1
|
Yeewa R, Pohsa S, Yamsri T, Wongkummool W, Jantaree P, Potikanond S, Nimlamool W, Shotelersuk V, Lo Piccolo L, Jantrapirom S. The histone acylation reader ENL/AF9 regulates aging in Drosophila melanogaster. Neurobiol Aging 2024; 144:153-162. [PMID: 39405796 DOI: 10.1016/j.neurobiolaging.2024.10.002] [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: 03/04/2024] [Revised: 10/05/2024] [Accepted: 10/05/2024] [Indexed: 10/21/2024]
Abstract
Histone acylation plays a pivotal role in modulating gene expression, ensuring proper neurogenesis and responsiveness to various signals. Recently, the evolutionary conserved YAF9, ENL, AF9, TAF41, SAS5 (YEATS) domain found in four human paralogs, has emerged as a new class of histone acylation reader with a preference for the bulkier crotonyl group lysine over acetylation. Despite advancements, the role of either histone crotonylation or its readers in neurons remains unclear. In this study, we employed Drosophila melanogaster to investigate the role of ENL/AF9 (dENL/AF9) in the nervous system. Pan-neuronal dENL/AF9 knockdown not only extended the lifespan of flies but also enhanced their overall fitness during aging, including improved sleep quality and locomotion. Moreover, a decreased activity of dENL/AF9 in neurons led to an up-regulation of catalase gene expression which combined with reduced levels of malondialdehyde (MDA) and an enhanced tolerance to oxidative stress in aging flies. This study unveiled a novel function of histone crotonylation readers in aging with potential implications for understanding age-related conditions in humans.
Collapse
Affiliation(s)
- Ranchana Yeewa
- Centre of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sureena Pohsa
- Centre of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Titaree Yamsri
- Centre of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Wasinee Wongkummool
- Centre of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Phatcharida Jantaree
- Centre of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Saranyapin Potikanond
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Wutigri Nimlamool
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Vorasuk Shotelersuk
- Centre of Excellence for Medical Genomics, Medical Genomics Cluster, Department of Paediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Excellence Centre for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand
| | - Luca Lo Piccolo
- Centre of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| | - Salinee Jantrapirom
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Drosophila Centre for Human Diseases and Drug Discovery (DHD), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
2
|
Zappia MP, Damschroder D, Westacott A, Wessells RJ, Frolov MV. The RU486-dependent activation of the GeneSwitch system in adult muscles leads to severe adverse effects in Drosophila. G3 (BETHESDA, MD.) 2024; 14:jkae039. [PMID: 38409337 PMCID: PMC11075533 DOI: 10.1093/g3journal/jkae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/15/2024] [Accepted: 02/20/2024] [Indexed: 02/28/2024]
Abstract
Robust genetic systems to control the expression of transgenes in a spatial and temporal manner are a valuable asset for researchers. The GeneSwitch system induced by the drug RU486 has gained widespread use in the Drosophila community. However, some concerns were raised as negative effects were seen depending on the stock, transgene, stage, and tissue under study. Here, we characterized the adverse effects triggered by activating the GeneSwitch system in adult muscles using the MHC-GS-GAL4 driver. When a control, mock UAS-RNAi transgene was induced by feeding adult flies with RU486, we found that the overall muscle structure, including myofibrils and mitochondrial shape, was significantly disrupted and led to a significant reduction in the lifespan. Remarkably, lifespan was even shorter when 2 copies of the driver were used even without the mock UAS-RNAi transgene. Thus, researchers should be cautious when interpreting the results given the adverse effects we found when inducing RU486-dependent MHC-GS-GAL4 in adult muscles. To account for the impact of these effects we recommend adjusting the dose of RU486, setting up additional control groups, such as a mock UAS-RNAi transgene, as comparing the phenotypes between RU486-treated and untreated animals could be insufficient.
Collapse
Affiliation(s)
- Maria Paula Zappia
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Deena Damschroder
- Department of Physiology, Wayne State School of Medicine, Detroit, MI 48201, USA
| | - Anton Westacott
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Robert J Wessells
- Department of Physiology, Wayne State School of Medicine, Detroit, MI 48201, USA
| | - Maxim V Frolov
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
3
|
Ureña E, Xu B, Regan JC, Atilano ML, Minkley LJ, Filer D, Lu YX, Bolukbasi E, Khericha M, Alic N, Partridge L. Trametinib ameliorates aging-associated gut pathology in Drosophila females by reducing Pol III activity in intestinal stem cells. Proc Natl Acad Sci U S A 2024; 121:e2311313121. [PMID: 38241436 PMCID: PMC10823232 DOI: 10.1073/pnas.2311313121] [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: 07/12/2023] [Accepted: 11/17/2023] [Indexed: 01/21/2024] Open
Abstract
Pharmacological therapies are promising interventions to slow down aging and reduce multimorbidity in the elderly. Studies in animal models are the first step toward translation of candidate molecules into human therapies, as they aim to elucidate the molecular pathways, cellular mechanisms, and tissue pathologies involved in the anti-aging effects. Trametinib, an allosteric inhibitor of MEK within the Ras/MAPK (Ras/Mitogen-Activated Protein Kinase) pathway and currently used as an anti-cancer treatment, emerged as a geroprotector candidate because it extended lifespan in the fruit fly Drosophila melanogaster. Here, we confirm that trametinib consistently and robustly extends female lifespan, and reduces intestinal stem cell (ISC) proliferation, tumor formation, tissue dysplasia, and barrier disruption in guts in aged flies. In contrast, pro-longevity effects of trametinib are weak and inconsistent in males, and it does not influence gut homeostasis. Inhibition of the Ras/MAPK pathway specifically in ISCs is sufficient to partially recapitulate the effects of trametinib. Moreover, in ISCs, trametinib decreases the activity of the RNA polymerase III (Pol III), a conserved enzyme synthesizing transfer RNAs and other short, non-coding RNAs, and whose inhibition also extends lifespan and reduces gut pathology. Finally, we show that the pro-longevity effect of trametinib in ISCs is partially mediated by Maf1, a repressor of Pol III, suggesting a life-limiting Ras/MAPK-Maf1-Pol III axis in these cells. The mechanism of action described in this work paves the way for further studies on the anti-aging effects of trametinib in mammals and shows its potential for clinical application in humans.
Collapse
Affiliation(s)
- Enric Ureña
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Bowen Xu
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Jennifer C. Regan
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Magda L. Atilano
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Lucy J. Minkley
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Danny Filer
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Yu-Xuan Lu
- Max Planck Institute for Biology of Ageing, CologneD-50931, Germany
| | - Ekin Bolukbasi
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Mobina Khericha
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Nazif Alic
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
| | - Linda Partridge
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, LondonWC1E 7JE, United Kingdom
- Max Planck Institute for Biology of Ageing, CologneD-50931, Germany
| |
Collapse
|
4
|
Brandt A, Petrovsky R, Kriebel M, Großhans J. Use of Farnesyl Transferase Inhibitors in an Ageing Model in Drosophila. J Dev Biol 2023; 11:40. [PMID: 37987370 PMCID: PMC10660854 DOI: 10.3390/jdb11040040] [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: 08/10/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/22/2023] Open
Abstract
The presence of farnesylated proteins at the inner nuclear membrane (INM), such as the Lamins or Kugelkern in Drosophila, leads to specific changes in the nuclear morphology and accelerated ageing on the organismal level reminiscent of the Hutchinson-Gilford progeria syndrome (HGPS). Farnesyl transferase inhibitors (FTIs) can suppress the phenotypes of the nuclear morphology in cultured fibroblasts from HGPS patients and cultured cells overexpressing farnesylated INM proteins. Similarly, FTIs have been reported to suppress the shortened lifespan in model organisms. Here, we report an experimental system combining cell culture and Drosophila flies for testing the activity of substances on the HGPS-like nuclear morphology and lifespan, with FTIs as an experimental example. Consistent with previous reports, we show that FTIs were able to ameliorate the nuclear phenotypes induced by the farnesylated nuclear proteins Progerin, Kugelkern, or truncated Lamin B in cultured cells. The subsequent validation in Drosophila lifespan assays demonstrated the applicability of the experimental system: treating adult Drosophila with the FTI ABT-100 reversed the nuclear phenotypes and extended the lifespan of experimentally induced short-lived flies. Since kugelkern-expressing flies have a significantly shorter average lifespan, half the time is needed for testing substances in the lifespan assay.
Collapse
Affiliation(s)
| | - Roman Petrovsky
- Department of Biology, Philipps University, Karl-von-Frisch-Straße 8, 35043 Marburg, Germany
| | - Maria Kriebel
- Department of Biology, Philipps University, Karl-von-Frisch-Straße 8, 35043 Marburg, Germany
| | - Jörg Großhans
- Department of Biology, Philipps University, Karl-von-Frisch-Straße 8, 35043 Marburg, Germany
| |
Collapse
|
5
|
Wang L, Lin J, Yang K, Wang W, Lv Y, Zeng X, Zhao Y, Yu J, Pan L. Perilipin 1 Deficiency Prompts Lipolysis in Lipid Droplets and Aggravates the Pathogenesis of Persistent Immune Activation in Drosophila. J Innate Immun 2023; 15:697-708. [PMID: 37742619 PMCID: PMC10601664 DOI: 10.1159/000534099] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 09/08/2023] [Indexed: 09/26/2023] Open
Abstract
Lipid droplets (LDs) are highly dynamic intracellular organelles, which are involved in lots of biological processes. However, the dynamic morphogenesis and functions of intracellular LDs during persistent innate immune responses remain obscure. In this study, we induce long-term systemic immune activation in Drosophila through genetic manipulation. Then, the dynamic pattern of LDs is traced in the Drosophila fat body. We find that deficiency of Plin1, a key regulator of LDs' reconfiguration, blocks LDs minimization at the initial stage of immune hyperactivation but enhances LDs breakdown at the later stage of sustained immune activation via recruiting the lipase Brummer (Bmm, homologous to human ATGL). The high wasting in LDs shortens the lifespan of flies with high-energy-cost immune hyperactivation. Therefore, these results suggest a critical function of LDs during long-term immune activation and provide a potential treatment for the resolution of persistent inflammation.
Collapse
Affiliation(s)
- Lei Wang
- Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection (Former Institut Pasteur of Shanghai), Chinese Academy of Sciences, Shanghai, China
| | - Jiaxin Lin
- Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection (Former Institut Pasteur of Shanghai), Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Kaiyan Yang
- Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection (Former Institut Pasteur of Shanghai), Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Weina Wang
- Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection (Former Institut Pasteur of Shanghai), Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yan Lv
- Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection (Former Institut Pasteur of Shanghai), Chinese Academy of Sciences, Shanghai, China
- Pasteurien College, Soochow University, Suzhou, China
| | - Xiangkang Zeng
- Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection (Former Institut Pasteur of Shanghai), Chinese Academy of Sciences, Shanghai, China
- Pasteurien College, Soochow University, Suzhou, China
| | - Yaya Zhao
- Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection (Former Institut Pasteur of Shanghai), Chinese Academy of Sciences, Shanghai, China
- The Joint Center for Infection and Immunity between Guangzhou Institute of Pediatrics and Institut Pasteur of Shanghai, Guangzhou Women and Children’s Medical Center, Guangzhou, China
| | - Junjing Yu
- Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Lei Pan
- Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection (Former Institut Pasteur of Shanghai), Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
- Pasteurien College, Soochow University, Suzhou, China
- The Joint Center for Infection and Immunity between Guangzhou Institute of Pediatrics and Institut Pasteur of Shanghai, Guangzhou Women and Children’s Medical Center, Guangzhou, China
| |
Collapse
|
6
|
Smith HJ, Lanjuin A, Sharma A, Prabhakar A, Nowak E, Stine PG, Sehgal R, Stojanovski K, Towbin BD, Mair WB. Neuronal mTORC1 inhibition promotes longevity without suppressing anabolic growth and reproduction in C. elegans. PLoS Genet 2023; 19:e1010938. [PMID: 37721956 PMCID: PMC10538657 DOI: 10.1371/journal.pgen.1010938] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 09/28/2023] [Accepted: 08/24/2023] [Indexed: 09/20/2023] Open
Abstract
mTORC1 (mechanistic target of rapamycin complex 1) is a metabolic sensor that promotes growth when nutrients are abundant. Ubiquitous inhibition of mTORC1 extends lifespan in multiple organisms but also disrupts several anabolic processes resulting in stunted growth, slowed development, reduced fertility, and disrupted metabolism. However, it is unclear if these pleiotropic effects of mTORC1 inhibition can be uncoupled from longevity. Here, we utilize the auxin-inducible degradation (AID) system to restrict mTORC1 inhibition to C. elegans neurons. We find that neuron-specific degradation of RAGA-1, an upstream activator of mTORC1, or LET-363, the ortholog of mammalian mTOR, is sufficient to extend lifespan in C. elegans. Unlike raga-1 loss of function genetic mutations or somatic AID of RAGA-1, neuronal AID of RAGA-1 robustly extends lifespan without impairing body size, developmental rate, brood size, or neuronal function. Moreover, while degradation of RAGA-1 in all somatic tissues alters the expression of thousands of genes, demonstrating the widespread effects of mTORC1 inhibition, degradation of RAGA-1 in neurons only results in around 200 differentially expressed genes with a specific enrichment in metabolism and stress response. Notably, our work demonstrates that targeting mTORC1 specifically in the nervous system in C. elegans uncouples longevity from growth and reproductive impairments, and that many canonical effects of low mTORC1 activity are not required to promote healthy aging. These data challenge previously held ideas about the mechanisms of mTORC1 lifespan extension and underscore the potential of promoting longevity by neuron-specific mTORC1 modulation.
Collapse
Affiliation(s)
- Hannah J. Smith
- Dept. Molecular Metabolism, Harvard TH Chan School of Public Health, Massachusetts, United States of America
| | - Anne Lanjuin
- Dept. Molecular Metabolism, Harvard TH Chan School of Public Health, Massachusetts, United States of America
| | - Arpit Sharma
- Dept. Molecular Metabolism, Harvard TH Chan School of Public Health, Massachusetts, United States of America
| | - Aditi Prabhakar
- Dept. Molecular Metabolism, Harvard TH Chan School of Public Health, Massachusetts, United States of America
| | - Ewelina Nowak
- Dept. Molecular Metabolism, Harvard TH Chan School of Public Health, Massachusetts, United States of America
| | - Peter G. Stine
- Dept. Molecular Metabolism, Harvard TH Chan School of Public Health, Massachusetts, United States of America
| | - Rohan Sehgal
- Dept. Molecular Metabolism, Harvard TH Chan School of Public Health, Massachusetts, United States of America
| | | | | | - William B. Mair
- Dept. Molecular Metabolism, Harvard TH Chan School of Public Health, Massachusetts, United States of America
| |
Collapse
|
7
|
Barwell T, Raina S, Page A, MacCharles H, Seroude L. Juvenile and adult expression of polyglutamine expanded huntingtin produce distinct aggregate distributions in Drosophila muscle. Hum Mol Genet 2023; 32:2656-2668. [PMID: 37369041 DOI: 10.1093/hmg/ddad098] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 05/09/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
While Huntington's disease (HD) is widely recognized as a disease affecting the nervous system, much evidence has accumulated to suggest peripheral or non-neuronal tissues are affected as well. Here, we utilize the UAS/GAL4 system to express a pathogenic HD construct in the muscle of the fly and characterize the effects. We observe detrimental phenotypes such as a reduced lifespan, decreased locomotion and accumulation of protein aggregates. Strikingly, depending on the GAL4 driver used to express the construct, we saw different aggregate distributions and severity of phenotypes. These different aggregate distributions were found to be dependent on the expression level and the timing of expression. Hsp70, a well-documented suppressor of polyglutamine aggregates, was found to strongly reduce the accumulation of aggregates in the eye, but in the muscle, it did not prevent the reduction of the lifespan. Therefore, the molecular mechanisms underlying the detrimental effects of aggregates in the muscle are distinct from the nervous system.
Collapse
Affiliation(s)
- Taylor Barwell
- Department of Biology, Queen's University, 116 Barrie St, Kingston, Ontario, K7L 3N6, Canada
| | - Sehaj Raina
- Department of Biology, Queen's University, 116 Barrie St, Kingston, Ontario, K7L 3N6, Canada
| | - Austin Page
- Department of Biology, Queen's University, 116 Barrie St, Kingston, Ontario, K7L 3N6, Canada
| | - Hayley MacCharles
- Department of Biology, Queen's University, 116 Barrie St, Kingston, Ontario, K7L 3N6, Canada
| | - Laurent Seroude
- Department of Biology, Queen's University, 116 Barrie St, Kingston, Ontario, K7L 3N6, Canada
| |
Collapse
|
8
|
Rozich E, Randolph LK, Insolera R. An optimized temporally controlled Gal4 system in Drosophila reveals degeneration caused by adult-onset neuronal Vps13D knockdown. Front Neurosci 2023; 17:1204068. [PMID: 37457002 PMCID: PMC10339317 DOI: 10.3389/fnins.2023.1204068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/09/2023] [Indexed: 07/18/2023] Open
Abstract
Mutations in the human gene VPS13D cause the adult-onset neurodegenerative disease ataxia. Our previous work showed that disruptions in the Vps13D gene in Drosophila neurons causes mitochondrial defects. However, developmental lethality caused by Vps13D loss limited our understanding of the long-term physiological effects of Vps13D perturbation in neurons. Here, we optimized a previously generated system to temporally knock down Vps13D expression precisely in adult Drosophila neurons using a modification to the Gal4/UAS system. Adult-onset activation of Gal4 was enacted using the chemically-inducible tool which fuses a destabilization-domain to the Gal4 repressor Gal80 (Gal80-DD). Optimization of the Gal80-DD tool shows that feeding animals the DD-stabilizing drug trimethoprim (TMP) during development and rearing at a reduced temperature maximally represses Gal4 activity. Temperature shift and removal of TMP from the food after eclosion robustly activates Gal4 expression in adult neurons. Using the optimized Gal80-DD system, we find that adult-onset Vps13D RNAi expression in neurons causes the accumulation of mitophagy intermediates, progressive deficits in locomotor activity, early lethality, and brain vacuolization characteristic of neurodegeneration. The development of this optimized system allows us to more precisely examine the degenerative phenotypes caused by Vps13D disruption, and can likely be utilized in the future for other genes associated with neurological diseases whose manipulation causes developmental lethality in Drosophila.
Collapse
Affiliation(s)
- Emily Rozich
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Lynsey K. Randolph
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Ryan Insolera
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
9
|
Ewen-Campen B, Luan H, Xu J, Singh R, Joshi N, Thakkar T, Berger B, White BH, Perrimon N. split-intein Gal4 provides intersectional genetic labeling that is repressible by Gal80. Proc Natl Acad Sci U S A 2023; 120:e2304730120. [PMID: 37276389 PMCID: PMC10268248 DOI: 10.1073/pnas.2304730120] [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: 03/22/2023] [Accepted: 05/10/2023] [Indexed: 06/07/2023] Open
Abstract
The split-Gal4 system allows for intersectional genetic labeling of highly specific cell types and tissues in Drosophila. However, the existing split-Gal4 system, unlike the standard Gal4 system, cannot be repressed by Gal80, and therefore cannot be controlled temporally. This lack of temporal control precludes split-Gal4 experiments in which a genetic manipulation must be restricted to specific timepoints. Here, we describe a split-Gal4 system based on a self-excising split-intein, which drives transgene expression as strongly as the current split-Gal4 system and Gal4 reagents, yet which is repressible by Gal80. We demonstrate the potent inducibility of "split-intein Gal4" in vivo using both fluorescent reporters and via reversible tumor induction in the gut. Further, we show that our split-intein Gal4 can be extended to the drug-inducible GeneSwitch system, providing an independent method for intersectional labeling with inducible control. We also show that the split-intein Gal4 system can be used to generate highly cell type-specific genetic drivers based on in silico predictions generated by single-cell RNAseq (scRNAseq) datasets, and we describe an algorithm ("Two Against Background" or TAB) to predict cluster-specific gene pairs across multiple tissue-specific scRNA datasets. We provide a plasmid toolkit to efficiently create split-intein Gal4 drivers based on either CRISPR knock-ins to target genes or using enhancer fragments. Altogether, the split-intein Gal4 system allows for the creation of highly specific intersectional genetic drivers that are inducible/repressible.
Collapse
Affiliation(s)
- Ben Ewen-Campen
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Haojiang Luan
- Laboratory of Molecular Biology, National Institute of Mental Health, NIH, Bethesda, MD20892
| | - Jun Xu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA02115
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai200032, China
| | - Rohit Singh
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Neha Joshi
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Tanuj Thakkar
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Bonnie Berger
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Mathematics, Massachusetts Institute of Technology, Cambridge, MA02143
| | - Benjamin H. White
- Laboratory of Molecular Biology, National Institute of Mental Health, NIH, Bethesda, MD20892
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA02115
- HHMI, Boston, MA02115
| |
Collapse
|
10
|
Ewen-Campen B, Luan H, Xu J, Singh R, Joshi N, Thakkar T, Berger B, White BH, Perrimon N. split-intein Gal4 provides intersectional genetic labeling that is fully repressible by Gal80. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.24.534001. [PMID: 36993523 PMCID: PMC10055387 DOI: 10.1101/2023.03.24.534001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/16/2023]
Abstract
The split-Gal4 system allows for intersectional genetic labeling of highly specific cell-types and tissues in Drosophila . However, the existing split-Gal4 system, unlike the standard Gal4 system, cannot be repressed by Gal80, and therefore cannot be controlled temporally. This lack of temporal control precludes split-Gal4 experiments in which a genetic manipulation must be restricted to specific timepoints. Here, we describe a new split-Gal4 system based on a self-excising split-intein, which drives transgene expression as strongly as the current split-Gal4 system and Gal4 reagents, yet which is fully repressible by Gal80. We demonstrate the potent inducibility of "split-intein Gal4" in vivo using both fluorescent reporters and via reversible tumor induction in the gut. Further, we show that our split-intein Gal4 can be extended to the drug-inducible GeneSwitch system, providing an independent method for intersectional labeling with inducible control. We also show that the split-intein Gal4 system can be used to generate highly cell-type specific genetic drivers based on in silico predictions generated by single cell RNAseq (scRNAseq) datasets, and we describe a new algorithm ("Two Against Background" or TAB) to predict cluster-specific gene pairs across multiple tissue-specific scRNA datasets. We provide a plasmid toolkit to efficiently create split-intein Gal4 drivers based on either CRISPR knock-ins to target genes or using enhancer fragments. Altogether, the split-intein Gal4 system allows for the creation of highly specific intersectional genetic drivers that are inducible/repressible. Significance statement The split-Gal4 system allows Drosophila researchers to drive transgene expression with extraordinary cell type specificity. However, the existing split-Gal4 system cannot be controlled temporally, and therefore cannot be applied to many important areas of research. Here, we present a new split-Gal4 system based on a self-excising split-intein, which is fully controllable by Gal80, as well as a related drug-inducible split GeneSwitch system. This approach can both leverage and inform single-cell RNAseq datasets, and we introduce an algorithm to identify pairs of genes that precisely and narrowly mark a desired cell cluster. Our split-intein Gal4 system will be of value to the Drosophila research community, and allow for the creation of highly specific genetic drivers that are also inducible/repressible.
Collapse
Affiliation(s)
- Ben Ewen-Campen
- These authors contributed equally
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Haojiang Luan
- These authors contributed equally
- Laboratory of Molecular Biology, National Institute of Mental Health, NIH, Bethesda, MD 20892, USA
| | - Jun Xu
- These authors contributed equally
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, China
| | - Rohit Singh
- These authors contributed equally
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Neha Joshi
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Tanuj Thakkar
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Bonnie Berger
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Mathematics, Massachusetts Institute of Technology, Cambridge MA 02143
| | - Benjamin H White
- Laboratory of Molecular Biology, National Institute of Mental Health, NIH, Bethesda, MD 20892, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
- HHMI, Boston, MA 02115, USA
| |
Collapse
|
11
|
Barwell T, Geld S, Seroude L. Comparison of GAL80ts and Tet-off GAL80 transgenes. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000770. [PMID: 37033706 PMCID: PMC10080293 DOI: 10.17912/micropub.biology.000770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 04/11/2023]
Abstract
The manipulation of gene expression requires complete control of space, time and level of expression. In Drosophila , the UAS/GAL4 system has been instrumental in achieving spatial control, but the pursuit for the ideal system for temporal control is ongoing. One strategy used by many groups is to use GAL80. Two different kinds of GAL80 transgenic constructs have been reported but have never been compared directly. Here, we characterize and compare the repression ability of the GAL80ts and Tet-off GAL80 transgenes by two different assays. We found that GAL80ts was generally more efficient than Tet-off GAL80 and that the level of repression was dependent on the GAL4 driver, and did not necessarily correlate with expression level. We also investigated the level of expression upon inactivation of GAL80ts. The level and kinetics of inactivation were found to differ depending on the GAL4 driver and the stage of the life cycle, and in many cases the inactivation was incomplete. These results emphasize that it is essential to measure experimentally the level of expression under repressed and unrepressed states when multiple GAL4 drivers are used with GAL80 transgenes.
Collapse
Affiliation(s)
| | - Sofia Geld
- Biology, Queen's University, Kingston, Ontario, Canada
| | | |
Collapse
|
12
|
Hwangbo DS, Kwon YJ, Iwanaszko M, Jiang P, Abbasi L, Wright N, Alli S, Hutchison AL, Dinner AR, Braun RI, Allada R. Dietary Restriction Impacts Peripheral Circadian Clock Output Important for Longevity in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.04.522718. [PMID: 36711760 PMCID: PMC9881908 DOI: 10.1101/2023.01.04.522718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Circadian clocks may mediate lifespan extension by caloric or dietary restriction (DR). We find that the core clock transcription factor Clock is crucial for a robust longevity and fecundity response to DR in Drosophila. To identify clock-controlled mediators, we performed RNA-sequencing from abdominal fat bodies across the 24 h day after just 5 days under control or DR diets. In contrast to more chronic DR regimens, we did not detect significant changes in the rhythmic expression of core clock genes. Yet we discovered that DR induced de novo rhythmicity or increased expression of rhythmic clock output genes. Network analysis revealed that DR increased network connectivity in one module comprised of genes encoding proteasome subunits. Adult, fat body specific RNAi knockdown demonstrated that proteasome subunits contribute to DR-mediated lifespan extension. Thus, clock control of output links DR-mediated changes in rhythmic transcription to lifespan extension.
Collapse
Affiliation(s)
- Dae-Sung Hwangbo
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
- Center for Sleep & Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL 60208, USA
- Department of Biology, University of Louisville, Louisville, 40292, KY, USA
| | - Yong-Jae Kwon
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Marta Iwanaszko
- Biostatistics Division, Department of Preventive Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Engineering Sciences and Applied Mathematics, Northwestern University, Evanston, IL 60208, USA
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL 60208, USA
| | - Peng Jiang
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
- Center for Sleep & Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Ladan Abbasi
- Department of Biology, University of Louisville, Louisville, 40292, KY, USA
| | - Nicholas Wright
- Department of Biology, University of Louisville, Louisville, 40292, KY, USA
| | - Sarayu Alli
- Department of Biology, University of Louisville, Louisville, 40292, KY, USA
| | - Alan L. Hutchison
- James Franck Institute, Department of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637, USA
| | - Aaron R. Dinner
- James Franck Institute, Department of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637, USA
| | - Rosemary I Braun
- Biostatistics Division, Department of Preventive Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Engineering Sciences and Applied Mathematics, Northwestern University, Evanston, IL 60208, USA
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL 60208, USA
| | - Ravi Allada
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
- Center for Sleep & Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
13
|
Merino MM. Azot expression in the Drosophila gut modulates organismal lifespan. Commun Integr Biol 2022; 16:2156735. [PMID: 36606245 PMCID: PMC9809965 DOI: 10.1080/19420889.2022.2156735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Cell Competition emerged in Drosophila as an unexpected phenomenon, when confronted clones of fit vs unfit cells genetically induced. During the last decade, it has been shown that this mechanism is physiologically active in Drosophila and higher organisms. In Drosophila, Flower (Fwe) eliminates unfit cells during development, regeneration and disease states. Furthermore, studies suggest that Fwe signaling is required to eliminate accumulated unfit cells during adulthood extending Drosophila lifespan. Indeed, ahuizotl (azot) mutants accumulate unfit cells during adulthood and after physical insults in the brain and other epithelial tissues, showing a decrease in organismal lifespan. On the contrary, flies carrying three functional copies of the gene, unfit cell culling seems to be more efficient and show an increase in lifespan. During aging, Azot is required for the elimination of unfit cells, however, the specific organs modulating organismal lifespan by Azot remain unknown. Here we found a potential connection between gut-specific Azot expression and lifespan which may uncover a more widespread organ-specific mechanism modulating organismal survival.
Collapse
Affiliation(s)
- Marisa M. Merino
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland,CONTACT Marisa M. Merino Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| |
Collapse
|
14
|
Martínez Corrales G, Li M, Svermova T, Goncalves A, Voicu D, Dobson AJ, Southall TD, Alic N. Transcriptional memory of dFOXO activation in youth curtails later-life mortality through chromatin remodeling and Xbp1. NATURE AGING 2022; 2:1176-1190. [PMID: 37118537 PMCID: PMC7614430 DOI: 10.1038/s43587-022-00312-x] [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: 12/10/2021] [Accepted: 10/19/2022] [Indexed: 12/03/2022]
Abstract
A transient, homeostatic transcriptional response can result in transcriptional memory, programming subsequent transcriptional outputs. Transcriptional memory has great but unappreciated potential to alter animal aging as animals encounter a multitude of diverse stimuli throughout their lifespan. Here we show that activating an evolutionarily conserved, longevity-promoting transcription factor, dFOXO, solely in early adulthood of female fruit flies is sufficient to improve their subsequent health and survival in midlife and late life. This youth-restricted dFOXO activation causes persistent changes to chromatin landscape in the fat body and requires chromatin remodelers such as the SWI/SNF and ISWI complexes to program health and longevity. Chromatin remodeling is accompanied by a long-lasting transcriptional program that is distinct from that observed during acute dFOXO activation and includes induction of Xbp1. We show that this later-life induction of Xbp1 is sufficient to curtail later-life mortality. Our study demonstrates that transcriptional memory can profoundly alter how animals age.
Collapse
Affiliation(s)
- Guillermo Martínez Corrales
- Institute of Healthy Ageing and the Research Department of Genetics, Evolution, and Environment, University College London, London, UK
| | - Mengjia Li
- Institute of Healthy Ageing and the Research Department of Genetics, Evolution, and Environment, University College London, London, UK
| | - Tatiana Svermova
- Institute of Healthy Ageing and the Research Department of Genetics, Evolution, and Environment, University College London, London, UK
| | - Alex Goncalves
- Institute of Healthy Ageing and the Research Department of Genetics, Evolution, and Environment, University College London, London, UK
| | - Diana Voicu
- Institute of Healthy Ageing and the Research Department of Genetics, Evolution, and Environment, University College London, London, UK
| | - Adam J Dobson
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Tony D Southall
- Department of Life Sciences, Imperial College London, London, UK
| | - Nazif Alic
- Institute of Healthy Ageing and the Research Department of Genetics, Evolution, and Environment, University College London, London, UK.
| |
Collapse
|
15
|
Barwell T, Seroude L. Protocol to measure ß-galactosidase in Drosophila extracts using a CPRG assay. STAR Protoc 2022; 3:101843. [PMID: 36595888 PMCID: PMC9667300 DOI: 10.1016/j.xpro.2022.101843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/14/2022] [Accepted: 10/19/2022] [Indexed: 11/14/2022] Open
Abstract
The quantification of ß-galactosidase activity is routinely required by laboratories worldwide. We present a cost-effective, highly replicable, simple technique for quantifying ß-galactosidase-specific activity from crude extracts made from whole organisms or dissected tissues or cells. Extracts are prepared and measured without the need for any specialized equipment, and tissue is ground manually by pestle and measured by colorimetric CPRG and Bradford assays. This protocol describes the assay using Drosophila extracts but could be applied to any biological system of interest. For complete details on the use and execution of this protocol, please refer to Seroude et al. (2002),1 Poirier et al. (2008),2 and Barwell et al. (2017).3.
Collapse
Affiliation(s)
- Taylor Barwell
- Queen’s University, Kingston, ON K7L 3N6, Canada,Corresponding author
| | - Laurent Seroude
- Queen’s University, Kingston, ON K7L 3N6, Canada,Corresponding author
| |
Collapse
|
16
|
Al-Sabri MH, Behare N, Alsehli AM, Berkins S, Arora A, Antoniou E, Moysiadou EI, Anantha-Krishnan S, Cosmen PD, Vikner J, Moulin TC, Ammar N, Boukhatmi H, Clemensson LE, Rask-Andersen M, Mwinyi J, Williams MJ, Fredriksson R, Schiöth HB. Statins Induce Locomotion and Muscular Phenotypes in Drosophila melanogaster That Are Reminiscent of Human Myopathy: Evidence for the Role of the Chloride Channel Inhibition in the Muscular Phenotypes. Cells 2022; 11:3528. [PMID: 36428957 PMCID: PMC9688544 DOI: 10.3390/cells11223528] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/17/2022] [Accepted: 11/01/2022] [Indexed: 11/09/2022] Open
Abstract
The underlying mechanisms for statin-induced myopathy (SIM) are still equivocal. In this study, we employ Drosophila melanogaster to dissect possible underlying mechanisms for SIM. We observe that chronic fluvastatin treatment causes reduced general locomotion activity and climbing ability. In addition, transmission microscopy of dissected skeletal muscles of fluvastatin-treated flies reveals strong myofibrillar damage, including increased sarcomere lengths and Z-line streaming, which are reminiscent of myopathy, along with fragmented mitochondria of larger sizes, most of which are round-like shapes. Furthermore, chronic fluvastatin treatment is associated with impaired lipid metabolism and insulin signalling. Mechanistically, knockdown of the statin-target Hmgcr in the skeletal muscles recapitulates fluvastatin-induced mitochondrial phenotypes and lowered general locomotion activity; however, it was not sufficient to alter sarcomere length or elicit myofibrillar damage compared to controls or fluvastatin treatment. Moreover, we found that fluvastatin treatment was associated with reduced expression of the skeletal muscle chloride channel, ClC-a (Drosophila homolog of CLCN1), while selective knockdown of skeletal muscle ClC-a also recapitulated fluvastatin-induced myofibril damage and increased sarcomere lengths. Surprisingly, exercising fluvastatin-treated flies restored ClC-a expression and normalized sarcomere lengths, suggesting that fluvastatin-induced myofibrillar phenotypes could be linked to lowered ClC-a expression. Taken together, these results may indicate the potential role of ClC-a inhibition in statin-associated muscular phenotypes. This study underlines the importance of Drosophila melanogaster as a powerful model system for elucidating the locomotion and muscular phenotypes, promoting a better understanding of the molecular mechanisms underlying SIM.
Collapse
Affiliation(s)
- Mohamed H. Al-Sabri
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
- Department of Pharmaceutical Biosciences, Uppsala University, 751 24 Uppsala, Sweden
| | - Neha Behare
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Ahmed M. Alsehli
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
- Faculty of Medicine, King Abdulaziz University and Hospital, Al Ehtifalat St., Jeddah 21589, Saudi Arabia
| | - Samuel Berkins
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Aadeya Arora
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Eirini Antoniou
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Eleni I. Moysiadou
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Sowmya Anantha-Krishnan
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Patricia D. Cosmen
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Johanna Vikner
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Thiago C. Moulin
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
- Faculty of Medicine, Department of Experimental Medical Science, Lund University, Sölvegatan 19, BMC F10, 221 84 Lund, Sweden
| | - Nourhene Ammar
- Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes, CNRS, UMR6290, 35065 Rennes, France
| | - Hadi Boukhatmi
- Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes, CNRS, UMR6290, 35065 Rennes, France
| | - Laura E. Clemensson
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Mathias Rask-Andersen
- Department of Immunology, Genetics and Pathology, Uppsala University, 752 37 Uppsala, Sweden
| | - Jessica Mwinyi
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Michael J. Williams
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| | - Robert Fredriksson
- Department of Pharmaceutical Biosciences, Uppsala University, 751 24 Uppsala, Sweden
| | - Helgi B. Schiöth
- Department of Surgical Sciences, Division of Functional Pharmacology and Neuroscience, Biomedical Center (BMC), Uppsala University, Husargatan 3, 751 24 Uppsala, Sweden
| |
Collapse
|
17
|
A photo-switchable assay system for dendrite degeneration and repair in Drosophila melanogaster. Proc Natl Acad Sci U S A 2022; 119:e2204577119. [PMID: 35969739 PMCID: PMC9407391 DOI: 10.1073/pnas.2204577119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Neurodegeneration arising from aging, injury, or diseases has devastating health consequences. Whereas neuronal survival and axon degeneration have been studied extensively, much less is known about how neurodegeneration affects dendrites, in part due to the limited assay systems available. To develop an assay for dendrite degeneration and repair, we used photo-switchable caspase-3 (caspase-Light-Oxygen-Voltage-sensing [caspase-LOV]) in peripheral class 4 dendrite arborization (c4da) neurons to induce graded neurodegeneration by adjusting illumination duration during development and adulthood in Drosophila melanogaster. We found that both developing and mature c4da neurons were able to survive while sustaining mild neurodegeneration induced by moderate caspase-LOV activation. Further, we observed active dendrite addition and dendrite regeneration in developing and mature c4da neurons, respectively. Using this assay, we found that the mouse Wallerian degeneration slow (WldS) protein can protect c4da neurons from caspase-LOV-induced dendrite degeneration and cell death. Furthermore, our data show that WldS can reduce dendrite elimination without affecting dendrite addition. In summary, we successfully established a photo-switchable assay system in both developing and mature neurons and used WldS as a test case to study the mechanisms underlying dendrite regeneration and repair.
Collapse
|
18
|
Schmid ET, Pyo JH, Walker DW. Neuronal induction of BNIP3-mediated mitophagy slows systemic aging in Drosophila. NATURE AGING 2022; 2:494-507. [PMID: 36213625 PMCID: PMC9540997 DOI: 10.1038/s43587-022-00214-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 03/25/2022] [Indexed: 02/07/2023]
Abstract
The effects of aging on the brain are widespread and can have dramatic implications on the overall health of an organism. Mitochondrial dysfunction is a hallmark of brain aging, but, the interplay between mitochondrial quality control, neuronal aging, and organismal health is not well understood. Here, we show that aging leads to a decline in mitochondrial autophagy (mitophagy) in the Drosophila brain with a concomitant increase in mitochondrial content. We find that induction of BCL2-interacting protein 3 (BNIP3), a mitochondrial outer membrane protein, in the adult nervous system induces mitophagy and prevents the accumulation of dysfunctional mitochondria in the aged brain. Importantly, neuronal induction of BNIP3-mediated mitophagy increases organismal longevity and healthspan. Furthermore, BNIP3-mediated mitophagy in the nervous system improves muscle and intestinal homeostasis in aged flies, indicating cell non-autonomous effects. Our findings identify BNIP3 as a therapeutic target to counteract brain aging and prolong overall organismal health with age.
Collapse
Affiliation(s)
- Edward T. Schmid
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Jung-Hoon Pyo
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - David W. Walker
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California 90095, USA
- Lead Contact
| |
Collapse
|
19
|
McClure CD, Hassan A, Aughey GN, Butt K, Estacio-Gómez A, Duggal A, Ying Sia C, Barber AF, Southall TD. An auxin-inducible, GAL4-compatible, gene expression system for Drosophila. eLife 2022; 11:e67598. [PMID: 35363137 PMCID: PMC8975555 DOI: 10.7554/elife.67598] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 03/27/2022] [Indexed: 01/04/2023] Open
Abstract
The ability to control transgene expression, both spatially and temporally, is essential for studying model organisms. In Drosophila, spatial control is primarily provided by the GAL4/UAS system, whilst temporal control relies on a temperature-sensitive GAL80 (which inhibits GAL4) and drug-inducible systems. However, these are not ideal. Shifting temperature can impact on many physiological and behavioural traits, and the current drug-inducible systems are either leaky, toxic, incompatible with existing GAL4-driver lines, or do not generate effective levels of expression. Here, we describe the auxin-inducible gene expression system (AGES). AGES relies on the auxin-dependent degradation of a ubiquitously expressed GAL80, and therefore, is compatible with existing GAL4-driver lines. Water-soluble auxin is added to fly food at a low, non-lethal, concentration, which induces expression comparable to uninhibited GAL4 expression. The system works in both larvae and adults, providing a stringent, non-lethal, cost-effective, and convenient method for temporally controlling GAL4 activity in Drosophila.
Collapse
Affiliation(s)
- Colin D McClure
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Amira Hassan
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Gabriel N Aughey
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Khushbakht Butt
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers, the State University of New JerseyNew BrunswickUnited States
| | | | - Aneisha Duggal
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Chee Ying Sia
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Annika F Barber
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers, the State University of New JerseyNew BrunswickUnited States
| | - Tony D Southall
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| |
Collapse
|
20
|
Manola MS, Gumeni S, Trougakos IP. Differential Dose- and Tissue-Dependent Effects of foxo on Aging, Metabolic and Proteostatic Pathways. Cells 2021; 10:3577. [PMID: 34944088 PMCID: PMC8700554 DOI: 10.3390/cells10123577] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/09/2021] [Accepted: 12/11/2021] [Indexed: 02/07/2023] Open
Abstract
Aging is the gradual deterioration of physiological functions that culminates in death. Several studies across a wide range of model organisms have revealed the involvement of FOXO (forkhead box, class O) transcription factors in orchestrating metabolic homeostasis, as well as in regulating longevity. To study possible dose- or tissue-dependent effects of sustained foxo overexpression, we utilized two different Drosophila transgenic lines expressing high and relatively low foxo levels and overexpressed foxo, either ubiquitously or in a tissue-specific manner. We found that ubiquitous foxo overexpression (OE) accelerated aging, induced the early onset of age-related phenotypes, increased sensitivity to thermal stress, and deregulated metabolic and proteostatic pathways; these phenotypes were more intense in transgenic flies expressing high levels of foxo. Interestingly, there is a defined dosage of foxo OE in muscles and cardiomyocytes that shifts energy resources into longevity pathways and thus ameliorates not only tissue but also organismal age-related defects. Further, we found that foxo OE stimulates in an Nrf2/cncC dependent-manner, counteracting proteostatic pathways, e.g., the ubiquitin-proteasome pathway, which is central in ameliorating the aberrant foxo OE-mediated toxicity. These findings highlight the differential dose- and tissue-dependent effects of foxo on aging, metabolic and proteostatic pathways, along with the foxo-Nrf2/cncC functional crosstalk.
Collapse
Affiliation(s)
| | | | - Ioannis P. Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Panepistimiopolis, 15784 Athens, Greece; (M.S.M.); (S.G.)
| |
Collapse
|
21
|
Wang L, Lin J, Yu J, Yang K, Sun L, Tang H, Pan L. Downregulation of Perilipin1 by the Immune Deficiency Pathway Leads to Lipid Droplet Reconfiguration and Adaptation to Bacterial Infection in Drosophila. THE JOURNAL OF IMMUNOLOGY 2021; 207:2347-2358. [PMID: 34588219 DOI: 10.4049/jimmunol.2100343] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 08/26/2021] [Indexed: 11/19/2022]
Abstract
Lipid droplets (LDs), the highly dynamic intracellular organelles, are critical for lipid metabolism. Dynamic alterations in the configurations and functions of LDs during innate immune responses to bacterial infections and the underlying mechanisms, however, remain largely unknown. In this study, we trace the time-course morphology of LDs in fat bodies of Drosophila after transient bacterial infection. Detailed analysis shows that perilipin1 (plin1), a core gene involved in the regulation of LDs, is suppressed by the immune deficiency signaling, one major innate immune pathway in Drosophila During immune activation, downregulated plin1 promotes the enlargement of LDs, which in turn alleviates immune reaction-associated reactive oxygen species stress. Thus, the growth of LDs is likely an active adaptation to maintain redox homeostasis in response to immune deficiency activation. Therefore, our study provides evidence that plin1 serves as a modulator on LDs' reconfiguration in regulating infection-induced pathogenesis, and plin1 might be a potential therapeutic target for coordinating inflammation resolution and lipid metabolism.
Collapse
Affiliation(s)
- Lei Wang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China.,The Center for Microbes, Development, and Health, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jiaxin Lin
- The Center for Microbes, Development, and Health, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Junjing Yu
- Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China; and
| | - Kaiyan Yang
- The Center for Microbes, Development, and Health, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Li Sun
- The Center for Microbes, Development, and Health, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Hong Tang
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China;
| | - Lei Pan
- The Center for Microbes, Development, and Health, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China; .,University of Chinese Academy of Sciences, Beijing, China.,Chinese Academy of Sciences Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
22
|
A genetic model of methionine restriction extends Drosophila health- and lifespan. Proc Natl Acad Sci U S A 2021; 118:2110387118. [PMID: 34588310 DOI: 10.1073/pnas.2110387118] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2021] [Indexed: 12/12/2022] Open
Abstract
Loss of metabolic homeostasis is a hallmark of aging and is characterized by dramatic metabolic reprogramming. To analyze how the fate of labeled methionine is altered during aging, we applied 13C5-Methionine labeling to Drosophila and demonstrated significant changes in the activity of different branches of the methionine metabolism as flies age. We further tested whether targeted degradation of methionine metabolism components would "reset" methionine metabolism flux and extend the fly lifespan. Specifically, we created transgenic flies with inducible expression of Methioninase, a bacterial enzyme capable of degrading methionine and revealed methionine requirements for normal maintenance of lifespan. We also demonstrated that microbiota-derived methionine is an alternative and important source in addition to food-derived methionine. In this genetic model of methionine restriction (MetR), we also demonstrate that either whole-body or tissue-specific Methioninase expression can dramatically extend Drosophila health- and lifespan and exerts physiological effects associated with MetR. Interestingly, while previous dietary MetR extended lifespan in flies only in low amino acid conditions, MetR from Methioninase expression extends lifespan independently of amino acid levels in the food. Finally, because impairment of the methionine metabolism has been previously associated with the development of Alzheimer's disease, we compared methionine metabolism reprogramming between aging flies and a Drosophila model relevant to Alzheimer's disease, and found that overexpression of human Tau caused methionine metabolism flux reprogramming similar to the changes found in aged flies. Altogether, our study highlights Methioninase as a potential agent for health- and lifespan extension.
Collapse
|
23
|
Weston RM, Schmitt RE, Grotewiel M, Miles MF. Transcriptome analysis of chloride intracellular channel knockdown in Drosophila identifies oxidation-reduction function as possible mechanism of altered sensitivity to ethanol sedation. PLoS One 2021; 16:e0246224. [PMID: 34228751 PMCID: PMC8259981 DOI: 10.1371/journal.pone.0246224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 06/18/2021] [Indexed: 01/22/2023] Open
Abstract
Chloride intracellular channels (CLICs) are a unique family of evolutionarily conserved metamorphic proteins, switching between stable conformations based on redox conditions. CLICs have been implicated in a wide variety biological processes including ion channel activity, apoptosis, membrane trafficking, and enzymatic oxidoreductase activity. Understanding the molecular mechanisms by which CLICs engage in these activities is an area of active research. Here, the sole Drosophila melanogaster ortholog, Clic, was targeted for RNAi knockdown to identify genes and biological processes associated with Clic expression. Clic knockdown had a substantial impact on global transcription, altering expression of over 7% of transcribed Drosophila genes. Overrepresentation analysis of differentially expressed genes identified enrichment of Gene Ontology terms including Cytoplasmic Translation, Oxidation-Reduction Process, Heme Binding, Membrane, Cell Junction, and Nucleolus. The top term, Cytoplasmic Translation, was enriched almost exclusively with downregulated genes. Drosophila Clic and vertebrate ortholog Clic4 have previously been tied to ethanol sensitivity and ethanol-regulated expression. Clic knockdown-responsive genes from the present study were found to overlap significantly with gene sets from 4 independently published studies related to ethanol exposure and sensitivity in Drosophila. Bioinformatic analysis of genes shared between these studies revealed an enrichment of genes related to amino acid metabolism, protein processing, oxidation-reduction processes, and lipid particles among others. To determine whether the modulation of ethanol sensitivity by Clic may be related to co-regulated oxidation-reduction processes, we evaluated the effect of hyperoxia on ethanol sedation in Clic knockdown flies. Consistent with previous findings, Clic knockdown reduced acute ethanol sedation sensitivity in flies housed under normoxia. However, this effect was reversed by exposure to hyperoxia, suggesting a common set of molecular-genetic mechanism may modulate each of these processes. This study suggests that Drosophila Clic has a major influence on regulation of oxidative stress signaling and that this function overlaps with the molecular mechanisms of acute ethanol sensitivity in the fly.
Collapse
Affiliation(s)
- Rory M. Weston
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, United States of America
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Rebecca E. Schmitt
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Mike Grotewiel
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Michael F. Miles
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, United States of America
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| |
Collapse
|
24
|
Ma P, Zhang Y, Liang Q, Yin Y, Wang S, Han R, Huo C, Deng H. Mifepristone (RU486) inhibits dietary lipid digestion by antagonizing the role of glucocorticoid receptor on lipase transcription. iScience 2021; 24:102507. [PMID: 34308280 PMCID: PMC8257970 DOI: 10.1016/j.isci.2021.102507] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 04/08/2021] [Accepted: 04/26/2021] [Indexed: 12/15/2022] Open
Abstract
Lipid digestion and absorption are tightly regulated to cope with metabolic demands among tissues. How these processes are coordinated is not well characterized. Here, we found that mifepristone (RU486) prevents lipid digestion both in flies and mice. In flies, RU486 administration suppresses lipid digestion by transcriptional downregulating Magro in guts. Similarly, intestinal lipid uptake in mice was also suppressed by RU486 through the glucocorticoid receptor (GR). Further studies showed that the pancreatic lipase Pnlip is a direct transcriptional target of GR in pancreas tissues. Glucocorticoid levels in mice fed a high fat diet (HFD) are significantly lower than those fed on a conventional diet, and RU486 administration inhibits HFD-induced obesity both in mice and flies. Our findings identified a novel mechanism of RU486 functions as a GR antagonist systematically regulating lipid metabolism, providing new insight on the role of Glucocorticoid/GR in Cushing disease, diabetes, and other related metabolic syndromes. RU486 suppresses lipid digestion both in mice and flies. In flies, lipase Magro is transcriptionally suppressed by RU486 through dERR. In mice, intestinal lipid digestion is inhibited by RU486 through (GR)/PTL pathway in pancreas. RU486 alleviates high fat diet-induced obesity both in flies and mice.
Collapse
Affiliation(s)
- Peng Ma
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 6B, Shixun Bldg, 1239 Siping Road, Yangpu District, Shanghai, 20092, China
| | - Yao Zhang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 6B, Shixun Bldg, 1239 Siping Road, Yangpu District, Shanghai, 20092, China
| | - Qiying Liang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 6B, Shixun Bldg, 1239 Siping Road, Yangpu District, Shanghai, 20092, China
| | - Youjie Yin
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 6B, Shixun Bldg, 1239 Siping Road, Yangpu District, Shanghai, 20092, China
| | - Saifei Wang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 6B, Shixun Bldg, 1239 Siping Road, Yangpu District, Shanghai, 20092, China
| | - Ruolei Han
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 6B, Shixun Bldg, 1239 Siping Road, Yangpu District, Shanghai, 20092, China
| | - Chunyu Huo
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 6B, Shixun Bldg, 1239 Siping Road, Yangpu District, Shanghai, 20092, China
| | - Hansong Deng
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 6B, Shixun Bldg, 1239 Siping Road, Yangpu District, Shanghai, 20092, China
| |
Collapse
|
25
|
Martínez Corrales G, Filer D, Wenz KC, Rogan A, Phillips G, Li M, Feseha Y, Broughton SJ, Alic N. Partial Inhibition of RNA Polymerase I Promotes Animal Health and Longevity. Cell Rep 2021; 30:1661-1669.e4. [PMID: 32049000 PMCID: PMC7013379 DOI: 10.1016/j.celrep.2020.01.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/12/2019] [Accepted: 01/03/2020] [Indexed: 12/19/2022] Open
Abstract
Health and survival in old age can be improved by changes in gene expression. RNA polymerase (Pol) I is the essential, conserved enzyme whose task is to generate the pre-ribosomal RNA (rRNA). We find that reducing the levels of Pol I activity is sufficient to extend lifespan in the fruit fly. This effect can be recapitulated by partial, adult-restricted inhibition, with both enterocytes and stem cells of the adult midgut emerging as important cell types. In stem cells, Pol I appears to act in the same longevity pathway as Pol III, implicating rRNA synthesis in these cells as the key lifespan determinant. Importantly, reduction in Pol I activity delays broad, age-related impairment and pathology, improving the function of diverse organ systems. Hence, our study shows that Pol I activity in the adult drives systemic, age-related decline in animal health and anticipates mortality. Partial inhibition of RNA polymerase I (Pol I) can extend lifespan in the fruit fly Reducing Pol I activity after development and only in the gut is sufficient Pol I activity affects aging from both post-mitotic and mitotically active cells Pol I activity affects the age-related decline in performance of multiple organs
Collapse
Affiliation(s)
- Guillermo Martínez Corrales
- Institute of Healthy Ageing and the Research Department of Genetics, Evolution, and Environment, University College London, WC1E 6BT London, UK
| | - Danny Filer
- Institute of Healthy Ageing and the Research Department of Genetics, Evolution, and Environment, University College London, WC1E 6BT London, UK
| | - Katharina C Wenz
- Institute of Healthy Ageing and the Research Department of Genetics, Evolution, and Environment, University College London, WC1E 6BT London, UK
| | - Abbie Rogan
- Institute of Healthy Ageing and the Research Department of Genetics, Evolution, and Environment, University College London, WC1E 6BT London, UK
| | - George Phillips
- Institute of Healthy Ageing and the Research Department of Genetics, Evolution, and Environment, University College London, WC1E 6BT London, UK
| | - Mengjia Li
- Institute of Healthy Ageing and the Research Department of Genetics, Evolution, and Environment, University College London, WC1E 6BT London, UK
| | - Yodit Feseha
- Institute of Healthy Ageing and the Research Department of Genetics, Evolution, and Environment, University College London, WC1E 6BT London, UK
| | - Susan J Broughton
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, LA1 4YQ Lancaster, UK
| | - Nazif Alic
- Institute of Healthy Ageing and the Research Department of Genetics, Evolution, and Environment, University College London, WC1E 6BT London, UK.
| |
Collapse
|
26
|
Driesschaert B, Mergan L, Temmerman L. Conditional gene expression in invertebrate animal models. J Genet Genomics 2021; 48:14-31. [PMID: 33814307 DOI: 10.1016/j.jgg.2021.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/11/2020] [Accepted: 01/08/2021] [Indexed: 10/22/2022]
Abstract
A mechanistic understanding of biology requires appreciating spatiotemporal aspects of gene expression and its functional implications. Conditional expression allows for (ir)reversible switching of genes on or off, with the potential of spatial and/or temporal control. This provides a valuable complement to the more often used constitutive gene (in)activation through mutagenesis, providing tools to answer a wider array of research questions across biological disciplines. Spatial and/or temporal control are granted primarily by (combinations of) specific promoters, temperature regimens, compound addition, or illumination. The use of such genetic tool kits is particularly widespread in invertebrate animal models because they can be applied to study biological processes in short time frames and on large scales, using organisms amenable to easy genetic manipulation. Recent years witnessed an exciting expansion and optimization of such tools, of which we provide a comprehensive overview and discussion regarding their use in invertebrates. The mechanism, applicability, benefits, and drawbacks of each of the systems, as well as further developments to be expected in the foreseeable future, are highlighted.
Collapse
Affiliation(s)
- Brecht Driesschaert
- Animal Physiology and Neurobiology, Department of Biology, University of Leuven (KU Leuven), Naamsestraat 59 - Box 2465, B-3000 Leuven, Belgium
| | - Lucas Mergan
- Animal Physiology and Neurobiology, Department of Biology, University of Leuven (KU Leuven), Naamsestraat 59 - Box 2465, B-3000 Leuven, Belgium
| | - Liesbet Temmerman
- Animal Physiology and Neurobiology, Department of Biology, University of Leuven (KU Leuven), Naamsestraat 59 - Box 2465, B-3000 Leuven, Belgium.
| |
Collapse
|
27
|
Parkhitko AA, Ramesh D, Wang L, Leshchiner D, Filine E, Binari R, Olsen AL, Asara JM, Cracan V, Rabinowitz JD, Brockmann A, Perrimon N. Downregulation of the tyrosine degradation pathway extends Drosophila lifespan. eLife 2020; 9:58053. [PMID: 33319750 PMCID: PMC7744100 DOI: 10.7554/elife.58053] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 11/28/2020] [Indexed: 12/31/2022] Open
Abstract
Aging is characterized by extensive metabolic reprogramming. To identify metabolic pathways associated with aging, we analyzed age-dependent changes in the metabolomes of long-lived Drosophila melanogaster. Among the metabolites that changed, levels of tyrosine were increased with age in long-lived flies. We demonstrate that the levels of enzymes in the tyrosine degradation pathway increase with age in wild-type flies. Whole-body and neuronal-specific downregulation of enzymes in the tyrosine degradation pathway significantly extends Drosophila lifespan, causes alterations of metabolites associated with increased lifespan, and upregulates the levels of tyrosine-derived neuromediators. Moreover, feeding wild-type flies with tyrosine increased their lifespan. Mechanistically, we show that suppression of ETC complex I drives the upregulation of enzymes in the tyrosine degradation pathway, an effect that can be rescued by tigecycline, an FDA-approved drug that specifically suppresses mitochondrial translation. In addition, tyrosine supplementation partially rescued lifespan of flies with ETC complex I suppression. Altogether, our study highlights the tyrosine degradation pathway as a regulator of longevity.
Collapse
Affiliation(s)
- Andrey A Parkhitko
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, United States.,Aging Institute of UPMC and the University of Pittsburgh, Pittsburgh, United States
| | - Divya Ramesh
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India.,Department of Biology, University of Konstanz, Konstanz, Germany
| | - Lin Wang
- Department of Chemistry, Princeton University, Princeton, United States.,Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, United States
| | - Dmitry Leshchiner
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, United States
| | - Elizabeth Filine
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, United States
| | - Richard Binari
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, United States.,Howard Hughes Medical Institute, Boston, United States
| | - Abby L Olsen
- Department of Neurology, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, United States
| | - John M Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center, and Department of Medicine, Harvard Medical School, Boston, United States
| | - Valentin Cracan
- Scintillon Institute, San Diego, United States.,Department of Chemistry, The Scripps Research Institute, La Jolla, United States
| | - Joshua D Rabinowitz
- Department of Chemistry, Princeton University, Princeton, United States.,Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, United States
| | - Axel Brockmann
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, United States.,Howard Hughes Medical Institute, Boston, United States
| |
Collapse
|
28
|
Bjedov I, Cochemé HM, Foley A, Wieser D, Woodling NS, Castillo-Quan JI, Norvaisas P, Lujan C, Regan JC, Toivonen JM, Murphy MP, Thornton J, Kinghorn KJ, Neufeld TP, Cabreiro F, Partridge L. Fine-tuning autophagy maximises lifespan and is associated with changes in mitochondrial gene expression in Drosophila. PLoS Genet 2020; 16:e1009083. [PMID: 33253201 PMCID: PMC7738165 DOI: 10.1371/journal.pgen.1009083] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 12/15/2020] [Accepted: 08/26/2020] [Indexed: 01/26/2023] Open
Abstract
Increased cellular degradation by autophagy is a feature of many interventions that delay ageing. We report here that increased autophagy is necessary for reduced insulin-like signalling (IIS) to extend lifespan in Drosophila and is sufficient on its own to increase lifespan. We first established that the well-characterised lifespan extension associated with deletion of the insulin receptor substrate chico was completely abrogated by downregulation of the essential autophagy gene Atg5. We next directly induced autophagy by over-expressing the major autophagy kinase Atg1 and found that a mild increase in autophagy extended lifespan. Interestingly, strong Atg1 up-regulation was detrimental to lifespan. Transcriptomic and metabolomic approaches identified specific signatures mediated by varying levels of autophagy in flies. Transcriptional upregulation of mitochondrial-related genes was the signature most specifically associated with mild Atg1 upregulation and extended lifespan, whereas short-lived flies, possessing strong Atg1 overexpression, showed reduced mitochondrial metabolism and up-regulated immune system pathways. Increased proteasomal activity and reduced triacylglycerol levels were features shared by both moderate and high Atg1 overexpression conditions. These contrasting effects of autophagy on ageing and differential metabolic profiles highlight the importance of fine-tuning autophagy levels to achieve optimal healthspan and disease prevention.
Collapse
Affiliation(s)
- Ivana Bjedov
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
- UCL Cancer Institute, Paul O'Gorman Building, London United Kingdom
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Helena M. Cochemé
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- MRC London Institute of Medical Sciences, Imperial College London, Du Cane Road, London, United Kingdom
| | - Andrea Foley
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
- MRC London Institute of Medical Sciences, Imperial College London, Du Cane Road, London, United Kingdom
| | - Daniela Wieser
- EMBL European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Nathaniel S. Woodling
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
| | - Jorge Iván Castillo-Quan
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Section on Islet Cell and Regenerative Biology, Research Division, Joslin Diabetes Center, Boston MA, United States of America
- Department of Genetics and Harvard Stem Cell Institute, Harvard Medical School, Boston MA, United States of America
| | - Povilas Norvaisas
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Celia Lujan
- UCL Cancer Institute, Paul O'Gorman Building, London United Kingdom
| | - Jennifer C. Regan
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Janne M. Toivonen
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
- LAGENBIO, Facultad de Veterinaria-IIS, IA2-CITA, CIBERNED, Universidad de Zaragoza, Zaragoza, Spain
| | - Michael P. Murphy
- MRC Mitochondrial Biology Unit, the Keith Peters Building, University of Cambridge, Cambridge, United Kingdom
| | - Janet Thornton
- EMBL European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Kerri J. Kinghorn
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
| | - Thomas P. Neufeld
- Department of Genetics, Cell Biology and Development, University of Minnesota, 321 Church St. SE, Minneapolis, MN, United States of America
| | - Filipe Cabreiro
- MRC London Institute of Medical Sciences, Imperial College London, Du Cane Road, London, United Kingdom
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Linda Partridge
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| |
Collapse
|
29
|
Smith HJ, Sharma A, Mair WB. Metabolic Communication and Healthy Aging: Where Should We Focus Our Energy? Dev Cell 2020; 54:196-211. [PMID: 32619405 DOI: 10.1016/j.devcel.2020.06.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/01/2020] [Accepted: 06/07/2020] [Indexed: 02/09/2023]
Abstract
Aging is associated with a loss of metabolic homeostasis and plasticity, which is causally linked to multiple age-onset pathologies. The majority of the interventions-genetic, dietary, and pharmacological-that have been found to slow aging and protect against age-related disease in various organisms do so by targeting central metabolic pathways. However, targeting metabolic pathways chronically and ubiquitously makes it difficult to define the downstream effects responsible for lifespan extension and often results in negative effects on growth and health, limiting therapeutic potential. Insight into how metabolic signals are relayed between tissues, cells, and organelles opens up new avenues to target metabolic regulators locally rather than globally for healthy aging. In this review, we discuss the pro-longevity effects of targeting metabolic pathways in specific tissues and how these interventions communicate with distal cells to modulate aging. These studies may be crucial in designing interventions that promote longevity without negative health consequences.
Collapse
Affiliation(s)
- Hannah J Smith
- Harvard T.H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - Arpit Sharma
- Harvard T.H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA
| | - William B Mair
- Harvard T.H. Chan School of Public Health, Department of Molecular Metabolism, Boston, MA, USA.
| |
Collapse
|
30
|
Schmitt RE, Messick MR, Shell BC, Dunbar EK, Fang H, Shelton KL, Venton BJ, Pletcher SD, Grotewiel M. Dietary yeast influences ethanol sedation in Drosophila via serotonergic neuron function. Addict Biol 2020; 25:e12779. [PMID: 31169340 DOI: 10.1111/adb.12779] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 03/23/2019] [Accepted: 05/02/2019] [Indexed: 01/10/2023]
Abstract
Abuse of alcohol is a major clinical problem with far-reaching health consequences. Understanding the environmental and genetic factors that contribute to alcohol-related behaviors is a potential gateway for developing novel therapeutic approaches for patients that abuse the drug. To this end, we have used Drosophila melanogaster as a model to investigate the effect of diet, an environmental factor, on ethanol sedation. Providing flies with diets high in yeast, a routinely used component of fly media, increased their resistance to ethanol sedation. The yeast-induced resistance to ethanol sedation occurred in several different genetic backgrounds, was observed in males and females, was elicited by yeast from different sources, was readily reversible, and was associated with increased nutrient intake as well as decreased internal ethanol levels. Inhibition of serotonergic neuron function using multiple independent genetic manipulations blocked the effect of yeast supplementation on ethanol sedation, nutrient intake, and internal ethanol levels. Our results demonstrate that yeast is a critical dietary component that influences ethanol sedation in flies and that serotonergic signaling is required for the effect of dietary yeast on nutrient intake, ethanol uptake/elimination, and ethanol sedation. Our studies establish the fly as a model for diet-induced changes in ethanol sedation and raise the possibility that serotonin might mediate the effect of diet on alcohol-related behavior in other species.
Collapse
Affiliation(s)
- Rebecca E. Schmitt
- Department of Human and Molecular Genetics Virginia Commonwealth University Richmond VA USA
| | - Monica R. Messick
- Department of Human and Molecular Genetics Virginia Commonwealth University Richmond VA USA
| | - Brandon C. Shell
- Department of Human and Molecular Genetics Virginia Commonwealth University Richmond VA USA
| | - Ellyn K. Dunbar
- Department of Human and Molecular Genetics Virginia Commonwealth University Richmond VA USA
| | - Huai‐Fang Fang
- Department of Chemistry and Neuroscience Graduate Program University of Virginia Charlottesville VA USA
| | - Keith L. Shelton
- Department of Pharmacology and Toxicology Virginia Commonwealth University Richmond VA USA
| | - B. Jill Venton
- Department of Chemistry and Neuroscience Graduate Program University of Virginia Charlottesville VA USA
| | - Scott D. Pletcher
- Department of Molecular and Integrative Physiology and Geriatrics Center University of Michigan Ann Arbor MI USA
| | - Mike Grotewiel
- Department of Human and Molecular Genetics Virginia Commonwealth University Richmond VA USA
- Virginia Commonwealth University Alcohol Research Center Richmond VA USA
| |
Collapse
|
31
|
Harrison BR, Wang L, Gajda E, Hoffman EV, Chung BY, Pletcher SD, Raftery D, Promislow DEL. The metabolome as a link in the genotype-phenotype map for peroxide resistance in the fruit fly, Drosophila melanogaster. BMC Genomics 2020; 21:341. [PMID: 32366330 PMCID: PMC7199327 DOI: 10.1186/s12864-020-6739-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 04/15/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Genetic association studies that seek to explain the inheritance of complex traits typically fail to explain a majority of the heritability of the trait under study. Thus, we are left with a gap in the map from genotype to phenotype. Several approaches have been used to fill this gap, including those that attempt to map endophenotype such as the transcriptome, proteome or metabolome, that underlie complex traits. Here we used metabolomics to explore the nature of genetic variation for hydrogen peroxide (H2O2) resistance in the sequenced inbred Drosophila Genetic Reference Panel (DGRP). RESULTS We first studied genetic variation for H2O2 resistance in 179 DGRP lines and along with identifying the insulin signaling modulator u-shaped and several regulators of feeding behavior, we estimate that a substantial amount of phenotypic variation can be explained by a polygenic model of genetic variation. We then profiled a portion of the aqueous metabolome in subsets of eight 'high resistance' lines and eight 'low resistance' lines. We used these lines to represent collections of genotypes that were either resistant or sensitive to the stressor, effectively modeling a discrete trait. Across the range of genotypes in both populations, flies exhibited surprising consistency in their metabolomic signature of resistance. Importantly, the resistance phenotype of these flies was more easily distinguished by their metabolome profiles than by their genotypes. Furthermore, we found a metabolic response to H2O2 in sensitive, but not in resistant genotypes. Metabolomic data further implicated at least two pathways, glycogen and folate metabolism, as determinants of sensitivity to H2O2. We also discovered a confounding effect of feeding behavior on assays involving supplemented food. CONCLUSIONS This work suggests that the metabolome can be a point of convergence for genetic variation influencing complex traits, and can efficiently elucidate mechanisms underlying trait variation.
Collapse
Affiliation(s)
- Benjamin R Harrison
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, 98195, USA.
| | - Lu Wang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, 98105, USA
| | - Erika Gajda
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Elise V Hoffman
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Brian Y Chung
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Scott D Pletcher
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Daniel Raftery
- Northwest Metabolomics Research Center, Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Daniel E L Promislow
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, 98195, USA
- Department of Biology, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
32
|
Expression of Heat Shock Protein 70 Is Insufficient To Extend Drosophila melanogaster Longevity. G3-GENES GENOMES GENETICS 2019; 9:4197-4207. [PMID: 31624139 PMCID: PMC6893204 DOI: 10.1534/g3.119.400782] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
It has been known for over 20 years that Drosophila melanogaster flies with twelve additional copies of the hsp70 gene encoding the 70 kD heat shock protein lives longer after a non-lethal heat treatment. Since the heat treatment also induces the expression of additional heat shock proteins, the biological effect can be due either to HSP70 acting alone or in combination. This study used the UAS/GAL4 system to determine whether hsp70 is sufficient to affect the longevity and the resistance to thermal, oxidative or desiccation stresses of the whole organism. We observed that HSP70 expression in the nervous system or muscles has no effect on longevity or stress resistance but ubiquitous expression reduces the life span of males. We also observed that the down-regulation of hsp70 using RNAi did not affect longevity.
Collapse
|
33
|
Coelho DS, Schwartz S, Merino MM, Hauert B, Topfel B, Tieche C, Rhiner C, Moreno E. Culling Less Fit Neurons Protects against Amyloid-β-Induced Brain Damage and Cognitive and Motor Decline. Cell Rep 2019; 25:3661-3673.e3. [PMID: 30590040 PMCID: PMC6315112 DOI: 10.1016/j.celrep.2018.11.098] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 07/05/2018] [Accepted: 11/28/2018] [Indexed: 01/25/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia, impairing cognitive and motor functions. One of the pathological hallmarks of AD is neuronal loss, which is not reflected in mouse models of AD. Therefore, the role of neuronal death is still uncertain. Here, we used a Drosophila AD model expressing a secreted form of human amyloid-β42 peptide and showed that it recapitulates key aspects of AD pathology, including neuronal death and impaired long-term memory. We found that neuronal apoptosis is mediated by cell fitness-driven neuronal culling, which selectively eliminates impaired neurons from brain circuits. We demonstrated that removal of less fit neurons delays β-amyloid-induced brain damage and protects against cognitive and motor decline, suggesting that contrary to common knowledge, neuronal death may have a beneficial effect in AD. Peptides linked to neurodegenerative diseases reduce neuronal fitness in Drosophila β-amyloid-induced neuronal death is mediated by fitness regulators flower and azot Suppression of fitness-based neuronal culling aggravates cognitive and motor decline Neuronal death related to fitness-based selection has a beneficial net effect
Collapse
Affiliation(s)
- Dina S Coelho
- Cell Fitness Lab, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal; Institute for Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Silvia Schwartz
- Stem Cells and Regeneration Lab, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal
| | - Marisa M Merino
- Institute for Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland; Department of Biochemistry, University of Geneva, Quai Ernest-Ansermet 30, 1211 Geneva 4, Switzerland
| | - Barbara Hauert
- Institute for Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Barbara Topfel
- Institute for Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Colin Tieche
- Institute for Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Christa Rhiner
- Institute for Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland; Stem Cells and Regeneration Lab, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal.
| | - Eduardo Moreno
- Cell Fitness Lab, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal; Institute for Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland.
| |
Collapse
|
34
|
Ke YT, Hsu HJ. Generation of Inducible Gene-Switched GAL4 Expressed in the Drosophila Female Germline Stem Cell Niche. G3 (BETHESDA, MD.) 2019; 9:2007-2016. [PMID: 31018943 PMCID: PMC6553524 DOI: 10.1534/g3.119.400246] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/17/2019] [Indexed: 12/14/2022]
Abstract
The stem cell niche, a regulatory microenvironment, houses and regulates stem cells for maintenance of tissues throughout an organism's lifespan. While it is known that stem cell function declines with age, the role of niche cells in this decline is not completely understood. Drosophila exhibits a short lifespan with well-characterized ovarian germline stem cells (GSCs) and niche compartments, providing a good model with which to study stem cell biology. However, no inducible tools for temporal and spatial control of gene expression in the GSC-niche unit have been previously developed for aging studies. The current UAS-GAL4 systems are not ideal for aging studies because fly physiological aging may be affected by the temperature shifts used to manipulate GAL4 activity. Additionally, the actual needs of the aged niche may be masked by continuously driven gene expression. Since GeneSwitch GAL4 is conveniently activated by the steroid RU486 (mifepristone), we conducted an enhancer-trap screen to isolate GeneSwitch GAL4 lines with expression in the GSC-niche unit. We identified six lines with expression in germarial somatic cells, and two lines (#2305 and #2261) with expression in niche cap cells, the major constituent of the GSC niche. The use of lines #2305 or #2261 to overexpress Drosophila insulin-like peptide 2, which maintains GSC lifespan, in aged niche cap cells significantly delayed age-dependent GSC loss. These results support the notion that insulin signaling is beneficial for maintaining aged stem cells and also validate the utility of our GeneSwitch GAL4 lines for studying stem cell aging.
Collapse
Affiliation(s)
- Yi-Teng Ke
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Hwei-Jan Hsu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| |
Collapse
|
35
|
Tissue-specific alteration of gene expression and function by RU486 and the GeneSwitch system. NPJ Aging Mech Dis 2019; 5:6. [PMID: 31123597 PMCID: PMC6529481 DOI: 10.1038/s41514-019-0036-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 05/06/2019] [Indexed: 12/11/2022] Open
Abstract
The GeneSwitch (GS) is a modified Gal4/UAS system, whereby transgene expression is induced in Drosophila by adding the drug RU486 to food. The GS system is routinely used in Drosophila aging and behavioral studies to avoid confounding effects related to genetic background mutations. Here, we report transcriptional and functional defects that are induced by RU486 in a stock- and tissue-dependent manner, such as defects in flight and mitochondrial gene expression. In addition to including proper controls, our findings suggest that context-specific side effects induced by RU486 should be considered in the experimental design and when interpreting the observed phenotypes.
Collapse
|
36
|
Moskalev A, Proshkina E, Zhavoronkov A, Shaposhnikov M. Effects of unpaired 1 gene overexpression on the lifespan of Drosophila melanogaster. BMC SYSTEMS BIOLOGY 2019; 13:16. [PMID: 30836998 PMCID: PMC6399815 DOI: 10.1186/s12918-019-0687-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Background The JAK/STAT signaling pathway is involved in many aging-related cellular functions. However, effects of overexpression of genes controlling JAK/STAT signal transduction on longevity of model organisms have not been studied. Here we evaluate the effect of overexpression of the unpaired 1 (upd1) gene, which encodes an activating ligand for JAK/STAT pathway, on the lifespan of Drosophila melanogaster. Results Overexpression of upd1 in the intestine caused a pronounced shortening of the median lifespan by 54.1–18.9%, and the age of 90% mortality by 40.9–19.1% in males and females, respectively. In fat body and in nervous system of male flies, an induction of upd1 overexpression increased the age of 90% mortality and median lifespan, respectively. An increase in upd1 expression enhanced mRNA levels of the JAK/STAT target genes domeless and Socs36E. Conclusions Conditional overexpression of upd1 in different tissues of Drosophila imago induces pro-aging or pro-longevity effects in tissue-dependent manner. The effects of upd1 overexpression on lifespan are accompanied by the transcription activation of genes for the components of JAK/STAT pathway.
Collapse
Affiliation(s)
- Alexey Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia. .,Institute of Biology, Komi Scientific Center, Ural Division, Russian Academy of Sciences, Syktyvkar, 167982, Russia. .,Moscow Institute of Physics and Technology, Dolgoprudny, 141700, Russia.
| | - Ekaterina Proshkina
- Institute of Biology, Komi Scientific Center, Ural Division, Russian Academy of Sciences, Syktyvkar, 167982, Russia
| | | | - Mikhail Shaposhnikov
- Institute of Biology, Komi Scientific Center, Ural Division, Russian Academy of Sciences, Syktyvkar, 167982, Russia
| |
Collapse
|
37
|
A New Tool for Inducible Gene Expression in Caenorhabditis elegans. Genetics 2018; 211:419-430. [PMID: 30504365 DOI: 10.1534/genetics.118.301705] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 11/29/2018] [Indexed: 02/07/2023] Open
Abstract
Controlling protein activity and localization is a key tool in modern biology. Mammalian steroid receptor ligand-binding domain (LBD) fusions have been used in a range of organisms and cell types to inactivate proteins of interest until the cognate steroid ligand is applied. Here, we demonstrate that the glucocorticoid receptor LBD confers ligand-gated control of a heterologous gene expression system (Q system) and the DAF-16 transcription factor in Caenorhabditis elegans These experiments provide a powerful tool for temporal control of protein activity, and will bolster existing tools used to modulate gene expression and protein activity in this animal.
Collapse
|
38
|
Solovev I, Shegoleva E, Fedintsev A, Shaposhnikov M, Moskalev A. Circadian clock genes' overexpression in Drosophila alters diet impact on lifespan. Biogerontology 2018; 20:159-170. [PMID: 30470951 DOI: 10.1007/s10522-018-9784-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/13/2018] [Indexed: 12/16/2022]
Abstract
Diet restriction is one of the most accurately confirmed interventions which extend lifespan. Genes coding circadian core clock elements are known to be the key controllers of cell metabolism especially in aging aspect. The molecular mechanisms standing behind the phenomenon of diet-restriction-mediated life extension are connected to circadian clock either. Here we investigate the effects of protein-rich and low-protein diets on lifespan observed in fruit flies overexpressing core clock genes (cry, per, Clk, cyc and tim). The majority of core clock genes being upregulated in peripheral tissues (muscles and fat body) on protein-rich diet significantly decrease the lifespan of male fruit flies from 5 to 61%. Nevertheless, positive increments of median lifespan were observed in both sexes, males overexpressing cry in fat body lived 20% longer on poor diet. Overexpression of per also on poor medium resulted in life extension in female fruit flies. Diet restriction reduces mortality caused by overexpression of core clock genes. Cox-regression model revealed that diet restriction seriously decreases mortality risks of flies which overexpress core clock genes. The hazard ratios are lower for flies overexpressing clock genes in fat body relatively to muscle-specific overexpression. The present work suggests a phenomenological view of how two peripheral circadian oscillators modify effects of rich and poor diets on lifespan and hazard ratios.
Collapse
Affiliation(s)
- Ilya Solovev
- Laboratory of Molecular Radiobiology and Gerontology, Komi Science Center, Institute of Biology, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya St., Syktyvkar, Komi Republic, Russian Federation, 167982.,Pitirim Sorokin Syktyvkar State University, Syktyvkar, Komi Republic, Russian Federation, 167001
| | - Eugenia Shegoleva
- Laboratory of Molecular Radiobiology and Gerontology, Komi Science Center, Institute of Biology, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya St., Syktyvkar, Komi Republic, Russian Federation, 167982
| | - Alexander Fedintsev
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russian Federation, 141701
| | - Mikhail Shaposhnikov
- Laboratory of Molecular Radiobiology and Gerontology, Komi Science Center, Institute of Biology, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya St., Syktyvkar, Komi Republic, Russian Federation, 167982
| | - Alexey Moskalev
- Laboratory of Molecular Radiobiology and Gerontology, Komi Science Center, Institute of Biology, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya St., Syktyvkar, Komi Republic, Russian Federation, 167982. .,Pitirim Sorokin Syktyvkar State University, Syktyvkar, Komi Republic, Russian Federation, 167001. .,Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russian Federation, 141701. .,Laboratory of Post-Genomic Research, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russian Federation, 119991.
| |
Collapse
|
39
|
Gáliková M, Klepsatel P. Obesity and Aging in the Drosophila Model. Int J Mol Sci 2018; 19:ijms19071896. [PMID: 29954158 PMCID: PMC6073435 DOI: 10.3390/ijms19071896] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 06/19/2018] [Accepted: 06/25/2018] [Indexed: 02/06/2023] Open
Abstract
Being overweight increases the risk of many metabolic disorders, but how it affects lifespan is not completely clear. Not all obese people become ill, and the exact mechanism that turns excessive fat storage into a health-threatening state remains unknown. Drosophila melanogaster has served as an excellent model for many diseases, including obesity, diabetes, and hyperglycemia-associated disorders, such as cardiomyopathy or nephropathy. Here, we review the connections between fat storage and aging in different types of fly obesity. Whereas obesity induced by high-fat or high-sugar diet is associated with hyperglycemia, cardiomyopathy, and in some cases, shortening of lifespan, there are also examples in which obesity correlates with longevity. Transgenic lines with downregulations of the insulin/insulin-like growth factor (IIS) and target of rapamycin (TOR) signaling pathways, flies reared under dietary restriction, and even certain longevity selection lines are obese, yet long-lived. The mechanisms that underlie the differential lifespans in distinct types of obesity remain to be elucidated, but fat turnover, inflammatory pathways, and dysregulations of glucose metabolism may play key roles. Altogether, Drosophila is an excellent model to study the physiology of adiposity in both health and disease.
Collapse
Affiliation(s)
- Martina Gáliková
- Department of Zoology, Stockholm University, Svante Arrhenius väg 18B, S-106 91 Stockholm, Sweden.
| | - Peter Klepsatel
- Institute of Zoology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 06 Bratislava, Slovakia.
| |
Collapse
|
40
|
Langerak S, Kim MJ, Lamberg H, Godinez M, Main M, Winslow L, O'Connor MB, Zhu CC. The Drosophila TGF-beta/Activin-like ligands Dawdle and Myoglianin appear to modulate adult lifespan through regulation of 26S proteasome function in adult muscle. Biol Open 2018; 7:bio.029454. [PMID: 29615416 PMCID: PMC5936056 DOI: 10.1242/bio.029454] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The Drosophila Activin signaling pathway employs at least three separate ligands – Activin-β (Actβ), Dawdle (Daw), and Myoglianin (Myo) – to regulate several general aspects of fruit fly larval development, including cell proliferation, neuronal remodeling, and metabolism. Here we provide experimental evidence indicating that both Daw and Myo are anti-ageing factors in adult fruit flies. Knockdown of Myo or Daw in adult fruit flies reduced mean lifespan, while overexpression of either ligand in adult muscle tissues but not in adipose tissues enhanced mean lifespan. An examination of ubiquitinated protein aggregates in adult muscles revealed a strong inverse correlation between Myo- or Daw-initiated Activin signaling and the amount of ubiquitinated protein aggregates. We show that this correlation has important functional implications by demonstrating that the lifespan extension effect caused by overexpression of wild-type Daw or Myo in adult muscle tissues can be completely abrogated by knockdown of a 26S proteasome regulatory subunit Rpn1 in adult fly muscle, and that the prolonged lifespan caused by overexpression of Daw or Myo in adult muscle could be due to enhanced protein levels of the key subunits of 26S proteasome. Overall, our data suggest that Activin signaling initiated by Myo and Daw in adult Drosophila muscles influences lifespan, in part, by modulation of protein homeostasis through either direct or indirect regulation of the 26S proteasome levels. Since Myo is closely related to the vertebrate muscle mass regulator Myostatin (GDF8) and the Myostatin paralog GDF11, our observations may offer a new experimental model for probing the roles of GDF11/8 in ageing regulation in vertebrates. This article has an associated First Person interview with the first author of the paper. Summary: This article has, for the first time, demonstrated that fruit fly TGF-beta, or Activin-type ligand Daw, or Myo-initiated Activin signaling in adult fruit fly muscle tissues works as an anti-ageing factor by regulating 26S proteasome activities in those tissues.
Collapse
Affiliation(s)
- Shaughna Langerak
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Myung-Jun Kim
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Hannah Lamberg
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Michael Godinez
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Mackenzie Main
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Lindsey Winslow
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Michael B O'Connor
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Changqi C Zhu
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| |
Collapse
|
41
|
Augustin H, McGourty K, Allen MJ, Adcott J, Wong CT, Boucrot E, Partridge L. Impact of insulin signaling and proteasomal activity on physiological output of a neuronal circuit in aging Drosophila melanogaster. Neurobiol Aging 2018; 66:149-157. [PMID: 29579685 PMCID: PMC5933513 DOI: 10.1016/j.neurobiolaging.2018.02.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 01/26/2018] [Accepted: 02/27/2018] [Indexed: 12/11/2022]
Abstract
The insulin family of growth factors plays an important role in development and function of the nervous system. Reduced insulin and insulin-growth-factor signaling (IIS), however, can improve symptoms of neurodegenerative diseases in laboratory model organisms and protect against age-associated decline in neuronal function. Recently, we showed that chronic, moderately lowered IIS rescues age-related decline in neurotransmission through the Drosophila giant fiber escape response circuit. Here, we expand our initial findings by demonstrating that reduced functional output in the giant fiber system of aging flies can be prevented by increasing proteasomal activity within the circuit. Manipulations of IIS in neurons can also affect longevity, underscoring the relevance of the nervous system for aging.
Collapse
Affiliation(s)
- Hrvoje Augustin
- Max Planck Institute for Biology of Ageing, Köln, Germany; Institute of Healthy Ageing, and GEE, University College London, London, UK
| | - Kieran McGourty
- Department of Structural and Molecular Biology, London, UK; The Bernal Institute, University of Limerick, Limerick, Ireland
| | - Marcus J Allen
- School of Biosciences, University of Kent, Canterbury, Kent, UK
| | - Jennifer Adcott
- Institute of Healthy Ageing, and GEE, University College London, London, UK
| | - Chi Tung Wong
- Institute of Healthy Ageing, and GEE, University College London, London, UK
| | | | - Linda Partridge
- Max Planck Institute for Biology of Ageing, Köln, Germany; Institute of Healthy Ageing, and GEE, University College London, London, UK.
| |
Collapse
|
42
|
Barwell T, DeVeale B, Poirier L, Zheng J, Seroude F, Seroude L. Regulating the UAS/GAL4 system in adult Drosophila with Tet-off GAL80 transgenes. PeerJ 2017; 5:e4167. [PMID: 29259847 PMCID: PMC5733373 DOI: 10.7717/peerj.4167] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 11/24/2017] [Indexed: 01/16/2023] Open
Abstract
The UAS/GAL4 system is the most used method in Drosophila melanogaster for directing the expression of a gene of interest to a specific tissue. However, the ability to control the temporal activity of GAL4 with this system is very limited. This study constructed and characterized Tet-off GAL80 transgenes designed to allow temporal control of GAL4 activity in aging adult muscles. By placing GAL80 under the control of a Tet-off promoter, GAL4 activity is regulated by the presence or absence of tetracycline in the diet. Almost complete inhibition of the expression of UAS transgenes during the pre-adult stages of the life cycle is obtained by using four copies and two types of Tet-off GAL80 transgenes. Upon treatment of newly emerged adults with tetracycline, induction of GAL4 activity is observed but the level of induction is influenced by the concentration of the inducer, the age, the sex and the anatomical location of the expression. The inhibition of GAL4 activity and the maintenance of induced expression are altered in old animals. This study reveals that the repressive ability of GAL80 is affected by the age and sex of the animal which is a major limitation to regulate gene expression with GAL80 in aged Drosophila.
Collapse
Affiliation(s)
- Taylor Barwell
- Department of Biology, Queen's University, Kingston, ON, Canada
| | - Brian DeVeale
- Department of Biology, Queen's University, Kingston, ON, Canada.,Department of Biology, Queen's University, Kingston, ON, Canada
| | - Luc Poirier
- Department of Biology, Queen's University, Kingston, ON, Canada
| | - Jie Zheng
- Department of Biology, Queen's University, Kingston, ON, Canada.,Department of Biology, Queen's University, Kingston, ON, Canada
| | | | - Laurent Seroude
- Department of Biology, Queen's University, Kingston, ON, Canada
| |
Collapse
|
43
|
Filer D, Thompson MA, Takhaveev V, Dobson AJ, Kotronaki I, Green JWM, Heinemann M, Tullet JMA, Alic N. RNA polymerase III limits longevity downstream of TORC1. Nature 2017; 552:263-267. [PMID: 29186112 PMCID: PMC5732570 DOI: 10.1038/nature25007] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 11/07/2017] [Indexed: 11/09/2022]
Abstract
Three distinct RNA polymerases (Pols) transcribe different classes of
genes in the eukaryotic nucleus1. Pol III
is the essential, evolutionarily conserved enzyme that generates short,
non-coding RNAs, including transfer RNAs (tRNAs) and 5S
ribosomal RNA (rRNA)2. Historical focus on
transcription of protein-coding genes has left the roles of Pol III in
organismal physiology relatively unexplored. The prominent regulator of Pol III
activity, Target of Rapamycin kinase Complex 1 (TORC1), is an important
longevity determinant3, raising the
question of Pol III’s involvement in ageing. Here we show that Pol III
limits lifespan downstream of TORC1. We find that a reduction in Pol III extends
chronological lifespan in yeast and organismal lifespan in worms and flies.
Inhibiting Pol III activity in the adult worm or fly gut is sufficient to extend
lifespan, and in flies, longevity can be achieved by Pol III inhibition
specifically in the intestinal stem cells (ISCs). The longevity phenotype is
associated with amelioration of age-related gut pathology and functional
decline, dampened protein synthesis and increased tolerance of proteostatic
stress. Importantly, Pol III acts downstream of TORC1 for lifespan and limiting
Pol III activity in the adult gut achieves the full longevity benefit of
systemic TORC1 inhibition. Hence, Pol III is a pivotal output of this key
nutrient signalling network for longevity; Pol III’s growth-promoting,
anabolic activity mediates the acceleration of ageing by TORC1. The evolutionary
conservation of Pol III affirms its potential as a therapeutic target.
Collapse
Affiliation(s)
- Danny Filer
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, Gower Street, London WC1E 6BT, UK
| | | | - Vakil Takhaveev
- Molecular Systems Biology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG Groningen, Netherlands
| | - Adam J Dobson
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, Gower Street, London WC1E 6BT, UK
| | - Ilektra Kotronaki
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, Gower Street, London WC1E 6BT, UK
| | - James W M Green
- School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK
| | - Matthias Heinemann
- Molecular Systems Biology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG Groningen, Netherlands
| | | | - Nazif Alic
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
44
|
Tain LS, Sehlke R, Jain C, Chokkalingam M, Nagaraj N, Essers P, Rassner M, Grönke S, Froelich J, Dieterich C, Mann M, Alic N, Beyer A, Partridge L. A proteomic atlas of insulin signalling reveals tissue-specific mechanisms of longevity assurance. Mol Syst Biol 2017; 13:939. [PMID: 28916541 PMCID: PMC5615923 DOI: 10.15252/msb.20177663] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/15/2017] [Accepted: 08/16/2017] [Indexed: 12/20/2022] Open
Abstract
Lowered activity of the insulin/IGF signalling (IIS) network can ameliorate the effects of ageing in laboratory animals and, possibly, humans. Although transcriptome remodelling in long-lived IIS mutants has been extensively documented, the causal mechanisms contributing to extended lifespan, particularly in specific tissues, remain unclear. We have characterized the proteomes of four key insulin-sensitive tissues in a long-lived Drosophila IIS mutant and control, and detected 44% of the predicted proteome (6,085 proteins). Expression of ribosome-associated proteins in the fat body was reduced in the mutant, with a corresponding, tissue-specific reduction in translation. Expression of mitochondrial electron transport chain proteins in fat body was increased, leading to increased respiration, which was necessary for IIS-mediated lifespan extension, and alone sufficient to mediate it. Proteasomal subunits showed altered expression in IIS mutant gut, and gut-specific over-expression of the RPN6 proteasomal subunit, was sufficient to increase proteasomal activity and extend lifespan, whilst inhibition of proteasome activity abolished IIS-mediated longevity. Our study thus uncovered strikingly tissue-specific responses of cellular processes to lowered IIS acting in concert to ameliorate ageing.
Collapse
Affiliation(s)
- Luke S Tain
- Max-Planck Institute for Biology of Ageing, Cologne, Germany
| | - Robert Sehlke
- Max-Planck Institute for Biology of Ageing, Cologne, Germany
- CECAD Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, Cologne, Germany
| | - Chirag Jain
- Max-Planck Institute for Biology of Ageing, Cologne, Germany
| | - Manopriya Chokkalingam
- CECAD Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, Cologne, Germany
| | - Nagarjuna Nagaraj
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, Martinsried, Germany
| | - Paul Essers
- Max-Planck Institute for Biology of Ageing, Cologne, Germany
| | - Mark Rassner
- Max-Planck Institute for Biology of Ageing, Cologne, Germany
| | | | - Jenny Froelich
- Max-Planck Institute for Biology of Ageing, Cologne, Germany
| | - Christoph Dieterich
- Section of Bioinformatics and Systems Cardiology, Department of Internal Medicine III and Klaus Tschira Institute for Integrative Computational Cardiology, University of Heidelberg, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, Martinsried, Germany
| | - Nazif Alic
- Institute of Healthy Ageing, and GEE, UCL, London, UK
| | - Andreas Beyer
- CECAD Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Linda Partridge
- Max-Planck Institute for Biology of Ageing, Cologne, Germany
- Institute of Healthy Ageing, and GEE, UCL, London, UK
| |
Collapse
|
45
|
Augustin H, McGourty K, Allen MJ, Madem SK, Adcott J, Kerr F, Wong CT, Vincent A, Godenschwege T, Boucrot E, Partridge L. Reduced insulin signaling maintains electrical transmission in a neural circuit in aging flies. PLoS Biol 2017; 15:e2001655. [PMID: 28902870 PMCID: PMC5597081 DOI: 10.1371/journal.pbio.2001655] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 08/04/2017] [Indexed: 12/11/2022] Open
Abstract
Lowered insulin/insulin-like growth factor (IGF) signaling (IIS) can extend healthy lifespan in worms, flies, and mice, but it can also have adverse effects (the “insulin paradox”). Chronic, moderately lowered IIS rescues age-related decline in neurotransmission through the Drosophila giant fiber system (GFS), a simple escape response neuronal circuit, by increasing targeting of the gap junctional protein innexin shaking-B to gap junctions (GJs). Endosomal recycling of GJs was also stimulated in cultured human cells when IIS was reduced. Furthermore, increasing the activity of the recycling small guanosine triphosphatases (GTPases) Rab4 or Rab11 was sufficient to maintain GJs upon elevated IIS in cultured human cells and in flies, and to rescue age-related loss of GJs and of GFS function. Lowered IIS thus elevates endosomal recycling of GJs in neurons and other cell types, pointing to a cellular mechanism for therapeutic intervention into aging-related neuronal disorders. Insulin and insulin-like growth factors play an important role in the nervous system development and function. Reduced insulin signaling, however, can improve symptoms of neurodegenerative diseases in different model organisms and protect against age-associated decline in neuronal function extending lifespan. Here, we analyze the effects of genetically attenuated insulin signaling on the escape response pathway in the fruit fly Drosophila melanogaster. This simple neuronal circuit is dominated by electrical synapses composed of the gap junctional shaking-B protein, which allows for the transfer of electrical impulses between cells. Transmission through the circuit is known to slow down with age. We show that this functional decline is prevented by systemic or circuit-specific suppression of insulin signaling due to the preservation of the number of gap junctional proteins in aging animals. Our experiments in a human cell culture system reveal increased membrane targeting of gap junctional proteins via small proteins Rab4 and Rab11 under reduced insulin conditions. We also find that increasing the level of these recycling-mediating proteins in flies preserves the escape response circuit output in old flies and suggests ways of improving the function of neuronal circuits dominated by electrical synapses during aging.
Collapse
Affiliation(s)
- Hrvoje Augustin
- Max Planck Institute for Biology of Aging, Köln, Germany
- Institute of Healthy Aging, and Genetics, Evolution, and Environment, University College London, London, United Kingdom
| | - Kieran McGourty
- Department of Structural and Molecular Biology, London, United Kingdom
| | - Marcus J. Allen
- School of Biosciences, University of Kent, Canterbury, Kent, United Kingdom
| | - Sirisha Kudumala Madem
- Department of Biological Sciences, Florida Atlantic University, Jupiter, Florida, United States of America
| | - Jennifer Adcott
- Max Planck Institute for Biology of Aging, Köln, Germany
- Institute of Healthy Aging, and Genetics, Evolution, and Environment, University College London, London, United Kingdom
| | - Fiona Kerr
- Max Planck Institute for Biology of Aging, Köln, Germany
- Institute of Healthy Aging, and Genetics, Evolution, and Environment, University College London, London, United Kingdom
| | - Chi Tung Wong
- Max Planck Institute for Biology of Aging, Köln, Germany
| | - Alec Vincent
- Max Planck Institute for Biology of Aging, Köln, Germany
| | - Tanja Godenschwege
- Department of Biological Sciences, Florida Atlantic University, Jupiter, Florida, United States of America
| | - Emmanuel Boucrot
- Department of Structural and Molecular Biology, London, United Kingdom
| | - Linda Partridge
- Max Planck Institute for Biology of Aging, Köln, Germany
- Institute of Healthy Aging, and Genetics, Evolution, and Environment, University College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
46
|
Rana A, Oliveira MP, Khamoui AV, Aparicio R, Rera M, Rossiter HB, Walker DW. Promoting Drp1-mediated mitochondrial fission in midlife prolongs healthy lifespan of Drosophila melanogaster. Nat Commun 2017; 8:448. [PMID: 28878259 PMCID: PMC5587646 DOI: 10.1038/s41467-017-00525-4] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 07/04/2017] [Indexed: 12/03/2022] Open
Abstract
The accumulation of dysfunctional mitochondria has been implicated in aging, but a deeper understanding of mitochondrial dynamics and mitophagy during aging is missing. Here, we show that upregulating Drp1—a Dynamin-related protein that promotes mitochondrial fission—in midlife, prolongs Drosophila lifespan and healthspan. We find that short-term induction of Drp1, in midlife, is sufficient to improve organismal health and prolong lifespan, and observe a midlife shift toward a more elongated mitochondrial morphology, which is linked to the accumulation of dysfunctional mitochondria in aged flight muscle. Promoting Drp1-mediated mitochondrial fission, in midlife, facilitates mitophagy and improves both mitochondrial respiratory function and proteostasis in aged flies. Finally, we show that autophagy is required for the anti-aging effects of midlife Drp1-mediated mitochondrial fission. Our findings indicate that interventions that promote mitochondrial fission could delay the onset of pathology and mortality in mammals when applied in midlife. Mitochondrial fission and fusion are important mechanisms to maintain mitochondrial function. Here, the authors report that middle-aged flies have more elongated, or ‘hyper-fused’ mitochondria, and show that induction of mitochondrial fission in midlife, but not in early life, extends the health and life of flies.
Collapse
Affiliation(s)
- Anil Rana
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA
| | - Matheus P Oliveira
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA.,Instituto de Bioquimica Medica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Cidade Universitaria, Rio de Janeiro, 21941-590, Brazil
| | - Andy V Khamoui
- Division of Respiratory & Critical Care Physiology & Medicine, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA.,Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Ricardo Aparicio
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA
| | - Michael Rera
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA.,Laboratory of Degenerative Processes, Stress and Aging, Université Paris Diderot, Paris, 75013, France
| | - Harry B Rossiter
- Division of Respiratory & Critical Care Physiology & Medicine, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA.,Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - David W Walker
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA. .,Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
47
|
Single vector non-leaky gene expression system for Drosophila melanogaster. Sci Rep 2017; 7:6899. [PMID: 28761084 PMCID: PMC5537222 DOI: 10.1038/s41598-017-07282-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 06/23/2017] [Indexed: 12/28/2022] Open
Abstract
An ideal transgenic gene expression system is inducible, non-leaky, and well tolerated by the target organism. While the former has been satisfactorily realized, leakiness and heavy physiological burden imposed by the existing systems are still prominent hurdles in their successful implementation. Here we describe a new system for non-leaky expression of transgenes in Drosophila. PRExpress is based on a single transgenic construct built from endogenous components, the inducible hsp70 promoter and a multimerized copy of a Polycomb response element (PRE) controlled by epigenetic chromatin regulators of the Polycomb group. We show that this system is non-leaky, rapidly and strongly inducible, and reversible. To make the application of PRExpress user-friendly, we deliver the construct via site-specific integration.
Collapse
|
48
|
Antimicrobial peptides extend lifespan in Drosophila. PLoS One 2017; 12:e0176689. [PMID: 28520752 PMCID: PMC5435158 DOI: 10.1371/journal.pone.0176689] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 04/16/2017] [Indexed: 02/07/2023] Open
Abstract
Antimicrobial peptides (AMPs) are important defense molecules of the innate immune system. High levels of AMPs are induced in response to infections to fight pathogens, whereas moderate levels induced by metabolic stress are thought to shape commensal microbial communities at barrier tissues. We expressed single AMPs in adult flies either ubiquitously or in the gut by using the inducible GeneSwitch system to tightly regulate AMP expression. We found that activation of single AMPs, including Drosocin, resulted in a significant extension of Drosophila lifespan. These animals showed reduced activity of immune pathways over lifetime, less intestinal regenerative processes, reduced stress response and a delayed loss of gut barrier integrity. Furthermore, intestinal Drosocin induction protected the animals against infections with the natural Drosophila pathogen Pseudomonas entomophila, whereas a germ-reduced environment prevented the lifespan extending effect of Drosocin. Our study provides new insights into the crosstalk of innate immunity, intestinal homeostasis and ageing.
Collapse
|
49
|
Mizielinska S, Ridler CE, Balendra R, Thoeng A, Woodling NS, Grässer FA, Plagnol V, Lashley T, Partridge L, Isaacs AM. Bidirectional nucleolar dysfunction in C9orf72 frontotemporal lobar degeneration. Acta Neuropathol Commun 2017; 5:29. [PMID: 28420437 PMCID: PMC5395972 DOI: 10.1186/s40478-017-0432-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 04/06/2017] [Indexed: 01/08/2023] Open
Abstract
An intronic GGGGCC expansion in C9orf72 is the most common known cause of both frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS). The repeat expansion leads to the generation of sense and antisense repeat RNA aggregates and dipeptide repeat (DPR) proteins, generated by repeat-associated non-ATG translation. The arginine-rich DPR proteins poly(glycine-arginine or GR) and poly(proline-arginine or PR) are potently neurotoxic and can localise to the nucleolus when expressed in cells, resulting in enlarged nucleoli with disrupted functionality. Furthermore, GGGGCC repeat RNA can bind nucleolar proteins in vitro. However, the relevance of nucleolar stress is unclear, as the arginine-rich DPR proteins do not localise to the nucleolus in C9orf72-associated FTLD/ALS (C9FTLD/ALS) patient brain. We measured nucleolar size in C9FTLD frontal cortex neurons using a three-dimensional, volumetric approach. Intriguingly, we found that C9FTLD brain exhibited bidirectional nucleolar stress. C9FTLD neuronal nucleoli were significantly smaller than control neuronal nucleoli. However, within C9FTLD brains, neurons containing poly(GR) inclusions had significantly larger nucleolar volumes than neurons without poly(GR) inclusions. In addition, expression of poly(GR) in adult Drosophila neurons led to significantly enlarged nucleoli. A small but significant increase in nucleolar volume was also observed in C9FTLD frontal cortex neurons containing GGGGCC repeat-containing RNA foci. These data show that nucleolar abnormalities are a consistent feature of C9FTLD brain, but that diverse pathomechanisms are at play, involving both DPR protein and repeat RNA toxicity.
Collapse
|
50
|
Zhang X, Schnorrer F. AIDing-targeted protein degradation in Drosophila. FEBS J 2017; 284:1178-1181. [PMID: 28382696 DOI: 10.1111/febs.14054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Conditional protein depletion is highly desirable for investigating protein functions in complex organisms. In this issue, Bence and colleagues combined auxin-inducible degradation with CRISPR, establishing an elegant tool to control protein levels. They achieve precise spatio-temporal control of protein degradation during Drosophila oogenesis and early embryogenesis by combining suitable GAL4 drivers (spatial control) with auxin feeding protocols (temporal control).
Collapse
Affiliation(s)
- Xu Zhang
- Muscle Dynamics Group, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Frank Schnorrer
- Muscle Dynamics Group, Max Planck Institute of Biochemistry, Martinsried, Germany.,Developmental Biology Institute of Marseille (IBDM), CNRS, UMR 7288, Aix-Marseille Université, France
| |
Collapse
|