ZurichZurichSwitzerland
| | - Brian H. Park
- Small Animal Surgery ClinicVetsuisse FacultyUniversity of ZurichZurichSwitzerland
| | - Frank Steffen
- Division of NeurologyVetsuisse FacultyUniversity of ZurichZurichSwitzerland
| |
Collapse
|
9
|
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
|
10
|
Kocher S, Kingwell C. The Molecular Substrates of Insect Eusociality. Annu Rev Genet 2024; 58:273-295. [PMID: 39146360 PMCID: PMC11588544 DOI: 10.1146/annurev-genet-111523-102510] [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] [Indexed: 08/17/2024]
Abstract
The evolution of eusociality in Hymenoptera-encompassing bees, ants, and wasps-is characterized by multiple gains and losses of social living, making this group a prime model to understand the mechanisms that underlie social behavior and social complexity. Our review synthesizes insights into the evolutionary history and molecular basis of eusociality. We examine new evidence for key evolutionary hypotheses and molecular pathways that regulate social behaviors, highlighting convergent evolution on a shared molecular toolkit that includes the insulin/insulin-like growth factor signaling (IIS) and target of rapamycin (TOR) pathways, juvenile hormone and ecdysteroid signaling, and epigenetic regulation. We emphasize how the crosstalk among these nutrient-sensing and endocrine signaling pathways enables social insects to integrate external environmental stimuli, including social cues, with internal physiology and behavior. We argue that examining these pathways as an integrated regulatory circuit and exploring how the regulatory architecture of this circuit evolves alongside eusociality can open the door to understanding the origin of the complex life histories and behaviors of this group.
Collapse
Affiliation(s)
- Sarah Kocher
- Department of Ecology and Evolutionary Biology, Lewis-Sigler Institute for Integrative Biology, and Howard Hughes Medical Institute, Princeton University, Princeton, New Jersey, USA;
| | - Callum Kingwell
- Smithsonian Tropical Research Institute, Ancon, Panama
- Department of Ecology and Evolutionary Biology, Lewis-Sigler Institute for Integrative Biology, and Howard Hughes Medical Institute, Princeton University, Princeton, New Jersey, USA;
| |
Collapse
|
11
|
Liu X, Jiang Z, Sun W, Lu J, He J, Wang Y, Li F, Li B, Wei J. Larval development of a parasitoid depends on host ecdysteroids. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2024; 174:104195. [PMID: 39419290 DOI: 10.1016/j.ibmb.2024.104195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
Parasitoids often exhibit high flexibility in their development depending on stages of their host at the parasitism, yet little is known about the mechanism underlying such flexibility. In the study, we evaluated the larval development time of the parasitoid Exorista sorbillans (Diptera: Tachinidae) on the lepidopteran model insect Bombyx mori (Lepidoptera: Bombycidae). We found that the development duration of E. sorbillans larvae parasitizing on the late-developmental silkworms was significantly shorter than that of the larvae parasitizing on the early-developmental hosts. Intriguingly, the 2nd-3rd instar molting of parasitoid always occurred when the ecdysteroid titers in the host were increased to higher levels. Furthermore, inhibiting the release of ecdysteroids to parasitic abdomen by thorax-abdomen ligation of the host only repressed the 2nd-instar growth and molting of E. sorbillans larvae, but had no effect on their pupation. Meanwhile, the ecdysone synthesis and 20-hydroxyecdysone (20 E) signaling in larval parasitoids were impeded after ligation treatment. Moreover, exogenous 20 E application could largely rescue the defect in 2nd instar growth and molting through stimulating ecdysone synthesis and signaling in E. sorbillans. Our results indicate that the parasitoid requires the host ecdysteroids to stimulate 20 E signaling and the subsequent 2nd-instar growth and molting. These findings will improve our understanding of the host utilization strategies of parasitoids, and contribute to the development of in vitro rearing procedures of tachinid parasitoids for biological control.
Collapse
Affiliation(s)
- Xinyi Liu
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China; Sericulture Institute of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Zhe Jiang
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Wei Sun
- College of Life Sciences, Chongqing University, Chongqing, 215123, China
| | - Jialei Lu
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Jiaru He
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yijie Wang
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Fanchi Li
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China; Sericulture Institute of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Bing Li
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China; Sericulture Institute of Soochow University, Suzhou, Jiangsu, 215123, China.
| | - Jing Wei
- School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, China; Sericulture Institute of Soochow University, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
12
|
Lafont R, Dinan L. Insect Sterols and Steroids. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024. [PMID: 39384701 DOI: 10.1007/5584_2024_823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/11/2024]
Abstract
Insects are incapable of biosynthesising sterols de novo so they need to obtain them from their diets or, in certain cases, from symbiotic microorganisms. Sterols serve a structural role in cellular membranes and act as precursors for signalling molecules and defence compounds. Many phytophagous insects dealkylate phytosterols to yield primarily cholesterol, which is also the main sterol that carnivorous and omnivorous insects obtain in their diets. Some phytophagous species have secondarily lost the capacity to dealkylate and consequently use phytosterols for structural and functional roles. The polyhydroxylated steroid hormones of insects, the ecdysteroids, are derived from cholesterol (or phytosterols in non-dealkylating phytophagous species) and regulate many crucial aspects of insect development and reproduction by means of precisely regulated titres resulting from controlled synthesis, storage and further metabolism/excretion. Ecdysteroids differ significantly from vertebrate steroid hormones in their chemical, biochemical and biological properties. Defensive steroids (cardenolides, bufadienolides, cucurbitacins and ecdysteroids) can be accumulated from host plants or biosynthesised within the insect, depending on species, stored in significant amounts in the insect and released when it is attacked. Other allelochemical steroids serve as pheromones. Vertebrate-type steroids have also been conclusively identified from insect sources, but debate continues about their significance. Side chain dealkylation of phytosterols, ecdysteroid metabolism and ecdysteroid mode of action are targets of potential insect control strategies.
Collapse
Affiliation(s)
- René Lafont
- BIOSIPE, Sorbonne Université, Paris, France.
| | | |
Collapse
|
13
|
Ohhara Y, Blick M, Park D, Yoon SE, Kim YJ, Pankratz MJ, O’Connor MB, Yamanaka N. A Neuropeptide Signaling Network That Regulates Developmental Timing and Systemic Growth in Drosophila. J Comp Neurol 2024; 532:e25677. [PMID: 39415613 PMCID: PMC11488662 DOI: 10.1002/cne.25677] [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: 06/21/2024] [Revised: 08/21/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024]
Abstract
Animals sense chemical cues such as nutritious and noxious stimuli through the chemosensory system and adapt their behavior, physiology, and developmental schedule to the environment. In the Drosophila central nervous system, chemosensory interneurons that produce neuropeptides called Hugin (Hug) peptides receive signals from gustatory receptor neurons and regulate feeding behavior. Because Hug neurons project their axons to the higher brain region within the protocerebrum where dendrites of multiple neurons producing developmentally important neuropeptides are extended, it has been postulated that Hug neurons regulate development through the neuroendocrine system. In this study, we show that Hug neurons interact with a subset of protocerebrum neurons that produce prothoracicotropic hormone (PTTH) and regulate the onset of metamorphosis and systemic growth. Loss of the hug gene and silencing of Hug neurons caused a delay in larval-to-prepupal transition and an increase in final body size. Furthermore, deletion of Hug receptor-encoding genes also caused developmental delay and body size increase, and the phenotype was restored by expressing Hug receptors in PTTH-producing neurons. These results indicate that Hug neurons regulate developmental timing and body size via PTTH-producing neurons. This study provides a basis for understanding how chemosensation is converted into neuroendocrine signaling to control insect growth and development.
Collapse
Affiliation(s)
- Yuya Ohhara
- School of Food and Nutritional Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Mikkal Blick
- Department of Entomology, Institute for Integrative Genome Biology, University of California, Riverside, CA 92521, USA
| | - Donghyun Park
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sung-Eun Yoon
- Korea Drosophila Resource Center (KDRC), Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Young-Joon Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Michael J. Pankratz
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, 53115 Bonn, Germany
| | - Michael B. O’Connor
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Naoki Yamanaka
- Department of Entomology, Institute for Integrative Genome Biology, University of California, Riverside, CA 92521, USA
| |
Collapse
|
14
|
Liu W, Yan M, King-Jones K. Soul is a master control gene governing the development of the Drosophila prothoracic gland. Proc Natl Acad Sci U S A 2024; 121:e2405469121. [PMID: 39312662 PMCID: PMC11459192 DOI: 10.1073/pnas.2405469121] [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: 04/10/2024] [Accepted: 07/31/2024] [Indexed: 09/25/2024] Open
Abstract
The prothoracic gland (PG) is a major insect endocrine organ. It is the principal source of insect steroid hormones, and critical for key developmental events such as the molts, the establishment of critical weight (CW), pupation, and sexual maturation. However, little is known about the developmental processes that regulate PG morphology. In this study, we identified soul, which encodes a PG-specific basic helix-loop-helix (bHLH) transcription factor. We demonstrate that Tap, also a bHLH protein, dimerizes with Soul. Both are expressed in the developing PG. Interfering with either soul or tap function caused strikingly similar phenotypes, resulting in small and fragmented PGs, the abolishment of steroid hormone-producing gene expression, larval arrest, and a failure to undergo metamorphosis. Furthermore, both soul and tap showed expression peaks just prior to the CW checkpoint. Disrupting soul- or tap-function before, but not after, the CW checkpoint caused larval arrest, and perturbed highly similar gene cohorts, which were enriched for regulators and components of the steroid hormone biosynthesis pathway. Intriguingly, a chitin-based cuticle gene, Cpr49Ah, and a POU domain transcription factor gene, pdm3, are direct target genes of the Soul/Tap complex, and disruption of either phenocopied key aspects of soul/tap loss-of-function phenotypes. Taken together, our findings demonstrate that the Soul/Tap heterodimer resides at the top of a complex gene hierarchy that drives PG development, CW establishment, and steroid hormone production.
Collapse
Affiliation(s)
- Wen Liu
- Department of Biological Sciences, University of Alberta, Edmonton, ABT6G 2E9, Canada
| | - Minyi Yan
- Department of Biological Sciences, University of Alberta, Edmonton, ABT6G 2E9, Canada
| | - Kirst King-Jones
- Department of Biological Sciences, University of Alberta, Edmonton, ABT6G 2E9, Canada
| |
Collapse
|
15
|
Viswanatha R, Entwisle S, Hu C, Reap K, Butnaru M, Mohr SE, Perrimon N. Higher resolution pooled genome-wide CRISPR knockout screening in Drosophila cells using Integration and Anti-CRISPR (IntAC). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613976. [PMID: 39345359 PMCID: PMC11429967 DOI: 10.1101/2024.09.19.613976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
CRISPR screens enable systematic, scalable genotype-to-phenotype mapping. We previously developed a pooled CRISPR screening method for Drosophila melanogaster and mosquito cell lines using plasmid transfection and site-specific integration to introduce single guide (sgRNA) libraries, followed by PCR and sequencing of integrated sgRNAs. While effective, the method relies on early constitutive Cas9 activity that potentially can lead to discrepancies between genome edits and sgRNAs detected by PCR, reducing screen accuracy. To address this issue, we introduce a new method to co-transfect a plasmid expressing the anti-CRISPR protein AcrIIa4 to suppress Cas9 activity during early sgRNA expression, which we term "IntAC" (integrase with anti-CRISPR). IntAC allowed us to construct a new CRISPR screening approach driven by the high strength dU6:3 promoter. This new library dramatically improved precision-recall of fitness genes across the genome, retrieving 90-95% of essential gene groups within 5% error, allowing us to generate the most comprehensive list of cell fitness genes yet assembled for Drosophila. Our analysis determined that elevated sgRNA levels, made permissible by the IntAC approach, drove much of the improvement. The Drosophila fitness genes show strong correlation with human fitness genes and underscore the effects of paralogs on gene essentiality. We further demonstrate that IntAC combined with a targeted sgRNA sub-library enabled precise positive selection of a transporter under solute overload. IntAC represents a straightforward enhancement to existing Drosophila CRISPR screening methods, dramatically increasing accuracy, and might also be broadly applicable to virus-free CRISPR screens in other cell types, including mosquito, lepidopteran, tick, and mammalian cells.
Collapse
Affiliation(s)
| | - Samuel Entwisle
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Claire Hu
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Kelly Reap
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Matthew Butnaru
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Stephanie E Mohr
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Howard Hughes Medical Institute, Boston, MA 02115
| |
Collapse
|
16
|
Ryniawec JM, Amoiroglou A, Rogers GC. Generating CRISPR-edited clonal lines of cultured Drosophila S2 cells. Biol Methods Protoc 2024; 9:bpae059. [PMID: 39206452 PMCID: PMC11357795 DOI: 10.1093/biomethods/bpae059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/26/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
CRISPR/Cas9 genome editing is a pervasive research tool due to its relative ease of use. However, some systems are not amenable to generating edited clones due to genomic complexity and/or difficulty in establishing clonal lines. For example, Drosophila Schneider 2 (S2) cells possess a segmental aneuploid genome and are challenging to single-cell select. Here, we describe a streamlined CRISPR/Cas9 methodology for knock-in and knock-out experiments in S2 cells, whereby an antibiotic resistance gene is inserted in-frame with the coding region of a gene-of-interest. By using selectable markers, we have improved the ease and efficiency for the positive selection of null cells using antibiotic selection in feeder layers followed by cell expansion to generate clonal lines. Using this method, we generated the first acentrosomal S2 cell lines by knocking-out centriole genes Polo-like Kinase 4/Plk4 or Ana2 as proof of concept. These strategies for generating gene-edited clonal lines will add to the collection of CRISPR tools available for cultured Drosophila cells by making CRISPR more practical and therefore improving gene function studies.
Collapse
Affiliation(s)
- John M Ryniawec
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, United States
| | - Anastasia Amoiroglou
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, United States
| | - Gregory C Rogers
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, United States
| |
Collapse
|
17
|
McPherson WK, Van Gorder EE, Hilovsky DL, Jamali LA, Keliinui CN, Suzawa M, Bland ML. Synchronizing Drosophila larvae with the salivary gland reporter Sgs3-GFP for discovery of phenotypes in the late third instar stage. Dev Biol 2024; 512:35-43. [PMID: 38710381 DOI: 10.1016/j.ydbio.2024.05.002] [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: 01/26/2024] [Revised: 04/23/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024]
Abstract
The larval stage of the Drosophila melanogaster life cycle is characterized by rapid growth and nutrient storage that occur over three instar stages separated by molts. In the third instar, the steroid hormone ecdysone drives key developmental processes and behaviors that occur in a temporally-controlled sequence and prepare the animal to undergo metamorphosis. Accurately staging Drosophila larvae within the final third instar is critical due to the rapid developmental progress at this stage, but it is challenging because the rate of development varies widely across a population of animals even if eggs are laid within a short period of time. Moreover, many methods to stage third instar larvae are cumbersome, and inherent variability in the rate of development confounds some of these approaches. Here we demonstrate the usefulness of the Sgs3-GFP transgene, a fusion of the Salivary gland secretion 3 (Sgs3) and GFP proteins, for staging third instar larvae. Sgs3-GFP is expressed in the salivary glands in an ecdysone-dependent manner from the midpoint of the third instar, and its expression pattern changes reproducibly as larvae progress through the third instar. We show that Sgs3-GFP can easily be incorporated into experiments, that it allows collection of developmentally-equivalent individuals from a mixed population of larvae, and that its use enables precise assessment of changing levels of hormones, metabolites, and gene expression during the second half of the third instar.
Collapse
Affiliation(s)
- W Kyle McPherson
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908-0875, USA
| | - Elizabeth E Van Gorder
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908-0875, USA
| | - Dalton L Hilovsky
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908-0875, USA
| | - Leila A Jamali
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908-0875, USA
| | - Cami N Keliinui
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908-0875, USA
| | - Miyuki Suzawa
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908-0875, USA
| | - Michelle L Bland
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 22908-0875, USA.
| |
Collapse
|
18
|
Grmai L, Jimenez E, Baxter E, Doren MV. Steroid signaling controls sex-specific development in an invertebrate. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.22.573099. [PMID: 38187640 PMCID: PMC10769319 DOI: 10.1101/2023.12.22.573099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
In vertebrate sexual development, two important steroid hormones, testosterone and estrogen, regulate the sex-specific development of many tissues. In contrast, invertebrates utilize a single steroid hormone, ecdysone, to regulate developmental timing in both sexes. However, here we show that in Drosophila melanogaster, sex-specific ecdysone (E) signaling controls important aspects of gonad sexual dimorphism. Rather than being regulated at the level of hormone production, hormone activity is regulated cell-autonomously through sex-specific hormone reception. Ecdysone receptor (EcR) expression is restricted to the developing ovary and is repressed in the testis at a time when ecdysone initiates ovary morphogenesis. Interestingly, EcR expression is regulated downstream of the sex determination factor Doublesex (Dsx), the founding member of the Dsx/Mab3 Related Transcription Factor (DMRT) family that regulates gonad development in all animals. E signaling is required for normal ovary development1,2, and ectopic activation of E signaling in the testis antagonized stem cell niche identity and feminized somatic support cells, which were transformed into follicle-like cells. This work demonstrates that invertebrates can also use steroid hormone signaling to control sex-specific development. Further, it may help explain recent work showing that vertebrate sexual development is surprisingly cell-autonomous. For example, chickens utilize testosterone and estrogen to control sex-specific development, but when they have a mixture of cells with male and female genotypes, the male cells develop as male and the female cells develop as female despite exposure to the same circulating hormones3. Sex-specific regulation of steroid hormone signaling may well underly such cell-autonomous sexual fate choices in vertebrates as it does in Drosophila.
Collapse
Affiliation(s)
- Lydia Grmai
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Erin Jimenez
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ellen Baxter
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Mark Van Doren
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
19
|
Leyria J. Endocrine factors modulating vitellogenesis and oogenesis in insects: An update. Mol Cell Endocrinol 2024; 587:112211. [PMID: 38494046 DOI: 10.1016/j.mce.2024.112211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 02/26/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
The endocrine system plays a pivotal role in shaping the mechanisms that ensure successful reproduction. With over a million known insect species, understanding the endocrine control of reproduction has become increasingly complex. Some of the key players include the classic insect lipid hormones juvenile hormone (JH) and ecdysteroids, and neuropeptides such as insulin-like peptides (ILPs). Individual endocrine factors not only modulate their own target tissue but also play crucial roles in crosstalk among themselves, ensuring successful vitellogenesis and oogenesis. Recent advances in omics, gene silencing, and genome editing approaches have accelerated research, offering both fundamental insights and practical applications for studying in-depth endocrine signaling pathways. This review provides an updated and integrated view of endocrine factors modulating vitellogenesis and oogenesis in insect females.
Collapse
Affiliation(s)
- Jimena Leyria
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, Canada.
| |
Collapse
|
20
|
Ying W, Wang Y, Wei H, Luo Y, Ma Q, Zhu H, Janssens H, Vukašinović N, Kvasnica M, Winne JM, Gao Y, Tan S, Friml J, Liu X, Russinova E, Sun L. Structure and function of the Arabidopsis ABC transporter ABCB19 in brassinosteroid export. Science 2024; 383:eadj4591. [PMID: 38513023 DOI: 10.1126/science.adj4591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 02/02/2024] [Indexed: 03/23/2024]
Abstract
Brassinosteroids are steroidal phytohormones that regulate plant development and physiology, including adaptation to environmental stresses. Brassinosteroids are synthesized in the cell interior but bind receptors at the cell surface, necessitating a yet to be identified export mechanism. Here, we show that a member of the ATP-binding cassette (ABC) transporter superfamily, ABCB19, functions as a brassinosteroid exporter. We present its structure in both the substrate-unbound and the brassinosteroid-bound states. Bioactive brassinosteroids are potent activators of ABCB19 ATP hydrolysis activity, and transport assays showed that ABCB19 transports brassinosteroids. In Arabidopsis thaliana, ABCB19 and its close homolog, ABCB1, positively regulate brassinosteroid responses. Our results uncover an elusive export mechanism for bioactive brassinosteroids that is tightly coordinated with brassinosteroid signaling.
Collapse
Affiliation(s)
- Wei Ying
- Department of Neurology of The First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Yaowei Wang
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
- Center for Plant Systems Biology, VIB, 9052 Ghent, Belgium
| | - Hong Wei
- Department of Neurology of The First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Yongming Luo
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
- Center for Plant Systems Biology, VIB, 9052 Ghent, Belgium
| | - Qian Ma
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
- Center for Plant Systems Biology, VIB, 9052 Ghent, Belgium
| | - Heyuan Zhu
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
- Center for Plant Systems Biology, VIB, 9052 Ghent, Belgium
| | - Hilde Janssens
- Department of Organic and Macromolecular Chemistry, Ghent University, 9000 Ghent, Belgium
| | - Nemanja Vukašinović
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
- Center for Plant Systems Biology, VIB, 9052 Ghent, Belgium
| | - Miroslav Kvasnica
- Laboratory of Growth Regulators, Institute of Experimental Botany, The Czech Academy of Sciences and Palacký University, 77900 Olomouc, Czech Republic
| | - Johan M Winne
- Department of Organic and Macromolecular Chemistry, Ghent University, 9000 Ghent, Belgium
| | - Yongxiang Gao
- Department of Neurology of The First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Shutang Tan
- Department of Neurology of The First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Jiří Friml
- Institute of Science and Technology Austria (ISTA), 3400 Klosterneuburg, Austria
| | - Xin Liu
- Department of Neurology of The First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Eugenia Russinova
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
- Center for Plant Systems Biology, VIB, 9052 Ghent, Belgium
| | - Linfeng Sun
- Department of Neurology of The First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| |
Collapse
|
21
|
Lv J, He QH, Shi P, Zhou F, Zhang TT, Zhang M, Zhang XY. RNAi-mediated silencing of the neverland gene inhibits molting in the migratory locust, Locusta migratoria. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 200:105845. [PMID: 38582577 DOI: 10.1016/j.pestbp.2024.105845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 04/08/2024]
Abstract
7-dehydrocholesterol (7-DHC) is a key intermediate product used for biosynthesis of molting hormone. This is achieved through a series of hydroxylation reactions catalyzed by the Halloween family of cytochrome P450s. Neverland is an enzyme catalyzes the first reaction of the ecdysteroidogenic pathway, which converts dietary cholesterol into 7-DHC. However, research on the physiological function of neverland in orthopteran insects is lacking. In this study, neverland from Locusta migratoria (LmNvd) was cloned and analyzed. LmNvd was mainly expressed in the prothoracic gland and highly expressed on days 6 and 7 of fifth instar nymphs. RNAi-mediated silencing of LmNvd resulted in serious molting delays and abnormal phenotypes, which could be rescued by 7-DHC and 20-hydroxyecdysone supplementation. Hematoxylin and eosin staining results showed that RNAi-mediated silencing of LmNvd disturbed the molting process by both promoting the synthesis of new cuticle and suppressing the degradation of the old cuticle. Quantitative real-time PCR results suggested that the mRNA expression of E75 early gene and chitinase 5 gene decreased and that of chitin synthase 1 gene was markedly upregulated after knockdown of LmNvd. Our results suggest that LmNvd participates in the biosynthesis process of molting hormone, which is involved in regulating chitin synthesis and degradation in molting cycles.
Collapse
Affiliation(s)
- Jia Lv
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi 030006, China; Shanxi Key Laboratory of Nucleic Acid Biopesticides, China; Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China
| | - Qi-Hui He
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi 030006, China; Shanxi Key Laboratory of Nucleic Acid Biopesticides, China; Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China
| | - Peng Shi
- Shanxi Academy of Forestry and Grassland, China
| | - Feng Zhou
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Ting-Ting Zhang
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi 030006, China; Shanxi Key Laboratory of Nucleic Acid Biopesticides, China; Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China
| | - Min Zhang
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi 030006, China; Shanxi Key Laboratory of Nucleic Acid Biopesticides, China; Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China.
| | - Xue-Yao Zhang
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi 030006, China; Shanxi Key Laboratory of Nucleic Acid Biopesticides, China; Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, China.
| |
Collapse
|
22
|
Sun H, Chen J, Wang R, Liu D, Zhang N, Zhang T, Jia L, Ma S, Xia Q. Genome-wide CRISPR screening reveals key genes and pathways associated with 20-hydroxyecdysone signal transduction in the silkworm (Bombyx mori). INSECT SCIENCE 2024; 31:47-58. [PMID: 37368860 DOI: 10.1111/1744-7917.13237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 06/29/2023]
Abstract
Metamorphosis is a complex developmental process involving multiple pathways and a large number of genes that are regulated by juvenile hormone (JH) and 20-hydroxyecdysone (20E). Despite important progress in understanding various aspects of silkworm biology, the hormone signaling pathway in the silkworm remains poorly understood. Genome-wide screening using clustered regularly interspaced short palindromic repeats (CRISPR) / CRISPR-associated protein 9 (Cas9)-based libraries has recently emerged as a novel method for analyzing genome function, enabling further research into essential genes, drug targets, and virus-host interaction. Previously, we constructed a genome-wide CRISPR/Cas9-based library of the silkworm (Bombyx mori) and successfully revealed the genes involved in biotic or abiotic stress factor responses. In this study, we used our silkworm CRISPR library and large-scale genome-wide screening to analyze the key genes in the silkworm 20E signaling pathway and their mechanisms of action. Functional annotation showed that 20E regulates key proteins in processes that mainly occur in the cytoplasm and nucleus. Pathway enrichment analysis showed that 20E can activate phosphorylation and may affect innate immunity, interfere with intracellular nutrition and energy metabolism, and eventually cause cell apoptosis. The screening results were experimentally validated by generating cells with knockout alleles of the relevant genes, which had increased tolerance to 20E. Our findings provide a panoramic overview of signaling in response to 20E in the silkworm, underscoring the utility of genome-wide CRISPR mutant libraries in deciphering hormone signaling pathways and the mechanisms that regulate metamorphosis in insects.
Collapse
Affiliation(s)
- Hao Sun
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
- Biological Science Research Center, Southwest University, Chongqing, China
| | - Jingya Chen
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
- Biological Science Research Center, Southwest University, Chongqing, China
| | - Ruolin Wang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
- Biological Science Research Center, Southwest University, Chongqing, China
| | - Dan Liu
- Biological Science Research Center, Southwest University, Chongqing, China
| | - Na Zhang
- Biological Science Research Center, Southwest University, Chongqing, China
| | - Tong Zhang
- Biological Science Research Center, Southwest University, Chongqing, China
| | - Ling Jia
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
- Biological Science Research Center, Southwest University, Chongqing, China
| | - Sanyuan Ma
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
- Biological Science Research Center, Southwest University, Chongqing, China
| | - Qingyou Xia
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
- Biological Science Research Center, Southwest University, Chongqing, China
| |
Collapse
|
23
|
Morrow H, Mirth CK. Timing Drosophila development through steroid hormone action. Curr Opin Genet Dev 2024; 84:102148. [PMID: 38271845 DOI: 10.1016/j.gde.2023.102148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/15/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024]
Abstract
Specifically timed pulses of the moulting hormone ecdysone are necessary for developmental progression in insects, guiding development through important milestones such as larval moults, pupation and metamorphosis. It also coordinates the acquisition of cell identities, known as cell patterning, and growth in a tissue-specific manner. In the absence of ecdysone, the ecdysone receptor heterodimer Ecdysone Receptor and Ultraspiracle represses expression of target primary response genes, which become de-repressed as the ecdysone titre rises. However, ecdysone signalling elicits both repressive and activating responses in a temporal and tissue-specific manner. To understand how ecdysone achieves such specificity, this review explores the layers of gene regulation involved in stage-appropriate ecdysone responses in Drosophila fruit flies.
Collapse
Affiliation(s)
- Hannah Morrow
- School of Biological Sciences, Monash University, Clayton, Victoria 3000, Australia.
| | - Christen K Mirth
- School of Biological Sciences, Monash University, Clayton, Victoria 3000, Australia
| |
Collapse
|
24
|
Zhang S, Wu S, Yao R, Wei X, Ohlstein B, Guo Z. Eclosion muscles secrete ecdysteroids to initiate asymmetric intestinal stem cell division in Drosophila. Dev Cell 2024; 59:125-140.e12. [PMID: 38096823 DOI: 10.1016/j.devcel.2023.11.016] [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: 06/29/2023] [Revised: 10/05/2023] [Accepted: 11/14/2023] [Indexed: 01/11/2024]
Abstract
During organ development, tissue stem cells first expand via symmetric divisions and then switch to asymmetric divisions to minimize the time to obtain a mature tissue. In the Drosophila midgut, intestinal stem cells switch their divisions from symmetric to asymmetric at midpupal development to produce enteroendocrine cells. However, the signals that initiate this switch are unknown. Here, we identify the signal as ecdysteroids. In the presence of ecdysone, EcR and Usp promote the expression of E93 to suppress Br expression, resulting in asymmetric divisions. Surprisingly, the primary source of pupal ecdysone is not from the prothoracic gland but from dorsal internal oblique muscles (DIOMs), a group of transient skeletal muscles that are required for eclosion. Genetic analysis shows that DIOMs secrete ecdysteroids during mTOR-mediated muscle remodeling. Our findings identify sequential endocrine and mechanical roles for skeletal muscle, which ensure the timely asymmetric divisions of intestinal stem cells.
Collapse
Affiliation(s)
- Song Zhang
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Song Wu
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ruining Yao
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xueying Wei
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Benjamin Ohlstein
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zheng Guo
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| |
Collapse
|
25
|
Li Z, Wang L, Yi T, Liu D, Li G, Jin DC. The nuclear receptor gene E75 plays a key role in regulating the molting process of the spider mite, Tetranychus urticae. EXPERIMENTAL & APPLIED ACAROLOGY 2024; 92:1-11. [PMID: 38112881 DOI: 10.1007/s10493-023-00868-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 11/13/2023] [Indexed: 12/21/2023]
Abstract
The nuclear receptor gene Ecdysone-induced protein 75 (E75), as the component of ecdysone response genes in the ecdysone signaling pathway, has important regulatory function for insect molting. However, the regulatory function of E75 during the molting process of spider mites is not yet clear. In this study, the expression pattern of E75 in the molting process of the spider mite Tetranychus urticae was analyzed. The results showed that there was a peak at 8 h post-molting, followed by a decline 8 h after entering each respective quiescent stage across various developmental stages. During the deutonymph stage, the expression dynamics of E75, observed at 4-h intervals, indicated that the transcript levels of TuE75 peaked at 24 h, coinciding with the onset of molting in the mites. To investigate the function of TuE75 during the molting process, silencing TuE75 through dsRNA injection into deutonymph mites at the age of 8 h yielded a notable outcome: 78% of the deutonymph mites were unable to progress to the adult stage. Among these phenotypic mites, 37% were incapable of transitioning into the quiescent state and eventually succumbed after a certain period. An additional 41% of the mites successfully entered the quiescent state but encountered difficulties in shedding the old epidermis, leading to eventual mortality. In summary, these results suggested that TuE75 plays a key role in the molting process of T. urticae.
Collapse
Affiliation(s)
- Zhuo Li
- Institute of Entomology, Guizhou University, Guiyang, 550025, China
- Guizhou Provincial Key Laboratory for Agricultural Pest Management of the Mountainous Region, Guiyang, 550025, China
| | - Liang Wang
- Institute of Entomology, Guizhou University, Guiyang, 550025, China
- Guizhou Provincial Key Laboratory for Agricultural Pest Management of the Mountainous Region, Guiyang, 550025, China
| | - Tianci Yi
- Institute of Entomology, Guizhou University, Guiyang, 550025, China
- Guizhou Provincial Key Laboratory for Agricultural Pest Management of the Mountainous Region, Guiyang, 550025, China
| | - Dongdong Liu
- Institute of Entomology, Guizhou University, Guiyang, 550025, China
- Guizhou Provincial Key Laboratory for Agricultural Pest Management of the Mountainous Region, Guiyang, 550025, China
| | - Gang Li
- Institute of Entomology, Guizhou University, Guiyang, 550025, China.
- Guizhou Provincial Key Laboratory for Agricultural Pest Management of the Mountainous Region, Guiyang, 550025, China.
| | - Dao-Chao Jin
- Institute of Entomology, Guizhou University, Guiyang, 550025, China.
- Guizhou Provincial Key Laboratory for Agricultural Pest Management of the Mountainous Region, Guiyang, 550025, China.
| |
Collapse
|
26
|
Perez-Mockus G, Cocconi L, Alexandre C, Aerne B, Salbreux G, Vincent JP. The Drosophila ecdysone receptor promotes or suppresses proliferation according to ligand level. Dev Cell 2023; 58:2128-2139.e4. [PMID: 37769663 PMCID: PMC7615657 DOI: 10.1016/j.devcel.2023.08.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 07/20/2023] [Accepted: 08/30/2023] [Indexed: 10/03/2023]
Abstract
The steroid hormone 20-hydroxy-ecdysone (20E) promotes proliferation in Drosophila wing precursors at low titer but triggers proliferation arrest at high doses. Remarkably, wing precursors proliferate normally in the complete absence of the 20E receptor, suggesting that low-level 20E promotes proliferation by overriding the default anti-proliferative activity of the receptor. By contrast, 20E needs its receptor to arrest proliferation. Dose-response RNA sequencing (RNA-seq) analysis of ex vivo cultured wing precursors identifies genes that are quantitatively activated by 20E across the physiological range, likely comprising positive modulators of proliferation and other genes that are only activated at high doses. We suggest that some of these "high-threshold" genes dominantly suppress the activity of the pro-proliferation genes. We then show mathematically and with synthetic reporters that combinations of basic regulatory elements can recapitulate the behavior of both types of target genes. Thus, a relatively simple genetic circuit can account for the bimodal activity of this hormone.
Collapse
Affiliation(s)
| | - Luca Cocconi
- The Francis Crick Institute, London NW1 1AT, UK.
| | | | | | - Guillaume Salbreux
- The Francis Crick Institute, London NW1 1AT, UK; Department of Genetics and Evolution, University of Geneva, Quai Ernest-Ansermet 30, 1205 Geneva, Switzerland.
| | | |
Collapse
|
27
|
Wang C, Cui C, Xu P, Zhu L, Xue H, Chen B, Jiang P. Targeting PDK2 rescues stress-induced impaired brain energy metabolism. Mol Psychiatry 2023; 28:4138-4150. [PMID: 37188779 DOI: 10.1038/s41380-023-02098-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/17/2023]
Abstract
Depression is a mental illness frequently accompanied by disordered energy metabolism. A dysregulated hypothalamus pituitary adrenal axis response with aberrant glucocorticoids (GCs) release is often observed in patients with depression. However, the associated etiology between GCs and brain energy metabolism remains poorly understood. Here, using metabolomic analysis, we showed that the tricarboxylic acid (TCA) cycle was inhibited in chronic social defeat stress (CSDS)-exposed mice and patients with first-episode depression. Decreased mitochondrial oxidative phosphorylation was concomitant with the impairment of the TCA cycle. In parallel, the activity of pyruvate dehydrogenase (PDH), the gatekeeper of mitochondrial TCA flux, was suppressed, which is associated with the CSDS-induced neuronal pyruvate dehydrogenase kinase 2 (PDK2) expression and consequently enhanced PDH phosphorylation. Considering the well-acknowledged role of GCs in energy metabolism, we further demonstrated that glucocorticoid receptors (GR) stimulated PDK2 expression by directly binding to its promoter region. Meanwhile, silencing PDK2 abrogated glucocorticoid-induced PDH inhibition, restored the neuronal oxidative phosphorylation, and improved the flux of isotope-labeled carbon (U-13C] glucose) into the TCA cycle. Additionally, in vivo, pharmacological inhibition and neuron-specific silencing of GR or PDK2 restored CSDS-induced PDH phosphorylation and exerted antidepressant activities against chronic stress exposure. Taken together, our findings reveal a novel mechanism of depression manifestation, whereby elevated GCs levels regulate PDK2 transcription via GR, thereby impairing brain energy metabolism and contributing to the onset of this condition.
Collapse
Affiliation(s)
- Changshui Wang
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272000, China
| | - Changmeng Cui
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272000, China
| | - Pengfei Xu
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
| | - Li Zhu
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China
| | - Hongjia Xue
- Faculty of Science and Engineering, University of Nottingham Ningbo China, Ningbo, 315100, China
| | - Beibei Chen
- ADFA School of Science, University of New South Wales, Canberra, ACT, Australia
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, 272000, China.
| |
Collapse
|
28
|
Kretschmer S, Perry N, Zhang Y, Kortemme T. Multi-input Drug-Controlled Switches of Mammalian Gene Expression Based on Engineered Nuclear Hormone Receptors. ACS Synth Biol 2023; 12:1924-1934. [PMID: 37315218 PMCID: PMC10367131 DOI: 10.1021/acssynbio.3c00080] [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: 02/02/2023] [Indexed: 06/16/2023]
Abstract
Protein-based switches that respond to different inputs to regulate cellular outputs, such as gene expression, are central to synthetic biology. For increased controllability, multi-input switches that integrate several cooperating and competing signals for the regulation of a shared output are of particular interest. The nuclear hormone receptor (NHR) superfamily offers promising starting points for engineering multi-input-controlled responses to clinically approved drugs. Starting from the VgEcR/RXR pair, we demonstrate that novel (multi)drug regulation can be achieved by exchange of the ecdysone receptor (EcR) ligand binding domain (LBD) for other human NHR-derived LBDs. For responses activated to saturation by an agonist for the first LBD, we show that outputs can be boosted by an agonist targeting the second LBD. In combination with an antagonist, output levels are tunable by up to three simultaneously present small-molecule drugs. Such high-level control validates NHRs as a versatile, engineerable platform for programming multidrug-controlled responses.
Collapse
Affiliation(s)
- Simon Kretschmer
- Department
of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California 94158, United States
- California
Quantitative Biosciences Institute (QBI) at UCSF, San Francisco, California 94158, United States
| | - Nicholas Perry
- Department
of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California 94158, United States
- California
Quantitative Biosciences Institute (QBI) at UCSF, San Francisco, California 94158, United States
- University
of California, Berkeley—University of California, San Francisco
Joint Graduate Program in Bioengineering, San Francisco, California 94158, United States
| | - Yang Zhang
- Department
of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California 94158, United States
- California
Quantitative Biosciences Institute (QBI) at UCSF, San Francisco, California 94158, United States
| | - Tanja Kortemme
- Department
of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California 94158, United States
- California
Quantitative Biosciences Institute (QBI) at UCSF, San Francisco, California 94158, United States
- University
of California, Berkeley—University of California, San Francisco
Joint Graduate Program in Bioengineering, San Francisco, California 94158, United States
| |
Collapse
|
29
|
Temporal control of neuronal wiring. Semin Cell Dev Biol 2023; 142:81-90. [PMID: 35644877 DOI: 10.1016/j.semcdb.2022.05.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 12/22/2022]
Abstract
Wiring an animal brain is a complex process involving a staggering number of cell-types born at different times and locations in the developing brain. Incorporation of these cells into precise circuits with high fidelity is critical for animal survival and behavior. Assembly of neuronal circuits is heavily dependent upon proper timing of wiring programs, requiring neurons to express specific sets of genes (sometimes transiently) at the right time in development. While cell-type specificity of genetic programs regulating wiring has been studied in detail, mechanisms regulating proper timing and coordination of these programs across cell-types are only just beginning to emerge. In this review, we discuss some temporal regulators of wiring programs and how their activity is controlled over time and space. A common feature emerges from these temporal regulators - they are induced by cell-extrinsic cues and control transcription factors capable of regulating a highly cell-type specific set of target genes. Target specificity in these contexts comes from cell-type specific transcription factors. We propose that the spatiotemporal specificity of wiring programs is controlled by the combinatorial activity of temporal programs and cell-type specific transcription factors. Going forward, a better understanding of temporal regulators will be key to understanding the mechanisms underlying brain wiring, and will be critical for the development of in vitro models like brain organoids.
Collapse
|
30
|
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
|
31
|
Wellmeyer B, Böhringer AC, Rösner J, Merzendorfer H. Analyses of ecdysteroid transporters in the fat body of Tribolium castaneum. INSECT MOLECULAR BIOLOGY 2023. [PMID: 36892191 DOI: 10.1111/imb.12839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/06/2023] [Indexed: 06/18/2023]
Abstract
The control of insect moulting and metamorphosis involves ecdysteroids that orchestrate the execution of developmental genetic programs by binding to dimeric hormone receptors consisting of the ecdysone receptor (EcR) and ultraspiracle (USP). In insects, the main ecdysteroids comprise ecdysone (E), which is synthesized in the prothoracic gland and secreted into the haemolymph, and 20-hydroxyecdysone (20E), which is considered the active form by binding to the nuclear receptor of the target cell. While biosynthesis of ecdysteroids has been studied in detail in different insects, the transport systems involved in guiding these steroid hormones across cellular membranes have just recently begun to be studied. By analysing RNAi phenotypes in the red flour beetle, Tribolium castaneum, we have identified three transporter genes, TcABCG-8A, TcABCG-4D and TcOATP4-C1, whose silencing results in phenotypes similar to that observed when the ecdysone receptor gene TcEcRA is silenced, that is, abortive moulting and abnormal development of adult compound eyes during the larval stage. The genes of all three transporters are expressed at higher levels in the larval fat body of T. castaneum. We analysed potential functions of these transporters by combining RNAi and mass spectrometry. However, the analysis of gene functions is challenged by mutual RNAi effects indicating interdependent gene regulation. Based on our findings, we propose that TcABCG-8A, TcABCG-4D and TcOATP4-C1 participate in the ecdysteroid transport in fat body cells, which are involved in E → 20E conversion catalysed by the P450 enzyme TcShade.
Collapse
Affiliation(s)
- Benedikt Wellmeyer
- Department of Chemistry-Biology, University of Siegen, Siegen, 57068, Germany
| | | | - Janin Rösner
- Department of Chemistry-Biology, University of Siegen, Siegen, 57068, Germany
| | - Hans Merzendorfer
- Department of Chemistry-Biology, University of Siegen, Siegen, 57068, Germany
| |
Collapse
|
32
|
Truman JW, Riddiford LM. Drosophila postembryonic nervous system development: a model for the endocrine control of development. Genetics 2023; 223:iyac184. [PMID: 36645270 PMCID: PMC9991519 DOI: 10.1093/genetics/iyac184] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/13/2022] [Indexed: 01/17/2023] Open
Abstract
During postembryonic life, hormones, including ecdysteroids, juvenile hormones, insulin-like peptides, and activin/TGFβ ligands act to transform the larval nervous system into an adult version, which is a fine-grained mosaic of recycled larval neurons and adult-specific neurons. Hormones provide both instructional signals that make cells competent to undergo developmental change and timing cues to evoke these changes across the nervous system. While touching on all the above hormones, our emphasis is on the ecdysteroids, ecdysone and 20-hydroxyecdysone (20E). These are the prime movers of insect molting and metamorphosis and are involved in all phases of nervous system development, including neurogenesis, pruning, arbor outgrowth, and cell death. Ecdysteroids appear as a series of steroid peaks that coordinate the larval molts and the different phases of metamorphosis. Each peak directs a stereotyped cascade of transcription factor expression. The cascade components then direct temporal programs of effector gene expression, but the latter vary markedly according to tissue and life stage. The neurons read the ecdysteroid titer through various isoforms of the ecdysone receptor, a nuclear hormone receptor. For example, at metamorphosis the pruning of larval neurons is mediated through the B isoforms, which have strong activation functions, whereas subsequent outgrowth is mediated through the A isoform through which ecdysteroids play a permissive role to allow local tissue interactions to direct outgrowth. The major circulating ecdysteroid can also change through development. During adult development ecdysone promotes early adult patterning and differentiation while its metabolite, 20E, later evokes terminal adult differentiation.
Collapse
Affiliation(s)
- James W Truman
- Friday Harbor Laboratories, University of Washington, Friday Harbor, WA 98250, USA
- Department of Biology, University of Washington, Box 351800, Seattle, WA 98195, USA
| | - Lynn M Riddiford
- Friday Harbor Laboratories, University of Washington, Friday Harbor, WA 98250, USA
- Department of Biology, University of Washington, Box 351800, Seattle, WA 98195, USA
| |
Collapse
|
33
|
Komori Y, Takayama K, Okamoto N, Kamiya M, Koizumi W, Ihara M, Misawa D, Kamiya K, Yoshinari Y, Seike K, Kondo S, Tanimoto H, Niwa R, Sattelle DB, Matsuda K. Functional impact of subunit composition and compensation on Drosophila melanogaster nicotinic receptors-targets of neonicotinoids. PLoS Genet 2023; 19:e1010522. [PMID: 36795653 PMCID: PMC9934367 DOI: 10.1371/journal.pgen.1010522] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/11/2022] [Indexed: 02/17/2023] Open
Abstract
Neonicotinoid insecticides target insect nicotinic acetylcholine receptors (nAChRs) and their adverse effects on non-target insects are of serious concern. We recently found that cofactor TMX3 enables robust functional expression of insect nAChRs in Xenopus laevis oocytes and showed that neonicotinoids (imidacloprid, thiacloprid, and clothianidin) exhibited agonist actions on some nAChRs of the fruit fly (Drosophila melanogaster), honeybee (Apis mellifera) and bumblebee (Bombus terrestris) with more potent actions on the pollinator nAChRs. However, other subunits from the nAChR family remain to be explored. We show that the Dα3 subunit co-exists with Dα1, Dα2, Dβ1, and Dβ2 subunits in the same neurons of adult D. melanogaster, thereby expanding the possible nAChR subtypes in these cells alone from 4 to 12. The presence of Dα1 and Dα2 subunits reduced the affinity of imidacloprid, thiacloprid, and clothianidin for nAChRs expressed in Xenopus laevis oocytes, whereas the Dα3 subunit enhanced it. RNAi targeting Dα1, Dα2 or Dα3 in adults reduced expression of targeted subunits but commonly enhanced Dβ3 expression. Also, Dα1 RNAi enhanced Dα7 expression, Dα2 RNAi reduced Dα1, Dα6, and Dα7 expression and Dα3 RNAi reduced Dα1 expression while enhancing Dα2 expression, respectively. In most cases, RNAi treatment of either Dα1 or Dα2 reduced neonicotinoid toxicity in larvae, but Dα2 RNAi enhanced neonicotinoid sensitivity in adults reflecting the affinity-reducing effect of Dα2. Substituting each of Dα1, Dα2, and Dα3 subunits by Dα4 or Dβ3 subunit mostly increased neonicotinoid affinity and reduced efficacy. These results are important because they indicate that neonicotinoid actions involve the integrated activity of multiple nAChR subunit combinations and counsel caution in interpreting neonicotinoid actions simply in terms of toxicity.
Collapse
Affiliation(s)
- Yuma Komori
- Department of Applied Biological Chemistry, Faculty of Agriculture, Kindai University, Nara, Japan
| | - Koichi Takayama
- Department of Applied Biological Chemistry, Faculty of Agriculture, Kindai University, Nara, Japan
| | - Naoki Okamoto
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
| | - Masaki Kamiya
- Department of Applied Biological Chemistry, Faculty of Agriculture, Kindai University, Nara, Japan
| | - Wataru Koizumi
- Department of Applied Biological Chemistry, Faculty of Agriculture, Kindai University, Nara, Japan
| | - Makoto Ihara
- Department of Applied Biological Chemistry, Faculty of Agriculture, Kindai University, Nara, Japan
| | | | | | - Yuto Yoshinari
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
| | - Kazuki Seike
- Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Japan
| | - Shu Kondo
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Tokyo, Japan
- Invertebrate Genetics Laboratory, National Institute of Genetics, Shizuoka, Japan
| | - Hiromu Tanimoto
- Graduate School of Life Sciences, Tohoku University, Miyagi, Japan
| | - Ryusuke Niwa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
| | - David B. Sattelle
- Centre for Respiratory Biology, Division of Medicine, University College London, London, United Kingdom
| | - Kazuhiko Matsuda
- Department of Applied Biological Chemistry, Faculty of Agriculture, Kindai University, Nara, Japan
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
- Agricultural Technology and Innovation Research Institute, Kindai University, Nara, Japan
- * E-mail:
| |
Collapse
|
34
|
Okamoto N, Fujinaga D, Yamanaka N. Steroid hormone signaling: What we can learn from insect models. VITAMINS AND HORMONES 2023; 123:525-554. [PMID: 37717997 DOI: 10.1016/bs.vh.2022.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Ecdysteroids are a group of steroid hormones in arthropods with pleiotropic functions throughout their life history. Ecdysteroid research in insects has made a significant contribution to our current understanding of steroid hormone signaling in metazoans, but how far can we extrapolate our findings in insects to other systems, such as mammals? In this chapter, we compare steroid hormone signaling in insects and mammals from multiple perspectives and discuss similarities and differences between the two lineages. We also highlight a few understudied areas and remaining questions of steroid hormone biology in metazoans and propose potential future research directions.
Collapse
Affiliation(s)
- Naoki Okamoto
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Daiki Fujinaga
- Department of Entomology, University of California, Riverside, CA, United States
| | - Naoki Yamanaka
- Department of Entomology, University of California, Riverside, CA, United States.
| |
Collapse
|
35
|
Devine K, Villalobos E, Kyle CJ, Andrew R, Reynolds RM, Stimson RH, Nixon M, Walker BR. The ATP-binding cassette proteins ABCB1 and ABCC1 as modulators of glucocorticoid action. Nat Rev Endocrinol 2023; 19:112-124. [PMID: 36221036 DOI: 10.1038/s41574-022-00745-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/23/2022] [Indexed: 01/24/2023]
Abstract
Responses to hormones that act through nuclear receptors are controlled by modulating hormone concentrations not only in the circulation but also within target tissues. The role of enzymes that amplify or reduce local hormone concentrations is well established for glucocorticoid and other lipophilic hormones; moreover, transmembrane transporters have proven critical in determining tissue responses to thyroid hormones. However, there has been less consideration of the role of transmembrane transport for steroid hormones. ATP-binding cassette (ABC) proteins were first shown to influence the accumulation of glucocorticoids in cells almost three decades ago, but observations over the past 10 years suggest that differential transport propensities of both exogenous and endogenous glucocorticoids by ABCB1 and ABCC1 transporters provide a mechanism whereby different tissues are preferentially sensitive to different steroids. This Review summarizes this evidence and the new insights provided for the physiology and pharmacology of glucocorticoid action, including new approaches to glucocorticoid replacement.
Collapse
Affiliation(s)
- Kerri Devine
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Elisa Villalobos
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Catriona J Kyle
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Ruth Andrew
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Rebecca M Reynolds
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Roland H Stimson
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Mark Nixon
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Brian R Walker
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
36
|
Kretschmer S, Perry N, Zhang Y, Kortemme T. Multi-input drug-controlled switches of mammalian gene expression based on engineered nuclear hormone receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.01.526549. [PMID: 36778233 PMCID: PMC9915577 DOI: 10.1101/2023.02.01.526549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Protein-based switches that respond to different inputs to regulate cellular outputs, such as gene expression, are central to synthetic biology. For increased controllability, multi-input switches that integrate several cooperating and competing signals for the regulation of a shared output are of particular interest. The nuclear hormone receptor (NHR) superfamily offers promising starting points for engineering multi-input-controlled responses to clinically approved drugs. Starting from the VgEcR/RXR pair, we demonstrate that novel (multi-)drug regulation can be achieved by exchange of the ecdysone receptor (EcR) ligand binding domain (LBD) for other human NHR-derived LBDs. For responses activated to saturation by an agonist for the first LBD, we show that outputs can be boosted by an agonist targeting the second LBD. In combination with an antagonist, output levels are tunable by up to three simultaneously present small-molecule drugs. Such high-level control validates NHRs as a versatile, engineerable platform for programming multi-drug-controlled responses.
Collapse
Affiliation(s)
- Simon Kretschmer
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA
- California Quantitative Biosciences Institute (QBI) at UCSF, San Francisco, CA 94158, USA
| | - Nicholas Perry
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA
- California Quantitative Biosciences Institute (QBI) at UCSF, San Francisco, CA 94158, USA
- University of California, Berkeley—University of California, San Francisco Joint Graduate Program in Bioengineering, San Francisco, CA, USA
| | - Yang Zhang
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA
- California Quantitative Biosciences Institute (QBI) at UCSF, San Francisco, CA 94158, USA
| | - Tanja Kortemme
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA
- California Quantitative Biosciences Institute (QBI) at UCSF, San Francisco, CA 94158, USA
- University of California, Berkeley—University of California, San Francisco Joint Graduate Program in Bioengineering, San Francisco, CA, USA
| |
Collapse
|
37
|
Li Y, Haynes P, Zhang SL, Yue Z, Sehgal A. Ecdysone acts through cortex glia to regulate sleep in Drosophila. eLife 2023; 12:e81723. [PMID: 36719183 PMCID: PMC9928426 DOI: 10.7554/elife.81723] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 01/30/2023] [Indexed: 02/01/2023] Open
Abstract
Steroid hormones are attractive candidates for transmitting long-range signals to affect behavior. These lipid-soluble molecules derived from dietary cholesterol easily penetrate the brain and act through nuclear hormone receptors (NHRs) that function as transcription factors. To determine the extent to which NHRs affect sleep:wake cycles, we knocked down each of the 18 highly conserved NHRs found in Drosophila adults and report that the ecdysone receptor (EcR) and its direct downstream NHR Eip75B (E75) act in glia to regulate the rhythm and amount of sleep. Given that ecdysone synthesis genes have little to no expression in the fly brain, ecdysone appears to act as a long-distance signal and our data suggest that it enters the brain more at night. Anti-EcR staining localizes to the cortex glia in the brain and functional screening of glial subtypes revealed that EcR functions in adult cortex glia to affect sleep. Cortex glia are implicated in lipid metabolism, which appears to be relevant for actions of ecdysone as ecdysone treatment mobilizes lipid droplets (LDs), and knockdown of glial EcR results in more LDs. In addition, sleep-promoting effects of exogenous ecdysone are diminished in lsd-2 mutant flies, which are lean and deficient in lipid accumulation. We propose that ecdysone is a systemic secreted factor that modulates sleep by stimulating lipid metabolism in cortex glia.
Collapse
Affiliation(s)
- Yongjun Li
- Howard Hughes Medical Institute and Chronobiology and Sleep Institute, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
- Department of Biology, University of PennsylvaniaPhiladelphiaUnited States
| | - Paula Haynes
- Howard Hughes Medical Institute and Chronobiology and Sleep Institute, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
- Department of Pharmacology, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Shirley L Zhang
- Howard Hughes Medical Institute and Chronobiology and Sleep Institute, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Zhifeng Yue
- Howard Hughes Medical Institute and Chronobiology and Sleep Institute, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Amita Sehgal
- Howard Hughes Medical Institute and Chronobiology and Sleep Institute, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
38
|
Velagala V, Soundarrajan DK, Unger MF, Gazzo D, Kumar N, Li J, Zartman J. The multimodal action of G alpha q in coordinating growth and homeostasis in the Drosophila wing imaginal disc. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.08.523049. [PMID: 36711848 PMCID: PMC9881979 DOI: 10.1101/2023.01.08.523049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Background G proteins mediate cell responses to various ligands and play key roles in organ development. Dysregulation of G-proteins or Ca 2+ signaling impacts many human diseases and results in birth defects. However, the downstream effectors of specific G proteins in developmental regulatory networks are still poorly understood. Methods We employed the Gal4/UAS binary system to inhibit or overexpress Gαq in the wing disc, followed by phenotypic analysis. Immunohistochemistry and next-gen RNA sequencing identified the downstream effectors and the signaling cascades affected by the disruption of Gαq homeostasis. Results Here, we characterized how the G protein subunit Gαq tunes the size and shape of the wing in the larval and adult stages of development. Downregulation of Gαq in the wing disc reduced wing growth and delayed larval development. Gαq overexpression is sufficient to promote global Ca 2+ waves in the wing disc with a concomitant reduction in the Drosophila final wing size and a delay in pupariation. The reduced wing size phenotype is further enhanced when downregulating downstream components of the core Ca 2+ signaling toolkit, suggesting that downstream Ca 2+ signaling partially ameliorates the reduction in wing size. In contrast, Gαq -mediated pupariation delay is rescued by inhibition of IP 3 R, a key regulator of Ca 2+ signaling. This suggests that Gαq regulates developmental phenotypes through both Ca 2+ -dependent and Ca 2+ -independent mechanisms. RNA seq analysis shows that disruption of Gαq homeostasis affects nuclear hormone receptors, JAK/STAT pathway, and immune response genes. Notably, disruption of Gαq homeostasis increases expression levels of Dilp8, a key regulator of growth and pupariation timing. Conclusion Gαq activity contributes to cell size regulation and wing metamorphosis. Disruption to Gαq homeostasis in the peripheral wing disc organ delays larval development through ecdysone signaling inhibition. Overall, Gαq signaling mediates key modules of organ size regulation and epithelial homeostasis through the dual action of Ca 2+ -dependent and independent mechanisms.
Collapse
|
39
|
Scanlan JL, Robin C, Mirth CK. Rethinking the ecdysteroid source during Drosophila pupal-adult development. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2023; 152:103891. [PMID: 36481381 DOI: 10.1016/j.ibmb.2022.103891] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/30/2022] [Accepted: 12/04/2022] [Indexed: 06/17/2023]
Abstract
Ecdysteroids, typified by 20-hydroxyecdysone (20E), are essential hormones for the development, reproduction and physiology of insects and other arthropods. For over half a century, the vinegar fly Drosophila melanogaster (Ephydroidea: Diptera) has been used as a model of ecdysteroid biology. Many aspects of the biosynthesis and regulation of ecdysteroids in this species are understood at the molecular level, particularly with respect to their secretion from the prothoracic gland (PG) cells of the ring gland, widely considered the dominant biosynthetic tissue during development. Discrete pulses of 20E orchestrate transitions during the D. melanogaster life cycle, the sources of which are generally well understood, apart from the large 20E pulse at the onset of pharate adult development, which has received little recent attention. As the source of this pharate adult pulse (PAP) is a curious blind spot in Drosophila endocrinology, we evaluate published biochemical and genetic data as they pertain to three hypotheses for the source of PAP 20E: the PG; an alternative biosynthetic tissue; or the recycling of stored 20E. Based on multiple lines of evidence, we contend the PAP cannot be derived from biosynthesis, with other data consistent with D. melanogaster able to recycle ecdysteroids before and during metamorphosis. Published data also suggest the PAP is conserved across Diptera, with evidence for pupal-adult ecdysteroid recycling occurring in other cyclorrhaphan flies. Further experimental work is required to test the ecdysteroid recycling hypothesis, which would establish fundamental knowledge of the function, regulation, and evolution of metamorphic hormones in dipterans and other insects.
Collapse
Affiliation(s)
- Jack L Scanlan
- School of BioSciences, The University of Melbourne, Parkville Campus, Melbourne, Victoria, 3010, Australia.
| | - Charles Robin
- School of BioSciences, The University of Melbourne, Parkville Campus, Melbourne, Victoria, 3010, Australia
| | - Christen K Mirth
- School of Biological Sciences, Monash University, Melbourne, Victoria, 3800, Australia
| |
Collapse
|
40
|
Guirado J, Carranza-Valencia J, Morante J. Mammalian puberty: a fly perspective. FEBS J 2023; 290:359-369. [PMID: 35607827 PMCID: PMC10084137 DOI: 10.1111/febs.16534] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/02/2022] [Accepted: 05/23/2022] [Indexed: 02/05/2023]
Abstract
Mammalian puberty and Drosophila metamorphosis, despite their evolutionary distance, exhibit similar design principles and conservation of molecular components. In this Viewpoint, we review recent advances in this area and the similarities between both processes in terms of the signaling pathways and neuroendocrine circuits involved. We argue that the detection and uptake of peripheral fat by Drosophila prothoracic endocrine cells induces endomembrane remodeling and ribosomal maturation, leading to the acquisition of high biosynthetic and secretory capacity. The absence of this fat-neuroendocrine interorgan communication leads to giant, obese, non-pupating larvae. Importantly, human leptin is capable of signaling the pupariation process in Drosophila, and its expression prevents obesity and triggers maturation in mutants that do not pupate. This implies that insect metamorphosis can be used to address issues related to the biology of leptin and puberty.
Collapse
Affiliation(s)
- Juan Guirado
- Instituto de Neurociencias, Consejo Superior de Investigaciones Cientificas (CSIC) and Universidad Miguel Hernández (UMH), San Juan de Alicante, Spain
| | - Juan Carranza-Valencia
- Instituto de Neurociencias, Consejo Superior de Investigaciones Cientificas (CSIC) and Universidad Miguel Hernández (UMH), San Juan de Alicante, Spain
| | - Javier Morante
- Instituto de Neurociencias, Consejo Superior de Investigaciones Cientificas (CSIC) and Universidad Miguel Hernández (UMH), San Juan de Alicante, Spain
| |
Collapse
|
41
|
Samantsidis GR, Fotiadou M, Tzavellas S, Geibel S, Nauen R, Swevers L, Denecke S, Vontas J. Functional characterization of putative ecdysone transporters in lepidopteran pests. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2022; 151:103830. [PMID: 36064128 DOI: 10.1016/j.ibmb.2022.103830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/26/2022] [Accepted: 08/27/2022] [Indexed: 06/15/2023]
Abstract
The insect steroid hormone ecdysone plays a critical role in insect development. Several recent studies have shown that ecdysone enters cells through Organic Anion Transporting Polypeptides (OATPs) in insects such as flies and mosquitoes. However, the conservation of this mechanism across other arthropods and the role of this transporter in canonical ecdysone pathways are less well studied. Herein we functionally characterized the putative ecdysone importer (EcI) from two major agricultural moth pests: Helicoverpa armigera (cotton bollworm) and Spodoptera frugiperda (fall armyworm). Phylogenetic analysis of OATP transporters across the superphylum Ecdysozoa revealed that EcI likely appeared only at the root of the arthropod lineage. Partial disruption of EcI in S. frugiperda decreased embryo hatching rate and larval survival, suggesting that this gene is essential for development in vivo. Depletion and re-expression of EcI in the lepidoptera cell line RP-HzGUT-AW1(MG) demonstrated this protein's ability to control ecdysone mediated signaling in gene regulation, its role in ecdysone mediated cell death, and its sensitivity to rifampicin, a well-known organic anion transporter inhibitor. Overall, this work sheds light on ecdysone uptake mechanisms across insect species and broadens our knowledge of the physiological roles of OATPs in the transportation of endogenous substrates.
Collapse
Affiliation(s)
- George-Rafael Samantsidis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece; Department of Biology, University of Crete, Vassilika Vouton, 71409, Heraklion, Crete, Greece
| | - Melina Fotiadou
- Department of Biology, University of Crete, Vassilika Vouton, 71409, Heraklion, Crete, Greece
| | - Savvas Tzavellas
- Department of Biology, University of Crete, Vassilika Vouton, 71409, Heraklion, Crete, Greece
| | - Sven Geibel
- R&D Pest Control, Bayer AG, Crop Science Division, Monheim, Germany
| | - Ralf Nauen
- R&D Pest Control, Bayer AG, Crop Science Division, Monheim, Germany
| | - Luc Swevers
- Insect Molecular Genetics and Biotechnology, National Centre for Scientific Research Demokritos, Institute of Biosciences and Applications, 15310, Athens, Greece
| | - Shane Denecke
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece; Department of Pathobiology, University of Pennsylvania, Philadelphia, United States.
| | - John Vontas
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece; Pesticide Science Lab, Department of Crop Science, Agricultural University of Athens, Greece.
| |
Collapse
|
42
|
Yoon S, Shin M, Shim J. Inter-organ regulation by the brain in Drosophila development and physiology. J Neurogenet 2022:1-13. [DOI: 10.1080/01677063.2022.2137162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Sunggyu Yoon
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul, Republic of Korea
| | - Mingyu Shin
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul, Republic of Korea
| | - Jiwon Shim
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul, Republic of Korea
- Research Institute for Natural Science, Hanyang University, Seoul, Republic of Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
43
|
Ohhara Y, Yamanaka N. Internal sensory neurons regulate stage-specific growth in Drosophila. Development 2022; 149:dev200440. [PMID: 36227580 PMCID: PMC10496149 DOI: 10.1242/dev.200440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 09/22/2022] [Indexed: 09/15/2023]
Abstract
Animals control their developmental schedule in accordance with internal states and external environments. In Drosophila larvae, it is well established that nutrient status is sensed by different internal organs, which in turn regulate production of insulin-like peptides and thereby control growth. In contrast, the impact of the chemosensory system on larval development remains largely unclear. Here, we performed a genetic screen to identify gustatory receptor (Gr) neurons regulating growth and development, and found that Gr28a-expressing neurons are required for proper progression of larval growth. Gr28a is expressed in a subset of peripheral internal sensory neurons, which directly extend their axons to insulin-producing cells (IPCs) in the central nervous system. Silencing of Gr28a-expressing neurons blocked insulin-like peptide release from IPCs and suppressed larval growth during the mid-larval period. These results indicate that Gr28a-expressing neurons promote larval development by directly regulating growth-promoting endocrine signaling in a stage-specific manner.
Collapse
Affiliation(s)
- Yuya Ohhara
- School of Food and Nutritional Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Naoki Yamanaka
- Department of Entomology, Institute for Integrative Genome Biology, University of California, Riverside, Riverside, CA 92521, USA
| |
Collapse
|
44
|
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: 15] [Impact Index Per Article: 5.0] [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
|
45
|
Dalton HM, Viswanatha R, Brathwaite R, Zuno JS, Berman AR, Rushforth R, Mohr SE, Perrimon N, Chow CY. A genome-wide CRISPR screen identifies DPM1 as a modifier of DPAGT1 deficiency and ER stress. PLoS Genet 2022; 18:e1010430. [PMID: 36166480 PMCID: PMC9543880 DOI: 10.1371/journal.pgen.1010430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 10/07/2022] [Accepted: 09/14/2022] [Indexed: 11/19/2022] Open
Abstract
Partial loss-of-function mutations in glycosylation pathways underlie a set of rare diseases called Congenital Disorders of Glycosylation (CDGs). In particular, DPAGT1-CDG is caused by mutations in the gene encoding the first step in N-glycosylation, DPAGT1, and this disorder currently lacks effective therapies. To identify potential therapeutic targets for DPAGT1-CDG, we performed CRISPR knockout screens in Drosophila cells for genes associated with better survival and glycoprotein levels under DPAGT1 inhibition. We identified hundreds of candidate genes that may be of therapeutic benefit. Intriguingly, inhibition of the mannosyltransferase Dpm1, or its downstream glycosylation pathways, could rescue two in vivo models of DPAGT1 inhibition and ER stress, even though impairment of these pathways alone usually causes CDGs. While both in vivo models ostensibly cause cellular stress (through DPAGT1 inhibition or a misfolded protein), we found a novel difference in fructose metabolism that may indicate glycolysis as a modulator of DPAGT1-CDG. Our results provide new therapeutic targets for DPAGT1-CDG, include the unique finding of Dpm1-related pathways rescuing DPAGT1 inhibition, and reveal a novel interaction between fructose metabolism and ER stress.
Collapse
Affiliation(s)
- Hans M. Dalton
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Raghuvir Viswanatha
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Roderick Brathwaite
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jae Sophia Zuno
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Alexys R. Berman
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Rebekah Rushforth
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Stephanie E. Mohr
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Boston, Massachusetts, United States of America
| | - Clement Y. Chow
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
- * E-mail:
| |
Collapse
|
46
|
Gjorgoska M, Rizner TL. Integration of androgen hormones in endometrial cancer biology. Trends Endocrinol Metab 2022; 33:639-651. [PMID: 35879182 DOI: 10.1016/j.tem.2022.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/27/2022] [Accepted: 06/26/2022] [Indexed: 12/03/2022]
Abstract
Endometrial cancer (EC) is a gynecological pathology that affects the uterine inner lining. In recent years, genomic studies revealed continually evolving mutational landscapes of endometrial tumors that hold great potential for tailoring therapeutic strategies. This review aims to broaden our knowledge of EC biology by focusing on the role of androgen hormones. First, we discuss epidemiological evidence implicating androgens with EC pathogenesis and cover their biosynthesis and metabolism to bioactive 11-oxyandrogens. Next, we explore the endometrial tumor tissue and the altered microbiota as alternative sources of androgens and their 11-oxymetabolites in EC patients. Finally, we discuss the biological significance of androgens' genomic and nongenomic signaling as part of a medley of pathways ultimately deciding the fate of cells.
Collapse
Affiliation(s)
- Marija Gjorgoska
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tea Lanisnik Rizner
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|
47
|
Essential functions of mosquito ecdysone importers in development and reproduction. Proc Natl Acad Sci U S A 2022; 119:e2202932119. [PMID: 35696563 PMCID: PMC9231622 DOI: 10.1073/pnas.2202932119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Steroid hormones control sexual maturation and reproduction in insects and humans alike. The insect steroid hormone ecdysone uses a membrane transporter named Ecdysone Importer (EcI) to enter cells and promote these physiological processes, but EcI is unexpectedly missing in mosquito genomes. Using the yellow fever mosquito Aedes aegypti, here we show that mosquitoes use alternative ecdysone importers to facilitate ecdysone-dependent development and reproduction. These transporters are also present in other insects, including fruit flies, but they are dispensable for fly development and reproduction likely due to their limited expression patterns. Our results thus indicate that differential expression of steroid hormone importers enables tissue- and stage-specific hormone responses, and some importers can obtain critical physiological functions only in certain species. The primary insect steroid hormone ecdysone requires a membrane transporter to enter its target cells. Although an organic anion-transporting polypeptide (OATP) named Ecdysone Importer (EcI) serves this role in the fruit fly Drosophila melanogaster and most likely in other arthropod species, this highly conserved transporter is apparently missing in mosquitoes. Here we report three additional OATPs that facilitate cellular incorporation of ecdysone in Drosophila and the yellow fever mosquito Aedes aegypti. These additional ecdysone importers (EcI-2, -3, and -4) are dispensable for development and reproduction in Drosophila, consistent with the predominant role of EcI. In contrast, in Aedes, EcI-2 is indispensable for ecdysone-mediated development, whereas EcI-4 is critical for vitellogenesis induced by ecdysone in adult females. Altogether, our results indicate unique and essential functions of these additional ecdysone importers in mosquito development and reproduction, making them attractive molecular targets for species- and stage-specific control of ecdysone signaling in mosquitoes.
Collapse
|
48
|
Tian S, Monteiro A. A transcriptomic atlas underlying developmental plasticity of seasonal forms of Bicyclus anynana butterflies. Mol Biol Evol 2022; 39:msac126. [PMID: 35679434 PMCID: PMC9218548 DOI: 10.1093/molbev/msac126] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/10/2022] [Accepted: 05/31/2022] [Indexed: 11/12/2022] Open
Abstract
Organisms residing in regions with alternating seasons often develop different phenotypes, or forms, in each season. These forms are often adaptations to each season and result from an altered developmental response to specific environmental cues such as temperature. While multiple studies have examined form-specific gene expression profiles in a diversity of species, little is known about how environments and developmental transitions, cued by hormone pulses, alter post-transcriptional patterns. In this study, we examine how gene expression, alternative splicing, and miRNA-mediated gene silencing in Bicyclus anynana butterfly hindwing tissue, varies across two rearing temperatures at four developmental timepoints. These timepoints flank two temperature-sensitive periods that coincide with two pulses of the insect hormone 20E. Our results suggest that developmental transitions, coincident with 20E pulses, elicit a greater impact on all these transcriptomic patterns than rearing temperatures per se. More similar transcriptomic patterns are observed pre-20E pulses than those observed post-20E pulses. We also found functionally distinct sets of differentially expressed and differentially spliced genes in the seasonal forms. Furthermore, around 10% of differentially expressed genes are predicted to be direct targets of, and regulated by, differentially expressed miRNAs between the seasonal forms. Many differentially expressed genes, miRNAs, or differentially spliced genes potentially regulate eyespot size plasticity, and we validated the differential splicing pattern of one such gene, daughterless. We present a comprehensive and interactive transcriptomic atlas of the hindwing tissue of both seasonal forms of B. anynana throughout development, a model organism of seasonal plasticity.
Collapse
Affiliation(s)
- Shen Tian
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Antónia Monteiro
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| |
Collapse
|
49
|
Chen SL, Liu BT, Lee WP, Liao SB, Deng YB, Wu CL, Ho SM, Shen BX, Khoo GH, Shiu WC, Chang CH, Shih HW, Wen JK, Lan TH, Lin CC, Tsai YC, Tzeng HF, Fu TF. WAKE-mediated modulation of cVA perception via a hierarchical neuro-endocrine axis in Drosophila male-male courtship behaviour. Nat Commun 2022; 13:2518. [PMID: 35523813 PMCID: PMC9076693 DOI: 10.1038/s41467-022-30165-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 04/19/2022] [Indexed: 12/18/2022] Open
Abstract
The nervous and endocrine systems coordinate with each other to closely influence physiological and behavioural responses in animals. Here we show that WAKE (encoded by wide awake, also known as wake) modulates membrane levels of GABAA receptor Resistance to Dieldrin (Rdl), in insulin-producing cells of adult male Drosophila melanogaster. This results in changes to secretion of insulin-like peptides which is associated with changes in juvenile hormone biosynthesis in the corpus allatum, which in turn leads to a decrease in 20-hydroxyecdysone levels. A reduction in ecdysone signalling changes neural architecture and lowers the perception of the male-specific sex pheromone 11-cis-vaccenyl acetate by odorant receptor 67d olfactory neurons. These finding explain why WAKE-deficient in Drosophila elicits significant male-male courtship behaviour. The authors show that the Drosophila master regulator WAKE modulates the secretion of insulin-like peptides, triggering a decrease in 20-hydroxyecdysone levels. This lowers the perception of a male-specific sex pheromone and explains why WAKE-deficient Drosophila flies show male-male courtship behaviour.
Collapse
Affiliation(s)
- Shiu-Ling Chen
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Bo-Ting Liu
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Wang-Pao Lee
- Department of Biochemistry and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Sin-Bo Liao
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan.,Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yao-Bang Deng
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Chia-Lin Wu
- Department of Biochemistry and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan.,Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan
| | - Shuk-Man Ho
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Bing-Xian Shen
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Guan-Hock Khoo
- Department of Life Science and Life Science Center, Tunghai University, Taichung, Taiwan
| | - Wei-Chiang Shiu
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Chih-Hsuan Chang
- Department of Life Science and Life Science Center, Tunghai University, Taichung, Taiwan.,Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, Taiwan.,National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Hui-Wen Shih
- Department of Life Science and Life Science Center, Tunghai University, Taichung, Taiwan
| | - Jung-Kun Wen
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Tsuo-Hung Lan
- Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Tsaotun Psychiatric Center, Ministry of Health and Welfare, Nantou, Taiwan.,Department of Psychiatry, Taichung Veterans General Hospital, Taichung, Taiwan.,Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Chih-Chien Lin
- Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Chen Tsai
- Department of Life Science and Life Science Center, Tunghai University, Taichung, Taiwan.
| | - Huey-Fen Tzeng
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan.
| | - Tsai-Feng Fu
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan.
| |
Collapse
|
50
|
Zirin J, Bosch J, Viswanatha R, Mohr SE, Perrimon N. State-of-the-art CRISPR for in vivo and cell-based studies in Drosophila. Trends Genet 2022; 38:437-453. [PMID: 34933779 PMCID: PMC9007876 DOI: 10.1016/j.tig.2021.11.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 12/31/2022]
Abstract
For more than 100 years, the fruit fly, Drosophila melanogaster, has served as a powerful model organism for biological and biomedical research due to its many genetic and physiological similarities to humans and the availability of sophisticated technologies used to manipulate its genome and genes. The Drosophila research community quickly adopted CRISPR technologies and, in the 8 years since the first clustered regularly interspaced short palindromic repeats (CRISPR) publications in flies, has explored and innovated methods for mutagenesis, precise genome engineering, and beyond. Moreover, the short lifespan and ease of genetics have made Drosophila an ideal testing ground for in vivo applications and refinements of the rapidly evolving set of CRISPR-associated (CRISPR-Cas) tools. Here, we review innovations in delivery of CRISPR reagents, increased efficiency of cutting and homology-directed repair (HDR), and alternatives to standard Cas9-based approaches. While the focus is primarily on in vivo systems, we also describe the role of Drosophila cultured cells as both an indispensable first step in the process of assessing new CRISPR technologies and a platform for genome-wide CRISPR pooled screens.
Collapse
Affiliation(s)
- Jonathan Zirin
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| | - Justin Bosch
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Raghuvir Viswanatha
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Stephanie E Mohr
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|