1
|
Rajan A, Karpac J. Inter-organ communication in Drosophila: Lipoproteins, adipokines, and immune-metabolic coordination. Curr Opin Cell Biol 2025; 94:102508. [PMID: 40187050 DOI: 10.1016/j.ceb.2025.102508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 04/07/2025]
Abstract
Inter-organ communication networks are essential for maintaining systemic homeostasis in multicellular organisms. In Drosophila melanogaster, studies of adipokines and lipoproteins reveal evolutionarily conserved mechanisms coordinating metabolism, immunity, and behavior. This mini-review focuses on two key pathways: the adipokine Unpaired 2 (Upd2) and lipoprotein-mediated signaling. Upd2, a leptin analog, mediates fat-brain communication to regulate insulin secretion, sleep, and feeding behavior. Recent work has uncovered an LC3/Atg8-dependent secretion mechanism for Upd2, linking nutrient sensing to systemic adaptation. Lipoproteins, particularly ApoLpp and LTP, function beyond lipid transport, orchestrating neural maintenance and immune responses. During infection, macrophage-derived signals trigger lipoprotein-mediated lipid redistribution to support host defense. Additionally, muscle tissue emerges as an unexpected mediator of immune-metabolic coordination through inter-organ signaling. These findings highlight the intricate cross-talk between organs required for organismal survival and suggest therapeutic strategies for metabolic disorders.
Collapse
Affiliation(s)
- Akhila Rajan
- Basic Sciences Division, Fred Hutch, Seattle, WA, USA.
| | - Jason Karpac
- Department of Biology, Texas A&M University, College Station, TX, USA; Department of Cell Biology and Genetics, Texas A&M University, College of Medicine, Bryan, TX, USA.
| |
Collapse
|
2
|
Brunetti K, Catalani E, Del Quondam S, Romano N, Ceci M, Clerici G, Gordin M, Bianchini G, Brandolini L, Aramini A, Cervia D. A novel free fatty acid receptor agonist improving metabolic health in Drosophila models. Pharmacol Res 2025; 216:107769. [PMID: 40348099 DOI: 10.1016/j.phrs.2025.107769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 05/06/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
The development of synthetic modulators for human free fatty acid receptors (FFARs) has gained attention for addressing diabetes, metabolic syndrome, and dyslipidaemia. A new dual FFAR1 (GPR40) and FFAR4 (GPR120) agonist, DFL23916, was recently identified to improve glucose homeostasis, prompting investigations into its effects on metabolic disorders. This study assessed the selectivity and toxicity of DFL23916 in vitro and evaluated its effects in vivo using Drosophila melanogaster models of high-sugar (HSD) and high-fat diets (HFD). DFL23916 showed no off-target activities with no toxicity/hepatotoxicity in cultured cells. In adult HSD-fed flies with defective mobility, the oral administration of DFL23916 enhanced climbing speed in a concentration-dependent manner and reduced the high content of glucose and triglyceride levels alongside markers of insulin resistance. The compound did not affect viability and food intake of flies exposed to chronic overcaloric conditions but it inhibited the progressive weight gain. Similarly, larvae development remained unaffected by DFL23916, while it counteracted glucose and triglyceride elevation and reduced lipid droplet size caused by HSD and HFD. Finally, in silico analysis highlighted the relevance of evolutionary conserved Drosophila receptors in fatty acid sensing, suggesting putative candidates for DFL23916 binding. Collectively, these findings indicated the safe profile of DFL23916 and its efficacy in ameliorating hyperglycaemic and hyperlipidaemic features in overcaloric fed flies. Our results not only suggest that DFL23916 could be a potential candidate for further investigation in the context of metabolic disorders but also reinforce D. melanogaster as a valuable model for the preclinical evaluation of metabolic interventions, including FFAR-targeting strategies.
Collapse
Affiliation(s)
- Kashi Brunetti
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy
| | - Elisabetta Catalani
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy
| | - Simona Del Quondam
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy
| | - Nicla Romano
- Department of Life Sciences, Health and Health Professions, Università degli Studi Link, Roma, Italy
| | - Marcello Ceci
- Department of Ecology and Biology (DEB), Università degli Studi della Tuscia, Viterbo, Italy
| | - Giuditta Clerici
- Department of Electronics, Information and Biomedical Engineering, Politecnico di Milano, Milano, Italy; Human Technopole, Milano, Italy
| | | | - Gianluca Bianchini
- Research and Early Development, Dompé farmaceutici S.p.A., L'Aquila, Italy
| | - Laura Brandolini
- Research and Early Development, Dompé farmaceutici S.p.A., L'Aquila, Italy
| | - Andrea Aramini
- Research and Early Development, Dompé farmaceutici S.p.A., L'Aquila, Italy
| | - Davide Cervia
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy.
| |
Collapse
|
3
|
Rai M, Li H, Policastro RA, Pepin R, Zentner GE, Nemkov T, D’Alessandro A, Tennessen JM. Glycolytic disruption restricts Drosophila melanogaster larval growth via the cytokine Upd3. PLoS Genet 2025; 21:e1011690. [PMID: 40315265 PMCID: PMC12068724 DOI: 10.1371/journal.pgen.1011690] [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: 12/05/2024] [Revised: 05/12/2025] [Accepted: 04/15/2025] [Indexed: 05/04/2025] Open
Abstract
Drosophila larval growth requires efficient conversion of dietary nutrients into biomass. Lactate dehydrogenase (Ldh) and glycerol-3-phosphate dehydrogenase (Gpdh1) support this larval metabolic program by cooperatively promoting glycolytic flux. Consistent with their cooperative functions, the loss of both enzymes, but not either single enzyme alone, induces a developmental arrest. However, Ldh and Gpdh1 exhibit complex and often mutually exclusive expression patterns, suggesting that the lethal phenotypes exhibited by Gpdh1; Ldh double mutants could be mediated non-autonomously. Supporting this possibility, we find that the developmental arrest displayed by double mutants extends beyond simple metabolic disruption and instead stems, in part, from changes in systemic growth factor signaling. Specifically, we demonstrate that the simultaneous loss of Gpdh1 and Ldh results in elevated expression of Upd3, a cytokine involved in Jak/Stat signaling. Furthermore, we show that upd3 loss-of-function mutations suppress the Gpdh1; Ldh larval arrest phenotype, indicating that Upd3 signaling restricts larval development in response to decreased glycolytic flux. Together, our findings reveal a mechanism by which metabolic disruptions can modulate systemic growth factor signaling.
Collapse
Affiliation(s)
- Madhulika Rai
- Department of Biology, Indiana University, Bloomington, Indiana, United States of America
| | - Hongde Li
- Department of Biology, Indiana University, Bloomington, Indiana, United States of America
| | - Robert A. Policastro
- Department of Biology, Indiana University, Bloomington, Indiana, United States of America
| | - Robert Pepin
- Department of Chemistry, Indiana University, Bloomington, Indiana, United States of America
| | - Gabriel E. Zentner
- Department of Biology, Indiana University, Bloomington, Indiana, United States of America
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Jason M. Tennessen
- Department of Biology, Indiana University, Bloomington, Indiana, United States of America
- Member, Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, United States of America
| |
Collapse
|
4
|
Wang C, Shen J. From metabolism to lifespan trade-offs: polyethylene microplastics induce circadian disruption and sex-specific aging in Drosophila melanogaster. Comp Biochem Physiol C Toxicol Pharmacol 2025; 295:110214. [PMID: 40300706 DOI: 10.1016/j.cbpc.2025.110214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 04/02/2025] [Accepted: 04/26/2025] [Indexed: 05/01/2025]
Abstract
Microplastics (MPs), particularly polyethylene microplastics (PE-MPs), are increasingly recognized as contaminants in both aquatic and terrestrial environments. However, the ecological impacts of PE-MPs on terrestrial organisms remain underexplored. This study investigates the physiological and behavioral effects of PE-MPs exposure in the terrestrial model organism Drosophila melanogaster, shedding light on the potential risks posed by PE-MPs in land-based ecosystems. After exposing the fruit flies to different concentrations of PE-MPs for 20 days, we assessed several physiological biomarkers, including spontaneous behavioral activity, starvation resistance, metabolic biomarkers, and lifespan. Our findings indicate that PE-MPs exposure significantly affects fruit fly physiology, with increased spontaneous activity, decreased starvation resistance, and reduced triglyceride (TG) and protein levels (in males), reflecting disruption of metabolic processes. While PE-MPs did not affect female reproductive capacity, they did result in sex-specific impacts on lifespan, with male fruit flies showing a significant reduction in both mean and median lifespan at higher PE-MPs concentrations. These results highlight the need to consider the sex-dependent nature of PE-MPs toxicity when assessing their ecological risks. This study contributes new insights into the potential for PE-MPs to disrupt terrestrial ecosystems and underscores the importance of investigating the effects of microplastics on terrestrial invertebrates, providing a foundation for future ecotoxicological research.
Collapse
Affiliation(s)
- Chengpeng Wang
- College of Artificial Intelligence, Hangzhou Dianzi University, Hangzhou 310018, China; Key Laboratory of Micro-nano Sensing and IoT of Wenzhou, Wenzhou Institute of Hangzhou Dianzi University, Wenzhou 325038, China
| | - Jie Shen
- College of Artificial Intelligence, Hangzhou Dianzi University, Hangzhou 310018, China; Key Laboratory of Micro-nano Sensing and IoT of Wenzhou, Wenzhou Institute of Hangzhou Dianzi University, Wenzhou 325038, China.
| |
Collapse
|
5
|
Akagi K, Jin YJ, Koizumi K, Oku M, Ito K, Shen X, Imura JI, Aihara K, Saito S. Integration of Dynamical Network Biomarkers, Control Theory and Drosophila Model Identifies Vasa/DDX4 as the Potential Therapeutic Targets for Metabolic Syndrome. Cells 2025; 14:415. [PMID: 40136664 PMCID: PMC11941168 DOI: 10.3390/cells14060415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/05/2025] [Accepted: 03/09/2025] [Indexed: 03/27/2025] Open
Abstract
Metabolic syndrome (MetS) is a subclinical disease, resulting in increased risk of type 2 diabetes (T2D), cardiovascular diseases, cancer, and mortality. Dynamical network biomarkers (DNB) theory has been developed to provide early-warning signals of the disease state during a preclinical stage. To improve the efficiency of DNB analysis for the target genes discovery, the DNB intervention analysis based on the control theory has been proposed. However, its biological validation in a specific disease such as MetS remains unexplored. Herein, we identified eight candidate genes from adipose tissue of MetS model mice at the preclinical stage by the DNB intervention analysis. Using Drosophila, we conducted RNAi-mediated knockdown screening of these candidate genes and identified vasa (also known as DDX4), encoding a DEAD-box RNA helicase, as a fat metabolism-associated gene. Fat body-specific knockdown of vasa abrogated high-fat diet (HFD)-induced enhancement of starvation resistance through up-regulation of triglyceride lipase. We also confirmed that DDX4 expressing adipocytes are increased in HFD-fed mice and high BMI patients using the public datasets. These results prove the potential of the DNB intervention analysis to search the therapeutic targets for diseases at the preclinical stage.
Collapse
Affiliation(s)
- Kazutaka Akagi
- Division of Presymptomatic Disease, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan;
- Research Center for Pre-Disease Science, University of Toyama, Toyama 930-8555, Japan; (M.O.); (S.S.)
| | - Ying-Jie Jin
- Graduate School of Pharma-Medical Sciences, University of Toyama, Toyama 930-0194, Japan;
| | - Keiichi Koizumi
- Division of Presymptomatic Disease, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan;
- Research Center for Pre-Disease Science, University of Toyama, Toyama 930-8555, Japan; (M.O.); (S.S.)
| | - Makito Oku
- Research Center for Pre-Disease Science, University of Toyama, Toyama 930-8555, Japan; (M.O.); (S.S.)
| | - Kaisei Ito
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan;
| | - Xun Shen
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, Tokyo 184-8588, Japan;
| | - Jun-ichi Imura
- Department of Systems and Control Engineering, School of Engineering, Institute of Science Tokyo, Tokyo 152-8552, Japan;
| | - Kazuyuki Aihara
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, Tokyo 113-0033, Japan;
| | - Shigeru Saito
- Research Center for Pre-Disease Science, University of Toyama, Toyama 930-8555, Japan; (M.O.); (S.S.)
| |
Collapse
|
6
|
Rojo AI, Buttari B, Cadenas S, Carlos AR, Cuadrado A, Falcão AS, López MG, Georgiev MI, Grochot-Przeczek A, Gumeni S, Jimenez-Villegas J, Horbanczuk JO, Konu O, Lastres-Becker I, Levonen AL, Maksimova V, Michaeloudes C, Mihaylova LV, Mickael ME, Milisav I, Miova B, Rada P, Santos M, Seabra MC, Strac DS, Tenreiro S, Trougakos IP, Dinkova-Kostova AT. Model organisms for investigating the functional involvement of NRF2 in non-communicable diseases. Redox Biol 2025; 79:103464. [PMID: 39709790 PMCID: PMC11733061 DOI: 10.1016/j.redox.2024.103464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/26/2024] [Accepted: 12/08/2024] [Indexed: 12/24/2024] Open
Abstract
Non-communicable chronic diseases (NCDs) are most commonly characterized by age-related loss of homeostasis and/or by cumulative exposures to environmental factors, which lead to low-grade sustained generation of reactive oxygen species (ROS), chronic inflammation and metabolic imbalance. Nuclear factor erythroid 2-like 2 (NRF2) is a basic leucine-zipper transcription factor that regulates the cellular redox homeostasis. NRF2 controls the expression of more than 250 human genes that share in their regulatory regions a cis-acting enhancer termed the antioxidant response element (ARE). The products of these genes participate in numerous functions including biotransformation and redox homeostasis, lipid and iron metabolism, inflammation, proteostasis, as well as mitochondrial dynamics and energetics. Thus, it is possible that a single pharmacological NRF2 modulator might mitigate the effect of the main hallmarks of NCDs, including oxidative, proteostatic, inflammatory and/or metabolic stress. Research on model organisms has provided tremendous knowledge of the molecular mechanisms by which NRF2 affects NCDs pathogenesis. This review is a comprehensive summary of the most commonly used model organisms of NCDs in which NRF2 has been genetically or pharmacologically modulated, paving the way for drug development to combat NCDs. We discuss the validity and use of these models and identify future challenges.
Collapse
Affiliation(s)
- Ana I Rojo
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain.
| | - Brigitta Buttari
- Department of Cardiovascular, Endocrine-Metabolic Diseases, and Aging, Italian National Institute of Health, 00161, Rome, Italy
| | - Susana Cadenas
- Centro de Biología Molecular Severo Ochoa (CSIC/UAM), Cantoblanco, Madrid, Spain
| | - Ana Rita Carlos
- CE3C-CHANGE, Department of Animal Biology, Faculty of Sciences, University of Lisbon, 1749-016, Lisbon, Portugal
| | - Antonio Cuadrado
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain
| | - Ana Sofia Falcão
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Manuela G López
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigación Sanitaria La Princesa, Hospital Universitario de la Princesa, Madrid, Spain
| | - Milen I Georgiev
- Department of Plant Cell Biotechnology, Center of Plant Systems Biology and Biotechnology, 4000, Plovdiv, Bulgaria; Laboratory of Metabolomics, Institute of Microbiology, Bulgarian Academy of Sciences, 139 Ruski Blvd., 4000, Plovdiv, Bulgaria
| | - Anna Grochot-Przeczek
- Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, 30-387, Krakow, Poland
| | - Sentiljana Gumeni
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, 15784, Greece
| | - José Jimenez-Villegas
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain
| | - Jarosław Olav Horbanczuk
- Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology, 36A Postępu, Jastrzębiec, 05-552, Poland
| | - Ozlen Konu
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey; Department of Neuroscience, Bilkent University, Ankara, Turkey; UNAM-Institute of Materials Science and Nanotechnology, Bilkent University, Ankara, Turkey
| | - Isabel Lastres-Becker
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Institute Teófilo Hernando for Drug Discovery, Universidad Autónoma de Madrid, 28029, Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain
| | - Anna-Liisa Levonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland
| | - Viktorija Maksimova
- Department of Applied Pharmacy, Division of Pharmacy, Faculty of Medical Sciences, Goce Delcev University, Stip, Krste Misirkov Str., No. 10-A, P.O. Box 201, 2000, Stip, Macedonia
| | | | - Liliya V Mihaylova
- Department of Plant Cell Biotechnology, Center of Plant Systems Biology and Biotechnology, 4000, Plovdiv, Bulgaria; Laboratory of Metabolomics, Institute of Microbiology, Bulgarian Academy of Sciences, 139 Ruski Blvd., 4000, Plovdiv, Bulgaria
| | - Michel Edwar Mickael
- Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology, 36A Postępu, Jastrzębiec, 05-552, Poland
| | - Irina Milisav
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloska 4, 1000, Ljubljana, Slovenia; Laboratory of oxidative stress research, Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, 1000, Ljubljana, Slovenia
| | - Biljana Miova
- Department of Experimental Physiology and Biochemistry, Institute of Biology, Faculty of Natural Sciences and Mathematics, University "St Cyril and Methodius", Skopje, Macedonia
| | - Patricia Rada
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Marlene Santos
- REQUIMTE/LAQV, Escola Superior de Saúde (E2S), Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida 400, 4200-072, Porto, Portugal; Molecular Oncology & Viral Pathology, IPO-Porto Research Center (CI-IPOP), Portuguese Institute of Oncology, 4200-072, Porto, Portugal
| | - Miguel C Seabra
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Dubravka Svob Strac
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Ruder Boskovic Institute, 10 000, Zagreb, Croatia
| | - Sandra Tenreiro
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Ioannis P Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, 15784, Greece
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cancer Research, School of Medicine, University of Dundee, Dundee, UK; Department of Pharmacology and Molecular Sciences and Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
7
|
Laghezza Masci V, Mezzani I, Alicandri E, Tomassi W, Paolacci AR, Covino S, Vinciguerra V, Catalani E, Cervia D, Ciaffi M, Garzoli S, Ovidi E. The Role of Extracts of Edible Parts and Production Wastes of Globe Artichoke ( Cynara cardunculus L. var. scolymus (L.)) in Counteracting Oxidative Stress. Antioxidants (Basel) 2025; 14:116. [PMID: 39857450 PMCID: PMC11761904 DOI: 10.3390/antiox14010116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
In addition to the immature edible flower heads, the cultivation of globe artichoke (Cynara cardunculus L. var. scolymus (L.) Fiori) generates substantial quantities of by-products, including leaves, stems, and roots, which constitute potential sources of bioactive compounds and prebiotic dietary fiber. Preserving agricultural biodiversity and promoting socioeconomic development are essential for enhancing domestic production and fostering innovation. In the search for new biomolecules with antioxidant properties, this research focused on a globe artichoke landrace at risk of genetic erosion, still cultivated in the northern part of the Lazio region, known as the "Carciofo Ortano". To investigate the antioxidant properties of various globe artichoke tissues from the "Carciofo Ortano" landrace, methanolic extracts were prepared from the immature main and secondary flower heads, stems, and leaves of representative genotypes of this landrace. Additionally, extracts were obtained from the same tissues of four landraces/clones included in the varietal platform of the PGI "Carciofo Romanesco del Lazio", which served as reference genotypes: Campagnano, Castellammare, C3, and Grato 1. The antioxidant properties of these extracts were assessed using FRAP, ABTS, DPPH assays, and total phenolic content (TPC). The stem and secondary flower head extracts of two representative "Carciofo Ortano" genotypes and the Grato 1 clone, which have higher phenolic content, demonstrated the highest antioxidant activity. These extracts were therefore studied for their chemical profile using HPLC-DAD and SPME-GC/MS analysis. Additionally, the same extracts were investigated in vitro for their antioxidant capacity in differentiated SH-SY5Y cells, assessing their effects on ROS levels and the restoration of GSH levels. Furthermore, the in vivo beneficial effects of counteracting oxidative stress were evaluated in high sucrose-fed Drosophila melanogaster, as oxidative stress is a typical hallmark of hyperglycemic status. Overall, the results indicated that the edible immature inflorescences of the "Carciofo Ortano" landrace, along with the byproducts of its cultivation, are sources of raw materials containing biomolecules whose properties can be exploited for further applications in the pharmaceutical and medical sectors.
Collapse
Affiliation(s)
- Valentina Laghezza Masci
- Department for Innovation in Biological, Agro-Food and Forest Systems, Tuscia University, 01100 Viterbo, Italy; (V.L.M.); (I.M.); (E.A.); (W.T.); (A.R.P.); (S.C.); (V.V.); (E.C.); (D.C.); (E.O.)
| | - Irene Mezzani
- Department for Innovation in Biological, Agro-Food and Forest Systems, Tuscia University, 01100 Viterbo, Italy; (V.L.M.); (I.M.); (E.A.); (W.T.); (A.R.P.); (S.C.); (V.V.); (E.C.); (D.C.); (E.O.)
| | - Enrica Alicandri
- Department for Innovation in Biological, Agro-Food and Forest Systems, Tuscia University, 01100 Viterbo, Italy; (V.L.M.); (I.M.); (E.A.); (W.T.); (A.R.P.); (S.C.); (V.V.); (E.C.); (D.C.); (E.O.)
| | - William Tomassi
- Department for Innovation in Biological, Agro-Food and Forest Systems, Tuscia University, 01100 Viterbo, Italy; (V.L.M.); (I.M.); (E.A.); (W.T.); (A.R.P.); (S.C.); (V.V.); (E.C.); (D.C.); (E.O.)
| | - Anna Rita Paolacci
- Department for Innovation in Biological, Agro-Food and Forest Systems, Tuscia University, 01100 Viterbo, Italy; (V.L.M.); (I.M.); (E.A.); (W.T.); (A.R.P.); (S.C.); (V.V.); (E.C.); (D.C.); (E.O.)
| | - Stefano Covino
- Department for Innovation in Biological, Agro-Food and Forest Systems, Tuscia University, 01100 Viterbo, Italy; (V.L.M.); (I.M.); (E.A.); (W.T.); (A.R.P.); (S.C.); (V.V.); (E.C.); (D.C.); (E.O.)
| | - Vittorio Vinciguerra
- Department for Innovation in Biological, Agro-Food and Forest Systems, Tuscia University, 01100 Viterbo, Italy; (V.L.M.); (I.M.); (E.A.); (W.T.); (A.R.P.); (S.C.); (V.V.); (E.C.); (D.C.); (E.O.)
| | - Elisabetta Catalani
- Department for Innovation in Biological, Agro-Food and Forest Systems, Tuscia University, 01100 Viterbo, Italy; (V.L.M.); (I.M.); (E.A.); (W.T.); (A.R.P.); (S.C.); (V.V.); (E.C.); (D.C.); (E.O.)
| | - Davide Cervia
- Department for Innovation in Biological, Agro-Food and Forest Systems, Tuscia University, 01100 Viterbo, Italy; (V.L.M.); (I.M.); (E.A.); (W.T.); (A.R.P.); (S.C.); (V.V.); (E.C.); (D.C.); (E.O.)
| | - Mario Ciaffi
- Department for Innovation in Biological, Agro-Food and Forest Systems, Tuscia University, 01100 Viterbo, Italy; (V.L.M.); (I.M.); (E.A.); (W.T.); (A.R.P.); (S.C.); (V.V.); (E.C.); (D.C.); (E.O.)
| | - Stefania Garzoli
- Department of Drug Chemistry and Technology, Sapienza University, 00185 Rome, Italy
| | - Elisa Ovidi
- Department for Innovation in Biological, Agro-Food and Forest Systems, Tuscia University, 01100 Viterbo, Italy; (V.L.M.); (I.M.); (E.A.); (W.T.); (A.R.P.); (S.C.); (V.V.); (E.C.); (D.C.); (E.O.)
| |
Collapse
|
8
|
Yoshinari Y, Nishimura T, Yoshii T, Kondo S, Tanimoto H, Kobayashi T, Matsuyama M, Niwa R. A high-protein diet-responsive gut hormone regulates behavioral and metabolic optimization in Drosophila melanogaster. Nat Commun 2024; 15:10819. [PMID: 39737959 PMCID: PMC11685984 DOI: 10.1038/s41467-024-55050-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 11/27/2024] [Indexed: 01/01/2025] Open
Abstract
Protein is essential for all living organisms; however, excessive protein intake can have adverse effects, such as hyperammonemia. Although mechanisms responding to protein deficiency are well-studied, there is a significant gap in our understanding of how organisms adaptively suppress excessive protein intake. In the present study, utilizing the fruit fly, Drosophila melanogaster, we discover that the peptide hormone CCHamide1 (CCHa1), secreted by enteroendocrine cells in response to a high-protein diet (HPD), is vital for suppressing overconsumption of protein. Gut-derived CCHa1 is received by a small subset of enteric neurons that produce short neuropeptide F, thereby modulating protein-specific satiety. Importantly, impairment of the CCHa1-mediated gut-enteric neuronal axis results in ammonia accumulation and a shortened lifespan under HPD conditions. Collectively, our findings unravel the crosstalk of gut hormone and neuronal pathways that orchestrate physiological responses to prevent and adapt to dietary protein overload.
Collapse
Affiliation(s)
- Yuto Yoshinari
- Metabolic Regulation and Genetics, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan.
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Takashi Nishimura
- Metabolic Regulation and Genetics, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan.
| | - Taishi Yoshii
- Graduate School of Environmental, Life, Natural Science and Technology, Okayama University, Okayama, 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, Mishima, Japan
| | - Hiromu Tanimoto
- Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Tomoe Kobayashi
- Division of Molecular Genetics, Shigei Medical Research Institute, Okayama, Japan
| | - Makoto Matsuyama
- Division of Molecular Genetics, Shigei Medical Research Institute, Okayama, Japan
| | - Ryusuke Niwa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, 305-8577, Japan.
| |
Collapse
|
9
|
Mahishi D, Agrawal N, Jiang W, Yapici N. From Mammals to Insects: Exploring the Genetic and Neural Basis of Eating Behavior. Annu Rev Genet 2024; 58:455-485. [PMID: 39585905 DOI: 10.1146/annurev-genet-111523-102202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Obesity and anorexia are life-threatening diseases that are still poorly understood at the genetic and neuronal levels. Patients suffering from these conditions experience disrupted regulation of food consumption, leading to extreme weight gain or loss and, in severe situations, death from metabolic dysfunction. Despite the development of various behavioral and pharmacological interventions, current treatments often yield limited and short-lived success. To address this, a deeper understanding of the genetic and neural mechanisms underlying food perception and appetite regulation is essential for identifying new drug targets and developing more effective treatment methods. This review summarizes the progress of past research in understanding the genetic and neural mechanisms controlling food consumption and appetite regulation, focusing on two key model organisms: the fruit fly Drosophila melanogaster and the mouse Mus musculus. These studies investigate how the brain senses energy and nutrient deficiency, how sensory signals trigger appetitive behaviors, and how food intake is regulated through interconnected neural circuits in the brain.
Collapse
Affiliation(s)
- Deepthi Mahishi
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA;
| | - Naman Agrawal
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA;
| | - Wenshuai Jiang
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA;
| | - Nilay Yapici
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA;
| |
Collapse
|
10
|
Schoofs A, Miroschnikow A, Schlegel P, Zinke I, Schneider-Mizell CM, Cardona A, Pankratz MJ. Serotonergic modulation of swallowing in a complete fly vagus nerve connectome. Curr Biol 2024; 34:4495-4512.e6. [PMID: 39270641 PMCID: PMC7616834 DOI: 10.1016/j.cub.2024.08.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 07/15/2024] [Accepted: 08/15/2024] [Indexed: 09/15/2024]
Abstract
How the body interacts with the brain to perform vital life functions, such as feeding, is a fundamental issue in physiology and neuroscience. Here, we use a whole-animal scanning transmission electron microscopy volume of Drosophila to map the neuronal circuits that connect the entire enteric nervous system to the brain via the insect vagus nerve at synaptic resolution. We identify a gut-brain feedback loop in which Piezo-expressing mechanosensory neurons in the esophagus convey food passage information to a cluster of six serotonergic neurons in the brain. Together with information on food value, these central serotonergic neurons enhance the activity of serotonin receptor 7-expressing motor neurons that drive swallowing. This elemental circuit architecture includes an axo-axonic synaptic connection from the glutamatergic motor neurons innervating the esophageal muscles onto the mechanosensory neurons that signal to the serotonergic neurons. Our analysis elucidates a neuromodulatory sensory-motor system in which ongoing motor activity is strengthened through serotonin upon completion of a biologically meaningful action, and it may represent an ancient form of motor learning.
Collapse
Affiliation(s)
- Andreas Schoofs
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Carl-Troll-Straße, Bonn 53115, Germany
| | - Anton Miroschnikow
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Carl-Troll-Straße, Bonn 53115, Germany
| | - Philipp Schlegel
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 TN1, UK; MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Trumpington, Cambridge CB2 0QH, UK
| | - Ingo Zinke
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Carl-Troll-Straße, Bonn 53115, Germany
| | | | - Albert Cardona
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Trumpington, Cambridge CB2 0QH, UK; Janelia Research Campus, Howard Hughes Medical Institute, Helix Drive, Ashburn, VA 20147, USA; Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Place, Cambridge CB2 3EL, UK
| | - Michael J Pankratz
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Carl-Troll-Straße, Bonn 53115, Germany.
| |
Collapse
|
11
|
Fritz García JHG, Keller Valsecchi CI, Basilicata MF. Sex as a biological variable in ageing: insights and perspectives on the molecular and cellular hallmarks. Open Biol 2024; 14:240177. [PMID: 39471841 PMCID: PMC11521605 DOI: 10.1098/rsob.240177] [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: 06/25/2024] [Revised: 08/28/2024] [Accepted: 09/05/2024] [Indexed: 11/01/2024] Open
Abstract
Sex-specific differences in lifespan and ageing are observed in various species. In humans, women generally live longer but are frailer and suffer from different age-related diseases compared to men. The hallmarks of ageing, such as genomic instability, telomere attrition or loss of proteostasis, exhibit sex-specific patterns. Sex chromosomes and sex hormones, as well as the epigenetic regulation of the inactive X chromosome, have been shown to affect lifespan and age-related diseases. Here we review the current knowledge on the biological basis of sex-biased ageing. While our review is focused on humans, we also discuss examples of model organisms such as the mouse, fruit fly or the killifish. Understanding these molecular differences is crucial as the elderly population is expected to double worldwide by 2050, making sex-specific approaches in the diagnosis, treatment, therapeutic development and prevention of age-related diseases a pressing need.
Collapse
Affiliation(s)
| | | | - M. Felicia Basilicata
- Institute of Molecular Biology (IMB), Mainz, Germany
- University Medical Center (UMC), Mainz, Germany
| |
Collapse
|
12
|
Alassaf M, Rajan A. Adipocyte metabolic state regulates glial phagocytic function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.614765. [PMID: 39386724 PMCID: PMC11463506 DOI: 10.1101/2024.09.24.614765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Obesity and type 2 diabetes are well-established risk factors for neurodegenerative disorders1-4, yet the underlying mechanisms remain poorly understood. The adipocyte-brain axis is crucial for brain function, as adipocytes secrete signaling molecules, including lipids and adipokines, that impinge on neural circuits to regulate feeding and energy expenditure5. Disruptions in the adipocyte-brain axis are associated with neurodegenerative conditions6, but the causal links are not fully understood. Neural debris accumulates with age and injury, and glial phagocytic function is crucial for clearing this debris and maintaining a healthy brain microenvironment7-9. Using adult Drosophila, we investigate how adipocyte metabolism influences glial phagocytic activity in the brain. We demonstrate that a prolonged obesogenic diet increases adipocyte fatty acid oxidation and ketogenesis. Genetic manipulations that mimic obesogenic diet-induced changes in adipocyte lipid and mitochondrial metabolism unexpectedly reduce the expression of the phagocytic receptor Draper in Drosophila microglia-like cells in the brain. We identify Apolpp-the Drosophila equivalent of human apolipoprotein B (ApoB)-as a critical adipocyte-derived signal that regulates glial phagocytosis. Additionally, we show that Lipoprotein Receptor 1 (LpR1), the LDL receptor on phagocytic glia, is required for glial capacity to clear injury-induced neuronal debris. Our findings establish that adipocyte-brain lipoprotein signaling regulates glial phagocytic function, revealing a novel pathway that links adipocyte metabolic disorders with neurodegeneration.
Collapse
Affiliation(s)
- Mroj Alassaf
- Basic Sciences Division, Fred Hutch, Seattle, WA-98109. The USA
| | - Akhila Rajan
- Basic Sciences Division, Fred Hutch, Seattle, WA-98109. The USA
| |
Collapse
|
13
|
Miyamoto T, Hedjazi S, Miyamoto C, Amrein H. Drosophila neuronal Glucose-6-Phosphatase is a modulator of neuropeptide release that regulates muscle glycogen stores via FMRFamide signaling. Proc Natl Acad Sci U S A 2024; 121:e2319958121. [PMID: 39008673 PMCID: PMC11287260 DOI: 10.1073/pnas.2319958121] [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: 11/28/2023] [Accepted: 06/07/2024] [Indexed: 07/17/2024] Open
Abstract
Neuropeptides (NPs) and their cognate receptors are critical effectors of diverse physiological processes and behaviors. We recently reported of a noncanonical function of the Drosophila Glucose-6-Phosphatase (G6P) gene in a subset of neurosecretory cells in the central nervous system that governs systemic glucose homeostasis in food-deprived flies. Here, we show that G6P-expressing neurons define six groups of NP-secreting cells, four in the brain and two in the thoracic ganglion. Using the glucose homeostasis phenotype as a screening tool, we find that neurons located in the thoracic ganglion expressing FMRFamide NPs (FMRFaG6P neurons) are necessary and sufficient to maintain systemic glucose homeostasis in starved flies. We further show that G6P is essential in FMRFaG6P neurons for attaining a prominent Golgi apparatus and secreting NPs efficiently. Finally, we establish that G6P-dependent FMRFa signaling is essential for the build-up of glycogen stores in the jump muscle which expresses the receptor for FMRFamides. We propose a general model in which the main role of G6P is to counteract glycolysis in peptidergic neurons for the purpose of optimizing the intracellular environment best suited for the expansion of the Golgi apparatus, boosting release of NPs and enhancing signaling to respective target tissues expressing cognate receptors.
Collapse
Affiliation(s)
- Tetsuya Miyamoto
- Department of Cell Biology and Genetics, School of Medicine, Texas A&M University, Bryan, TX77807
| | - Sheida Hedjazi
- Department of Cell Biology and Genetics, School of Medicine, Texas A&M University, Bryan, TX77807
| | - Chika Miyamoto
- Department of Cell Biology and Genetics, School of Medicine, Texas A&M University, Bryan, TX77807
| | - Hubert Amrein
- Department of Cell Biology and Genetics, School of Medicine, Texas A&M University, Bryan, TX77807
| |
Collapse
|
14
|
Rai M, Li H, Policastro RA, Zentner GE, Nemkov T, D’Alessandro A, Tennessen JM. Glycolytic Disruption Triggers Interorgan Signaling to Nonautonomously Restrict Drosophila Larval Growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597835. [PMID: 38895259 PMCID: PMC11185712 DOI: 10.1101/2024.06.06.597835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Drosophila larval growth requires efficient conversion of dietary nutrients into biomass. Lactate Dehydrogenase (Ldh) and Glycerol-3-phosphate dehydrogenase (Gpdh1) support larval biosynthetic metabolism by maintaining NAD+/NADH redox balance and promoting glycolytic flux. Consistent with the cooperative functions of Ldh and Gpdh1, the loss of both enzymes, but neither single enzyme, induces a developmental arrest. However, Ldh and Gpdh1 exhibit complex and often mutually exclusive expression patterns, suggesting that the Gpdh1; Ldh double mutant lethal phenotype could be mediated nonautonomously. Here we find that the developmental arrest displayed by the double mutants extends beyond simple metabolic disruption and instead stems, in part, from changes in systemic growth factor signaling. Specifically, we demonstrate that this synthetic lethality is linked to the upregulation of Upd3, a cytokine involved in the Jak/Stat signaling pathway. Moreover, we demonstrate that either loss of the Upd3 or dietary administration of the steroid hormone 20-hydroxyecdysone (20E) rescue the synthetic lethal phenotype of Gpdh1; Ldh double mutants. Together, these findings demonstrate that metabolic disruptions within a single tissue can nonautonomously modulate interorgan signaling to ensure synchronous developmental growth.
Collapse
Affiliation(s)
- Madhulika Rai
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Hongde Li
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | | | | | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Colorado, USA
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Colorado, USA
| | | |
Collapse
|
15
|
Singh A, Abhilasha KV, Acharya KR, Liu H, Nirala NK, Parthibane V, Kunduri G, Abimannan T, Tantalla J, Zhu LJ, Acharya JK, Acharya UR. A nutrient responsive lipase mediates gut-brain communication to regulate insulin secretion in Drosophila. Nat Commun 2024; 15:4410. [PMID: 38782979 PMCID: PMC11116528 DOI: 10.1038/s41467-024-48851-8] [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: 06/23/2022] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
Pancreatic β cells secrete insulin in response to glucose elevation to maintain glucose homeostasis. A complex network of inter-organ communication operates to modulate insulin secretion and regulate glucose levels after a meal. Lipids obtained from diet or generated intracellularly are known to amplify glucose-stimulated insulin secretion, however, the underlying mechanisms are not completely understood. Here, we show that a Drosophila secretory lipase, Vaha (CG8093), is synthesized in the midgut and moves to the brain where it concentrates in the insulin-producing cells in a process requiring Lipid Transfer Particle, a lipoprotein originating in the fat body. In response to dietary fat, Vaha stimulates insulin-like peptide release (ILP), and Vaha deficiency results in reduced circulatory ILP and diabetic features including hyperglycemia and hyperlipidemia. Our findings suggest Vaha functions as a diacylglycerol lipase physiologically, by being a molecular link between dietary fat and lipid amplified insulin secretion in a gut-brain axis.
Collapse
Affiliation(s)
- Alka Singh
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA, 01605, USA
| | | | - Kathya R Acharya
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA, 01605, USA
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, 21702, USA
- University of Cincinnati College of Medicine, 3230 Eden Ave, Cincinnati, OH, 45267, USA
| | - Haibo Liu
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Niraj K Nirala
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Velayoudame Parthibane
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, 21702, USA
| | - Govind Kunduri
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, 21702, USA
| | - Thiruvaimozhi Abimannan
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, 21702, USA
| | - Jacob Tantalla
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, 21702, USA
| | - Lihua Julie Zhu
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Jairaj K Acharya
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, 21702, USA.
| | - Usha R Acharya
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, 21702, USA.
| |
Collapse
|
16
|
Gao J, Zhang S, Deng P, Wu Z, Lemaitre B, Zhai Z, Guo Z. Dietary L-Glu sensing by enteroendocrine cells adjusts food intake via modulating gut PYY/NPF secretion. Nat Commun 2024; 15:3514. [PMID: 38664401 PMCID: PMC11045819 DOI: 10.1038/s41467-024-47465-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Amino acid availability is monitored by animals to adapt to their nutritional environment. Beyond gustatory receptors and systemic amino acid sensors, enteroendocrine cells (EECs) are believed to directly percept dietary amino acids and secrete regulatory peptides. However, the cellular machinery underlying amino acid-sensing by EECs and how EEC-derived hormones modulate feeding behavior remain elusive. Here, by developing tools to specifically manipulate EECs, we find that Drosophila neuropeptide F (NPF) from mated female EECs inhibits feeding, similar to human PYY. Mechanistically, dietary L-Glutamate acts through the metabotropic glutamate receptor mGluR to decelerate calcium oscillations in EECs, thereby causing reduced NPF secretion via dense-core vesicles. Furthermore, two dopaminergic enteric neurons expressing NPFR perceive EEC-derived NPF and relay an anorexigenic signal to the brain. Thus, our findings provide mechanistic insights into how EECs assess food quality and identify a conserved mode of action that explains how gut NPF/PYY modulates food intake.
Collapse
Affiliation(s)
- Junjun Gao
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Song Zhang
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pan Deng
- State Key Laboratory of Digital Manufacturing Equipment and Technology, Huazhong University of Science and Technology, Wuhan, PR China
- Department of Mechanical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Zhigang Wu
- State Key Laboratory of Digital Manufacturing Equipment and Technology, Huazhong University of Science and Technology, Wuhan, PR China
| | - Bruno Lemaitre
- Global Health Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Zongzhao Zhai
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan, PR China.
| | - Zheng Guo
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
17
|
Miyamoto T, Hedjazi S, Miyamoto C, Amrein H. Drosophila Neuronal Glucose 6 Phosphatase is a Modulator of Neuropeptide Release that Regulates Muscle Glycogen Stores via FMRFamide Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.28.568950. [PMID: 38077084 PMCID: PMC10705280 DOI: 10.1101/2023.11.28.568950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
Neuropeptides (NPs) and their cognate receptors are critical effectors of diverse physiological processes and behaviors. We recently reported of a non-canonical function of the Drosophila Glucose-6-Phosphatase ( G6P ) gene in a subset of neurosecretory cells in the CNS that governs systemic glucose homeostasis in food deprived flies. Here, we show that G6P expressing neurons define 6 groups of neuropeptide secreting cells, 4 in the brain and 2 in the thoracic ganglion. Using the glucose homeostasis phenotype as a screening tool, we find that neurons located in the thoracic ganglion expressing FMRFamide neuropeptides ( FMRFa G6P neurons) are necessary and sufficient to maintain systemic glucose homeostasis in starved flies. We further show that G6P is essential in FMRFa G6P neurons for attaining a prominent Golgi apparatus and secreting neuropeptides efficiently. Finally, we establish that G6P dependent FMRFa signaling is essential for the build-up of glycogen stores in the jump muscle which expresses the receptor for FMRFamides. We propose a general model in which the main role of G6P is to counteract glycolysis in peptidergic neurons for the purpose of optimizing the intracellular environment best suited for the expansion of the Golgi apparatus, boosting release of neuropeptides and enhancing signaling to respective target tissues expressing cognate receptors. SIGNIFICANCE STATEMENT Glucose-6-phosphtase (G6P) is a critical enzyme in sugar synthesis and catalyzes the final step in glucose production. In Drosophila - and insects in general - where trehalose is the circulating sugar and Trehalose phosphate synthase, and not G6P, is used for sugar production, G6P has adopted a novel and unique role in peptidergic neurons in the CNS. Interestingly, flies lacking G6P show diminished Neuropeptide secretions and have a smaller Golgi apparatus in peptidergic neurons. It is hypothesized that the role of G6P is to counteract glycolysis, thereby creating a cellular environment that is more amenable to efficient neuropeptide secretion.
Collapse
|
18
|
Zandawala M, Gera J. Leptin- and cytokine-like unpaired signaling in Drosophila. Mol Cell Endocrinol 2024; 584:112165. [PMID: 38266772 DOI: 10.1016/j.mce.2024.112165] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/13/2024] [Accepted: 01/19/2024] [Indexed: 01/26/2024]
Abstract
Animals have evolved a multitude of signaling pathways that enable them to orchestrate diverse physiological processes to tightly regulate systemic homeostasis. This signaling is mediated by various families of peptide hormones and cytokines that are conserved across the animal kingdom. In this review, we primarily focus on the unpaired (Upd) family of proteins in Drosophila which are evolutionarily related to mammalian leptin and the cytokine interleukin 6. We summarize expression patterns of Upd in Drosophila and discuss the parallels in structure, signaling pathway, and functions between Upd and their mammalian counterparts. In particular, we focus on the roles of Upd in governing metabolic homeostasis, growth and development, and immune responses. We aim to stimulate future studies on leptin-like signaling in other phyla which can help bridge the evolutionary gap between insect Upd and vertebrate leptin and cytokines like interleukin 6.
Collapse
Affiliation(s)
- Meet Zandawala
- Neurobiology and Genetics, Theodor-Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany; Department of Biochemistry and Molecular Biology, University of Nevada, Reno, NV, 89557, USA.
| | - Jayati Gera
- Neurobiology and Genetics, Theodor-Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany
| |
Collapse
|
19
|
Li J, Dong Y, Zhou T, Tian H, Huang X, Zhang YQ, Wang Y, Lam SM, Shui G. Long-chain acyl-CoA synthetase regulates systemic lipid homeostasis via glycosylation-dependent lipoprotein production. LIFE METABOLISM 2024; 3:loae004. [PMID: 39872215 PMCID: PMC11749247 DOI: 10.1093/lifemeta/loae004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 01/04/2024] [Accepted: 01/16/2024] [Indexed: 01/30/2025]
Abstract
Interorgan lipid transport is crucial for organism development and the maintenance of physiological function. Here, we demonstrate that Drosophila long-chain acyl-CoA synthetase (dAcsl), which catalyzes the conversion of fatty acids into acyl-coenzyme As (acyl-CoAs), plays a critical role in regulating systemic lipid homeostasis. dAcsl deficiency in the fat body led to the ectopic accumulation of neutral lipids in the gut, along with significantly reduced lipoprotein contents in both the fat body and hemolymph. The aberrant phenotypes were rescued by fat body-specific overexpression of apolipophorin. A multi-omics investigation comprising lipidomics, metabolomics, and proteomics in conjunction with genetic screening revealed that glycosylation processes were suppressed in dAcsl knockdown flies. Overexpression of CG9035, human ortholog of which is implicated in the congenital disorder of glycosylation, ameliorated gut lipid accumulation in Drosophila. Aberrant lipoprotein glycosylation led to accelerated proteasome-related degradation and induced ER stress in dAcsl knockdown flies, impairing lipoprotein release into the circulation which compromised interorgan lipid transport between the fat body and the gut. Inhibition of ubiquitin-proteasome-dependent degradation alleviated the phenotype of gut ectopic fat accumulation in dAcsl knockdown flies. Finally, we verified that ACSL4, the human homolog of dAcsl, also regulated lipoprotein levels in HepG2 cells, indicating that the role of dAcsl in modulating lipoprotein secretion and systemic lipid homeostasis is possibly conserved in humans.
Collapse
Affiliation(s)
- Jie Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences , Beijing 100101, China
| | - Yue Dong
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences , Beijing 100101, China
| | - Tianxing Zhou
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences , Beijing 100101, China
| | - He Tian
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiahe Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yong Q Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences , Beijing 100101, China
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences , Beijing 100101, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- Lipidall Technologies Company Limited, Changzhou, Jiangsu 213000, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences , Beijing 100101, China
| |
Collapse
|
20
|
Kockel L, Zhang V, Wang J, Gulick C, Laws ME, Rajan A, Lantz N, Asgarova A, Dai L, Garcia K, Kim C, Li M, Ordonez-Acosta P, Peng D, Shull H, Tse L, Wang Y, Yu W, Zhou Z, Rankin A, Park S, Kim SK. CRISPR/Cas9 gene editing in Drosophila via visual selection in a summer classroom. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.28.587232. [PMID: 38585736 PMCID: PMC10996655 DOI: 10.1101/2024.03.28.587232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
CRISPR/Cas9 methods are a powerful in vivo approach to edit the genome of Drosophila melanogaster. To convert existing Drosophila GAL4 lines to LexA driver lines in a secondary school classroom setting, we applied the CRISPR-based genetic approach to a collection of Gal4 'driver' lines. The integration of the yellow+ coat color marker into homology-assisted CRISPR knock-in (HACK) enabled visual selection of Gal4-to-LexA conversions using brightfield stereo-microscopy available in a broader set of standard classrooms. Here, we report the successful conversion of eleven Gal4 lines with expression in neuropeptide-expressing cells into corresponding, novel LexA drivers. The conversion was confirmed by LexA- and Gal4-specific GFP reporter gene expression. This curriculum was successfully implemented in a summer course running 16 hours/week for seven weeks. The modularity, flexibility, and compactness of this course should enable development of similar classes in secondary schools and undergraduate curricula, to provide opportunities for experience-based science instruction, and university-secondary school collaborations that simultaneously fulfill research needs in the community of science.
Collapse
Affiliation(s)
- Lutz Kockel
- Stanford University, Stanford, CA 94305, USA
- Dept. of Developmental Biology, Stanford University SOM, Stanford, CA 94305, USA
| | | | - Jenna Wang
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | | | | | - Arjun Rajan
- Stanford University, Stanford, CA 94305, USA
- Dept. of Developmental Biology, Stanford University SOM, Stanford, CA 94305, USA
| | - Nicole Lantz
- The Lawrenceville School, Lawrenceville, NJ 08648, USA
| | | | - Lillian Dai
- Lexington High School, Lexington, MA 02421, USA
| | | | - Charlene Kim
- Busan International High School, Busan, South Korea
| | - Michelle Li
- Shenzhen Foreign Language School, Shenzhen, China
| | | | - Dongshen Peng
- University of North Carolina, Chapel Hill, NC 27599, USA
| | - Henry Shull
- Harvard University, Cambridge, MA 02138, USA
| | - Lauren Tse
- Hong Kong International School, Hong Kong, China
| | | | - Wenxin Yu
- Guangdong Country Garden School, Guangdong, China
| | - Zee Zhou
- Thomas Jefferson High School for Science and Technology, Alexandria, VA 22312, USA
| | - Anne Rankin
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | - Sangbin Park
- Grier School, Tyrone, PA 16686, USA
- Dept. of Developmental Biology, Stanford University SOM, Stanford, CA 94305, USA
| | - Seung K Kim
- Stanford University, Stanford, CA 94305, USA
- Dept. of Developmental Biology, Stanford University SOM, Stanford, CA 94305, USA
- Lexington High School, Lexington, MA 02421, USA
- Stanford Diabetes Research Center, Stanford, CA 94305, USA
| |
Collapse
|
21
|
Martelli F, Quig A, Mele S, Lin J, Fulton TL, Wansbrough M, Barlow CK, Schittenhelm RB, Johnson TK, Piper MDW. A defined diet for pre-adult Drosophila melanogaster. Sci Rep 2024; 14:6974. [PMID: 38521863 PMCID: PMC10960813 DOI: 10.1038/s41598-024-57681-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 03/20/2024] [Indexed: 03/25/2024] Open
Abstract
Drosophila melanogaster is unique among animal models because it has a fully defined synthetic diet available to study nutrient-gene interactions. However, use of this diet is limited to adult studies due to impaired larval development and survival. Here, we provide an adjusted formula that reduces the developmental period, restores fat levels, enhances body mass, and fully rescues survivorship without compromise to adult lifespan. To demonstrate an application of this formula, we explored pre-adult diet compositions of therapeutic potential in a model of an inherited metabolic disorder affecting the metabolism of branched-chain amino acids. We reveal rapid, specific, and predictable nutrient effects on the disease state consistent with observations from mouse and patient studies. Together, our diet provides a powerful means with which to examine the interplay between diet and metabolism across all life stages in an animal model.
Collapse
Affiliation(s)
- Felipe Martelli
- School of Biological Sciences, Monash University, Clayton, VIC, 3800, Australia
- School of BioSciences, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Annelise Quig
- School of Biological Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Sarah Mele
- School of Biological Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Jiayi Lin
- School of Biological Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Tahlia L Fulton
- School of Biological Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Mia Wansbrough
- School of Biological Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Christopher K Barlow
- Monash Proteomics and Metabolomics Platform, Monash Biomedicine Discovery Institute & Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Ralf B Schittenhelm
- Monash Proteomics and Metabolomics Platform, Monash Biomedicine Discovery Institute & Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Travis K Johnson
- School of Biological Sciences, Monash University, Clayton, VIC, 3800, Australia.
- Department of Biochemistry and Chemistry and La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia.
| | - Matthew D W Piper
- School of Biological Sciences, Monash University, Clayton, VIC, 3800, Australia.
| |
Collapse
|
22
|
Wang L, Zhang G, Li Q, Lu F, Yang K, Dai X. Carrageenan oligosaccharide alleviates intestinal damage via gut microflora through activating IMD/relish pathway in female Drosophila melanogaster. FASEB J 2024; 38:e23455. [PMID: 38308636 DOI: 10.1096/fj.202301218r] [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: 06/18/2023] [Revised: 01/07/2024] [Accepted: 01/17/2024] [Indexed: 02/05/2024]
Abstract
Recent evidence suggests the anti-inflammatory effect of carrageenan oligosaccharides (COS). The effects of COS on intestinal injury induced by 0.6% sodium dodecyl sulfate (SDS) and the molecular mechanisms involved were investigated in this study. 0.625, 1.25, and 2.5 mg/mL COS in diet had no toxic effect in flies, and they could all prolong SDS-treated female flies' survival rate. 1.25 mg/mL COS prevented the development of inflammation by improving the intestinal barrier integrity and maintaining the intestinal morphology stability, inhibited the proliferation of intestine stem cells (ISCs), and the production of lysosomes induced by SDS, accompanied by a decrease in the expression of autophagy-related genes. Moreover, COS decreased the active oxygen species (ROS) content in gut and increased the antioxidant activity in SDS-induced female flies, while COS still played a role in increasing survival rate and decreasing intestinal leakage in CncC-RNAi flies. The improvement of anti-inflammation capacity may be associated with the regulation of intestinal microflora with COS supplementation for Drosophila melanogaster. COS changed the gut microbiota composition, and COS had no effect on germ-free (GF) flies. It is highlighted that COS could not work in Relish-RNAi flies, indicating relish is required for COS to perform beneficial effects. These results provide insights into the study of gut microbiota interacting with COS to modulate intestinal inflammation in specific hosts.
Collapse
Affiliation(s)
- Lu Wang
- College of Life Sciences, China Jiliang University, Hangzhou, Zhejiang, China
| | - Guocai Zhang
- College of Life Sciences, China Jiliang University, Hangzhou, Zhejiang, China
| | - Qiaowei Li
- College of Life Sciences, China Jiliang University, Hangzhou, Zhejiang, China
| | - Fangyuan Lu
- College of Life Sciences, China Jiliang University, Hangzhou, Zhejiang, China
| | - Kun Yang
- College of Life Sciences, China Jiliang University, Hangzhou, Zhejiang, China
| | - Xianjun Dai
- College of Life Sciences, China Jiliang University, Hangzhou, Zhejiang, China
| |
Collapse
|
23
|
Rankin AE, Fox E, Chisholm T, Lantz N, Rajan A, Phillips W, Griffin E, Harper J, Suhr C, Tan M, Wang J, Yang A, Kim ES, Ankrah NKA, Chakraborty P, Lam ACK, Laws ME, Lee J, Park KK, Wesel E, Covert PH, Kockel L, Park S, Kim SK. Simplified homology-assisted CRISPR for gene editing in Drosophila. G3 (BETHESDA, MD.) 2024; 14:jkad277. [PMID: 38058125 PMCID: PMC10849607 DOI: 10.1093/g3journal/jkad277] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 08/28/2023] [Accepted: 10/29/2023] [Indexed: 12/08/2023]
Abstract
In vivo genome editing with clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 generates powerful tools to study gene regulation and function. We revised the homology-assisted CRISPR knock-in method to convert Drosophila GAL4 lines to LexA lines using a new universal knock-in donor strain. A balancer chromosome-linked donor strain with both body color (yellow) and eye red fluorescent protein (RFP) expression markers simplified the identification of LexA knock-in using light or fluorescence microscopy. A second balancer chromosome-linked donor strain readily converted the second chromosome-linked GAL4 lines regardless of target location in the cis-chromosome but showed limited success for the third chromosome-linked GAL4 lines. We observed a consistent and robust expression of the yellow transgene in progeny harboring a LexA knock-in at diverse genomic locations. Unexpectedly, the expression of the 3xP3-RFP transgene in the "dual transgene" cassette was significantly increased compared with that of the original single 3xP3-RFP transgene cassette in all tested genomic locations. Using this improved screening approach, we generated 16 novel LexA lines; tissue expression by the derived LexA and originating GAL4 lines was similar or indistinguishable. In collaboration with 2 secondary school classes, we also established a systematic workflow to generate a collection of LexA lines from frequently used GAL4 lines.
Collapse
Affiliation(s)
| | - Elizabeth Fox
- The Lawrenceville School, Lawrenceville, NJ 08648, USA
| | | | - Nicole Lantz
- The Lawrenceville School, Lawrenceville, NJ 08648, USA
| | - Arjun Rajan
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | | | | | | | | | - Max Tan
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | - Jason Wang
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | - Alana Yang
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | - Ella S Kim
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | | | | | | | | | - Jackson Lee
- The Lawrenceville School, Lawrenceville, NJ 08648, USA
| | - Kyle K Park
- The Lawrenceville School, Lawrenceville, NJ 08648, USA
| | - Emily Wesel
- Stanford University, Stanford, CA 94305, USA
| | | | - Lutz Kockel
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sangbin Park
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Seung K Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Diabetes Research Center, Stanford, CA 94305, USA
| |
Collapse
|
24
|
Saedi H, Waro G, Giacchetta L, Tsunoda S. miR-137 regulates PTP61F, affecting insulin signaling, metabolic homeostasis, and starvation resistance in Drosophila. Proc Natl Acad Sci U S A 2024; 121:e2319475121. [PMID: 38252824 PMCID: PMC10835047 DOI: 10.1073/pnas.2319475121] [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: 11/07/2023] [Accepted: 12/13/2023] [Indexed: 01/24/2024] Open
Abstract
miR-137 is a highly conserved brain-enriched microRNA (miRNA) that has been associated with neuronal function and proliferation. Here, we show that Drosophila miR-137 null mutants display increased body weight with enhanced triglyceride content and decreased locomotor activity. In addition, when challenged by nutrient deprivation, miR-137 mutants exhibit reduced motivation to feed and prolonged survival. We show through genetic epistasis and rescue experiments that this starvation resistance is due to a disruption in insulin signaling. Our studies further show that miR-137 null mutants exhibit a drastic reduction in levels of the phosphorylated/activated insulin receptor, InR (InR-P). We investigated if this is due to the predicted miR-137 target, Protein Tyrosine Phosphatase 61F (PTP61F), ortholog of mammalian TC-PTP/PTP1B, which are known to dephosphorylate InR-P. Indeed, levels of an endogenously tagged GFP-PTP61F are significantly elevated in miR-137 null mutants, and we show that overexpression of PTP61F alone is sufficient to mimic many of the metabolic phenotypes of miR-137 mutants. Finally, we knocked-down elevated levels of PTP61F in the miR-137 null mutant background and show that this rescues levels of InR-P, restores normal body weight and triglyceride content, starvation sensitivity, as well as attenuates locomotor and starvation-induced feeding defects. Our study supports a model in which miR-137 is critical for dampening levels of PTP61F, thereby maintaining normal insulin signaling and energy homeostasis.
Collapse
Affiliation(s)
- Hana Saedi
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO80523
| | - Girma Waro
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO80523
| | - Lea Giacchetta
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO80523
| | - Susan Tsunoda
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO80523
| |
Collapse
|
25
|
Heidarian Y, Tourigny JP, Fasteen TD, Mahmoudzadeh NH, Hurlburt AJ, Nemkov T, Reisz JA, D’Alessandro A, Tennessen JM. Metabolomic analysis of Drosophila melanogaster larvae lacking pyruvate kinase. G3 (BETHESDA, MD.) 2023; 14:jkad228. [PMID: 37792629 PMCID: PMC10755183 DOI: 10.1093/g3journal/jkad228] [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: 06/02/2023] [Revised: 06/02/2023] [Accepted: 09/24/2023] [Indexed: 10/06/2023]
Abstract
Pyruvate kinase (Pyk) is a rate-limiting enzyme that catalyzes the final metabolic reaction in glycolysis. The importance of this enzyme, however, extends far beyond ATP production, as Pyk is also known to regulate tissue growth, cell proliferation, and development. Studies of this enzyme in Drosophila melanogaster are complicated by the fact that the fly genome encodes 6 Pyk paralogs whose functions remain poorly defined. To address this issue, we used sequence distance and phylogenetic approaches to demonstrate that the gene Pyk encodes the enzyme most similar to the mammalian Pyk orthologs, while the other 5 Drosophila Pyk paralogs have significantly diverged from the canonical enzyme. Consistent with this observation, metabolomic studies of 2 different Pyk mutant strains revealed that larvae lacking Pyk exhibit a severe block in glycolysis, with a buildup of glycolytic intermediates upstream of pyruvate. However, our analysis also unexpectedly reveals that pyruvate levels are unchanged in Pyk mutants, indicating that larval metabolism maintains pyruvate pool size despite severe metabolic limitations. Consistent with our metabolomic findings, a complementary RNA-seq analysis revealed that genes involved in lipid metabolism and protease activity are elevated in Pyk mutants, again indicating that loss of this glycolytic enzyme induces compensatory changes in other aspects of metabolism. Overall, our study provides both insight into how Drosophila larval metabolism adapts to disruption of glycolytic metabolism as well as immediate clinical relevance, considering that Pyk deficiency is the most common congenital enzymatic defect in humans.
Collapse
Affiliation(s)
- Yasaman Heidarian
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Jason P Tourigny
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Tess D Fasteen
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | | | | | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Jason M Tennessen
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
26
|
Holcombe J, Weavers H. Functional-metabolic coupling in distinct renal cell types coordinates organ-wide physiology and delays premature ageing. Nat Commun 2023; 14:8405. [PMID: 38110414 PMCID: PMC10728150 DOI: 10.1038/s41467-023-44098-x] [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: 05/26/2023] [Accepted: 11/30/2023] [Indexed: 12/20/2023] Open
Abstract
Precise coupling between cellular physiology and metabolism is emerging as a vital relationship underpinning tissue health and longevity. Nevertheless, functional-metabolic coupling within heterogenous microenvironments in vivo remains poorly understood due to tissue complexity and metabolic plasticity. Here, we establish the Drosophila renal system as a paradigm for linking mechanistic analysis of metabolism, at single-cell resolution, to organ-wide physiology. Kidneys are amongst the most energetically-demanding organs, yet exactly how individual cell types fine-tune metabolism to meet their diverse, unique physiologies over the life-course remains unclear. Integrating live-imaging of metabolite and organelle dynamics with spatio-temporal genetic perturbation within intact functional tissue, we uncover distinct cellular metabolic signatures essential to support renal physiology and healthy ageing. Cell type-specific programming of glucose handling, PPP-mediated glutathione regeneration and FA β-oxidation via dynamic lipid-peroxisomal networks, downstream of differential ERR receptor activity, precisely match cellular energetic demands whilst limiting damage and premature senescence; however, their dramatic dysregulation may underlie age-related renal dysfunction.
Collapse
Affiliation(s)
- Jack Holcombe
- School of Biochemistry, Biomedical Sciences, University of Bristol, Bristol, BS8 1TD, UK
| | - Helen Weavers
- School of Biochemistry, Biomedical Sciences, University of Bristol, Bristol, BS8 1TD, UK.
| |
Collapse
|
27
|
Li X, Karpac J. A distinct Acyl-CoA binding protein (ACBP6) shapes tissue plasticity during nutrient adaptation in Drosophila. Nat Commun 2023; 14:7599. [PMID: 37989752 PMCID: PMC10663470 DOI: 10.1038/s41467-023-43362-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 11/08/2023] [Indexed: 11/23/2023] Open
Abstract
Nutrient availability is a major selective force in the evolution of metazoa, and thus plasticity in tissue function and morphology is shaped by adaptive responses to nutrient changes. Utilizing Drosophila, we reveal that distinct calibration of acyl-CoA metabolism, mediated by Acbp6 (Acyl-CoA binding-protein 6), is critical for nutrient-dependent tissue plasticity. Drosophila Acbp6, which arose by evolutionary duplication and binds acyl-CoA to tune acetyl-CoA metabolism, is required for intestinal resizing after nutrient deprivation through activating intestinal stem cell proliferation from quiescence. Disruption of acyl-CoA metabolism by Acbp6 attenuation drives aberrant 'switching' of metabolic networks in intestinal enterocytes during nutrient adaptation, impairing acetyl-CoA metabolism and acetylation amid intestinal resizing. We also identified STAT92e, whose function is influenced by acetyl-CoA levels, as a key regulator of acyl-CoA and nutrient-dependent changes in stem cell activation. These findings define a regulatory mechanism, shaped by acyl-CoA metabolism, that adjusts proliferative homeostasis to coordinately regulate tissue plasticity during nutrient adaptation.
Collapse
Affiliation(s)
- Xiaotong Li
- Department of Cell Biology and Genetics, Texas A&M University, School of Medicine, Bryan, TX, USA
| | - Jason Karpac
- Department of Cell Biology and Genetics, Texas A&M University, School of Medicine, Bryan, TX, USA.
| |
Collapse
|
28
|
Catalani E, Del Quondam S, Brunetti K, Cherubini A, Bongiorni S, Taddei AR, Zecchini S, Giovarelli M, De Palma C, Perrotta C, Clementi E, Prantera G, Cervia D. Neuroprotective role of plumbagin on eye damage induced by high-sucrose diet in adult fruit fly Drosophila melanogaster. Biomed Pharmacother 2023; 166:115298. [PMID: 37597318 DOI: 10.1016/j.biopha.2023.115298] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/03/2023] [Accepted: 08/05/2023] [Indexed: 08/21/2023] Open
Abstract
The natural compound plumbagin has a wide range of pharmacological and potential therapeutic activities, although its role in neuroretina degeneration is unknown. Here we evaluated the effects of plumbagin on retina homeostasis of the fruit fly Drosophila melanogaster fed with high glucose diet, a model of hyperglycemia-induced eye impairment to study the pathophysiology of diabetic retinopathy at the early stages. To this aim, the visual system of flies orally administered with plumbagin has been analyzed at structural, functional, and molecular/cellular level as for instance neuronal apoptosis/autophagy dysregulation and oxidative stress-related signals. Our results demonstrated that plumbagin ameliorates the visual performance of hyperglycemic flies. Drosophila eye-structure, clearly altered by hyperglycemia, i.e. defects of the pattern of ommatidia, irregular rhabdomeres, vacuoles, damaged mitochondria, and abnormal phototransduction units were rescued, at least in part, by plumbagin. In addition, it reactivated autophagy, decreased the presence of cell death/apoptotic features, and exerted antioxidant effects in the retina. In terms of mechanisms favoring death/survival ratio, Nrf2 signaling activation may be one of the strategies by which plumbagin reduced redox unbalance mainly increasing the levels of glutathione-S-transferase. Likewise, plumbagin may act additively and/or synergistically inhibiting the mitochondrial-endoplasmic reticulum stress and unfolded protein response pathways, which prevented neuronal impairment and eye damage induced by reactive oxygen species. These results provide an avenue for further studies, which may be helpful to develop novel therapeutic candidates and drug targets against eye neurotoxicity by high glucose, a key aspect in retinal complications of diabetes.
Collapse
Affiliation(s)
- Elisabetta Catalani
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Simona Del Quondam
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Kashi Brunetti
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Agnese Cherubini
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Silvia Bongiorni
- Department of Ecological and Biological Sciences (DEB), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Anna Rita Taddei
- Section of Electron Microscopy, Great Equipment Center, Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Silvia Zecchini
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, via G.B. Grassi 74, 20157 Milano, Italy
| | - Matteo Giovarelli
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, via G.B. Grassi 74, 20157 Milano, Italy
| | - Clara De Palma
- Department of Medical Biotechnology and Translational Medicine (BioMeTra), Università degli Studi di Milano, via L. Vanvitelli 32, 20129 Milano, Italy
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, via G.B. Grassi 74, 20157 Milano, Italy
| | - Emilio Clementi
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, via G.B. Grassi 74, 20157 Milano, Italy; Scientific Institute IRCCS "Eugenio Medea", via Don Luigi Monza 20, 23842 Bosisio Parini, Italy
| | - Giorgio Prantera
- Department of Ecological and Biological Sciences (DEB), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Davide Cervia
- Department for Innovation in Biological, Agro-food and Forest systems (DIBAF), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy.
| |
Collapse
|
29
|
Kim ES, Rajan A, Chang K, Govindarajan S, Gulick C, English E, Rodriguez B, Bloomfield O, Nakada S, Beard C, O’Connor S, Mastroianni S, Downey E, Feigenbaum M, Tolentino C, Pace A, Khan M, Moon S, DiPrima J, Syed A, Lin F, Abukhadra Y, Bacon I, Beckerle J, Cho S, Donkor NE, Garberg L, Harrington A, Hoang M, Lawani N, Noori A, Park E, Parsons E, Oravitan P, Chen M, Molina C, Richmond C, Reddi A, Huang J, Shugrue C, Coviello R, Unver S, Indelicarto M, Islamovic E, McIlroy R, Yang A, Hamad M, Griffin E, Ahmed Z, Alla A, Fitzgerald P, Choi A, Das T, Cheng Y, Yu J, Roderiques T, Lee E, Liu L, Harper J, Wang J, Suhr C, Tan M, Luque J, Tam AR, Chen E, Triff M, Zimmermann L, Zhang E, Wood J, Clark K, Kpodonu N, Dey A, Ecker A, Chuang M, López RKS, Sun H, Wei Z, Stone H, Chi CYJ, Silvestri A, Orloff P, Nedumaran N, Zou A, Ünver L, Page O, Kim M, Chan TYT, Tulloch A, Hernandez A, Pillai A, Chen C, Chowdhury N, Huang L, Mudide A, Paik G, Wingate A, Quinn L, Conybere C, Baumgardt LL, Buckley R, et alKim ES, Rajan A, Chang K, Govindarajan S, Gulick C, English E, Rodriguez B, Bloomfield O, Nakada S, Beard C, O’Connor S, Mastroianni S, Downey E, Feigenbaum M, Tolentino C, Pace A, Khan M, Moon S, DiPrima J, Syed A, Lin F, Abukhadra Y, Bacon I, Beckerle J, Cho S, Donkor NE, Garberg L, Harrington A, Hoang M, Lawani N, Noori A, Park E, Parsons E, Oravitan P, Chen M, Molina C, Richmond C, Reddi A, Huang J, Shugrue C, Coviello R, Unver S, Indelicarto M, Islamovic E, McIlroy R, Yang A, Hamad M, Griffin E, Ahmed Z, Alla A, Fitzgerald P, Choi A, Das T, Cheng Y, Yu J, Roderiques T, Lee E, Liu L, Harper J, Wang J, Suhr C, Tan M, Luque J, Tam AR, Chen E, Triff M, Zimmermann L, Zhang E, Wood J, Clark K, Kpodonu N, Dey A, Ecker A, Chuang M, López RKS, Sun H, Wei Z, Stone H, Chi CYJ, Silvestri A, Orloff P, Nedumaran N, Zou A, Ünver L, Page O, Kim M, Chan TYT, Tulloch A, Hernandez A, Pillai A, Chen C, Chowdhury N, Huang L, Mudide A, Paik G, Wingate A, Quinn L, Conybere C, Baumgardt LL, Buckley R, Kolberg Z, Pattison R, Shazli AA, Ganske P, Sfragara L, Strub A, Collier B, Tamana H, Ravindran D, Howden J, Stewart M, Shimizu S, Braniff J, Fong M, Gutman L, Irvine D, Malholtra S, Medina J, Park J, Yin A, Abromavage H, Barrett B, Chen J, Cho R, Dilatush M, Gaw G, Gu C, Huang J, Kilby H, Markel E, McClure K, Phillips W, Polaski B, Roselli A, Saint-Cyr S, Shin E, Tatum K, Tumpunyawat T, Wetherill L, Ptaszynska S, Zeleznik M, Pesendorfer A, Nolan A, Tao J, Sammeta D, Nicholson L, Dinh GV, Foltz M, Vo A, Ross M, Tokarski A, Hariharan S, Wang E, Baziuk M, Tay A, Wong YHM, Floyd J, Cui A, Pierre K, Coppisetti N, Kutam M, Khurjekar D, Gadzi A, Gubbay B, Pedretti S, Belovich S, Yeung T, Fey M, Shaffer L, Li A, Beritela G, Huyghue K, Foster G, Durso-Finley G, Thierfelder Q, Kiernan H, Lenkowsky A, Thomas T, Cheng N, Chao O, L’Etoile-Goga P, King A, McKinley P, Read N, Milberg D, Lin L, Wong M, Gilman I, Brown S, Chen L, Kosai J, Verbinsky M, Belshaw-Hood A, Lee H, Zhou C, Lobo M, Tse A, Tran K, Lewis K, Sonawane P, Ngo J, Zuzga S, Chow L, Huynh V, Yang W, Lim S, Stites B, Chang S, Cruz-Balleza R, Pelta M, Kujawski S, Yuan C, Standen-Bloom E, Witt O, Anders K, Duane A, Huynh N, Lester B, Fung-Lee S, Fung M, Situ M, Canigiula P, Dijkgraaf M, Romero W, Baula SK, Wong K, Xu I, Martinez B, Nuygen R, Norris L, Nijensohn N, Altman N, Maajid E, Burkhardt O, Chanda J, Doscher C, Gopal A, Good A, Good J, Herrera N, Lanting L, Liem S, Marks A, McLaughlin E, Lee A, Mohr C, Patton E, Pyarali N, Oczon C, Richards D, Good N, Goss S, Khan A, Madonia R, Mitchell V, Sun N, Vranka T, Garcia D, Arroyo F, Morales E, Camey S, Cano G, Bernabe A, Arroyo J, Lopez Y, Gonzalez E, Zumba B, Garcia J, Vargas E, Trinidad A, Candelaria N, Valdez V, Campuzano F, Pereznegron E, Medrano J, Gutierrez J, Gutierrez E, Abrego ET, Gutierrez D, Ortiz C, Barnes A, Arms E, Mitchell L, Balanzá C, Bradford J, Detroy H, Ferguson D, Guillermo E, Manapragada A, Nanula D, Serna B, Singh K, Sramaty E, Wells B, Wiggins M, Dowling M, Schmadeke G, Cafferky S, Good S, Reese M, Fleig M, Gannett A, Cain C, Lee M, Oberto P, Rinehart J, Pan E, Mathis SA, Joiner J, Barr L, Evans CJ, Baena-Lopez A, Beatty A, Collette J, Smullen R, Suttie J, Chisholm T, Rotondo C, Lewis G, Turner V, Stark L, Fox E, Amirapu A, Park S, Lantz N, Rankin AE, Kim SK, Kockel L. Generation of LexA enhancer-trap lines in Drosophila by an international scholastic network. G3 (BETHESDA, MD.) 2023; 13:jkad124. [PMID: 37279923 PMCID: PMC10468311 DOI: 10.1093/g3journal/jkad124] [Show More Authors] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 05/05/2023] [Accepted: 05/10/2023] [Indexed: 06/08/2023]
Abstract
Conditional gene regulation in Drosophila through binary expression systems like the LexA-LexAop system provides a superb tool for investigating gene and tissue function. To increase the availability of defined LexA enhancer trap insertions, we present molecular, genetic, and tissue expression studies of 301 novel Stan-X LexA enhancer traps derived from mobilization of the index SX4 line. This includes insertions into distinct loci on the X, II, and III chromosomes that were not previously associated with enhancer traps or targeted LexA constructs, an insertion into ptc, and seventeen insertions into natural transposons. A subset of enhancer traps was expressed in CNS neurons known to produce and secrete insulin, an essential regulator of growth, development, and metabolism. Fly lines described here were generated and characterized through studies by students and teachers in an international network of genetics classes at public, independent high schools, and universities serving a diversity of students, including those underrepresented in science. Thus, a unique partnership between secondary schools and university-based programs has produced and characterized novel resources in Drosophila, establishing instructional paradigms devoted to unscripted experimental science.
Collapse
Affiliation(s)
- Ella S Kim
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | - Arjun Rajan
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kathleen Chang
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | - Eva English
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | | | | | | | | | - Sarah O’Connor
- Commack High School, 1 Scholar Ln, Commack, NY 11725, USA
| | | | - Emma Downey
- Commack High School, 1 Scholar Ln, Commack, NY 11725, USA
| | | | | | - Abigail Pace
- Commack High School, 1 Scholar Ln, Commack, NY 11725, USA
| | - Marina Khan
- Commack High School, 1 Scholar Ln, Commack, NY 11725, USA
| | - Soyoun Moon
- Commack High School, 1 Scholar Ln, Commack, NY 11725, USA
| | - Jordan DiPrima
- Commack High School, 1 Scholar Ln, Commack, NY 11725, USA
| | - Amber Syed
- Commack High School, 1 Scholar Ln, Commack, NY 11725, USA
| | - Flora Lin
- Commack High School, 1 Scholar Ln, Commack, NY 11725, USA
| | | | | | | | - Sophia Cho
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | | | | | | | - Mai Hoang
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | - Nosa Lawani
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | - Ayush Noori
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | - Euwie Park
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | | | | | | | | | | | - Adith Reddi
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | - Jason Huang
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | | | | | - Selma Unver
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | | | | | | | - Alana Yang
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | - Mahdi Hamad
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | | | - Zara Ahmed
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | - Asha Alla
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | | | - Audrey Choi
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | - Tanya Das
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | | | - Joshua Yu
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | | | - Ethan Lee
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | | | | | - Jason Wang
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | - Chris Suhr
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | - Max Tan
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | | | | | - Emma Chen
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | - Max Triff
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | | | - Eric Zhang
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | - Jackie Wood
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | | | - Nat Kpodonu
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | - Antar Dey
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | | | | | | | - Harry Sun
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | - Zijing Wei
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | - Henry Stone
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | | | | | | | | | | | - Leyla Ünver
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | - Oscair Page
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | - Minseo Kim
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | | | | | | | | | | | | | - Lina Huang
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | | | | | | | - Lily Quinn
- Haileybury School, Hertford SG13 7NU, UK
| | | | | | | | | | | | | | - Pia Ganske
- Haileybury School, Hertford SG13 7NU, UK
| | | | | | | | | | | | | | | | | | - Julia Braniff
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Melanie Fong
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Lucy Gutman
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Danny Irvine
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Sahil Malholtra
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Jillian Medina
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - John Park
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Alicia Yin
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | | | - Breanna Barrett
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Jacqueline Chen
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Rachelle Cho
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Mac Dilatush
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Gabriel Gaw
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Caitlin Gu
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Jupiter Huang
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Houston Kilby
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Ethan Markel
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Katie McClure
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - William Phillips
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Benjamin Polaski
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Amelia Roselli
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Soleil Saint-Cyr
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Ellie Shin
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Kylan Tatum
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Tai Tumpunyawat
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Lucia Wetherill
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Sara Ptaszynska
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Maddie Zeleznik
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | | | - Anna Nolan
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Jeffrey Tao
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Divya Sammeta
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Laney Nicholson
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Giao Vu Dinh
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Merrin Foltz
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - An Vo
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Maggie Ross
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Andrew Tokarski
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Samika Hariharan
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Elaine Wang
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Martha Baziuk
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Ashley Tay
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | | | - Jax Floyd
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Aileen Cui
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Kieran Pierre
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Nikita Coppisetti
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Matthew Kutam
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Dhruv Khurjekar
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Anthony Gadzi
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Ben Gubbay
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Sophia Pedretti
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Sofiya Belovich
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Tiffany Yeung
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Mercy Fey
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Layla Shaffer
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Arthur Li
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | | | - Kyle Huyghue
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Greg Foster
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | | | - Quinn Thierfelder
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Holly Kiernan
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Andrew Lenkowsky
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Tesia Thomas
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Nicole Cheng
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Olivia Chao
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Pia L’Etoile-Goga
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Alexa King
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Paris McKinley
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Nicole Read
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - David Milberg
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Leila Lin
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Melinda Wong
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Io Gilman
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Samantha Brown
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Lila Chen
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Jordyn Kosai
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Mark Verbinsky
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | | | - Honon Lee
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Cathy Zhou
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Maya Lobo
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Asia Tse
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Kyle Tran
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Kira Lewis
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Pratmesh Sonawane
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Jonathan Ngo
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Sophia Zuzga
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Lillian Chow
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Vianne Huynh
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Wenyi Yang
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Samantha Lim
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Brandon Stites
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Shannon Chang
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | | | - Michaela Pelta
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Stella Kujawski
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Christopher Yuan
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | | | - Oliver Witt
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Karina Anders
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Audrey Duane
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Nancy Huynh
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Benjamin Lester
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Samantha Fung-Lee
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Melanie Fung
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Mandy Situ
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Paolo Canigiula
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Matijs Dijkgraaf
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Wilbert Romero
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | | | - Kimberly Wong
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Ivana Xu
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | | | - Reena Nuygen
- Latin School of Chicago, 59 W North Blvd, Chicago, IL 60610, USA
| | - Lucy Norris
- Latin School of Chicago, 59 W North Blvd, Chicago, IL 60610, USA
| | - Noah Nijensohn
- Latin School of Chicago, 59 W North Blvd, Chicago, IL 60610, USA
| | - Naomi Altman
- Latin School of Chicago, 59 W North Blvd, Chicago, IL 60610, USA
| | - Elise Maajid
- Latin School of Chicago, 59 W North Blvd, Chicago, IL 60610, USA
| | | | | | | | - Alex Gopal
- Albuquerque Academy, Albuquerque, NM 87109, USA
| | - Aaron Good
- Albuquerque Academy, Albuquerque, NM 87109, USA
| | - Jonah Good
- Albuquerque Academy, Albuquerque, NM 87109, USA
| | | | | | - Sophia Liem
- Albuquerque Academy, Albuquerque, NM 87109, USA
| | - Anila Marks
- Albuquerque Academy, Albuquerque, NM 87109, USA
| | | | - Audrey Lee
- Albuquerque Academy, Albuquerque, NM 87109, USA
| | - Collin Mohr
- Albuquerque Academy, Albuquerque, NM 87109, USA
| | - Emma Patton
- Albuquerque Academy, Albuquerque, NM 87109, USA
| | | | | | | | - Nathan Good
- Albuquerque Academy, Albuquerque, NM 87109, USA
| | | | - Adeeb Khan
- Albuquerque Academy, Albuquerque, NM 87109, USA
| | | | | | - Natasha Sun
- Albuquerque Academy, Albuquerque, NM 87109, USA
| | | | | | | | | | | | | | | | | | | | | | - Bryan Zumba
- Pritzker College Prep, Chicago, IL 60639, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Jake Bradford
- Loyola Marymount University, Los Angeles, CA 90045, USA
| | | | | | | | | | | | | | - Khushi Singh
- Loyola Marymount University, Los Angeles, CA 90045, USA
| | - Emily Sramaty
- Loyola Marymount University, Los Angeles, CA 90045, USA
| | - Brian Wells
- Loyola Marymount University, Los Angeles, CA 90045, USA
| | | | - Melissa Dowling
- Latin School of Chicago, 59 W North Blvd, Chicago, IL 60610, USA
| | | | | | | | | | | | | | - Cory Cain
- Pritzker College Prep, Chicago, IL 60639, USA
| | - Melody Lee
- Harvard-Westlake School, Los Angeles, CA 90077, USA
| | | | | | | | | | | | - Leslie Barr
- Westtown School, West Chester, PA 19382, USA
| | - Cory J Evans
- Loyola Marymount University, Los Angeles, CA 90045, USA
| | | | - Andrea Beatty
- Commack High School, 1 Scholar Ln, Commack, NY 11725, USA
| | | | - Robert Smullen
- Commack High School, 1 Scholar Ln, Commack, NY 11725, USA
| | - Jeanne Suttie
- Commack High School, 1 Scholar Ln, Commack, NY 11725, USA
| | | | | | | | | | | | - Elizabeth Fox
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | - Anjana Amirapu
- Lowell High School, 1101 Eucalyptus Dr, San Francisco, CA 94132, USA
| | - Sangbin Park
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nicole Lantz
- The Lawrenceville School, 2500 Main St, Lawrenceville, NJ 08648, USA
| | | | - Seung K Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lutz Kockel
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
30
|
Demir E, Kacew S. Drosophila as a Robust Model System for Assessing Autophagy: A Review. TOXICS 2023; 11:682. [PMID: 37624187 PMCID: PMC10458868 DOI: 10.3390/toxics11080682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/07/2023] [Accepted: 08/07/2023] [Indexed: 08/26/2023]
Abstract
Autophagy is the process through which a body breaks down and recycles its own cellular components, primarily inside lysosomes. It is a cellular response to starvation and stress, which plays decisive roles in various biological processes such as senescence, apoptosis, carcinoma, and immune response. Autophagy, which was first discovered as a survival mechanism during starvation in yeast, is now known to serve a wide range of functions in more advanced organisms. It plays a vital role in how cells respond to stress, starvation, and infection. While research on yeast has led to the identification of many key components of the autophagy process, more research into autophagy in more complex systems is still warranted. This review article focuses on the use of the fruit fly Drosophila melanogaster as a robust testing model in further research on autophagy. Drosophila provides an ideal environment for exploring autophagy in a living organism during its development. Additionally, Drosophila is a well-suited compact tool for genetic analysis in that it serves as an intermediate between yeast and mammals because evolution conserved the molecular machinery required for autophagy in this species. Experimental tractability of host-pathogen interactions in Drosophila also affords great convenience in modeling human diseases on analogous structures and tissues.
Collapse
Affiliation(s)
- Esref Demir
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
- Medical Laboratory Techniques Program, Department of Medical Services and Techniques, Vocational School of Health Services, Antalya Bilim University, 07190 Antalya, Turkey
| | - Sam Kacew
- R. Samuel McLaughllin Center for Population Health Risk Assessment, Institute of Population Health, University of Ottawa, 1 Stewart (320), Ottawa, ON K1N 6N5, Canada;
| |
Collapse
|
31
|
Heidarian Y, Tourigny JP, Fasteen TD, Mahmoudzadeh NH, Hurlburt AJ, Nemkov T, Reisz JA, D'Alessandro A, Tennessen JM. Metabolomic analysis of Drosophila melanogaster larvae lacking Pyruvate kinase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.05.543743. [PMID: 37333180 PMCID: PMC10274742 DOI: 10.1101/2023.06.05.543743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Pyruvate kinase (Pyk) is a rate-limiting enzyme that catalyzes the final metabolic reaction in glycolysis. The importance of this enzyme, however, extends far beyond ATP production, as Pyk is also known to regulate tissue growth, cell proliferation, and development. Studies of this enzyme in Drosophila melanogaster , however, are complicated by the fact that the fly genome encodes six Pyk paralogs whose functions remain poorly defined. To address this issue, we used sequence distance and phylogenetic approaches to demonstrate that the gene Pyk encodes the enzyme most similar to the mammalian Pyk orthologs, while the other five Drosophila Pyk paralogs have significantly diverged from the canonical enzyme. Consistent with this observation, metabolomic studies of two different Pyk mutant backgrounds revealed that larvae lacking Pyk exhibit a severe block in glycolysis, with a buildup of glycolytic intermediates upstream of pyruvate. However, our analysis also unexpectedly reveals that steady state pyruvate levels are unchanged in Pyk mutants, indicating that larval metabolism maintains pyruvate pool size despite severe metabolic limitations. Consistent with our metabolomic findings, a complementary RNA-seq analysis revealed that genes involved in lipid metabolism and peptidase activity are elevated in Pyk mutants, again indicating that loss of this glycolytic enzyme induces compensatory changes in other aspects of metabolism. Overall, our study provides both insight into how Drosophila larval metabolism adapts to disruption of glycolytic metabolism as well as immediate clinical relevance, considering that Pyk deficiency is the most common congenital enzymatic defect in humans.
Collapse
Affiliation(s)
- Yasaman Heidarian
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Jason P Tourigny
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Tess D Fasteen
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | | | | | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Jason M Tennessen
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
32
|
Song T, Qin W, Lai Z, Li H, Li D, Wang B, Deng W, Wang T, Wang L, Huang R. Dietary cysteine drives body fat loss via FMRFamide signaling in Drosophila and mouse. Cell Res 2023; 33:434-447. [PMID: 37055592 PMCID: PMC10235132 DOI: 10.1038/s41422-023-00800-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 03/17/2023] [Indexed: 04/15/2023] Open
Abstract
Obesity imposes a global health threat and calls for safe and effective therapeutic options. Here, we found that protein-rich diet significantly reduced body fat storage in fruit flies, which was largely attributed to dietary cysteine intake. Mechanistically, dietary cysteine increased the production of a neuropeptide FMRFamide (FMRFa). Enhanced FMRFa activity simultaneously promoted energy expenditure and suppressed food intake through its cognate receptor (FMRFaR), both contributing to the fat loss effect. In the fat body, FMRFa signaling promoted lipolysis by increasing PKA and lipase activity. In sweet-sensing gustatory neurons, FMRFa signaling suppressed appetitive perception and hence food intake. We also demonstrated that dietary cysteine worked in a similar way in mice via neuropeptide FF (NPFF) signaling, a mammalian RFamide peptide. In addition, dietary cysteine or FMRFa/NPFF administration provided protective effect against metabolic stress in flies and mice without behavioral abnormalities. Therefore, our study reveals a novel target for the development of safe and effective therapies against obesity and related metabolic diseases.
Collapse
Affiliation(s)
- Tingting Song
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Wusa Qin
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Zeliang Lai
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, China
| | - Haoyu Li
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, China
| | - Daihan Li
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, China
| | - Baojia Wang
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Wuquan Deng
- Department of Endocrinology and Nephrology, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, China
| | - Tingzhang Wang
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Liming Wang
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China.
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Rui Huang
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, China.
| |
Collapse
|
33
|
Nikolopoulos N, Matos RC, Ravaud S, Courtin P, Akherraz H, Palussiere S, Gueguen-Chaignon V, Salomon-Mallet M, Guillot A, Guerardel Y, Chapot-Chartier MP, Grangeasse C, Leulier F. Structure-function analysis of Lactiplantibacillus plantarum DltE reveals D-alanylated lipoteichoic acids as direct cues supporting Drosophila juvenile growth. eLife 2023; 12:e84669. [PMID: 37042660 PMCID: PMC10241514 DOI: 10.7554/elife.84669] [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: 11/03/2022] [Accepted: 04/11/2023] [Indexed: 04/13/2023] Open
Abstract
Metazoans establish mutually beneficial interactions with their resident microorganisms. However, our understanding of the microbial cues contributing to host physiology remains elusive. Previously, we identified a bacterial machinery encoded by the dlt operon involved in Drosophila melanogaster's juvenile growth promotion by Lactiplantibacillus plantarum. Here, using crystallography combined with biochemical and cellular approaches, we investigate the physiological role of an uncharacterized protein (DltE) encoded by this operon. We show that lipoteichoic acids (LTAs) but not wall teichoic acids are D-alanylated in Lactiplantibacillus plantarumNC8 cell envelope and demonstrate that DltE is a D-Ala carboxyesterase removing D-Ala from LTA. Using the mutualistic association of L. plantarumNC8 and Drosophila melanogaster as a symbiosis model, we establish that D-alanylated LTAs (D-Ala-LTAs) are direct cues supporting intestinal peptidase expression and juvenile growth in Drosophila. Our results pave the way to probing the contribution of D-Ala-LTAs to host physiology in other symbiotic models.
Collapse
Affiliation(s)
- Nikos Nikolopoulos
- Molecular Microbiology and Structural Biochemistry, CNRS UMR 5086, Université Claude Bernard Lyon 1LyonFrance
| | - Renata C Matos
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, CNRS UMR 5242, Université Claude Bernard Lyon 1LyonFrance
| | - Stephanie Ravaud
- Molecular Microbiology and Structural Biochemistry, CNRS UMR 5086, Université Claude Bernard Lyon 1LyonFrance
| | - Pascal Courtin
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis InstituteJouy-en-JosasFrance
| | - Houssam Akherraz
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, CNRS UMR 5242, Université Claude Bernard Lyon 1LyonFrance
| | - Simon Palussiere
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis InstituteJouy-en-JosasFrance
| | - Virginie Gueguen-Chaignon
- Protein Science Facility, CNRS UAR3444, INSERM US8, Université Claude Bernard Lyon 1, Ecole Normale Supérieur de LyonLyonFrance
| | - Marie Salomon-Mallet
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis InstituteJouy-en-JosasFrance
| | - Alain Guillot
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis InstituteJouy-en-JosasFrance
| | - Yann Guerardel
- Institute for Glyco-core Research (iGCORE), Gifu UniversityGifuJapan
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et FonctionnelleLilleFrance
| | | | - Christophe Grangeasse
- Molecular Microbiology and Structural Biochemistry, CNRS UMR 5086, Université Claude Bernard Lyon 1LyonFrance
| | - François Leulier
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, CNRS UMR 5242, Université Claude Bernard Lyon 1LyonFrance
| |
Collapse
|
34
|
Schoofs A, Pankratz MJ. Neuroscience: Moving thoughts control insulin release. Curr Biol 2023; 33:R274-R276. [PMID: 37040711 DOI: 10.1016/j.cub.2023.02.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
Insulin release has mostly been studied in the context of metabolic signals. An electrophysiology approach in Drosophila now reveals regulation of insulin-producing cell activity by neuronal circuits controlling locomotion. Even without actual movement, activating these circuits is sufficient to inhibit neuropeptide release.
Collapse
Affiliation(s)
- Andreas Schoofs
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Michael J Pankratz
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany.
| |
Collapse
|
35
|
Rideout EJ, Tennessen JM. Editorial. Semin Cell Dev Biol 2023; 138:81-82. [PMID: 35970667 DOI: 10.1016/j.semcdb.2022.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Affiliation(s)
- Elizabeth J Rideout
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver V6T 1Z3, Canada
| | - Jason M Tennessen
- Department of Biology, Indiana University, Bloomington, IN 47404, USA.
| |
Collapse
|
36
|
Mele S, Martelli F, Lin J, Kanca O, Christodoulou J, Bellen HJ, Piper MDW, Johnson TK. Drosophila as a diet discovery tool for treating amino acid disorders. Trends Endocrinol Metab 2023; 34:85-105. [PMID: 36567227 DOI: 10.1016/j.tem.2022.12.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
Amino acid disorders (AADs) are a large group of rare inherited conditions that collectively impact one in 6500 live births, often resulting in rapid neurological decline and death during infancy. For several AADs, including phenylketonuria, dietary modification prevents physiological deterioration and ameliorates symptoms. Despite this remarkable potential for treatment success, dietary therapy for most AADs remains largely unexplored. Although animal models have provided novel insights into AAD mechanisms, few have been used for therapeutic diet discovery. Here, we find that of all the animal models, Drosophila is particularly well suited for nutrigenomic disease modelling, having amino acid pathways conserved with humans, exceptional genetic tractability, and the unique availability of a synthetic customisable diet.
Collapse
Affiliation(s)
- Sarah Mele
- School of Biological Sciences, Monash University, Clayton, Victoria 3800, Australia
| | - Felipe Martelli
- School of Biological Sciences, Monash University, Clayton, Victoria 3800, Australia
| | - Jiayi Lin
- School of Biological Sciences, Monash University, Clayton, Victoria 3800, Australia
| | - Oguz Kanca
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Duncan Neurological Research Institute of Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - John Christodoulou
- Murdoch Children's Research Institute, Parkville, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Duncan Neurological Research Institute of Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Matthew D W Piper
- School of Biological Sciences, Monash University, Clayton, Victoria 3800, Australia.
| | - Travis K Johnson
- School of Biological Sciences, Monash University, Clayton, Victoria 3800, Australia.
| |
Collapse
|
37
|
Tsao DD, Chang KR, Kockel L, Park S, Kim SK. A genetic strategy to measure insulin signaling regulation and physiology in Drosophila. PLoS Genet 2023; 19:e1010619. [PMID: 36730473 PMCID: PMC9928101 DOI: 10.1371/journal.pgen.1010619] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 02/14/2023] [Accepted: 01/15/2023] [Indexed: 02/04/2023] Open
Abstract
Insulin regulation is a hallmark of health, and impaired insulin signaling promotes metabolic diseases like diabetes mellitus. However, current assays for measuring insulin signaling in all animals remain semi-quantitative and lack the sensitivity, tissue-specificity or temporal resolution needed to quantify in vivo physiological signaling dynamics. Insulin signal transduction is remarkably conserved across metazoans, including insulin-dependent phosphorylation and regulation of Akt/Protein kinase B. Here, we generated transgenic fruit flies permitting tissue-specific expression of an immunoepitope-labelled Akt (AktHF). We developed enzyme-linked immunosorption assays (ELISA) to quantify picomolar levels of phosphorylated (pAktHF) and total AktHF in single flies, revealing dynamic tissue-specific physiological regulation of pAktHF in response to fasting and re-feeding, exogenous insulin, or targeted genetic suppression of established insulin signaling regulators. Genetic screening revealed Pp1-87B as an unrecognized regulator of Akt and insulin signaling. Tools and concepts here provide opportunities to discover tissue-specific regulators of in vivo insulin signaling responses.
Collapse
Affiliation(s)
- Deborah D. Tsao
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Kathleen R. Chang
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Lutz Kockel
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Sangbin Park
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Seung K. Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Medicine (Division of Endocrinology, Metabolism, Gerontology), Stanford University School of Medicine, Stanford, California, United States of America
- Department of Pediatrics (Division of Endocrinology), Stanford University School of Medicine, Stanford, California, United States of America
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
38
|
Demir E, Turna Demir F. Drosophila melanogaster as a dynamic in vivo model organism reveals the hidden effects of interactions between microplastic/nanoplastic and heavy metals. J Appl Toxicol 2023; 43:212-219. [PMID: 35644834 DOI: 10.1002/jat.4353] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 05/26/2022] [Accepted: 05/26/2022] [Indexed: 01/17/2023]
Abstract
Plastic waste in different environments has been constantly transforming into microplastic/nanoplastic (MNPLs). As they may coexist with other contaminants, they may behave as vectors that transport various toxic trace elements, including metals. Because the impact of exposure to such matter on health still remains elusive, the abundant presence of MNPLs has lately become a pressing environmental issue. Researchers have been utilizing Drosophila melanogaster as a dynamic in vivo model in genetic research for some time. The fly has also recently gained wider recognition in toxicology and nanogenotoxicity studies. The use of nanoparticles in numerous medical and consumer products raises serious concern, since many in vitro studies have shown their toxic potential. However, there is rather limited in vivo research into nanomaterial genotoxicity using mice or other mammalians owing to high costs and ethical concerns. In this context, Drosophila, thanks to its genetic tractability, short life span, with its entire life cycle lasting about 10 days, and distinct developmental stages, renders this organism an excellent model in testing toxic effects mediated by MNPLs. This review therefore aims to encourage research entities to employ Drosophila as a model in their nanogenotoxicity experiments focusing on impact of MNPLs at the molecular level.
Collapse
Affiliation(s)
- Eşref Demir
- Department of Medical Services and Techniques, Medical Laboratory Techniques Programme, Vocational School of Health Services, Antalya Bilim University, Dosemealti, Antalya, Turkey
| | - Fatma Turna Demir
- Department of Medical Services and Techniques, Medical Laboratory Techniques Programme, Vocational School of Health Services, Antalya Bilim University, Dosemealti, Antalya, Turkey
| |
Collapse
|
39
|
Okamoto N, Watanabe A. Interorgan communication through peripherally derived peptide hormones in Drosophila. Fly (Austin) 2022; 16:152-176. [PMID: 35499154 PMCID: PMC9067537 DOI: 10.1080/19336934.2022.2061834] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/21/2022] [Accepted: 03/29/2022] [Indexed: 02/06/2023] Open
Abstract
In multicellular organisms, endocrine factors such as hormones and cytokines regulate development and homoeostasis through communication between different organs. For understanding such interorgan communications through endocrine factors, the fruit fly Drosophila melanogaster serves as an excellent model system due to conservation of essential endocrine systems between flies and mammals and availability of powerful genetic tools. In Drosophila and other insects, functions of neuropeptides or peptide hormones from the central nervous system have been extensively studied. However, a series of recent studies conducted in Drosophila revealed that peptide hormones derived from peripheral tissues also play critical roles in regulating multiple biological processes, including growth, metabolism, reproduction, and behaviour. Here, we summarise recent advances in understanding target organs/tissues and functions of peripherally derived peptide hormones in Drosophila and describe how these hormones contribute to various biological events through interorgan communications.
Collapse
Affiliation(s)
- Naoki Okamoto
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akira Watanabe
- Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
40
|
Deshpande R, Lee B, Grewal SS. Enteric bacterial infection in Drosophila induces whole-body alterations in metabolic gene expression independently of the immune deficiency signaling pathway. G3 GENES|GENOMES|GENETICS 2022; 12:6628587. [PMID: 35781508 PMCID: PMC9635644 DOI: 10.1093/g3journal/jkac163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/22/2022] [Indexed: 12/04/2022]
Abstract
When infected by intestinal pathogenic bacteria, animals initiate both local and systemic defence responses. These responses are required to reduce pathogen burden and also to alter host physiology and behavior to promote infection tolerance, and they are often mediated through alterations in host gene expression. Here, we have used transcriptome profiling to examine gene expression changes induced by enteric infection with the Gram-negative bacteria Pseudomonas entomophila in adult female Drosophila. We find that infection induces a strong upregulation of metabolic gene expression, including gut and fat body-enriched genes involved in lipid transport, lipolysis, and beta-oxidation, as well as glucose and amino acid metabolism genes. Furthermore, we find that the classic innate immune deficiency (Imd)/Relish/NF-KappaB pathway is not required for, and in some cases limits, these infection-mediated increases in metabolic gene expression. We also see that enteric infection with Pseudomonas entomophila downregulates the expression of many transcription factors and cell–cell signaling molecules, particularly those previously shown to be involved in gut-to-brain and neuronal signaling. Moreover, as with the metabolic genes, these changes occurred largely independent of the Imd pathway. Together, our study identifies many metabolic, signaling, and transcription factor gene expression changes that may contribute to organismal physiological and behavioral responses to enteric pathogen infection.
Collapse
Affiliation(s)
- Rujuta Deshpande
- Clark H Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Alberta Children’s Hospital Research Institute, University of Calgary , Alberta T2N 4N1, Canada
- Department of Biochemistry and Molecular Biology Calgary, University of Calgary , Alberta T2N 4N1, Canada
| | - Byoungchun Lee
- Clark H Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Alberta Children’s Hospital Research Institute, University of Calgary , Alberta T2N 4N1, Canada
- Department of Biochemistry and Molecular Biology Calgary, University of Calgary , Alberta T2N 4N1, Canada
| | - Savraj S Grewal
- Clark H Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Alberta Children’s Hospital Research Institute, University of Calgary , Alberta T2N 4N1, Canada
- Department of Biochemistry and Molecular Biology Calgary, University of Calgary , Alberta T2N 4N1, Canada
| |
Collapse
|
41
|
Gáliková M, Klepsatel P. Endocrine control of glycogen and triacylglycerol breakdown in the fly model. Semin Cell Dev Biol 2022; 138:104-116. [PMID: 35393234 DOI: 10.1016/j.semcdb.2022.03.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 03/15/2022] [Accepted: 03/28/2022] [Indexed: 12/12/2022]
Abstract
Over the last decade, the combination of genetics, transcriptomic and proteomic approaches yielded substantial insights into the mechanisms behind the synthesis and breakdown of energy stores in the model organisms. The fruit fly Drosophila melanogaster has been particularly useful to unravel genetic regulations of energy metabolism. Despite the considerable evolutionary distance between humans and flies, the energy storage organs, main metabolic pathways, and even their genetic regulations remained relatively conserved. Glycogen and fat are universal energy reserves used in all animal phyla and several of their endocrine regulators, such as the insulin pathway, are highly evolutionarily conserved. Nevertheless, some of the factors inducing catabolism of energy stores have diverged significantly during evolution. Moreover, even within a single insect species, D. melanogaster, there are substantial developmental and context-dependent variances in the regulation of energy stores. These differences include, among others, the endocrine pathways that govern the catabolic events or the predominant fuel which is utilized for the given process. For example, many catabolic regulators that control energy reserves in adulthood seem to be largely dispensable for energy mobilization during development. In this review, we focus on a selection of the most important catabolic regulators from the group of peptide hormones (Adipokinetic hormone, Corazonin), catecholamines (octopamine), steroid hormones (20-hydroxyecdysone), and other factors (extracellular adenosine, regulators of lipase Brummer). We discuss their roles in the mobilization of energy reserves for processes such as development through non-feeding stages, flight or starvation survival. Finally, we conclude with future perspectives on the energy balance research in the fly model.
Collapse
Affiliation(s)
- Martina Gáliková
- Institute of Zoology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 06 Bratislava, Slovakia.
| | - Peter Klepsatel
- Institute of Zoology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 06 Bratislava, Slovakia; Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05 Bratislava, Slovakia
| |
Collapse
|
42
|
Cui X, Gruzdeva A, Kim H, Yapici N. Of flies, mice and neural control of food intake: lessons to learn from both models. Curr Opin Neurobiol 2022; 73:102531. [PMID: 35390643 PMCID: PMC9167741 DOI: 10.1016/j.conb.2022.102531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/20/2022] [Accepted: 03/02/2022] [Indexed: 11/03/2022]
Abstract
In her book, A Room of One's Own, the famous author Virginia Woolf writes "One cannot think well, love well, sleep well if one has not dined well". This is true. All animals need to forage for food and consume specific nutrients to maintain their physiological homeostasis, maximize their fitness and their reproduction. After decades of research in humans and many model organisms, we now know that our brain is one of the key players that control what, when, and how much we eat. In this review, we discuss the recent literature on neural control of food intake behaviors in mice and flies with the view that these two model organisms complement one another in efforts to uncover conserved principles brains use to regulate energy metabolism and food ingestion.
Collapse
Affiliation(s)
- Xinyue Cui
- Department of Neurobiology and Behavior, Cornell University, 14853, Ithaca, NY, USA
| | - Anna Gruzdeva
- Department of Neurobiology and Behavior, Cornell University, 14853, Ithaca, NY, USA
| | - Haein Kim
- Department of Neurobiology and Behavior, Cornell University, 14853, Ithaca, NY, USA
| | - Nilay Yapici
- Department of Neurobiology and Behavior, Cornell University, 14853, Ithaca, NY, USA.
| |
Collapse
|
43
|
Chang KR, Tsao DD, Bennett C, Wang E, Floyd JF, Tay ASY, Greenwald E, Kim ES, Griffin C, Morse E, Chisholm T, Rankin AE, Baena-Lopez LA, Lantz N, Fox E, Kockel L, Kim SK, Park S. Transgenic Drosophila lines for LexA-dependent gene and growth regulation. G3 (BETHESDA, MD.) 2022; 12:jkac018. [PMID: 35100369 PMCID: PMC8895989 DOI: 10.1093/g3journal/jkac018] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/05/2022] [Indexed: 11/12/2022]
Abstract
Conditional expression of short hairpin RNAs with binary genetic systems is an indispensable tool for studying gene function. Addressing mechanisms underlying cell-cell communication in vivo benefits from simultaneous use of 2 independent gene expression systems. To complement the abundance of existing Gal4/UAS-based resources in Drosophila, we and others have developed LexA/LexAop-based genetic tools. Here, we describe experimental and pedagogical advances that promote the efficient conversion of Drosophila Gal4 lines to LexA lines, and the generation of LexAop-short hairpin RNA lines to suppress gene function. We developed a CRISPR/Cas9-based knock-in system to replace Gal4 coding sequences with LexA, and a LexAop-based short hairpin RNA expression vector to achieve short hairpin RNA-mediated gene silencing. We demonstrate the use of these approaches to achieve targeted genetic loss-of-function in multiple tissues. We also detail our development of secondary school curricula that enable students to create transgenic flies, thereby magnifying the production of well-characterized LexA/LexAop lines for the scientific community. The genetic tools and teaching methods presented here provide LexA/LexAop resources that complement existing resources to study intercellular communication coordinating metazoan physiology and development.
Collapse
Affiliation(s)
- Kathleen R Chang
- Stanford University, Stanford, CA 94305, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Deborah D Tsao
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Celine Bennett
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Elaine Wang
- The Lawrenceville School, Lawrenceville, NJ 08648, USA
| | - Jax F Floyd
- The Lawrenceville School, Lawrenceville, NJ 08648, USA
| | | | - Emily Greenwald
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ella S Kim
- Phillips Exeter Academy, Exeter, NH 03833, USA
| | | | | | | | | | | | - Nicole Lantz
- The Lawrenceville School, Lawrenceville, NJ 08648, USA
| | - Elizabeth Fox
- The Lawrenceville School, Lawrenceville, NJ 08648, USA
| | - Lutz Kockel
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Seung K Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Departments of Medicine and of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Diabetes Research Center, Stanford, CA 94305, USA
| | - Sangbin Park
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
44
|
Medina A, Bellec K, Polcowñuk S, Cordero JB. Investigating local and systemic intestinal signalling in health and disease with Drosophila. Dis Model Mech 2022; 15:274860. [PMID: 35344037 PMCID: PMC8990086 DOI: 10.1242/dmm.049332] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Whole-body health relies on complex inter-organ signalling networks that enable organisms to adapt to environmental perturbations and to changes in tissue homeostasis. The intestine plays a major role as a signalling centre by producing local and systemic signals that are relayed to the body and that maintain intestinal and organismal homeostasis. Consequently, disruption of intestinal homeostasis and signalling are associated with systemic diseases and multi-organ dysfunction. In recent years, the fruit fly Drosophila melanogaster has emerged as a prime model organism to study tissue-intrinsic and systemic signalling networks of the adult intestine due to its genetic tractability and functional conservation with mammals. In this Review, we highlight Drosophila research that has contributed to our understanding of how the adult intestine interacts with its microenvironment and with distant organs. We discuss the implications of these findings for understanding intestinal and whole-body pathophysiology, and how future Drosophila studies might advance our knowledge of the complex interplay between the intestine and the rest of the body in health and disease. Summary: We outline work in the fruit fly Drosophila melanogaster that has contributed knowledge on local and whole-body signalling coordinated by the adult intestine, and discuss its implications in intestinal pathophysiology and associated systemic dysfunction.
Collapse
Affiliation(s)
- Andre Medina
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK.,CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Karen Bellec
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Sofia Polcowñuk
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Julia B Cordero
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK.,CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| |
Collapse
|
45
|
Abstract
Cancer continues to be a leading cause of death worldwide, largely due to metastases and cachexia. It is a complex disease that is commonly associated with a variety of comorbidities. With global increases in ageing populations and obesity, multimorbidity is a rapidly growing clinical issue in the context of cancer. Cancer is also genetically heterogeneous, with a tumour's unique profile determining its incidence of metastasis, degree of cachexia and response to therapeutics. These complexities of human cancer are difficult to replicate in animal models and are, in part, responsible for the failures in translational cancer research. In this Perspective, we highlight the fruit fly, Drosophila melanogaster, as a powerful model organism to investigate multimorbidity and tumour diversity. We also highlight how harnessing these complexities in Drosophila can, potentially, enhance cancer research and advance therapeutic discoveries.
Collapse
Affiliation(s)
- Courtney Choutka
- Medical Research Council London Institute of Medical Sciences, Du Cane Road, London W12 0NN, United Kingdom
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom
| | - Cecilia Cabrera
- Medical Research Council London Institute of Medical Sciences, Du Cane Road, London W12 0NN, United Kingdom
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom
| | - Susumu Hirabayashi
- Medical Research Council London Institute of Medical Sciences, Du Cane Road, London W12 0NN, United Kingdom
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom
| |
Collapse
|
46
|
Amelioration of hepatic steatosis by dietary essential amino acid-induced ubiquitination. Mol Cell 2022; 82:1528-1542.e10. [PMID: 35245436 DOI: 10.1016/j.molcel.2022.01.021] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 12/14/2021] [Accepted: 01/25/2022] [Indexed: 12/30/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a global health concern with no approved drugs. High-protein dietary intervention is currently the most effective treatment. However, its underlying mechanism is unknown. Here, using Drosophila oenocytes, the specialized hepatocyte-like cells, we find that dietary essential amino acids ameliorate hepatic steatosis by inducing polyubiquitination of Plin2, a lipid droplet-stabilizing protein. Leucine and isoleucine, two branched-chain essential amino acids, strongly bind to and activate the E3 ubiquitin ligase Ubr1, targeting Plin2 for degradation. We further show that the amino acid-induced Ubr1 activity is necessary to prevent steatosis in mouse livers and cultured human hepatocytes, providing molecular insight into the anti-NAFLD effects of dietary protein/amino acids. Importantly, split-intein-mediated trans-splicing expression of constitutively active UBR2, an Ubr1 family member, significantly ameliorates obesity-induced and high fat diet-induced hepatic steatosis in mice. Together, our results highlight activation of Ubr1 family proteins as a promising strategy in NAFLD treatment.
Collapse
|