1
|
Hanson KM, Macdonald SJ. Dynamic changes in gene expression through aging in Drosophila melanogaster heads. G3 (BETHESDA, MD.) 2025; 15:jkaf039. [PMID: 39992875 PMCID: PMC12005168 DOI: 10.1093/g3journal/jkaf039] [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: 12/10/2024] [Revised: 02/07/2025] [Accepted: 02/14/2025] [Indexed: 02/26/2025]
Abstract
Work in many systems has shown large-scale changes in gene expression during aging. However, many studies employ just 2 arbitrarily chosen timepoints to measure expression and can only observe an increase or a decrease in expression between "young" and "old" animals, failing to capture any dynamic, nonlinear changes that occur throughout the aging process. We used RNA sequencing to measure expression in male head tissue at 15 timepoints through the lifespan of an inbred Drosophila melanogaster strain. We detected >6,000 significant, age-related genes, nearly all of which have been seen in previous Drosophila aging expression studies and that include several known to harbor lifespan-altering mutations. We grouped our gene set into 28 clusters via their temporal expression change, observing a diversity of trajectories; some clusters show a linear change over time, while others show more complex, nonlinear patterns. Notably, reanalysis of our dataset comparing the earliest and latest timepoints-mimicking a 2-timepoint design-revealed fewer differentially expressed genes (around 4,500). Additionally, those genes exhibiting complex expression trajectories in our multitimepoint analysis were most impacted in this reanalysis; their identification, and the inferred change in gene expression with age, was often dependent on the timepoints chosen. Informed by our trajectory-based clusters, we executed a series of gene enrichment analyses, identifying enriched functions/pathways in all clusters, including the commonly seen increase in stress- and immune-related gene expression with age. Finally, we developed a pair of accessible Shiny apps to enable exploration of our differential expression and gene enrichment results.
Collapse
Affiliation(s)
- Katherine M Hanson
- Department of Molecular Biosciences and Center for Genomics, University of Kansas, 1200 Sunnyside Avenue, Lawrence, KS 66045, USA
| | - Stuart J Macdonald
- Department of Molecular Biosciences and Center for Genomics, University of Kansas, 1200 Sunnyside Avenue, Lawrence, KS 66045, USA
| |
Collapse
|
2
|
Tatar M, Zheng W, Yadav S, Yamamoto R, Curtis-Joseph N, Li S, Wang L, Parkhitko AA. Mutation of an insulin-sensitive Drosophila insulin-like receptor mutant requires methionine metabolism reprogramming to extend lifespan. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640731. [PMID: 40093182 PMCID: PMC11908128 DOI: 10.1101/2025.02.28.640731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Insulin/insulin growth factor signaling is a conserved pathway that regulates lifespan across many species. Multiple mechanisms are proposed for how this altered signaling slows aging. To elaborate these causes, we recently developed a series of Drosophila insulin-like receptor (dInr) mutants with single amino acid substitutions that extend lifespan but differentially affect insulin sensitivity, growth and reproduction. Transheterozygotes of canonical dInr mutants (Type I) extend longevity and are insulin-resistant, small and weakly fecund. In contrast, a dominant mutation (dInr 353, Type II) within the Kinase Insert Domain (KID) robustly extends longevity but is insulin-sensitive, full-sized, and highly fecund. We applied transcriptome and metabolome analyses to explore how dInr 353 slows aging without insulin resistance. Type I and II mutants overlap in many pathways but also produce distinct transcriptomic profiles that include differences in innate immune and reproductive functions. In metabolomic analyses, the KID mutant dInr 353 reprograms methionine metabolism in a way that phenocopies dietary methionine restriction, in contrast to canonical mutants which are characterized by upregulation of the transsulfuration pathway. Because abrogation of S-adenosylhomocysteine hydrolase blocks the longevity benefit conferred by dInr 353, we conclude the methionine cycle reprogramming of Type II is sufficient to slow aging. Metabolomic analysis further revealed the Type II mutant is metabolically flexible: unlike aged wildtype, aged dInr 353 adults can reroute methionine toward the transsulfuration pathway, while Type I mutant flies upregulate the trassulfuration pathway continuously from young age. Altered insulin/insulin growth factor signaling has the potential to slow aging without the complications of insulin resistance by modulating methionine cycle dynamics.
Collapse
Affiliation(s)
- Marc Tatar
- Department of Ecology, Evolution and Organismal Biology, and The Center for the Biology of Aging, Brown University, Providence, RI, USA
| | - Wenjing Zheng
- Department of Ecology, Evolution and Organismal Biology, and The Center for the Biology of Aging, Brown University, Providence, RI, USA
| | - Shweta Yadav
- Aging Institute of UPMC and the University of Pittsburgh, Pittsburgh, PA, USA
| | - Rochele Yamamoto
- Department of Ecology, Evolution and Organismal Biology, and The Center for the Biology of Aging, Brown University, Providence, RI, USA
| | - Noelle Curtis-Joseph
- Department of Ecology, Evolution and Organismal Biology, and The Center for the Biology of Aging, Brown University, Providence, RI, USA
| | - Shengxi Li
- State Key Laboratory of Common Mechanism Research for Major Disease, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Wang
- State Key Laboratory of Common Mechanism Research for Major Disease, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Andrey A Parkhitko
- Aging Institute of UPMC and the University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
3
|
Hanson KM, Macdonald SJ. Dynamic Changes in Gene Expression Through Aging in Drosophila melanogaster Heads. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.11.627977. [PMID: 39764034 PMCID: PMC11702523 DOI: 10.1101/2024.12.11.627977] [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: 01/14/2025]
Abstract
Work in many systems has shown large-scale changes in gene expression during aging. However, many studies employ just two, arbitrarily-chosen timepoints at which to measure expression, and can only observe an increase or a decrease in expression between "young" and "old" animals, failing to capture any dynamic, non-linear changes that occur throughout the aging process. We used RNA sequencing to measure expression in male head tissue at 15 timepoints through the lifespan of an inbred Drosophila melanogaster strain. We detected >6,000 significant, age-related genes, nearly all of which have been seen in previous fly aging expression studies, and which include several known to harbor lifespan-altering mutations. We grouped our gene set into 28 clusters via their temporal expression change, observing a diversity of trajectories; some clusters show a linear change over time, while others show more complex, non-linear patterns. Notably, re-analysis of our dataset comparing the earliest and latest timepoints - mimicking a two-timepoint design - revealed fewer differentially-expressed genes (around 4,500). Additionally, those genes exhibiting complex expression trajectories in our multi-timepoint analysis were most impacted in this re-analysis; Their identification, and the inferred change in gene expression with age, was often dependent on the timepoints chosen. Informed by our trajectory-based clusters, we executed a series of gene enrichment analyses, identifying enriched functions/pathways in all clusters, including the commonly seen increase in stress- and immune-related gene expression with age. Finally, we developed a pair of accessible shiny apps to enable exploration of our differential expression and gene enrichment results.
Collapse
Affiliation(s)
- Katherine M Hanson
- Department of Molecular Biosciences and Center for Genomics, University of Kansas, 1200 Sunnyside Avenue, Lawrence, KS 66045, USA
| | - Stuart J Macdonald
- Department of Molecular Biosciences and Center for Genomics, University of Kansas, 1200 Sunnyside Avenue, Lawrence, KS 66045, USA
| |
Collapse
|
4
|
Jang S, Choi B, Lim C, Kim M, Lee JE, Lee H, Baek E, Cho KS. Neuronal fatty acid-binding protein enhances autophagy and suppresses amyloid-β pathology in a Drosophila model of Alzheimer's disease. PLoS Genet 2024; 20:e1011475. [PMID: 39561115 PMCID: PMC11575808 DOI: 10.1371/journal.pgen.1011475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 10/28/2024] [Indexed: 11/21/2024] Open
Abstract
Fatty acid-binding proteins (FABPs) are small cytoplasmic proteins involved in intracellular lipid transport and bind free fatty acids, cholesterol, and retinoids. FABP3, the major neuronal FABP in the adult brain, is upregulated in the CSF of patients with Alzheimer's disease (AD). However, the precise role of neuronal FABPs in AD pathogenesis remains unclear. This study investigates the contribution of fabp, the Drosophila homolog of FABP3 and FABP7, to amyloid β (Aβ) pathology using a Drosophila model. Neuronal knockdown of fabp shortened the lifespan of flies and increased age-related protein aggregates in the brain. In an AD model, fabp knockdown in neurons increased Aβ accumulation and Aβ-induced neurodegeneration, whereas fabp overexpression ameliorated Aβ pathology. Notably, fabp overexpression stimulated autophagy, which was inhibited by the knockdown of Eip75B, the Drosophila homolog of the peroxisome proliferator-activated receptor (PPAR). The PPAR activator rosiglitazone restored autophagy impaired by fabp knockdown and reduced fabp knockdown-induced increased Aβ aggregation and cell death. Furthermore, knockdown of either fabp or Eip75B in the wing imaginal disc or adult fly brain reduced the expression of Atg6 and Atg8a. Additionally, treatment of the fabp knockdown AD model flies with polyunsaturated fatty acids, such as docosahexaenoic acid or linoleic acid, partially alleviated cell death in the developing eye, restored impaired autophagy flux, reduced Aβ aggregation, and attenuated Aβ-induced cell death. Our results suggest that Drosophila fabp plays an important role in maintaining protein homeostasis during aging and protects neurons from Aβ-induced cell death by enhancing autophagy through the PPAR pathway. These findings highlight the potential importance of neuronal FABP function in AD pathogenesis.
Collapse
Affiliation(s)
- Seokhui Jang
- Department of Biological Sciences, Konkuk University, Seoul, Republic of Korea
| | - Byoungyun Choi
- Department of Biological Sciences, Konkuk University, Seoul, Republic of Korea
| | - Chaejin Lim
- Department of Biological Sciences, Konkuk University, Seoul, Republic of Korea
| | - Minkyoung Kim
- Department of Biological Sciences, Konkuk University, Seoul, Republic of Korea
| | - Ji-Eun Lee
- Department of Biological Sciences, Konkuk University, Seoul, Republic of Korea
| | - Hyungi Lee
- Department of Biological Sciences, Konkuk University, Seoul, Republic of Korea
| | - Eunji Baek
- Department of Biological Sciences, Konkuk University, Seoul, Republic of Korea
| | - Kyoung Sang Cho
- Department of Biological Sciences, Konkuk University, Seoul, Republic of Korea
- Korea Hemp Institute, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
5
|
Jamerson LE, Bradshaw PC. The Roles of White Adipose Tissue and Liver NADPH in Dietary Restriction-Induced Longevity. Antioxidants (Basel) 2024; 13:820. [PMID: 39061889 PMCID: PMC11273496 DOI: 10.3390/antiox13070820] [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: 05/29/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Dietary restriction (DR) protocols frequently employ intermittent fasting. Following a period of fasting, meal consumption increases lipogenic gene expression, including that of NADPH-generating enzymes that fuel lipogenesis in white adipose tissue (WAT) through the induction of transcriptional regulators SREBP-1c and CHREBP. SREBP-1c knockout mice, unlike controls, did not show an extended lifespan on the DR diet. WAT cytoplasmic NADPH is generated by both malic enzyme 1 (ME1) and the pentose phosphate pathway (PPP), while liver cytoplasmic NADPH is primarily synthesized by folate cycle enzymes provided one-carbon units through serine catabolism. During the daily fasting period of the DR diet, fatty acids are released from WAT and are transported to peripheral tissues, where they are used for beta-oxidation and for phospholipid and lipid droplet synthesis, where monounsaturated fatty acids (MUFAs) may activate Nrf1 and inhibit ferroptosis to promote longevity. Decreased WAT NADPH from PPP gene knockout stimulated the browning of WAT and protected from a high-fat diet, while high levels of NADPH-generating enzymes in WAT and macrophages are linked to obesity. But oscillations in WAT [NADPH]/[NADP+] from feeding and fasting cycles may play an important role in maintaining metabolic plasticity to drive longevity. Studies measuring the WAT malate/pyruvate as a proxy for the cytoplasmic [NADPH]/[NADP+], as well as studies using fluorescent biosensors expressed in the WAT of animal models to monitor the changes in cytoplasmic [NADPH]/[NADP+], are needed during ad libitum and DR diets to determine the changes that are associated with longevity.
Collapse
Affiliation(s)
| | - Patrick C. Bradshaw
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| |
Collapse
|
6
|
Yang M, Zinkgraf M, Fitzgerald-Cook C, Harrison BR, Putzier A, Promislow DEL, Wang AM. Using Drosophila to identify naturally occurring genetic modifiers of amyloid beta 42- and tau-induced toxicity. G3 (BETHESDA, MD.) 2023; 13:jkad132. [PMID: 37311212 PMCID: PMC10468303 DOI: 10.1093/g3journal/jkad132] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/15/2023] [Accepted: 05/15/2023] [Indexed: 06/15/2023]
Abstract
Alzheimer's disease is characterized by 2 pathological proteins, amyloid beta 42 and tau. The majority of Alzheimer's disease cases in the population are sporadic and late-onset Alzheimer's disease, which exhibits high levels of heritability. While several genetic risk factors for late-onset Alzheimer's disease have been identified and replicated in independent studies, including the ApoE ε4 allele, the great majority of the heritability of late-onset Alzheimer's disease remains unexplained, likely due to the aggregate effects of a very large number of genes with small effect size, as well as to biases in sample collection and statistical approaches. Here, we present an unbiased forward genetic screen in Drosophila looking for naturally occurring modifiers of amyloid beta 42- and tau-induced ommatidial degeneration. Our results identify 14 significant SNPs, which map to 12 potential genes in 8 unique genomic regions. Our hits that are significant after genome-wide correction identify genes involved in neuronal development, signal transduction, and organismal development. Looking more broadly at suggestive hits (P < 10-5), we see significant enrichment in genes associated with neurogenesis, development, and growth as well as significant enrichment in genes whose orthologs have been identified as significantly or suggestively associated with Alzheimer's disease in human GWAS studies. These latter genes include ones whose orthologs are in close proximity to regions in the human genome that are associated with Alzheimer's disease, but where a causal gene has not been identified. Together, our results illustrate the potential for complementary and convergent evidence provided through multitrait GWAS in Drosophila to supplement and inform human studies, helping to identify the remaining heritability and novel modifiers of complex diseases.
Collapse
Affiliation(s)
- Ming Yang
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Matthew Zinkgraf
- Department of Biology, Western Washington University, Bellingham, WA 98225, USA
| | - Cecilia Fitzgerald-Cook
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Benjamin R Harrison
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Alexandra Putzier
- Department of Biology, Western Washington University, Bellingham, WA 98225, USA
| | - Daniel E L Promislow
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA 98195, USA
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Adrienne M Wang
- Department of Biology, Western Washington University, Bellingham, WA 98225, USA
| |
Collapse
|
7
|
Iino S, Oya S, Kakutani T, Kohno H, Kubo T. Identification of ecdysone receptor target genes in the worker honey bee brains during foraging behavior. Sci Rep 2023; 13:10491. [PMID: 37380789 DOI: 10.1038/s41598-023-37001-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 06/14/2023] [Indexed: 06/30/2023] Open
Abstract
Ecdysone signaling plays central roles in morphogenesis and female ovarian development in holometabolous insects. In the European honey bee (Apis mellifera L.), however, ecdysone receptor (EcR) is expressed in the brains of adult workers, which have already undergone metamorphosis and are sterile with shrunken ovaries, during foraging behavior. Aiming at unveiling the significance of EcR signaling in the worker brain, we performed chromatin-immunoprecipitation sequencing of EcR to search for its target genes using the brains of nurse bees and foragers. The majority of the EcR targets were common between the nurse bee and forager brains and some of them were known ecdysone signaling-related genes. RNA-sequencing analysis revealed that some EcR target genes were upregulated in forager brains during foraging behavior and some were implicated in the repression of metabolic processes. Single-cell RNA-sequencing analysis revealed that EcR and its target genes were expressed mostly in neurons and partly in glial cells in the optic lobes of the forager brain. These findings suggest that in addition to its role during development, EcR transcriptionally represses metabolic processes during foraging behavior in the adult worker honey bee brain.
Collapse
Affiliation(s)
- Shiori Iino
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Satoyo Oya
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Tetsuji Kakutani
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Hiroki Kohno
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Takeo Kubo
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
8
|
Suh GSB, Yu K, Kim YJ, Oh Y, Park JJ. History of Drosophila neurogenetic research in South Korea. J Neurogenet 2022:1-7. [PMID: 36165786 DOI: 10.1080/01677063.2022.2115040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Neurogenetic research using the Drosophila model has immensely expanded around the world. Likewise, scientists in South Korea have leveraged the advantages of Drosophila genetic tools to understand various neurobiological processes. In this special issue, we will overview the history of Drosophila neurogenetic research in South Korea that led to significant discoveries and notably implications. We will describe how Drosophila system was first introduced to elevate neural developmental studies in 1990s. Establishing Drosophila-related resources has been a key venture, which led to the generation of over 100,000 mutant lines and the launch of the K-Gut initiative with Korea Drosophila Research Center (KDRC). These resources have supported the pioneer studies in modeling human disease and understanding genes and neural circuits that regulate animal behavior and physiology.
Collapse
Affiliation(s)
- Greg S B Suh
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Kweon Yu
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Metabolism and Neurophysiology Research Group, Daejeon, Republic of Korea
| | - Young-Joon Kim
- Department of Biological Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Yangkyun Oh
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Joong-Jean Park
- Department of Physiology, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
9
|
Wang H, Lei L, Chen W, Chi X, Han K, Wang Y, Ma L, Liu Z, Xu B. The Comparison of Antioxidant Performance, Immune Performance, IIS Activity and Gut Microbiota Composition between Queen and Worker Bees Revealed the Mechanism of Different Lifespan of Female Casts in the Honeybee. INSECTS 2022; 13:772. [PMID: 36135473 PMCID: PMC9506344 DOI: 10.3390/insects13090772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/20/2022] [Accepted: 08/20/2022] [Indexed: 06/16/2023]
Abstract
Queen bees and worker bees both develop from fertilized eggs, whereas queens live longer than workers. The mechanism of this phenomenon is worth exploring. Antioxidant capacity, immune and IIS are the conserved mechanisms of aging. The importance of gut bacteria for health prompted us to connect with bee aging. Therefore, the differences of antioxidant, immune, IIS and gut microflora between queen and worker bees were compared to find potential mechanisms of queens' longevity. The results showed queens had stronger antioxidant capacity and lower immune pathway and IIS activity than workers. The higher expression level of catalase and SOD1/2 in queens resulted in the stronger ROS scavenging ability, which leads to the lower ROS level and the reduced accumulation of oxidative damage products in queens. The lower IMD expression and higher antimicrobial peptides (AMPs) expressions in queens suggested that queens maintain lower immune pathway activity and stronger immune capacity than workers. Gut bacteria composition analysis indicated that queens had supernal Acetobacteraceae (notably Commensalibacter and Bombella), Lactobacillus and Bifidobacterium over workers. In conclusion, antioxidant, immune, IIS, and gut symbiotic bacteria all contribute to the longevity of queens. This study provides more insights into revealing the mechanisms of queens' longevity.
Collapse
|
10
|
Doyle T, Jimenez‐Guri E, Hawkes WLS, Massy R, Mantica F, Permanyer J, Cozzuto L, Hermoso Pulido T, Baril T, Hayward A, Irimia M, Chapman JW, Bass C, Wotton KR. Genome-wide transcriptomic changes reveal the genetic pathways involved in insect migration. Mol Ecol 2022; 31:4332-4350. [PMID: 35801824 PMCID: PMC9546057 DOI: 10.1111/mec.16588] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/21/2022] [Accepted: 06/27/2022] [Indexed: 11/29/2022]
Abstract
Insects are capable of extraordinary feats of long-distance movement that have profound impacts on the function of terrestrial ecosystems. The ability to undertake these movements arose multiple times through the evolution of a suite of traits that make up the migratory syndrome, however the underlying genetic pathways involved remain poorly understood. Migratory hoverflies (Diptera: Syrphidae) are an emerging model group for studies of migration. They undertake seasonal movements in huge numbers across large parts of the globe and are important pollinators, biological control agents and decomposers. Here, we assembled a high-quality draft genome of the marmalade hoverfly (Episyrphus balteatus). We leveraged this genomic resource to undertake a genome-wide transcriptomic comparison of actively migrating Episyrphus, captured from a high mountain pass as they flew south to overwinter, with the transcriptomes of summer forms which were non-migratory. We identified 1543 genes with very strong evidence for differential expression. Interrogation of this gene set reveals a remarkable range of roles in metabolism, muscle structure and function, hormonal regulation, immunity, stress resistance, flight and feeding behaviour, longevity, reproductive diapause and sensory perception. These features of the migrant phenotype have arisen by the integration and modification of pathways such as insulin signalling for diapause and longevity, JAK/SAT for immunity, and those leading to octopamine production and fuelling to boost flight capabilities. Our results provide a powerful genomic resource for future research, and paint a comprehensive picture of global expression changes in an actively migrating insect, identifying key genomic components involved in this important life-history strategy.
Collapse
Affiliation(s)
- Toby Doyle
- Centre for Ecology and ConservationUniversity of Exeter, Cornwall CampusPenrynUK
| | - Eva Jimenez‐Guri
- Centre for Ecology and ConservationUniversity of Exeter, Cornwall CampusPenrynUK
| | - Will L. S. Hawkes
- Centre for Ecology and ConservationUniversity of Exeter, Cornwall CampusPenrynUK
| | - Richard Massy
- Centre for Ecology and ConservationUniversity of Exeter, Cornwall CampusPenrynUK
| | - Federica Mantica
- Centre for Genomic RegulationBarcelona Institute of Science and TechnologyBarcelonaSpain
| | - Jon Permanyer
- Centre for Genomic RegulationBarcelona Institute of Science and TechnologyBarcelonaSpain
| | - Luca Cozzuto
- Centre for Genomic RegulationBarcelona Institute of Science and TechnologyBarcelonaSpain
| | - Toni Hermoso Pulido
- Centre for Genomic RegulationBarcelona Institute of Science and TechnologyBarcelonaSpain
| | - Tobias Baril
- Centre for Ecology and ConservationUniversity of Exeter, Cornwall CampusPenrynUK
| | - Alex Hayward
- Centre for Ecology and ConservationUniversity of Exeter, Cornwall CampusPenrynUK
| | - Manuel Irimia
- Centre for Genomic RegulationBarcelona Institute of Science and TechnologyBarcelonaSpain
- Universitat Pompeu FabraBarcelonaSpain
- ICREABarcelonaSpain
| | - Jason W. Chapman
- Centre for Ecology and ConservationUniversity of Exeter, Cornwall CampusPenrynUK
- Environment and Sustainability InstituteUniversity of Exeter, Cornwall CampusPenrynUK
- Department of Entomology, College of Plant ProtectionNanjing Agricultural UniversityNanjingPeople's Republic of China
| | - Chris Bass
- Centre for Ecology and ConservationUniversity of Exeter, Cornwall CampusPenrynUK
| | - Karl R. Wotton
- Centre for Ecology and ConservationUniversity of Exeter, Cornwall CampusPenrynUK
| |
Collapse
|
11
|
Cho Y, Kim G, Park J. Mitochondrial aconitase 1 regulates age-related memory impairment via autophagy/mitophagy-mediated neural plasticity in middle-aged flies. Aging Cell 2021; 20:e13520. [PMID: 34799973 PMCID: PMC8672789 DOI: 10.1111/acel.13520] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/06/2021] [Accepted: 11/03/2021] [Indexed: 01/24/2023] Open
Abstract
Age‐related memory impairment (AMI) occurs in many species, including humans. The underlying mechanisms are not fully understood. In wild‐type Drosophila (w1118), AMI appears in the form of a decrease in learning (3‐min memory) from middle age (30 days after eclosion [DAE]). We performed in vivo, DNA microarray, and behavioral screen studies to identify genes controlling both lifespan and AMI and selected mitochondrial Acon1 (mAcon1). mAcon1 expression in the head of w1118 decreased with age. Neuronal overexpression of mAcon1 extended its lifespan and improved AMI. Neuronal or mushroom body expression of mAcon1 regulated the learning of young (10 DAE) and middle‐aged flies. Interestingly, acetyl‐CoA and citrate levels increased in the heads of middle‐aged and neuronal mAcon1 knockdown flies. Acetyl‐CoA, as a cellular energy sensor, is related to autophagy. Autophagy activity and efficacy determined by the positive and negative changes in the expression levels of Atg8a‐II and p62 were proportional to the expression level of mAcon1. Levels of the presynaptic active zone scaffold protein Bruchpilot were inversely proportional to neuronal mAcon1 levels in the whole brain. Furthermore, mAcon1 overexpression in Kenyon cells induced mitophagy labeled with mt‐Keima and improved learning ability. Both processes were blocked by pink1 knockdown. Taken together, our results imply that the regulation of learning and AMI by mAcon1 occurs via autophagy/mitophagy‐mediated neural plasticity.
Collapse
Affiliation(s)
- Yun‐Ho Cho
- Department of Physiology Korea University College of Medicine Seoul Republic of Korea
| | - Gye‐Hyeong Kim
- Department of Physiology Korea University College of Medicine Seoul Republic of Korea
| | - Joong‐Jean Park
- Department of Physiology Korea University College of Medicine Seoul Republic of Korea
| |
Collapse
|
12
|
Fabian DK, Fuentealba M, Dönertaş HM, Partridge L, Thornton JM. Functional conservation in genes and pathways linking ageing and immunity. IMMUNITY & AGEING 2021; 18:23. [PMID: 33990202 PMCID: PMC8120713 DOI: 10.1186/s12979-021-00232-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 04/06/2021] [Indexed: 12/31/2022]
Abstract
At first glance, longevity and immunity appear to be different traits that have not much in common except the fact that the immune system promotes survival upon pathogenic infection. Substantial evidence however points to a molecularly intertwined relationship between the immune system and ageing. Although this link is well-known throughout the animal kingdom, its genetic basis is complex and still poorly understood. To address this question, we here provide a compilation of all genes concomitantly known to be involved in immunity and ageing in humans and three well-studied model organisms, the nematode worm Caenorhabditis elegans, the fruit fly Drosophila melanogaster, and the house mouse Mus musculus. By analysing human orthologs among these species, we identified 7 evolutionarily conserved signalling cascades, the insulin/TOR network, three MAPK (ERK, p38, JNK), JAK/STAT, TGF-β, and Nf-κB pathways that act pleiotropically on ageing and immunity. We review current evidence for these pathways linking immunity and lifespan, and their role in the detrimental dysregulation of the immune system with age, known as immunosenescence. We argue that the phenotypic effects of these pathways are often context-dependent and vary, for example, between tissues, sexes, and types of pathogenic infection. Future research therefore needs to explore a higher temporal, spatial and environmental resolution to fully comprehend the connection between ageing and immunity.
Collapse
Affiliation(s)
- Daniel K Fabian
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, UK. .,Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK.
| | - Matías Fuentealba
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, UK.,Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Handan Melike Dönertaş
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, UK
| | - Linda Partridge
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK.,Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Janet M Thornton
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, UK
| |
Collapse
|
13
|
Alexander MS, Hightower RM, Reid AL, Bennett AH, Iyer L, Slonim DK, Saha M, Kawahara G, Kunkel LM, Kopin AS, Gupta VA, Kang PB, Draper I. hnRNP L is essential for myogenic differentiation and modulates myotonic dystrophy pathologies. Muscle Nerve 2021; 63:928-940. [PMID: 33651408 DOI: 10.1002/mus.27216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 02/25/2021] [Accepted: 02/28/2021] [Indexed: 12/12/2022]
Abstract
INTRODUCTION RNA-binding proteins (RBPs) play an important role in skeletal muscle development and disease by regulating RNA splicing. In myotonic dystrophy type 1 (DM1), the RBP MBNL1 (muscleblind-like) is sequestered by toxic CUG repeats, leading to missplicing of MBNL1 targets. Mounting evidence from the literature has implicated other factors in the pathogenesis of DM1. Herein we sought to evaluate the functional role of the splicing factor hnRNP L in normal and DM1 muscle cells. METHODS Co-immunoprecipitation assays using hnRNPL and MBNL1 expression constructs and splicing profiling in normal and DM1 muscle cell lines were performed. Zebrafish morpholinos targeting hnrpl and hnrnpl2 were injected into one-cell zebrafish for developmental and muscle analysis. In human myoblasts downregulation of hnRNP L was achieved with shRNAi. Ascochlorin administration to DM1 myoblasts was performed and expression of the CUG repeats, DM1 splicing biomarkers, and hnRNP L expression levels were evaluated. RESULTS Using DM1 patient myoblast cell lines we observed the formation of abnormal hnRNP L nuclear foci within and outside the expanded CUG repeats, suggesting a role for this factor in DM1 pathology. We showed that the antiviral and antitumorigenic isoprenoid compound ascochlorin increased MBNL1 and hnRNP L expression levels. Drug treatment of DM1 muscle cells with ascochlorin partially rescued missplicing of established early biomarkers of DM1 and improved the defective myotube formation displayed by DM1 muscle cells. DISCUSSION Together, these studies revealed that hnRNP L can modulate DM1 pathologies and is a potential therapeutic target.
Collapse
Affiliation(s)
- Matthew S Alexander
- Division of Neurology, Department of Pediatrics, University of Alabama at Birmingham and Children's of Alabama, Birmingham, Alabama, USA.,Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Civitan International Research Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rylie M Hightower
- Division of Neurology, Department of Pediatrics, University of Alabama at Birmingham and Children's of Alabama, Birmingham, Alabama, USA.,Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Andrea L Reid
- Division of Neurology, Department of Pediatrics, University of Alabama at Birmingham and Children's of Alabama, Birmingham, Alabama, USA
| | - Alexis H Bennett
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Lakshmanan Iyer
- Department of Neuroscience, Tufts University, Boston, Massachusetts, USA
| | - Donna K Slonim
- Department of Computer Science, Tufts University, Medford, Massachusetts, USA
| | - Madhurima Saha
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Genri Kawahara
- Department of Pathophysiology, Tokyo Medical University, Tokyo, Japan
| | - Louis M Kunkel
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA.,The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Alan S Kopin
- Department of Medicine, Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Vandana A Gupta
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Peter B Kang
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida, USA.,Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, Florida, USA.,Department of Neurology, University of Florida College of Medicine, Gainesville, Florida, USA.,Genetics Institute and Myology Institute, University of Florida, Gainesville, Florida, USA.,Paul and Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, Minnesota, USA.,Neurology Department, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Isabelle Draper
- Department of Medicine, Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Liu G, Liu W, Zhao R, He J, Dong Z, Chen L, Wan W, Chang Z, Wang W, Li X. Genome-wide identification and gene-editing of pigment transporter genes in the swallowtail butterfly Papilio xuthus. BMC Genomics 2021; 22:120. [PMID: 33596834 PMCID: PMC7891156 DOI: 10.1186/s12864-021-07400-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 01/19/2021] [Indexed: 02/03/2023] Open
Abstract
Background Insect body coloration often functions as camouflage to survive from predators or mate selection. Transportation of pigment precursors or related metabolites from cytoplasm to subcellular pigment granules is one of the key steps in insect pigmentation and usually executed via such transporter proteins as the ATP-binding cassette (ABC) transmembrane transporters and small G-proteins (e.g. Rab protein). However, little is known about the copy numbers of pigment transporter genes in the butterfly genomes and about the roles of pigment transporters in the development of swallowtail butterflies. Results Here, we have identified 56 ABC transporters and 58 Rab members in the genome of swallowtail butterfly Papilio xuthus. This is the first case of genome-wide gene copy number identification of ABC transporters in swallowtail butterflies and Rab family in lepidopteran insects. Aiming to investigate the contribution of the five genes which are orthologous to well-studied pigment transporters (ABCG: white, scarlet, brown and ok; Rab: lightoid) of fruit fly or silkworm during the development of swallowtail butterflies, we performed CRISPR/Cas9 gene-editing of these genes using P. xuthus as a model and sequenced the transcriptomes of their morphological mutants. Our results indicate that the disruption of each gene produced mutated phenotypes in the colors of larvae (cuticle, testis) and/or adult eyes in G0 individuals but have no effect on wing color. The transcriptomic data demonstrated that mutations induced by CRISPR/Cas9 can lead to the accumulation of abnormal transcripts and the decrease or dosage compensation of normal transcripts at gene expression level. Comparative transcriptomes revealed 606 ~ 772 differentially expressed genes (DEGs) in the mutants of four ABCG transporters and 1443 DEGs in the mutants of lightoid. GO and KEGG enrichment analysis showed that DEGs in ABCG transporter mutants enriched to the oxidoreductase activity, heme binding, iron ion binding process possibly related to the color display, and DEGs in lightoid mutants are enriched in glycoprotein binding and protein kinases. Conclusions Our data indicated these transporter proteins play an important role in body color of P. xuthus. Our study provides new insights into the function of ABC transporters and small G-proteins in the morphological development of butterflies. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07400-z.
Collapse
Affiliation(s)
- Guichun Liu
- School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, 710072, Shanxi, China.,State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China
| | - Wei Liu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China
| | - Ruoping Zhao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China
| | - Jinwu He
- School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, 710072, Shanxi, China.,State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China
| | - Zhiwei Dong
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China
| | - Lei Chen
- School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, 710072, Shanxi, China
| | - Wenting Wan
- School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, 710072, Shanxi, China.,State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China
| | - Zhou Chang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China
| | - Wen Wang
- School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, 710072, Shanxi, China. .,State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China. .,Center for Excellence in Animal Evolution and Genetics, Kunming, 650223, Yunnan, China.
| | - Xueyan Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China.
| |
Collapse
|
15
|
Tatar M. Aging Regulated Through a Stability Model of Insulin/Insulin Growth Factor Receptor Function. Front Endocrinol (Lausanne) 2021; 12:649880. [PMID: 33776941 PMCID: PMC7991905 DOI: 10.3389/fendo.2021.649880] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 02/08/2021] [Indexed: 01/04/2023] Open
Abstract
Mutations of the insulin-like receptor in Drosophila extend lifespan. New research suggests this receptor operates in two modes. The first extends lifespan while slowing reproduction and reducing growth. The second strongly extends lifespan without impairing growth or reproduction; it confers longevity assurance. The mutation that confers longevity assurance resides in the kinase insert domain, which contains a potential SH2 binding site for substrate proteins. We apply a recent model for the function of receptor tyrosine kinases to propose how insulin receptor structure can modulate aging. This concept hypothesizes that strong insulin-like ligands promote phosphorylation of high threshold substrate binding sites to robustly induce reproduction, which impairs survival as a consequence of trade-offs. Lower levels of receptor stimulation provide less kinase dimer stability, which reduces reproduction and extends lifespan by avoiding reproductive costs. Environmental conditions that favor diapause alter the expression of insulin ligands to further repress the stability of the interacting kinase domains, block phosphorylation of low threshold substrates and thus induce a unique molecular program that confers longevity assurance. Mutations of the insulin receptor that block low-phosphorylation site interactions, such as within the kinase insert domain, can extend lifespan while maintaining overall dimer stability. These flies are long-lived while maintaining reproduction and growth. The kinase insert domain of Drosophila provides a novel avenue from which to seek signaling of the insulin/insulin-like growth factor system of humans that modulate aging without impacting reproduction and growth, or incurring insulin resistance pathology.
Collapse
|
16
|
Targeting metabolic pathways for extension of lifespan and healthspan across multiple species. Ageing Res Rev 2020; 64:101188. [PMID: 33031925 DOI: 10.1016/j.arr.2020.101188] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/20/2020] [Accepted: 09/21/2020] [Indexed: 12/16/2022]
Abstract
Metabolism plays a significant role in the regulation of aging at different levels, and metabolic reprogramming represents a major driving force in aging. Metabolic reprogramming leads to impaired organismal fitness, an age-dependent increase in susceptibility to diseases, decreased ability to mount a stress response, and increased frailty. The complexity of age-dependent metabolic reprogramming comes from the multitude of levels on which metabolic changes can be connected to aging and regulation of lifespan. This is further complicated by the different metabolic requirements of various tissues, cross-organ communication via metabolite secretion, and direct effects of metabolites on epigenetic state and redox regulation; however, not all of these changes are causative to aging. Studies in yeast, flies, worms, and mice have played a crucial role in identifying mechanistic links between observed changes in various metabolic traits and their effects on lifespan. Here, we review how changes in the organismal and organ-specific metabolome are associated with aging and how targeting of any one of over a hundred different targets in specific metabolic pathways can extend lifespan. An important corollary is that restriction or supplementation of different metabolites can change activity of these metabolic pathways in ways that improve healthspan and extend lifespan in different organisms. Due to the high levels of conservation of metabolism in general, translating findings from model systems to human beings will allow for the development of effective strategies for human health- and lifespan extension.
Collapse
|
17
|
The Drosophila Post-mating Response: Gene Expression and Behavioral Changes Reveal Perdurance and Variation in Cross-Tissue Interactions. G3-GENES GENOMES GENETICS 2020; 10:967-983. [PMID: 31907222 PMCID: PMC7056969 DOI: 10.1534/g3.119.400963] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Examining cross-tissue interactions is important for understanding physiology and homeostasis. In animals, the female gonad produces signaling molecules that act distally. We examine gene expression in Drosophila melanogaster female head tissues in 1) virgins without a germline compared to virgins with a germline, 2) post-mated females with and without a germline compared to virgins, and 3) post-mated females mated to males with and without a germline compared to virgins. In virgins, the absence of a female germline results in expression changes in genes with known roles in nutrient homeostasis. At one- and three-day(s) post-mating, genes that change expression are enriched with those that function in metabolic pathways, in all conditions. We systematically examine female post-mating impacts on sleep, food preference and re-mating, in the strains and time points used for gene expression analyses and compare to published studies. We show that post-mating, gene expression changes vary by strain, prompting us to examine variation in female re-mating. We perform a genome-wide association study that identifies several DNA polymorphisms, including four in/near Wnt signaling pathway genes. Together, these data reveal how gene expression and behavior in females are influenced by cross-tissue interactions, by examining the impact of mating, fertility, and genotype.
Collapse
|
18
|
Xinxin Z, Shuang Y, Xunming Z, Shang W, Juhong Z, Jinghui X. TMT-Based Quantitative Proteomic Profiling of Overwintering Lissorhoptrus oryzophilus. Front Physiol 2020; 10:1623. [PMID: 32038298 PMCID: PMC6985562 DOI: 10.3389/fphys.2019.01623] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 12/24/2019] [Indexed: 01/05/2023] Open
Abstract
Adaptations to low temperature play a critical role in restricting the geographical distribution of insects. Decreasing day lengths and temperatures trigger seasonal cold adaptations in insects. These adaptions include changes in expression at the miRNA, mRNA and protein levels. The rice water weevil (RWW), Lissorhoptrus oryzophilus, introduced from the Mississippi River, is a globally invasive pest of wetland rice that can survive at the northern border of China. To investigate the changes in expression at the protein level in overwintering female RWW adults, 6-plex tandem mass tags (TMTs) were used in overwintering and summer adults. By using a proteome database available for Curculionidae, 1077 proteins were quantified, 183 of which differed significantly between the overwintering and summer samples. To further understand these differentially expressed proteins (DEPs), bioinformatics analyses such as gene ontology (GO) enrichment analyses were performed. DEPs associated with the terms binding, structural molecule activity, catalytic activity, multicellular organismal process, extracellular region, chitin binding, metabolic process, intracellular part and organic cyclic compound binding were altered by selection during winter. The changes in the expression of these proteins suggest that the proteins are important for RWW survival in winter.
Collapse
Affiliation(s)
- Zhang Xinxin
- College of Plant Sciences, Jilin University, Changchun, China
| | - Yang Shuang
- College of Plant Sciences, Jilin University, Changchun, China
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, China
| | - Zhang Xunming
- College of Plant Sciences, Jilin University, Changchun, China
| | - Wang Shang
- College of Plant Sciences, Jilin University, Changchun, China
| | - Zhang Juhong
- College of Plant Sciences, Jilin University, Changchun, China
| | - Xi Jinghui
- College of Plant Sciences, Jilin University, Changchun, China
| |
Collapse
|
19
|
Three Quantitative Trait Loci Explain More than 60% of Variation for Chill Coma Recovery Time in a Natural Population of Drosophila ananassae. G3-GENES GENOMES GENETICS 2019; 9:3715-3725. [PMID: 31690597 PMCID: PMC6829138 DOI: 10.1534/g3.119.400453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Ectothermic species such as insects are particularly vulnerable to climatic fluctuations. Nevertheless, many insects that evolved and diversified in the tropics have successfully colonized temperate regions all over the globe. To shed light on the genetic basis of cold tolerance in such species, we conducted a quantitative trait locus (QTL) mapping experiment for chill coma recovery time (CCRT) in Drosophila ananassae, a cosmopolitan species that has expanded its range from tropical to temperate regions. We created a mapping population of recombinant inbred advanced intercross lines (RIAILs) from two founder strains with diverging CCRT phenotypes. The RIAILs were phenotyped for their CCRT and, together with the founder strains, genotyped for polymorphic markers with double-digest restriction site-associated DNA (ddRAD) sequencing. Using a hierarchical mapping approach that combined standard interval mapping and a multiple-QTL model, we mapped three QTL which altogether explained 64% of the phenotypic variance. For two of the identified QTL, we found evidence of epistasis. To narrow down the list of cold tolerance candidate genes, we cross-referenced the QTL intervals with genes that we previously identified as differentially expressed in response to cold in D. ananassae, and with thermotolerance candidate genes of D. melanogaster. Among the 58 differentially expressed genes that were contained within the QTL, GF15058 showed a significant interaction of the CCRT phenotype and gene expression. Further, we identified the orthologs of four D. melanogaster thermotolerance candidate genes, MtnA, klarsicht, CG5246 (D.ana/GF17132) and CG10383 (D.ana/GF14829) as candidates for cold tolerance in D. ananassae.
Collapse
|
20
|
Yamada T, Habara O, Yoshii Y, Matsushita R, Kubo H, Nojima Y, Nishimura T. The role of glycogen in development and adult fitness in Drosophila. Development 2019; 146:dev.176149. [PMID: 30918052 DOI: 10.1242/dev.176149] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 03/21/2019] [Indexed: 12/20/2022]
Abstract
The polysaccharide glycogen is an evolutionarily conserved storage form of glucose. However, the physiological significance of glycogen metabolism on homeostatic control throughout the animal life cycle remains incomplete. Here, we describe Drosophila mutants that have defective glycogen metabolism. Null mutants of glycogen synthase (GlyS) and glycogen phosphorylase (GlyP) displayed growth defects and larval lethality, indicating that glycogen plays a crucial role in larval development. Unexpectedly, however, a certain population of larvae developed into adults with normal morphology. Semi-lethality in glycogen mutants during the larval period can be attributed to the presence of circulating sugar trehalose. Homozygous glycogen mutants produced offspring, indicating that glycogen stored in oocytes is dispensable for embryogenesis. GlyS and GlyP mutants showed distinct metabolic defects in the levels of circulating sugars and triglycerides in a life stage-specific manner. In adults, glycogen as an energy reserve is not crucial for physical fitness and lifespan under nourished conditions, but glycogen becomes important under energy stress conditions. This study provides a fundamental understanding of the stage-specific requirements for glycogen metabolism in the fruit fly.
Collapse
Affiliation(s)
- Takayuki Yamada
- Laboratory for Growth Control Signaling, RIKEN Center for Biosystems Dynamics Research (BDR), 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Okiko Habara
- Laboratory for Growth Control Signaling, RIKEN Center for Biosystems Dynamics Research (BDR), 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Yuka Yoshii
- Laboratory for Growth Control Signaling, RIKEN Center for Biosystems Dynamics Research (BDR), 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.,Graduate School of Biological Science, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0101, Japan
| | - Ryota Matsushita
- Laboratory for Growth Control Signaling, RIKEN Center for Biosystems Dynamics Research (BDR), 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.,Graduate School of Biological Science, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0101, Japan
| | - Hitomi Kubo
- Laboratory for Growth Control Signaling, RIKEN Center for Biosystems Dynamics Research (BDR), 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Yosui Nojima
- Laboratory for Growth Control Signaling, RIKEN Center for Biosystems Dynamics Research (BDR), 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Takashi Nishimura
- Laboratory for Growth Control Signaling, RIKEN Center for Biosystems Dynamics Research (BDR), 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan .,Graduate School of Biological Science, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0101, Japan
| |
Collapse
|
21
|
Everman ER, McNeil CL, Hackett JL, Bain CL, Macdonald SJ. Dissection of Complex, Fitness-Related Traits in Multiple Drosophila Mapping Populations Offers Insight into the Genetic Control of Stress Resistance. Genetics 2019; 211:1449-1467. [PMID: 30760490 PMCID: PMC6456312 DOI: 10.1534/genetics.119.301930] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 02/06/2019] [Indexed: 12/11/2022] Open
Abstract
We leverage two complementary Drosophila melanogaster mapping panels to genetically dissect starvation resistance-an important fitness trait. Using >1600 genotypes from the multiparental Drosophila Synthetic Population Resource (DSPR), we map numerous starvation stress QTL that collectively explain a substantial fraction of trait heritability. Mapped QTL effects allowed us to estimate DSPR founder phenotypes, predictions that were correlated with the actual phenotypes of these lines. We observe a modest phenotypic correlation between starvation resistance and triglyceride level, traits that have been linked in previous studies. However, overlap among QTL identified for each trait is low. Since we also show that DSPR strains with extreme starvation phenotypes differ in desiccation resistance and activity level, our data imply multiple physiological mechanisms contribute to starvation variability. We additionally exploited the Drosophila Genetic Reference Panel (DGRP) to identify sequence variants associated with starvation resistance. Consistent with prior work these sites rarely fall within QTL intervals mapped in the DSPR. We were offered a unique opportunity to directly compare association mapping results across laboratories since two other groups previously measured starvation resistance in the DGRP. We found strong phenotypic correlations among studies, but extremely low overlap in the sets of genomewide significant sites. Despite this, our analyses revealed that the most highly associated variants from each study typically showed the same additive effect sign in independent studies, in contrast to otherwise equivalent sets of random variants. This consistency provides evidence for reproducible trait-associated sites in a widely used mapping panel, and highlights the polygenic nature of starvation resistance.
Collapse
Affiliation(s)
- Elizabeth R Everman
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66045
| | - Casey L McNeil
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66045
| | - Jennifer L Hackett
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66045
| | - Clint L Bain
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66045
| | - Stuart J Macdonald
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66045
| |
Collapse
|
22
|
Cytoplasmic and Mitochondrial NADPH-Coupled Redox Systems in the Regulation of Aging. Nutrients 2019; 11:nu11030504. [PMID: 30818813 PMCID: PMC6471790 DOI: 10.3390/nu11030504] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 12/20/2022] Open
Abstract
The reduced form of nicotinamide adenine dinucleotide phosphate (NADPH) protects against redox stress by providing reducing equivalents to antioxidants such as glutathione and thioredoxin. NADPH levels decline with aging in several tissues, but whether this is a major driving force for the aging process has not been well established. Global or neural overexpression of several cytoplasmic enzymes that synthesize NADPH have been shown to extend lifespan in model organisms such as Drosophila suggesting a positive relationship between cytoplasmic NADPH levels and longevity. Mitochondrial NADPH plays an important role in the protection against redox stress and cell death and mitochondrial NADPH-utilizing thioredoxin reductase 2 levels correlate with species longevity in cells from rodents and primates. Mitochondrial NADPH shuttles allow for some NADPH flux between the cytoplasm and mitochondria. Since a decline of nicotinamide adenine dinucleotide (NAD+) is linked with aging and because NADP+ is exclusively synthesized from NAD+ by cytoplasmic and mitochondrial NAD+ kinases, a decline in the cytoplasmic or mitochondrial NADPH pool may also contribute to the aging process. Therefore pro-longevity therapies should aim to maintain the levels of both NAD+ and NADPH in aging tissues.
Collapse
|
23
|
Everman ER, Morgan TJ. Antagonistic pleiotropy and mutation accumulation contribute to age-related decline in stress response. Evolution 2018; 72:303-317. [PMID: 29214647 DOI: 10.1111/evo.13408] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 11/10/2017] [Accepted: 11/13/2017] [Indexed: 01/17/2023]
Abstract
As organisms age, the effectiveness of natural selection weakens, leading to age-related decline in fitness-related traits. The evolution of age-related changes associated with senescence is likely influenced by mutation accumulation (MA) and antagonistic pleiotropy (AP). MA predicts that age-related decline in fitness components is driven by age-specific sets of alleles, nonnegative genetic correlations within trait across age, and an increase in the coefficient of genetic variance. AP predicts that age-related decline in a trait is driven by alleles with positive effects on fitness in young individuals and negative effects in old individuals, and is expected to lead to negative genetic correlations within traits across age. We build on these predictions using an association mapping approach to investigate the change in additive effects of SNPs across age and among traits for multiple stress-response fitness-related traits, including cold stress with and without acclimation and starvation resistance. We found support for both MA and AP theories of aging in the age-related decline in stress tolerance. Our study demonstrates that the evolution of age-related decline in stress tolerance is driven by a combination of alleles that have age-specific additive effects, consistent with MA, as well as nonindependent and antagonistic genetic architectures characteristic of AP.
Collapse
Affiliation(s)
- Elizabeth R Everman
- Division of Biology, Kansas State University, Manhattan, Kansas 66506
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66045
| | - Theodore J Morgan
- Division of Biology, Kansas State University, Manhattan, Kansas 66506
| |
Collapse
|
24
|
Short-term exposure to dim light at night disrupts rhythmic behaviors and causes neurodegeneration in fly models of tauopathy and Alzheimer's disease. Biochem Biophys Res Commun 2017; 495:1722-1729. [PMID: 29217196 DOI: 10.1016/j.bbrc.2017.12.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 12/04/2017] [Indexed: 12/14/2022]
Abstract
The accumulation and aggregation of phosphorylated tau proteins in the brain are the hallmarks for the onset of Alzheimer's disease (AD). In addition, disruptions in circadian rhythms (CRs) with altered sleep-wake cycles, dysregulation of locomotion, and increased memory defects have been reported in patients with AD. Drosophila flies that have an overexpression of human tau protein in neurons exhibit most of the symptoms of human patients with AD, including locomotion defects and neurodegeneration. Using the fly model for tauopathy/AD, we investigated the effects of an exposure to dim light at night on AD symptoms. We used a light intensity of 10 lux, which is considered the lower limit of light pollution in many countries. After the tauopathy flies were exposed to the dim light at night for 3 days, the flies showed disrupted CRs, altered sleep-wake cycles due to increased pTau proteins and neurodegeneration, in the brains of the AD flies. The results indicate that the nighttime exposure of tauopathy/AD model Drosophila flies to dim light disrupted CR and sleep-wake behavior and promoted neurodegeneration.
Collapse
|
25
|
Yu S, Lee E, Tsogbadrakh B, Son GI, Kim M. Prenatal hyperbaric normoxia treatment improves healthspan and regulates chitin metabolic genes in Drosophila melanogaster. Aging (Albany NY) 2017; 8:2538-2550. [PMID: 27777382 PMCID: PMC5115905 DOI: 10.18632/aging.101084] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 10/10/2016] [Indexed: 12/30/2022]
Abstract
Aging is a universal, irreversible process accompanied by physiological declines that culminate in death. Rapid progress in gerontology research has revealed that aging can be slowed through mild stress-induced hormesis. We previously reported that hyperbaric normoxia (HN, 2 atm absolute pressure with 10% O2) induces a cytoprotective response in vitro by regulating fibronectin. In the present study, we investigated the hormetic effects of prenatal HN exposure on Drosophila healthspan related to molecular defense mechanisms. HN exposure had no disruptive effect on developmental rate or adult body weight. However, lifespan was clearly enhanced, as was resistance to oxidative and heat stress. In addition, levels of reactive oxygen species were significantly decreased and motor performance was increased. HN stress has been shown to trigger molecular changes in the heat shock response and ROS scavenging system, including hsp70, catalase, glutathione synthase, and MnSOD. Furthermore, to determine the hormetic mechanism underlying these phenotypic and molecular changes, we performed a genome-wide profiling in HN-exposed and control flies. Genes encoding chitin metabolism were highly up-regulated, which could possibly serve to scavenge free radicals. These results identify prenatal HN exposure as a potential hormetic factor that may improve longevity and healthspan by enhancing defense mechanisms in Drosophila.
Collapse
Affiliation(s)
- Suyeun Yu
- Department of Preventive Medicine, College of Medicine, Korea University, Seoul, 136-701, Republic of Korea
| | - Eunil Lee
- Department of Preventive Medicine, College of Medicine, Korea University, Seoul, 136-701, Republic of Korea
| | - Bodokhsuren Tsogbadrakh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, 151-742, Republic of Korea
| | - Gwang-Ic Son
- Department of Preventive Medicine, College of Medicine, Korea University, Seoul, 136-701, Republic of Korea
| | - Mari Kim
- Department of Preventive Medicine, College of Medicine, Korea University, Seoul, 136-701, Republic of Korea
| |
Collapse
|
26
|
Abstract
Cancer immunotherapy is an increasingly successful strategy for the treatment of patients who have advanced or conventional therapy-resistant cancers. T cells are key mediators of tumor destruction and their specificity for tumor-expressed antigens is of paramount importance, but other T cell-intrinsic qualities, such as durability, longevity, and functionality also play important roles in determining the efficacy of immunotherapy. The cellular energetic pathways that are utilized by T cells play a key role in regulating each of these qualities. Metabolic activity, which both regulates and is regulated by cellular signaling pathways and epigenetics, also profoundly influences the trajectories of T cell differentiation and fate. In this Review, we discuss how cell metabolism influences T cell anti-tumor activity, the metabolic qualities of highly-functional T cells, and strategies to modulate metabolism for improving the immune response to tumors.
Collapse
Affiliation(s)
- Rigel J Kishton
- Center for Cell-Based Therapy, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA; Surgery Branch, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| | - Madhusudhanan Sukumar
- Center for Cell-Based Therapy, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA; Surgery Branch, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Nicholas P Restifo
- Center for Cell-Based Therapy, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA; Surgery Branch, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
27
|
Garlapow ME, Everett LJ, Zhou S, Gearhart AW, Fay KA, Huang W, Morozova TV, Arya GH, Turlapati L, St Armour G, Hussain YN, McAdams SE, Fochler S, Mackay TFC. Genetic and Genomic Response to Selection for Food Consumption in Drosophila melanogaster. Behav Genet 2017; 47:227-243. [PMID: 27704301 PMCID: PMC5305434 DOI: 10.1007/s10519-016-9819-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 09/16/2016] [Indexed: 12/21/2022]
Abstract
Food consumption is an essential component of animal fitness; however, excessive food intake in humans increases risk for many diseases. The roles of neuroendocrine feedback loops, food sensing modalities, and physiological state in regulating food intake are well understood, but not the genetic basis underlying variation in food consumption. Here, we applied ten generations of artificial selection for high and low food consumption in replicate populations of Drosophila melanogaster. The phenotypic response to selection was highly asymmetric, with significant responses only for increased food consumption and minimal correlated responses in body mass and composition. We assessed the molecular correlates of selection responses by DNA and RNA sequencing of the selection lines. The high and low selection lines had variants with significantly divergent allele frequencies within or near 2081 genes and 3526 differentially expressed genes in one or both sexes. A total of 519 genes were both genetically divergent and differentially expressed between the divergent selection lines. We performed functional analyses of the effects of RNAi suppression of gene expression and induced mutations for 27 of these candidate genes that have human orthologs and the strongest statistical support, and confirmed that 25 (93 %) affected the mean and/or variance of food consumption.
Collapse
Affiliation(s)
- Megan E Garlapow
- Program in Genetics, North Carolina State University, Raleigh, NC, 27695-7614, USA
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695-7614, USA
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, 27695-7614, USA
| | - Logan J Everett
- Program in Genetics, North Carolina State University, Raleigh, NC, 27695-7614, USA
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695-7614, USA
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, 27695-7614, USA
- Initiative for Biological Complexity, North Carolina State University, Raleigh, NC, 27695-7614, USA
| | - Shanshan Zhou
- Program in Genetics, North Carolina State University, Raleigh, NC, 27695-7614, USA
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695-7614, USA
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, 27695-7614, USA
- Initiative for Biological Complexity, North Carolina State University, Raleigh, NC, 27695-7614, USA
| | - Alexander W Gearhart
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695-7614, USA
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, 27695-7614, USA
| | - Kairsten A Fay
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695-7614, USA
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, 27695-7614, USA
| | - Wen Huang
- Program in Genetics, North Carolina State University, Raleigh, NC, 27695-7614, USA
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695-7614, USA
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, 27695-7614, USA
- Initiative for Biological Complexity, North Carolina State University, Raleigh, NC, 27695-7614, USA
| | - Tatiana V Morozova
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695-7614, USA
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, 27695-7614, USA
| | - Gunjan H Arya
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695-7614, USA
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, 27695-7614, USA
| | - Lavanya Turlapati
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695-7614, USA
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, 27695-7614, USA
| | - Genevieve St Armour
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695-7614, USA
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, 27695-7614, USA
| | - Yasmeen N Hussain
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695-7614, USA
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, 27695-7614, USA
| | - Sarah E McAdams
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695-7614, USA
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, 27695-7614, USA
| | - Sophia Fochler
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695-7614, USA
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, 27695-7614, USA
- School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Trudy F C Mackay
- Program in Genetics, North Carolina State University, Raleigh, NC, 27695-7614, USA.
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695-7614, USA.
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, 27695-7614, USA.
- Initiative for Biological Complexity, North Carolina State University, Raleigh, NC, 27695-7614, USA.
| |
Collapse
|
28
|
Ligand-Bound GeneSwitch Causes Developmental Aberrations in Drosophila that Are Alleviated by the Alternative Oxidase. G3-GENES GENOMES GENETICS 2016; 6:2839-46. [PMID: 27412986 PMCID: PMC5015941 DOI: 10.1534/g3.116.030882] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Culture of Drosophila expressing the steroid-dependent GeneSwitch transcriptional activator under the control of the ubiquitous α-tubulin promoter was found to produce extensive pupal lethality, as well as a range of dysmorphic adult phenotypes, in the presence of high concentrations of the inducing drug RU486. Prominent among these was cleft thorax, seen previously in flies bearing mutant alleles of the nuclear receptor Ultraspiracle and many other mutants, as well as notched wings, leg malformations, and bristle abnormalities. Neither the α-tubulin-GeneSwitch driver nor the inducing drug on their own produced any of these effects. A second GeneSwitch driver, under the control of the daughterless promoter, which gave much lower and more tissue-restricted transgene expression, exhibited only mild bristle abnormalities in the presence of high levels of RU486. Coexpression of the alternative oxidase (AOX) from Ciona intestinalis produced a substantial shift in the developmental outcome toward a wild-type phenotype, which was dependent on the AOX expression level. Neither an enzymatically inactivated variant of AOX, nor GFP, or the alternative NADH dehydrogenase Ndi1 from yeast gave any such rescue. Users of the GeneSwitch system should be aware of the potential confounding effects of its application in developmental studies.
Collapse
|
29
|
Longevity-modulating effects of symbiosis: insights from Drosophila–Wolbachia interaction. Biogerontology 2016; 17:785-803. [DOI: 10.1007/s10522-016-9653-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 05/18/2016] [Indexed: 01/30/2023]
|
30
|
Gajan A, Barnes VL, Liu M, Saha N, Pile LA. The histone demethylase dKDM5/LID interacts with the SIN3 histone deacetylase complex and shares functional similarities with SIN3. Epigenetics Chromatin 2016; 9:4. [PMID: 26848313 PMCID: PMC4740996 DOI: 10.1186/s13072-016-0053-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 01/14/2016] [Indexed: 01/01/2023] Open
Abstract
Background Regulation of gene expression by histone-modifying enzymes is essential to control cell fate decisions and developmental processes. Two histone-modifying enzymes, RPD3, a deacetylase, and dKDM5/LID, a demethylase, are present in a single complex, coordinated through the SIN3 scaffold protein. While the SIN3 complex has been demonstrated to have functional histone deacetylase activity, the role of the demethylase dKDM5/LID as part of the complex has not been investigated. Results Here, we analyzed the developmental and transcriptional activities of dKDM5/LID in relation to SIN3. Knockdown of either Sin3A or lid resulted in decreased cell proliferation in S2 cells and wing imaginal discs. Conditional knockdown of either Sin3A or lid resulted in flies that displayed wing developmental defects. Interestingly, overexpression of dKDM5/LID rescued the wing developmental defect due to reduced levels of SIN3 in female flies, indicating a major role for dKDM5/LID in cooperation with SIN3 during development. Together, these observed phenotypes strongly suggest that dKDM5/LID as part of the SIN3 complex can impact previously uncharacterized transcriptional networks. Transcriptome analysis revealed that SIN3 and dKDM5/LID regulate many common genes. While several genes implicated in cell cycle and wing developmental pathways were affected upon altering the level of these chromatin factors, a significant affect was also observed on genes required to mount an effective stress response. Further, under conditions of induced oxidative stress, reduction of SIN3 and/or dKDM5/LID altered the expression of a greater number of genes involved in cell cycle-related processes relative to normal conditions. This highlights an important role for SIN3 and dKDM5/LID proteins to maintain proper progression through the cell cycle in environments of cellular stress. Further, we find that target genes are bound by both SIN3 and dKDM5/LID, however, histone acetylation, not methylation, plays a predominant role in gene regulation by the SIN3 complex. Conclusions We have provided genetic evidence to demonstrate functional cooperation between the histone demethylase dKDM5/LID and SIN3. Biochemical and transcriptome data further support functional links between these proteins. Together, the data provide a solid framework for analyzing the gene regulatory pathways through which SIN3 and dKDM5/LID control diverse biological processes in the organism. Electronic supplementary material The online version of this article (doi:10.1186/s13072-016-0053-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ambikai Gajan
- Department of Biological Sciences, Wayne State University, Detroit, MI USA
| | - Valerie L Barnes
- Department of Biological Sciences, Wayne State University, Detroit, MI USA
| | - Mengying Liu
- Department of Biological Sciences, Wayne State University, Detroit, MI USA
| | - Nirmalya Saha
- Department of Biological Sciences, Wayne State University, Detroit, MI USA
| | - Lori A Pile
- Department of Biological Sciences, Wayne State University, Detroit, MI USA
| |
Collapse
|
31
|
Basic mechanisms of longevity: A case study of Drosophila pro-longevity genes. Ageing Res Rev 2015; 24:218-31. [PMID: 26318059 DOI: 10.1016/j.arr.2015.08.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 08/11/2015] [Accepted: 08/20/2015] [Indexed: 12/13/2022]
Abstract
Drosophila is one of the most convenient model organisms in the genetics of aging and longevity. Unlike the nematodes, which allow for the detection of new pro-aging genes by knockout and RNAi-mediated knock-down, Drosophila also provides an opportunity to find new pro-longevity genes by driver-induced overexpression. Similar studies on other models are extremely rare. In this review, we focused on genes whose overexpression prolongs the life of fruit flies. The majority of longevity-associated genes regulates metabolism and stress resistance, and belongs to the IGF-1R, PI3K, PKB, AMPK and TOR metabolic regulation cluster and the FOXO, HDAC, p53 stress response cluster.
Collapse
|
32
|
Carnes MU, Campbell T, Huang W, Butler DG, Carbone MA, Duncan LH, Harbajan SV, King EM, Peterson KR, Weitzel A, Zhou S, Mackay TFC. The Genomic Basis of Postponed Senescence in Drosophila melanogaster. PLoS One 2015; 10:e0138569. [PMID: 26378456 PMCID: PMC4574564 DOI: 10.1371/journal.pone.0138569] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 09/01/2015] [Indexed: 12/30/2022] Open
Abstract
Natural populations harbor considerable genetic variation for lifespan. While evolutionary theory provides general explanations for the existence of this variation, our knowledge of the genes harboring naturally occurring polymorphisms affecting lifespan is limited. Here, we assessed the genetic divergence between five Drosophila melanogaster lines selected for postponed senescence for over 170 generations (O lines) and five lines from the same base population maintained at a two week generation interval for over 850 generations (B lines). On average, O lines live 70% longer than B lines, are more productive at all ages, and have delayed senescence for other traits than reproduction. We performed population sequencing of pools of individuals from all B and O lines and identified 6,394 genetically divergent variants in or near 1,928 genes at a false discovery rate of 0.068. A 2.6 Mb region at the tip of the X chromosome contained many variants fixed for alternative alleles in the two populations, suggestive of a hard selective sweep. We also assessed genome wide gene expression of O and B lines at one and five weeks of age using RNA sequencing and identified genes with significant (false discovery rate < 0.05) effects on gene expression with age, population and the age by population interaction, separately for each sex. We identified transcripts that exhibited the transcriptional signature of postponed senescence and integrated the gene expression and genetic divergence data to identify 98 (175) top candidate genes in females (males) affecting postponed senescence and increased lifespan. While several of these genes have been previously associated with Drosophila lifespan, most are novel and constitute a rich resource for future functional validation.
Collapse
Affiliation(s)
- Megan Ulmer Carnes
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
| | - Terry Campbell
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
| | - Wen Huang
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
- Program in Genetics, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
| | - Daniel G. Butler
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
| | - Mary Anna Carbone
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
- Program in Genetics, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
| | - Laura H. Duncan
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
| | - Sasha V. Harbajan
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
| | - Edward M. King
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
| | - Kara R. Peterson
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
| | - Alexander Weitzel
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
| | - Shanshan Zhou
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
- Program in Genetics, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
| | - Trudy F. C. Mackay
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
- Program in Genetics, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
| |
Collapse
|
33
|
Nmdmc overexpression extends Drosophila lifespan and reduces levels of mitochondrial reactive oxygen species. Biochem Biophys Res Commun 2015; 465:845-50. [PMID: 26319556 DOI: 10.1016/j.bbrc.2015.08.098] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 08/22/2015] [Indexed: 12/18/2022]
Abstract
NAD-dependent methylenetetrahydrofolate dehydrogenase-methenyltetrahydrofolate cyclohydrolase (NMDMC) is a bifunctional enzyme involved in folate-dependent metabolism and highly expressed in rapidly proliferating cells. However, Nmdmc physiological roles remain unveiled. We found that ubiquitous Nmdmc overexpression enhanced Drosophila lifespan and stress resistance. Interestingly, Nmdmc overexpression in the fat body was sufficient to increase lifespan and tolerance against oxidative stress. In addition, these conditions coincided with significant decreases in the levels of mitochondrial ROS and Hsp22 as well as with a significant increase in the copy number of mitochondrial DNA. These results suggest that Nmdmc overexpression should be beneficial for mitochondrial homeostasis and increasing lifespan.
Collapse
|
34
|
Tower J. Mitochondrial maintenance failure in aging and role of sexual dimorphism. Arch Biochem Biophys 2015; 576:17-31. [PMID: 25447815 PMCID: PMC4409928 DOI: 10.1016/j.abb.2014.10.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 10/08/2014] [Accepted: 10/18/2014] [Indexed: 12/31/2022]
Abstract
Gene expression changes during aging are partly conserved across species, and suggest that oxidative stress, inflammation and proteotoxicity result from mitochondrial malfunction and abnormal mitochondrial-nuclear signaling. Mitochondrial maintenance failure may result from trade-offs between mitochondrial turnover versus growth and reproduction, sexual antagonistic pleiotropy and genetic conflicts resulting from uni-parental mitochondrial transmission, as well as mitochondrial and nuclear mutations and loss of epigenetic regulation. Aging phenotypes and interventions are often sex-specific, indicating that both male and female sexual differentiation promote mitochondrial failure and aging. Studies in mammals and invertebrates implicate autophagy, apoptosis, AKT, PARP, p53 and FOXO in mediating sex-specific differences in stress resistance and aging. The data support a model where the genes Sxl in Drosophila, sdc-2 in Caenorhabditis elegans, and Xist in mammals regulate mitochondrial maintenance across generations and in aging. Several interventions that increase life span cause a mitochondrial unfolded protein response (UPRmt), and UPRmt is also observed during normal aging, indicating hormesis. The UPRmt may increase life span by stimulating mitochondrial turnover through autophagy, and/or by inhibiting the production of hormones and toxic metabolites. The data suggest that metazoan life span interventions may act through a common hormesis mechanism involving liver UPRmt, mitochondrial maintenance and sexual differentiation.
Collapse
Affiliation(s)
- John Tower
- Molecular and Computational Biology Program, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089-2910, United States.
| |
Collapse
|
35
|
Barnes VL, Bhat A, Unnikrishnan A, Heydari AR, Arking R, Pile LA. SIN3 is critical for stress resistance and modulates adult lifespan. Aging (Albany NY) 2015; 6:645-60. [PMID: 25133314 PMCID: PMC4169859 DOI: 10.18632/aging.100684] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Coordinate control of gene activity is critical for fitness and longevity of an organism. The SIN3 histone deacetylase (HDAC) complex functions as a transcriptional repressor of many genes. SIN3-regulated genes include those that encode proteins affecting multiple aspects of mitochondrial function, such as energy production and stress responsiveness, important for health maintenance. Here we used Drosophila melanogaster as a model organism to examine the role of SIN3 in the regulation of fitness and longevity. Adult flies with RNA interference (RNAi) induced knockdown expression of Sin3A have reduced climbing ability; an activity that likely requires fully functional mitochondria. Additionally, compared to wild type, adult Sin3A knockdown flies were more sensitive to oxidative stress. Interestingly, media supplementation with the antioxidant glutathione largely restored fly tolerance to oxidative stress. Although Sin3A knockdown flies exhibited decreased longevity compared to wild type, no significant changes in expression of many well-categorized aging genes were observed. We found, however, that Sin3A knockdown corresponded to a significant reduction in expression of genes encoding proteins involved in the de novo synthesis of glutathione. Taken together, the data support a model whereby SIN3 regulates a gene expression program required for proper mitochondrial function and effective stress response during adulthood.
Collapse
Affiliation(s)
- Valerie L Barnes
- Department of Biological Sciences, Wayne State University, Detroit, Michigan, 48202, USA
| | - Abhineeth Bhat
- Department of Biological Sciences, Wayne State University, Detroit, Michigan, 48202, USA
| | - Archana Unnikrishnan
- Department of Nutrition and Food Science Wayne State University, Detroit, Michigan, 48202,USA
| | - Ahmad R Heydari
- Department of Nutrition and Food Science Wayne State University, Detroit, Michigan, 48202,USA
| | - Robert Arking
- Department of Biological Sciences, Wayne State University, Detroit, Michigan, 48202, USA
| | - Lori A Pile
- Department of Biological Sciences, Wayne State University, Detroit, Michigan, 48202, USA
| |
Collapse
|
36
|
Omelyanchuk LV, Shaposhnikov MV, Moskalev AA. Drosophila nervous system as a target of aging and anti-aging interventions. Front Genet 2015; 6:89. [PMID: 25806047 PMCID: PMC4354387 DOI: 10.3389/fgene.2015.00089] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 02/19/2015] [Indexed: 12/18/2022] Open
Affiliation(s)
- Leonid V. Omelyanchuk
- Institute of Molecular and Cellular Biology, Siberian Branch of Russian Academy of SciencesNovosibirsk, Russia
- Department of Cytology and Genetics, Novosibirsk State UniversityNovosibirsk, Russia
| | - Mikhail V. Shaposhnikov
- Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of SciencesSyktyvkar, Russia
- Moscow Institute of Physics and TechnologyDolgoprudny, Russia
| | - Alexey A. Moskalev
- Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of SciencesSyktyvkar, Russia
- Moscow Institute of Physics and TechnologyDolgoprudny, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of SciencesMoscow, Russia
| |
Collapse
|
37
|
Kim GH, Lee YE, Lee GH, Cho YH, Lee YN, Jang Y, Paik D, Park JJ. Overexpression of malic enzyme in the larval stage extends Drosophila lifespan. Biochem Biophys Res Commun 2014; 456:676-82. [PMID: 25511696 DOI: 10.1016/j.bbrc.2014.12.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 12/05/2014] [Indexed: 02/01/2023]
Abstract
Metabolic modifications during the developmental period can extend longevity. We found that malic enzyme (Men) overexpression during the larval period lengthened the lifespan of Drosophila. Men overexpression by S106-GeneSwitch-Gal4 driver increased pyruvate content and NADPH/NADP(+) ratio but reduced triglyceride, glycogen, and ATP levels in the larvae. ROS levels increased unexpectedly in Men-overexpressing larvae. Interestingly, adults exposed to larval Men-overexpression maintained ROS tolerance with enhanced expression levels of glutathione-S-transferase D2 and thioredoxin-2. Our results suggest that metabolic changes mediated by Men during development might be related to the control of ROS tolerance and the longevity of Drosophila.
Collapse
Affiliation(s)
- Gye-Hyeong Kim
- Department of Physiology, College of Medicine, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul 136-705, Republic of Korea
| | - Young-Eun Lee
- Department of Physiology, College of Medicine, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul 136-705, Republic of Korea
| | - Gun-Ho Lee
- Department of Physiology, College of Medicine, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul 136-705, Republic of Korea
| | - Youn-Ho Cho
- Department of Physiology, College of Medicine, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul 136-705, Republic of Korea
| | - Young-Nam Lee
- Department of Physiology, College of Medicine, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul 136-705, Republic of Korea
| | - Yeogil Jang
- Department of Physiology, College of Medicine, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul 136-705, Republic of Korea
| | - Donggi Paik
- Department of Physiology, College of Medicine, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul 136-705, Republic of Korea
| | - Joong-Jean Park
- Department of Physiology, College of Medicine, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul 136-705, Republic of Korea.
| |
Collapse
|
38
|
Emerging roles for hnRNPs in post-transcriptional regulation: what can we learn from flies? Chromosoma 2014; 123:515-27. [PMID: 24913828 DOI: 10.1007/s00412-014-0470-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 05/21/2014] [Accepted: 05/23/2014] [Indexed: 12/13/2022]
Abstract
Heterogeneous nuclear ribonucleoproteins (hnRNPs) are a highly conserved family of RNA-binding proteins able to associate with nascent RNAs in order to support their localization, maturation and translation. Research over this last decade has remarked the importance of gene regulatory processes at post-transcriptional level, highlighting the emerging roles of hnRNPs in several essential biological events. Indeed, hnRNPs are key factors in regulating gene expression, thus, having a number of roles in many biological pathways. Moreover, failure of the activities catalysed by hnRNPs affects various biological processes and may underlie several human diseases including cancer, diabetes and neurodegenerative syndromes. In this review, we summarize some of hnRNPs' roles in the model organism Drosophila melanogaster, particularly focusing on their participation in all aspects of post-transcriptional regulation as well as their conserved role and involvement in the aetiology of human pathologies.
Collapse
|
39
|
Park S, Sonn JY, Oh Y, Lim C, Choe J. SIFamide and SIFamide receptor defines a novel neuropeptide signaling to promote sleep in Drosophila. Mol Cells 2014; 37:295-301. [PMID: 24658384 PMCID: PMC4012077 DOI: 10.14348/molcells.2014.2371] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 02/25/2014] [Accepted: 03/04/2014] [Indexed: 01/16/2023] Open
Abstract
SIFamide receptor (SIFR) is a Drosophila G protein-coupled receptor for the neuropeptide SIFamide (SIFa). Although the sequence and spatial expression of SIFa are evolutionarily conserved among insect species, the physiological function of SIFa/SIFR signaling remains elusive. Here, we provide genetic evidence that SIFa and SIFR promote sleep in Drosophila. Either genetic ablation of SIFa-expressing neurons in the pars intercerebralis (PI) or pan-neuronal depletion of SIFa expression shortened baseline sleep and reduced sleep-bout length, suggesting that it caused sleep fragmentation. Consistently, RNA interference- mediated knockdown of SIFR expression caused short sleep phenotypes as observed in SIFa-ablated or depleted flies. Using a panel of neuron-specific Gal4 drivers, we further mapped SIFR effects to subsets of PI neurons. Taken together, these results reveal a novel physiological role of the neuropeptide SIFa/SIFR pathway to regulate sleep through sleep-promoting neural circuits in the PI of adult fly brains.
Collapse
Affiliation(s)
- Sangjin Park
- Department of Biological Sciences, College of Life Science and Bioengineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
| | - Jun Young Sonn
- Department of Biological Sciences, College of Life Science and Bioengineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
| | - Yangkyun Oh
- Department of Biological Sciences, College of Life Science and Bioengineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
| | - Chunghun Lim
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan 689-798,
Korea
| | - Joonho Choe
- Department of Biological Sciences, College of Life Science and Bioengineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
| |
Collapse
|
40
|
Moskalev A, Shaposhnikov M, Snezhkina A, Kogan V, Plyusnina E, Peregudova D, Melnikova N, Uroshlev L, Mylnikov S, Dmitriev A, Plusnin S, Fedichev P, Kudryavtseva A. Mining gene expression data for pollutants (dioxin, toluene, formaldehyde) and low dose of gamma-irradiation. PLoS One 2014; 9:e86051. [PMID: 24475070 PMCID: PMC3901678 DOI: 10.1371/journal.pone.0086051] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 12/04/2013] [Indexed: 12/28/2022] Open
Abstract
General and specific effects of molecular genetic responses to adverse environmental factors are not well understood. This study examines genome-wide gene expression profiles of Drosophila melanogaster in response to ionizing radiation, formaldehyde, toluene, and 2,3,7,8-tetrachlorodibenzo-p-dioxin. We performed RNA-seq analysis on 25,415 transcripts to measure the change in gene expression in males and females separately. An analysis of the genes unique to each treatment yielded a list of genes as a gene expression signature. In the case of radiation exposure, both sexes exhibited a reproducible increase in their expression of the transcription factors sugarbabe and tramtrack. The influence of dioxin up-regulated metabolic genes, such as anachronism, CG16727, and several genes with unknown function. Toluene activated a gene involved in the response to the toxins, Cyp12d1-p; the transcription factor Fer3's gene; the metabolic genes CG2065, CG30427, and CG34447; and the genes Spn28Da and Spn3, which are responsible for reproduction and immunity. All significantly differentially expressed genes, including those shared among the stressors, can be divided into gene groups using Gene Ontology Biological Process identifiers. These gene groups are related to defense response, biological regulation, the cell cycle, metabolic process, and circadian rhythms. KEGG molecular pathway analysis revealed alteration of the Notch signaling pathway, TGF-beta signaling pathway, proteasome, basal transcription factors, nucleotide excision repair, Jak-STAT signaling pathway, circadian rhythm, Hippo signaling pathway, mTOR signaling pathway, ribosome, mismatch repair, RNA polymerase, mRNA surveillance pathway, Hedgehog signaling pathway, and DNA replication genes. Females and, to a lesser extent, males actively metabolize xenobiotics by the action of cytochrome P450 when under the influence of dioxin and toluene. Finally, in this work we obtained gene expression signatures pollutants (dioxin, toluene), low dose of gamma-irradiation and common molecular pathways for different kind of stressors.
Collapse
Affiliation(s)
- Alexey Moskalev
- Laboratory of Molecular Radiobiology and Gerontology, Institute of Biology of Komi Science Center of RAS, Syktyvkar, Russia
- Ecological Department, Syktyvkar State University, Syktyvkar, Russia
- Laboratory of Genetics of Aging and Longevity, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Mikhail Shaposhnikov
- Laboratory of Molecular Radiobiology and Gerontology, Institute of Biology of Komi Science Center of RAS, Syktyvkar, Russia
- Ecological Department, Syktyvkar State University, Syktyvkar, Russia
| | - Anastasia Snezhkina
- Group of Postgenomic Studies, Engelhardt Institute of Molecular Biology of RAS, Moscow, Russia
| | - Valeria Kogan
- Laboratory of Genetics of Aging and Longevity, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Quantum Pharmaceuticals, Moscow, Russia
| | - Ekaterina Plyusnina
- Laboratory of Molecular Radiobiology and Gerontology, Institute of Biology of Komi Science Center of RAS, Syktyvkar, Russia
- Ecological Department, Syktyvkar State University, Syktyvkar, Russia
| | - Darya Peregudova
- Laboratory of Molecular Radiobiology and Gerontology, Institute of Biology of Komi Science Center of RAS, Syktyvkar, Russia
| | - Nataliya Melnikova
- Group of Postgenomic Studies, Engelhardt Institute of Molecular Biology of RAS, Moscow, Russia
| | - Leonid Uroshlev
- Group of Postgenomic Studies, Engelhardt Institute of Molecular Biology of RAS, Moscow, Russia
- Department of Computational Systems Biology, Vavilov Institute of General Genetics, Moscow, Russia
| | - Sergey Mylnikov
- Department of Genetics, St. Petersburg State University, St. Petersburg, Russia
| | - Alexey Dmitriev
- Group of Postgenomic Studies, Engelhardt Institute of Molecular Biology of RAS, Moscow, Russia
| | - Sergey Plusnin
- Laboratory of Molecular Radiobiology and Gerontology, Institute of Biology of Komi Science Center of RAS, Syktyvkar, Russia
- Ecological Department, Syktyvkar State University, Syktyvkar, Russia
| | - Peter Fedichev
- Laboratory of Genetics of Aging and Longevity, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Quantum Pharmaceuticals, Moscow, Russia
| | - Anna Kudryavtseva
- Group of Postgenomic Studies, Engelhardt Institute of Molecular Biology of RAS, Moscow, Russia
| |
Collapse
|
41
|
McCormick MA, Kennedy BK. Genome-scale studies of aging: challenges and opportunities. Curr Genomics 2013; 13:500-7. [PMID: 23633910 PMCID: PMC3468883 DOI: 10.2174/138920212803251454] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2012] [Revised: 06/08/2012] [Accepted: 07/25/2012] [Indexed: 12/21/2022] Open
Abstract
Whole-genome studies involving a phenotype of interest are increasingly prevalent, in part due to a dramatic increase in speed at which many high throughput technologies can be performed coupled to simultaneous decreases in cost. This type of genome-scale methodology has been applied to the phenotype of lifespan, as well as to whole-transcriptome changes during the aging process or in mutants affecting aging. The value of high throughput discovery-based science in this field is clearly evident, but will it yield a true systems-level understanding of the aging process? Here we review some of this work to date, focusing on recent findings and the unanswered puzzles to which they point. In this context, we also discuss recent technological advances and some of the likely future directions that they portend.
Collapse
|
42
|
Overexpression of fatty-acid-β-oxidation-related genes extends the lifespan of Drosophila melanogaster. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2012; 2012:854502. [PMID: 22997544 PMCID: PMC3446750 DOI: 10.1155/2012/854502] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 07/23/2012] [Accepted: 08/03/2012] [Indexed: 11/21/2022]
Abstract
A better understanding of the aging process is necessary to ensure that the healthcare needs of an aging population are met. With the trend toward increased human life expectancies, identification of candidate genes affecting the regulation of lifespan and its relationship to environmental factors is essential. Through misexpression screening of EP mutant lines, we previously isolated several genes extending lifespan when ubiquitously overexpressed, including the two genes encoding the fatty-acid-binding protein and dodecenoyl-CoA delta-isomerase involved in fatty-acid β-oxidation, which is the main energy resource pathway in eukaryotic cells. In this study, we analyzed flies overexpressing the two main components of fatty-acid β-oxidation, and found that overexpression of fatty-acid-β-oxidation-related genes extended the Drosophila lifespan. Furthermore, we found that the ability of dietary restriction to extend lifespan was reduced by the overexpression of fatty-acid-β-oxidation-related genes. Moreover, the overexpression of fatty-acid-β-oxidation-related genes enhanced stress tolerance to oxidative and starvation stresses and activated the dFOXO signal, indicating translocation to the nucleus and transcriptional activation of the dFOXO target genes. Overall, the results of this study suggest that overexpression of fatty-acid-β-oxidation-related genes extends lifespan in a dietary-restriction-related manner, and that the mechanism of this process may be related to FOXO activation.
Collapse
|