1
|
Li H, Seugnet L. Decoding the nexus: branched-chain amino acids and their connection with sleep, circadian rhythms, and cardiometabolic health. Neural Regen Res 2025; 20:1350-1363. [PMID: 39075896 PMCID: PMC11624887 DOI: 10.4103/nrr.nrr-d-23-02020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/18/2024] [Accepted: 05/12/2024] [Indexed: 07/31/2024] Open
Abstract
The sleep-wake cycle stands as an integrative process essential for sustaining optimal brain function and, either directly or indirectly, overall body health, encompassing metabolic and cardiovascular well-being. Given the heightened metabolic activity of the brain, there exists a considerable demand for nutrients in comparison to other organs. Among these, the branched-chain amino acids, comprising leucine, isoleucine, and valine, display distinctive significance, from their contribution to protein structure to their involvement in overall metabolism, especially in cerebral processes. Among the first amino acids that are released into circulation post-food intake, branched-chain amino acids assume a pivotal role in the regulation of protein synthesis, modulating insulin secretion and the amino acid sensing pathway of target of rapamycin. Branched-chain amino acids are key players in influencing the brain's uptake of monoamine precursors, competing for a shared transporter. Beyond their involvement in protein synthesis, these amino acids contribute to the metabolic cycles of γ-aminobutyric acid and glutamate, as well as energy metabolism. Notably, they impact GABAergic neurons and the excitation/inhibition balance. The rhythmicity of branched-chain amino acids in plasma concentrations, observed over a 24-hour cycle and conserved in rodent models, is under circadian clock control. The mechanisms underlying those rhythms and the physiological consequences of their disruption are not fully understood. Disturbed sleep, obesity, diabetes, and cardiovascular diseases can elevate branched-chain amino acid concentrations or modify their oscillatory dynamics. The mechanisms driving these effects are currently the focal point of ongoing research efforts, since normalizing branched-chain amino acid levels has the ability to alleviate the severity of these pathologies. In this context, the Drosophila model, though underutilized, holds promise in shedding new light on these mechanisms. Initial findings indicate its potential to introduce novel concepts, particularly in elucidating the intricate connections between the circadian clock, sleep/wake, and metabolism. Consequently, the use and transport of branched-chain amino acids emerge as critical components and orchestrators in the web of interactions across multiple organs throughout the sleep/wake cycle. They could represent one of the so far elusive mechanisms connecting sleep patterns to metabolic and cardiovascular health, paving the way for potential therapeutic interventions.
Collapse
Affiliation(s)
- Hui Li
- Department of Neurology, Xijing Hospital, Xi’an, Shaanxi Province, China
| | - Laurent Seugnet
- Centre de Recherche en Neurosciences de Lyon, Integrated Physiology of the Brain Arousal Systems (WAKING), Université Claude Bernard Lyon 1, INSERM U1028, CNRS UMR 5292, Bron, France
| |
Collapse
|
2
|
Huang Y, He M, Zhang J, Cheng S, Cheng X, Chen H, Wu G, Wang F, Zeng S. White Tea Aqueous Extract: A Potential Anti-Aging Agent Against High-Fat Diet-Induced Senescence in Drosophila melanogaster. Foods 2024; 13:4034. [PMID: 39766976 PMCID: PMC11728359 DOI: 10.3390/foods13244034] [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: 11/01/2024] [Revised: 11/28/2024] [Accepted: 12/04/2024] [Indexed: 01/15/2025] Open
Abstract
White tea has been scientifically proven to exhibit positive biological effects in combating chronic diseases, including cancer, metabolic syndrome, etc. Nevertheless, the anti-aging activity and mechanism of white tea on organisms exposed to a high-fat diet remain unexplored. Herein, we prepared a white tea aqueous extract (WTAE) from white peony in Fuding and assessed its in vivo antioxidant and anti-aging effects by employing a Drosophila melanogaster senescence model induced by lard, delving into the underlying molecular mechanisms through which the WTAE contributes to lifespan improvement. Notably, the WTAE significantly extended the lifespan of Drosophila fed a high-fat diet and partially restored the climbing ability of Drosophila on a high-fat diet, accompanied by increased activities of copper-zinc superoxide dismutase, manganese-superoxide dismutase, and catalase and decreased lipid hydroperoxide levels in Drosophila. Furthermore, transcriptomic analysis indicated that the WTAE countered aging triggered by a high-fat diet via activating oxidative phosphorylation, neuroactive ligand-receptor interactions, and more pathways, as well as inhibiting circadian rhythm-fly, protein processing in the endoplasmic reticulum, and more pathways. Our findings suggest that WTAE exhibits excellent inhibitory activity against high-fat diet-induced senescence and holds promising potential as an anti-aging agent that can be further developed.
Collapse
Affiliation(s)
- Yan Huang
- College of Tea and Food Science, Wuyi University, Wuyishan 354300, China; (Y.H.); (M.H.); (J.Z.); (S.C.); (X.C.)
| | - Miaoyuan He
- College of Tea and Food Science, Wuyi University, Wuyishan 354300, China; (Y.H.); (M.H.); (J.Z.); (S.C.); (X.C.)
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China;
- Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Jianming Zhang
- College of Tea and Food Science, Wuyi University, Wuyishan 354300, China; (Y.H.); (M.H.); (J.Z.); (S.C.); (X.C.)
| | - Shilong Cheng
- College of Tea and Food Science, Wuyi University, Wuyishan 354300, China; (Y.H.); (M.H.); (J.Z.); (S.C.); (X.C.)
| | - Xi Cheng
- College of Tea and Food Science, Wuyi University, Wuyishan 354300, China; (Y.H.); (M.H.); (J.Z.); (S.C.); (X.C.)
| | - Haoran Chen
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China;
- Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Guangheng Wu
- Fujian Provincial Key Laboratory of Eco-Industrial Green Technology, College of Ecology and Resources Engineering, Wuyi University, Wuyishan 354300, China;
| | - Fang Wang
- College of Food Science and Technology, Ningde Normal University, Ningde 352100, China;
| | - Shaoxiao Zeng
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China;
- Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| |
Collapse
|
3
|
Haynes PR, Pyfrom ES, Li Y, Stein C, Cuddapah VA, Jacobs JA, Yue Z, Sehgal A. A neuron-glia lipid metabolic cycle couples daily sleep to mitochondrial homeostasis. Nat Neurosci 2024; 27:666-678. [PMID: 38360946 PMCID: PMC11001586 DOI: 10.1038/s41593-023-01568-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 12/29/2023] [Indexed: 02/17/2024]
Abstract
Sleep is thought to be restorative to brain energy homeostasis, but it is not clear how this is achieved. We show here that Drosophila glia exhibit a daily cycle of glial mitochondrial oxidation and lipid accumulation that is dependent on prior wake and requires the Drosophila APOE orthologs NLaz and GLaz, which mediate neuron-glia lipid transfer. In turn, a full night of sleep is required for glial lipid clearance, mitochondrial oxidative recovery and maximal neuronal mitophagy. Knockdown of neuronal NLaz causes oxidative stress to accumulate in neurons, and the neuronal mitochondrial integrity protein, Drp1, is required for daily glial lipid accumulation. These data suggest that neurons avoid accumulation of oxidative mitochondrial damage during wake by using mitophagy and passing damage to glia in the form of lipids. We propose that a mitochondrial lipid metabolic cycle between neurons and glia reflects a fundamental function of sleep relevant for brain energy homeostasis.
Collapse
Affiliation(s)
- Paula R Haynes
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Elana S Pyfrom
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Yongjun Li
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Carly Stein
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Howard Hughes Medical Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Vishnu Anand Cuddapah
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - Jack A Jacobs
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Zhifeng Yue
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Howard Hughes Medical Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Amita Sehgal
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA.
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
- Howard Hughes Medical Institute, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
4
|
Yang X, Xiaoping W, Nan D, Jian Z, Xiaofeng L, Liwei Y, Zhao M, Wang F. Proteomic and bioinformatic analysis of human endometrium from polycystic ovarian syndrome with and without insulin resistance. Gynecol Endocrinol 2023; 39:2173948. [PMID: 36750132 DOI: 10.1080/09513590.2023.2173948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Objective: The aim of this study was to investigate the endometrial proteomic profiles of patients with polycystic ovary syndrome (PCOS) with and without insulin resistance (IR). Method of Study: We collected 40 endometrial samples, including PCOS-IR (n = 21), PCOS-non-IR (n = 12), and control (n = 7). Data-independent acquisition (DIA)-based proteomics method is used to identify the expressed proteins among the three groups. The correlation between pregnancy outcomes and identified proteins was analyzed by Lasso regression. Results: A total of 5331 proteins were identified, while 275 proteins were differentially expressed in the PCOS vs. control group and 215 proteins were differentially expressed in the PCOS-IR vs. PCOS-non-IR group. Platelet degranulation, neutrophil degranulation, and very long-chain fatty acid catabolic processes have been found to play important roles in the endometrium of patients with PCOS-IR. Lasso regression analysis found that ACTR1A, TSC22D2, CKB, ABRAXAS2, and TAGLN2 were associated with miscarriage in patients with PCOS. ACTR1A and CKB were higher in the PCOS-IR group and were positively correlated with HOMA-IR (p < .05). Conclusion: In this study, a panel of proteins was found to be differently expressed in the endometrium. ACTR1A and CKB may be considered as PCOS-IR candidate biomarkers.
Collapse
Affiliation(s)
- Xin Yang
- Reproductive Medicine Center, Second Hospital of Lanzhou University, Lanzhou, China
| | - Wang Xiaoping
- Reproductive Medicine Center, Second Hospital of Lanzhou University, Lanzhou, China
| | - Ding Nan
- Reproductive Medicine Center, Second Hospital of Lanzhou University, Lanzhou, China
| | - Zhang Jian
- Reproductive Medicine Center, Second Hospital of Lanzhou University, Lanzhou, China
| | - Li Xiaofeng
- Reproductive Medicine Center, Second Hospital of Lanzhou University, Lanzhou, China
| | - Yuan Liwei
- Reproductive Medicine Center, Second Hospital of Lanzhou University, Lanzhou, China
| | - Mengni Zhao
- Reproductive Medicine Center, Second Hospital of Lanzhou University, Lanzhou, China
| | - Fang Wang
- Reproductive Medicine Center, Second Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
5
|
Diaz AV, Stephenson D, Nemkov T, D’Alessandro A, Reis T. Spenito-dependent metabolic sexual dimorphism intrinsic to fat storage cells. Genetics 2023; 225:iyad164. [PMID: 37738330 PMCID: PMC10627258 DOI: 10.1093/genetics/iyad164] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/16/2023] [Indexed: 09/24/2023] Open
Abstract
Metabolism in males and females is distinct. Differences are usually linked to sexual reproduction, with circulating signals (e.g. hormones) playing major roles. In contrast, sex differences prior to sexual maturity and intrinsic to individual metabolic tissues are less understood. We analyzed Drosophila melanogaster larvae and find that males store more fat than females, the opposite of the sexual dimorphism in adults. We show that metabolic differences are intrinsic to the major fat storage tissue, including many differences in the expression of metabolic genes. Our previous work identified fat storage roles for Spenito (Nito), a conserved RNA-binding protein and regulator of sex determination. Nito knockdown specifically in the fat storage tissue abolished fat differences between males and females. We further show that Nito is required for sex-specific expression of the master regulator of sex determination, Sex-lethal (Sxl). "Feminization" of fat storage cells via tissue-specific overexpression of a Sxl target gene made larvae lean, reduced the fat differences between males and females, and induced female-like metabolic gene expression. Altogether, this study supports a model in which Nito autonomously controls sexual dimorphisms and differential expression of metabolic genes in fat cells in part through its regulation of the sex determination pathway.
Collapse
Affiliation(s)
- Arely V Diaz
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Daniel Stephenson
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Tânia Reis
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
6
|
Gerstner JR, Flores CC, Lefton M, Rogers B, Davis CJ. FABP7: a glial integrator of sleep, circadian rhythms, plasticity, and metabolic function. Front Syst Neurosci 2023; 17:1212213. [PMID: 37404868 PMCID: PMC10315501 DOI: 10.3389/fnsys.2023.1212213] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/02/2023] [Indexed: 07/06/2023] Open
Abstract
Sleep and circadian rhythms are observed broadly throughout animal phyla and influence neural plasticity and cognitive function. However, the few phylogenetically conserved cellular and molecular pathways that are implicated in these processes are largely focused on neuronal cells. Research on these topics has traditionally segregated sleep homeostatic behavior from circadian rest-activity rhythms. Here we posit an alternative perspective, whereby mechanisms underlying the integration of sleep and circadian rhythms that affect behavioral state, plasticity, and cognition reside within glial cells. The brain-type fatty acid binding protein, FABP7, is part of a larger family of lipid chaperone proteins that regulate the subcellular trafficking of fatty acids for a wide range of cellular functions, including gene expression, growth, survival, inflammation, and metabolism. FABP7 is enriched in glial cells of the central nervous system and has been shown to be a clock-controlled gene implicated in sleep/wake regulation and cognitive processing. FABP7 is known to affect gene transcription, cellular outgrowth, and its subcellular localization in the fine perisynaptic astrocytic processes (PAPs) varies based on time-of-day. Future studies determining the effects of FABP7 on behavioral state- and circadian-dependent plasticity and cognitive processes, in addition to functional consequences on cellular and molecular mechanisms related to neural-glial interactions, lipid storage, and blood brain barrier integrity will be important for our knowledge of basic sleep function. Given the comorbidity of sleep disturbance with neurological disorders, these studies will also be important for our understanding of the etiology and pathophysiology of how these diseases affect or are affected by sleep.
Collapse
Affiliation(s)
- Jason R. Gerstner
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Steve Gleason Institute for Neuroscience, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Carlos C. Flores
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Micah Lefton
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Brooke Rogers
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Christopher J. Davis
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Steve Gleason Institute for Neuroscience, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| |
Collapse
|
7
|
Li Y, Zhou X, Cheng C, Ding G, Zhao P, Tan K, Chen L, Perrimon N, Veenstra JA, Zhang L, Song W. Gut AstA mediates sleep deprivation-induced energy wasting in Drosophila. Cell Discov 2023; 9:49. [PMID: 37221172 DOI: 10.1038/s41421-023-00541-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 03/13/2023] [Indexed: 05/25/2023] Open
Abstract
Severe sleep deprivation (SD) has been highly associated with systemic energy wasting, such as lipid loss and glycogen depletion. Despite immune dysregulation and neurotoxicity observed in SD animals, whether and how the gut-secreted hormones participate in SD-induced disruption of energy homeostasis remains largely unknown. Using Drosophila as a conserved model organism, we characterize that production of intestinal Allatostatin A (AstA), a major gut-peptide hormone, is robustly increased in adult flies bearing severe SD. Interestingly, the removal of AstA production in the gut using specific drivers significantly improves lipid loss and glycogen depletion in SD flies without affecting sleep homeostasis. We reveal the molecular mechanisms whereby gut AstA promotes the release of an adipokinetic hormone (Akh), an insulin counter-regulatory hormone functionally equivalent to mammalian glucagon, to mobilize systemic energy reserves by remotely targeting its receptor AstA-R2 in Akh-producing cells. Similar regulation of glucagon secretion and energy wasting by AstA/galanin is also observed in SD mice. Further, integrating single-cell RNA sequencing and genetic validation, we uncover that severe SD results in ROS accumulation in the gut to augment AstA production via TrpA1. Altogether, our results demonstrate the essential roles of the gut-peptide hormone AstA in mediating SD-associated energy wasting.
Collapse
Affiliation(s)
- Yingge Li
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Xiaoya Zhou
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Chen Cheng
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Guangming Ding
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Peng Zhao
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Kai Tan
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
| | - Lixia Chen
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Norbert Perrimon
- Department of Genetics, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - Jan A Veenstra
- INCIA, UMR 5287 CNRS, University of Bordeaux, Talence, France
| | - Luoying Zhang
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Song
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China.
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
8
|
Maruoka N, Makino T, Urabe J. RNA-seq analysis to identify genes related to resting egg production of panarctic Daphnia pulex. BMC Genomics 2023; 24:262. [PMID: 37198540 PMCID: PMC10190107 DOI: 10.1186/s12864-023-09369-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/08/2023] [Indexed: 05/19/2023] Open
Abstract
BACKGROUND The genus Daphnia switches its reproductive mode from subitaneous egg production to resting egg production in response to environmental stimuli. Although this life history trait is essential for surviving unsuitable environments, the molecular mechanism of resting egg production is little understood. In this study, we examined genes related to induction of resting egg production using two genotypes of panarctic Daphnia pulex, the JPN1 and JPN2 lineages, which differ genetically in the frequency of resting egg production. We reared these genotypes under high and low food levels. At the high food level, individuals of both genotypes continually produced subitaneous eggs, whereas at the low food level, only the JPN2 genotype produced resting eggs. Then, we performed RNA-seq analysis on specimens of three instars, including before and after egg production. RESULTS These results showed that expressed genes differed significantly between individuals grown under high and low food levels and among individuals of different instars and genotypes. Among these differentially expressed genes (DEGs), we found 16 that changed expression level before resting egg production. Some of these genes showed high-level expression only before resting egg production and one gene was an ortholog of bubblegum (bgm), which is reportedly up-regulated before diapause in bumblebees. According to gene ontology (GO) enrichment analysis, one GO term annotated as long-chain fatty acid biosynthetic process was enriched among these 16 genes. In addition, GO terms related to glycometabolism were enriched among down-regulated genes of individuals holding resting eggs, compared to those before resting egg production. CONCLUSIONS We found candidate genes highly expressed only before resting egg production. Although functions of candidate genes found in this study have not been reported previously in Daphnia, catabolism of long-chain fatty acids and metabolism of glycerates are related to diapause in other organisms. Thus, it is highly probable that candidate genes identified in this study are related to the molecular mechanism regulating resting egg production in Daphnia.
Collapse
Affiliation(s)
- Natsumi Maruoka
- Graduate School of Life sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, 980-8578, 022-795-6686, Miyagi, Japan.
- Center for Bioscience Research and Education, Utsunomiya University, 350 Mine-machi, Utsunomiya, 321-8505, 028-649-5129, Tochigi, Japan.
| | - Takashi Makino
- Graduate School of Life sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, 980-8578, 022-795-6686, Miyagi, Japan
| | - Jotaro Urabe
- Graduate School of Life sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, 980-8578, 022-795-6686, Miyagi, Japan
| |
Collapse
|
9
|
Bedont JL, Kolesnik A, Pivarshev P, Malik D, Hsu CT, Weljie A, Sehgal A. Chronic sleep loss sensitizes Drosophila melanogaster to nitrogen stress. Curr Biol 2023; 33:1613-1623.e5. [PMID: 36965479 PMCID: PMC10133188 DOI: 10.1016/j.cub.2023.03.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/22/2022] [Accepted: 03/03/2023] [Indexed: 03/27/2023]
Abstract
Chronic sleep loss profoundly impacts metabolic health and shortens lifespan, but studies of the mechanisms involved have focused largely on acute sleep deprivation.1,2 To identify metabolic consequences of chronically reduced sleep, we conducted unbiased metabolomics on heads of three adult Drosophila short-sleeping mutants with very different mechanisms of sleep loss: fumin (fmn), redeye (rye), and sleepless (sss).3,4,5,6,7 Common features included elevated ornithine and polyamines, with lipid, acyl-carnitine, and TCA cycle changes suggesting mitochondrial dysfunction. Studies of excretion demonstrate inefficient nitrogen elimination in adult sleep mutants, likely contributing to their polyamine accumulation. Increasing levels of polyamines, particularly putrescine, promote sleep in control flies but poison sleep mutants. This parallels the broadly enhanced toxicity of high dietary nitrogen load from protein in chronically sleep-restricted Drosophila, including both sleep mutants and flies with hyper-activated wake-promoting neurons. Together, our results implicate nitrogen stress as a novel mechanism linking chronic sleep loss to adverse health outcomes-and perhaps for linking food and sleep homeostasis at the cellular level in healthy organisms.
Collapse
Affiliation(s)
- Joseph L Bedont
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Anna Kolesnik
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Pavel Pivarshev
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Dania Malik
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Cynthia T Hsu
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Aalim Weljie
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Amita Sehgal
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA; Howard Hughes Medical Institute, 4000 Jones Bridge Road, Chevy Chase, MD 20815, USA.
| |
Collapse
|
10
|
Mariano V, Kanellopoulos AK, Aiello G, Lo AC, Legius E, Achsel T, Bagni C. SREBP modulates the NADP +/NADPH cycle to control night sleep in Drosophila. Nat Commun 2023; 14:763. [PMID: 36808152 PMCID: PMC9941135 DOI: 10.1038/s41467-022-35577-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 12/12/2022] [Indexed: 02/22/2023] Open
Abstract
Sleep behavior is conserved throughout evolution, and sleep disturbances are a frequent comorbidity of neuropsychiatric disorders. However, the molecular basis underlying sleep dysfunctions in neurological diseases remains elusive. Using a model for neurodevelopmental disorders (NDDs), the Drosophila Cytoplasmic FMR1 interacting protein haploinsufficiency (Cyfip85.1/+), we identify a mechanism modulating sleep homeostasis. We show that increased activity of the sterol regulatory element-binding protein (SREBP) in Cyfip85.1/+ flies induces an increase in the transcription of wakefulness-associated genes, such as the malic enzyme (Men), causing a disturbance in the daily NADP+/NADPH ratio oscillations and reducing sleep pressure at the night-time onset. Reduction in SREBP or Men activity in Cyfip85.1/+ flies enhances the NADP+/NADPH ratio and rescues the sleep deficits, indicating that SREBP and Men are causative for the sleep deficits in Cyfip heterozygous flies. This work suggests modulation of the SREBP metabolic axis as a new avenue worth exploring for its therapeutic potential in sleep disorders.
Collapse
Affiliation(s)
- Vittoria Mariano
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, 1005, Switzerland.,Department of Human Genetics, KU Leuven, Leuven, 3000, Belgium
| | | | - Giuseppe Aiello
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, 1005, Switzerland
| | - Adrian C Lo
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, 1005, Switzerland
| | - Eric Legius
- Department of Human Genetics, KU Leuven, Leuven, 3000, Belgium
| | - Tilmann Achsel
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, 1005, Switzerland
| | - Claudia Bagni
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, 1005, Switzerland. .,Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, 00133, Italy.
| |
Collapse
|
11
|
Diaz AV, Matheny T, Stephenson D, Nemkov T, D’Alessandro A, Reis T. Spenito-dependent metabolic sexual dimorphism intrinsic to fat storage cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.17.528952. [PMID: 36824729 PMCID: PMC9949119 DOI: 10.1101/2023.02.17.528952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Metabolism in males and females is distinct. Differences are usually linked to sexual reproduction, with circulating signals (e.g. hormones) playing major roles. By contrast, sex differences prior to sexual maturity and intrinsic to individual metabolic tissues are less understood. We analyzed Drosophila melanogaster larvae and find that males store more fat than females, the opposite of the sexual dimorphism in adults. We show that metabolic differences are intrinsic to the major fat storage tissue, including many differences in the expression of metabolic genes. Our previous work identified fat storage roles for Spenito (Nito), a conserved RNA-binding protein and regulator of sex determination. Nito knockdown specifically in the fat storage tissue abolished fat differences between males and females. We further show that Nito is required for sex-specific expression of the master regulator of sex determination, Sex-lethal (Sxl). "Feminization" of fat storage cells via tissue-specific overexpression of a Sxl target gene made larvae lean, reduced the fat differences between males and females, and induced female-like metabolic gene expression. Altogether, this study supports a model in which Nito autonomously controls sexual dimorphisms and differential expression of metabolic genes in fat cells in part through its regulation of the sex determination pathway.
Collapse
Affiliation(s)
- Arely V. Diaz
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Tyler Matheny
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Daniel Stephenson
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Tânia Reis
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| |
Collapse
|
12
|
Dissel S, Klose MK, van Swinderen B, Cao L, Ford M, Periandri EM, Jones JD, Li Z, Shaw PJ. Sleep-promoting neurons remodel their response properties to calibrate sleep drive with environmental demands. PLoS Biol 2022; 20:e3001797. [PMID: 36173939 PMCID: PMC9521806 DOI: 10.1371/journal.pbio.3001797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 08/16/2022] [Indexed: 01/29/2023] Open
Abstract
Falling asleep at the wrong time can place an individual at risk of immediate physical harm. However, not sleeping degrades cognition and adaptive behavior. To understand how animals match sleep need with environmental demands, we used live-brain imaging to examine the physiological response properties of the dorsal fan-shaped body (dFB) following interventions that modify sleep (sleep deprivation, starvation, time-restricted feeding, memory consolidation) in Drosophila. We report that dFB neurons change their physiological response-properties to dopamine (DA) and allatostatin-A (AstA) in response to different types of waking. That is, dFB neurons are not simply passive components of a hard-wired circuit. Rather, the dFB neurons intrinsically regulate their response to the activity from upstream circuits. Finally, we show that the dFB appears to contain a memory trace of prior exposure to metabolic challenges induced by starvation or time-restricted feeding. Together, these data highlight that the sleep homeostat is plastic and suggests an underlying mechanism.
Collapse
Affiliation(s)
- Stephane Dissel
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
- * E-mail: (SD); (PJS)
| | - Markus K. Klose
- University of Pittsburgh School of Medicine, Department of Pharmacology & Chemical Biology, Pittsburgh, Pennsylvania, United States of America
| | - Bruno van Swinderen
- Queensland Brain Institute, The University of Queensland, St Lucia, Australia
| | - Lijuan Cao
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Melanie Ford
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Erica M. Periandri
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Joseph D. Jones
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Zhaoyi Li
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Paul J. Shaw
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail: (SD); (PJS)
| |
Collapse
|
13
|
Ukita Y, Okumura M, Chihara T. Ubiquitin proteasome system in circadian rhythm and sleep homeostasis: Lessons from Drosophila. Genes Cells 2022; 27:381-391. [PMID: 35438236 PMCID: PMC9322287 DOI: 10.1111/gtc.12935] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/27/2022] [Accepted: 03/28/2022] [Indexed: 11/30/2022]
Abstract
Sleep is regulated by two main processes: the circadian clock and sleep homeostasis. Circadian rhythms have been well studied at the molecular level. In the Drosophila circadian clock neurons, the core clock proteins are precisely regulated by post-translational modifications and degraded via the ubiquitin-proteasome system (UPS). Sleep homeostasis, however, is less understood; nevertheless, recent reports suggest that proteasome-mediated degradation of core clock proteins or synaptic proteins contributes to the regulation of sleep amount. Here, we review the molecular mechanism of the UPS and summarize the role of protein degradation in the regulation of circadian clock and homeostatic sleep in Drosophila. Moreover, we discuss the potential interaction between circadian clock and homeostatic sleep regulation with a prime focus on E3 ubiquitin ligases.
Collapse
Affiliation(s)
- Yumiko Ukita
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Misako Okumura
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan.,Program of Basic Biology, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Takahiro Chihara
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan.,Program of Basic Biology, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
14
|
Weiss JT, Donlea JM. Roles for Sleep in Neural and Behavioral Plasticity: Reviewing Variation in the Consequences of Sleep Loss. Front Behav Neurosci 2022; 15:777799. [PMID: 35126067 PMCID: PMC8810646 DOI: 10.3389/fnbeh.2021.777799] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/16/2021] [Indexed: 12/13/2022] Open
Abstract
Sleep is a vital physiological state that has been broadly conserved across the evolution of animal species. While the precise functions of sleep remain poorly understood, a large body of research has examined the negative consequences of sleep loss on neural and behavioral plasticity. While sleep disruption generally results in degraded neural plasticity and cognitive function, the impact of sleep loss can vary widely with age, between individuals, and across physiological contexts. Additionally, several recent studies indicate that sleep loss differentially impacts distinct neuronal populations within memory-encoding circuitry. These findings indicate that the negative consequences of sleep loss are not universally shared, and that identifying conditions that influence the resilience of an organism (or neuron type) to sleep loss might open future opportunities to examine sleep's core functions in the brain. Here, we discuss the functional roles for sleep in adaptive plasticity and review factors that can contribute to individual variations in sleep behavior and responses to sleep loss.
Collapse
Affiliation(s)
- Jacqueline T. Weiss
- Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
- Neuroscience Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jeffrey M. Donlea
- Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
- *Correspondence: Jeffrey M. Donlea
| |
Collapse
|
15
|
Zarubin M, Gangapshev A, Gavriljuk Y, Kazalov V, Kravchenko E. First transcriptome profiling of D. melanogaster after development in a deep underground low radiation background laboratory. PLoS One 2021; 16:e0255066. [PMID: 34351964 PMCID: PMC8341612 DOI: 10.1371/journal.pone.0255066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 07/08/2021] [Indexed: 11/19/2022] Open
Abstract
Natural background radiation is a permanent multicomponent factor. It has an influence on biological organisms, but effects of its deprivation still remain unclear. The aim of our work was to study for the first time responses of D. melanogaster to conditions of the Deep Underground Low-Background Laboratory DULB-4900 (BNO, INR, RAS, Russia) at the transcriptome level by RNA-seq profiling. Overall 77 transcripts demonstrated differential abundance between flies exposed to low and natural background radiation. Enriched biological process functional categories were established for all genes with differential expression. The results showed down-regulation of primary metabolic processes and up-regulation of both the immune system process and the response to stimuli. The comparative analysis of our data and publicly available transcriptome data on D. melanogaster exposed to low and high doses of ionizing radiation did not reveal common DEGs in them. We hypothesize that the observed changes in gene expression can be explained by the influence of the underground conditions in DULB-4900, in particular, by the lack of stimuli. Thus, our study challenges the validity of the LNT model for the region of background radiation doses below a certain level (~16.4 nGy h-1) and the presence of a dose threshold for D. melanogaster.
Collapse
Affiliation(s)
| | - Albert Gangapshev
- Institute for Nuclear Research, Russian Academy of Sciences, Moscow, Russia
| | - Yuri Gavriljuk
- Institute for Nuclear Research, Russian Academy of Sciences, Moscow, Russia
| | - Vladimir Kazalov
- Institute for Nuclear Research, Russian Academy of Sciences, Moscow, Russia
| | - Elena Kravchenko
- Joint Institute for Nuclear Research, DLNP, Dubna, Russia
- * E-mail:
| |
Collapse
|
16
|
Klose MK, Shaw PJ. Sleep drive reconfigures wake-promoting clock circuitry to regulate adaptive behavior. PLoS Biol 2021; 19:e3001324. [PMID: 34191802 PMCID: PMC8277072 DOI: 10.1371/journal.pbio.3001324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 07/13/2021] [Accepted: 06/15/2021] [Indexed: 11/19/2022] Open
Abstract
Circadian rhythms help animals synchronize motivated behaviors to match environmental demands. Recent evidence indicates that clock neurons influence the timing of behavior by differentially altering the activity of a distributed network of downstream neurons. Downstream circuits can be remodeled by Hebbian plasticity, synaptic scaling, and, under some circumstances, activity-dependent addition of cell surface receptors; the role of this receptor respecification phenomena is not well studied. We demonstrate that high sleep pressure quickly reprograms the wake-promoting large ventrolateral clock neurons to express the pigment dispersing factor receptor (PDFR). The addition of this signaling input into the circuit is associated with increased waking and early mating success. The respecification of PDFR in both young and adult large ventrolateral neurons requires 2 dopamine (DA) receptors and activation of the transcriptional regulator nejire (cAMP response element-binding protein [CREBBP]). These data identify receptor respecification as an important mechanism to sculpt circuit function to match sleep levels with demand.
Collapse
Affiliation(s)
- Markus K. Klose
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Paul J. Shaw
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
17
|
Epstein NR, Saez K, Polat A, Davis SR, Aardema ML. The urban-adapted underground mosquito Culex pipiens form molestus maintains exogenously influenceable circadian rhythms. J Exp Biol 2021; 224:268332. [PMID: 34027981 DOI: 10.1242/jeb.242231] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/12/2021] [Indexed: 01/01/2023]
Abstract
Genes known to affect circadian rhythms (i.e. 'clock genes') also influence the photoperiodic induction of overwintering reproductive diapause in the northern house mosquito, Culex pipiens f. pipiens. This suggests that molecular changes in one or more clock genes could contribute to the inability to diapause in a second form of this mosquito, Culex pipiens f. molestus. Temperate populations of Cx. pipiens f. molestus inhabit underground locations generally devoid of predictable photoperiods. For this reason, there could be limited fitness consequences if the hypothesized molecular changes to its clock genes also eliminated this mosquito's ability to regulate circadian rhythms in response to photoperiod variation. Here, we demonstrate that in contrast to this prediction, underground derived Cx. pipiens f. molestus retain exogenously influenceable circadian rhythms. Nonetheless, our genetic analyses indicate that the gene Helicase domino (dom) has a nine-nucleotide, in-frame deletion specific to Cx. pipiens f. molestus. Previous work has shown that splice variants in this gene differentially influence circadian behavior in Drosophila melanogaster. We also find derived, non-synonymous single nucleotide polymorphisms (SNPs) in eight genes that may also affect circadian rhythms and/or diapause induction in Cx. pipiens f. molestus. Finally, four putative circadian genes were found to have no quantifiable expression during any examined life stage, suggesting potential regulatory effects. Collectively, our findings indicate that the distinct, but molecularly interconnected life-history traits of diapause induction and circadian rhythms are decoupled in Cx. pipiens f. molestus and suggest this taxon may be a valuable tool for exploring exogenously influenced phenotypes in mosquitoes more broadly.
Collapse
Affiliation(s)
- Natalie R Epstein
- Department of Biology, Montclair State University, 1 Normal Avenue, Montclair, NJ 07043, USA
| | - Kevin Saez
- Department of Biology, Montclair State University, 1 Normal Avenue, Montclair, NJ 07043, USA
| | - Asya Polat
- Department of Biology, Montclair State University, 1 Normal Avenue, Montclair, NJ 07043, USA
| | - Steven R Davis
- Division of Invertebrate Zoology, American Museum of Natural History, 200 Central Park West New York, NY 10024-5102, USA
| | - Matthew L Aardema
- Department of Biology, Montclair State University, 1 Normal Avenue, Montclair, NJ 07043, USA.,Sackler Institute for Comparative Genomics , American Museum of Natural History, 200 Central Park West, New York, NY 10024-5102, USA
| |
Collapse
|
18
|
Barroso IG, Cardoso C, Ferreira C, Terra WR. Transcriptomic and proteomic analysis of the underlying mechanisms of digestion of triacylglycerols and phosphatides and absorption and fate of fatty acids along the midgut of Musca domestica. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2021; 39:100826. [PMID: 33839527 DOI: 10.1016/j.cbd.2021.100826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 03/23/2021] [Accepted: 03/23/2021] [Indexed: 11/24/2022]
Abstract
Most dietary lipids are triacylglycerols (TAGs) and phosphatides that are digested by TAG lipases and phospholipases (PLIPs), respectively, originating fatty acids (FA). The genome of Musca domestica has genes coding for phospholipases A1 (1PLIP), A2 (2PLIP), B (BPLIP), and acid lipases (ALIP), as for proteins involved in activation, binding, and metabolism of FA, which expression in the larval midgut was evaluated by RNA-seq. Some of the codified proteins were identified in midgut microvillar-enriched membrane by proteomics. 1PLIPs are the most expressed PLIPs, mainly in anterior midgut whereas 2PLIPs, and BPLIP in middle and posterior midgut, and ALIPs between middle and posterior regions. Absorption of FAs is putatively accomplished by proteins involved in FA activation (acyl-CoA synthetases) found in microvillar-enriched membrane preparations. Furthermore, FA uptake could be enhanced by proteins that bind FAs (FA-binding proteins) and its activated form (acyl-CoA binding proteins) mainly expressed in posterior midgut. Activated FAs could have different fates: synthesis of diacylglycerol (DAG) and TAG through monoacylglycerol and glycerol-3-phosphate pathways; synthesis of phosphatides; energy source by β-oxidation. Most genes coding for enzymes of those routes is expressed mainly at the end of posterior midgut. Data suggest that phosphatides are digested in anterior midgut by Md1PLIPs, releasing lysophosphatides that emulsify fats to be digested by MdALIPs in the middle and posterior midgut. Most resulting FAs is absorbed in the posterior midgut, where they follow the synthesis of DAG, TAG, and phosphatides or are oxidized along the midgut, mainly in highly metabolic middle and posterior midgut regions.
Collapse
Affiliation(s)
- Ignacio G Barroso
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil
| | - Christiane Cardoso
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil
| | - Clelia Ferreira
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil
| | - Walter R Terra
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, 05508-000 São Paulo, Brazil.
| |
Collapse
|
19
|
Dissel S, Morgan E, Duong V, Chan D, van Swinderen B, Shaw P, Zars T. Sleep restores place learning to the adenylyl cyclase mutant rutabaga. J Neurogenet 2020; 34:83-91. [PMID: 31997683 PMCID: PMC7250152 DOI: 10.1080/01677063.2020.1720674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 01/20/2020] [Indexed: 01/29/2023]
Abstract
Sleep plays an important role in regulating plasticity. In Drosophila, the relationship between sleep and learning and memory has primarily focused on mushroom body dependent operant-learning assays such as aversive phototaxic suppression and courtship conditioning. In this study, sleep was increased in the classic mutant rutabaga (rut2080) and dunce (dnc1) by feeding them the GABA-A agonist gaboxadol (Gab). Performance was evaluated in each mutant in response to social enrichment and place learning, tasks that do not require the mushroom body. Gab-induced sleep did not restore behavioral plasticity to either rut2080 or dnc1 mutants following social enrichment. However, increased sleep restored place learning to rut2080 mutants. These data extend the positive effects of enhanced sleep to place learning and highlight the utility of Gab for elucidating the beneficial effects of sleep on brain functioning.
Collapse
Affiliation(s)
- Stephane Dissel
- School of Biological and Chemical Sciences, University of Missouri-Kansas City, 5007 Rockhill Rd, Kansas City, MO 64110
- Department of Neuroscience, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, U.S.A
| | - Ellen Morgan
- Department of Neuroscience, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, U.S.A
| | - Vincent Duong
- Department of Neuroscience, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, U.S.A
| | - Dorothy Chan
- Department of Neuroscience, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, U.S.A
| | - Bruno van Swinderen
- The Queensland Brain Institute, University of Queensland, Brisbane Qld 4072 Australia
| | - Paul Shaw
- Department of Neuroscience, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, U.S.A
| | - Troy Zars
- Division of Biological Sciences, University of Missouri, Columbia, Missouri, United States of America
| |
Collapse
|
20
|
Aboudhiaf S, Alves G, Parrot S, Amri M, Simonnet MM, Grosjean Y, Manière G, Seugnet L. LAT1-like transporters regulate dopaminergic transmission and sleep in Drosophila. Sleep 2019; 41:5054580. [PMID: 30016498 DOI: 10.1093/sleep/zsy137] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Indexed: 02/06/2023] Open
Abstract
Amino acid transporters are involved in functions reportedly linked to the sleep/wake cycle: neurotransmitter synthesis and recycling, the regulation of synaptic strength, protein synthesis, and energy metabolism. In addition, the existence of bidirectional relationships among extracellular content, transport systems, and sleep/wake states is receiving emerging support. Nevertheless, the connection between amino acid transport and sleep/wake regulation remains elusive. To address this question, we used Drosophila melanogaster and investigated the role of LAT1 (large neutral amino acid transporter 1) transporters. We show that the two Drosophila LAT1-like transporters: Juvenile hormone Inducible-21 and minidiscs (Mnd) are required in dopaminergic neurons for sleep/wake regulation. Down-regulating either gene in dopaminergic neurons resulted in higher daily sleep and longer sleep bout duration during the night, suggesting a defect in dopaminergic transmission. Since LAT1 transporters can mediate in mammals the uptake of L-DOPA, a precursor of dopamine, we assessed amino acid transport efficiency by L-DOPA feeding. We find that downregulation of JhI-21, but not Mnd, reduced the sensitivity to L-DOPA as measured by sleep loss. JhI-21 downregulation also attenuated the sleep loss induced by continuous activation of dopaminergic neurons. Since LAT1 transporters are known to regulate target of rapamycin (TOR) signaling, we investigated the role of this amino acid sensing pathway in dopaminergic neurons. Consistently, we report that TOR activity in dopaminergic neurons modulates sleep/wake states. Altogether, this study provides evidence that LAT1-mediated amino acid transport in dopaminergic neurons is playing a significant role in sleep/wake regulation and is providing several entry points to elucidate the role of nutrients such as amino acids in sleep/wake regulation.
Collapse
Affiliation(s)
- Sami Aboudhiaf
- Centre de Recherche en Neurosciences de Lyon - INSERM U1028 - CNRS UMR 5292 - Université de Lyon - WAKING group, Lyon, France.,Université de Tunis El Manar, Faculté des Sciences de Tunis, UR/11ES09 Laboratory of Functional Neurophysiology and Pathology, Tunis, Tunisia
| | - Georges Alves
- Centre des Sciences du Goût et de l'Alimentation, Agrosup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, Dijon, France
| | - Sandrine Parrot
- Centre de Recherche en Neurosciences de Lyon - INSERM U1028 - UMR 5292 - Université de Lyon - NeuroDialytics Unit, Lyon, France
| | - Mohamed Amri
- Université de Tunis El Manar, Faculté des Sciences de Tunis, UR/11ES09 Laboratory of Functional Neurophysiology and Pathology, Tunis, Tunisia
| | - Mégane M Simonnet
- Centre des Sciences du Goût et de l'Alimentation, Agrosup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, Dijon, France
| | - Yael Grosjean
- Centre des Sciences du Goût et de l'Alimentation, Agrosup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, Dijon, France
| | - Gérard Manière
- Centre des Sciences du Goût et de l'Alimentation, Agrosup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, Dijon, France
| | - Laurent Seugnet
- Centre de Recherche en Neurosciences de Lyon - INSERM U1028 - CNRS UMR 5292 - Université de Lyon - WAKING group, Lyon, France
| |
Collapse
|
21
|
Moskalev A, Guvatova Z, Shaposhnikov M, Lashmanova E, Proshkina E, Koval L, Zhavoronkov A, Krasnov G, Kudryavtseva A. The Neuronal Overexpression of Gclc in Drosophila melanogaster Induces Life Extension With Longevity-Associated Transcriptomic Changes in the Thorax. Front Genet 2019; 10:149. [PMID: 30891062 PMCID: PMC6411687 DOI: 10.3389/fgene.2019.00149] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 02/12/2019] [Indexed: 01/24/2023] Open
Abstract
Some effects of aging in animals are tissue-specific. In D. melanogaster neuronal overexpression of Gclc increases lifespan and improves certain physiological parameters associated with health benefits such as locomotor activity, circadian rhythmicity, and stress resistance. Our previous transcriptomic analyses of Drosophila heads, primarily composed of neuronal tissue, revealed significant changes in expression levels of genes involved in aging-related signaling pathways (Jak-STAT, MAPK, FOXO, Notch, mTOR, TGF-beta), translation, protein processing in endoplasmic reticulum, proteasomal degradation, glycolysis, oxidative phosphorylation, apoptosis, regulation of circadian rhythms, differentiation of neurons, synaptic plasticity, and transmission. Considering that various tissues age differently and age-related gene expression changes are tissue-specific, we investigated the effects of neuronal Gclc overexpression on gene expression levels in the imago thorax, which is primarily composed of muscles. A total of 58 genes were found to be differentially expressed between thoraces of control and Gclc overexpressing flies. The Gclc level demonstrated associations with expression of genes involved in the circadian rhythmicity, the genes in categories related to the muscle system process and the downregulation of genes involved in proteolysis. Most of the functional categories altered by Gclc overexpression related to metabolism including Drug metabolism, Metabolism of xenobiotics by cytochrome P450, Glutathione metabolism, Starch and sucrose metabolism, Citrate cycle (TCA cycle), One carbon pool by folate. Thus, the transcriptomic changes caused by neuron-specific Gclc overexpression in the thorax were less pronounced than in the head and affected pathways also differed from previous results. Although these pathways don't belong to the canonical longevity pathways, we suggest that they could participate in the delay of aging of Gclc overexpressing flies.
Collapse
Affiliation(s)
- Alexey Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of Sciences, Syktyvkar, Russia
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Zulfiya Guvatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Mikhail Shaposhnikov
- Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of Sciences, Syktyvkar, Russia
| | - Ekaterina Lashmanova
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Ekaterina Proshkina
- Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of Sciences, Syktyvkar, Russia
| | - Liubov Koval
- Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of Sciences, Syktyvkar, Russia
| | | | - George Krasnov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Anna Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
22
|
Nagy S, Maurer GW, Hentze JL, Rose M, Werge TM, Rewitz K. AMPK signaling linked to the schizophrenia-associated 1q21.1 deletion is required for neuronal and sleep maintenance. PLoS Genet 2018; 14:e1007623. [PMID: 30566533 PMCID: PMC6317821 DOI: 10.1371/journal.pgen.1007623] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 01/03/2019] [Accepted: 11/20/2018] [Indexed: 12/31/2022] Open
Abstract
The human 1q21.1 deletion of ten genes is associated with increased risk of schizophrenia. This deletion involves the β-subunit of the AMP-activated protein kinase (AMPK) complex, a key energy sensor in the cell. Although neurons have a high demand for energy and low capacity to store nutrients, the role of AMPK in neuronal physiology is poorly defined. Here we show that AMPK is important in the nervous system for maintaining neuronal integrity and for stress survival and longevity in Drosophila. To understand the impact of this signaling system on behavior and its potential contribution to the 1q21.1 deletion syndrome, we focused on sleep, an important role of which is proposed to be the reestablishment of neuronal energy levels that are diminished during energy-demanding wakefulness. Sleep disturbances are one of the most common problems affecting individuals with psychiatric disorders. We show that AMPK is required for maintenance of proper sleep architecture and for sleep recovery following sleep deprivation. Neuronal AMPKβ loss specifically leads to sleep fragmentation and causes dysregulation of genes believed to play a role in sleep homeostasis. Our data also suggest that AMPKβ loss may contribute to the increased risk of developing mental disorders and sleep disturbances associated with the human 1q21.1 deletion. The human 1q21.1 chromosomal deletion is associated with increased risk of schizophrenia. Because this deletion affects only a small number of genes, it provides a unique opportunity to identify the specific disease-causing gene(s) using animal models. Here, we report the use of a Drosophila model to identify the potential contribution of one gene affected by the 1q21.1 deletion–PRKAB2 –to the pathology of the 1q21.1 deletion syndrome. PRKAB2 encodes a subunit of the AMP-activated protein kinase (AMPK) complex, the main cellular energy sensor. We show that AMPK deficiency reduces lifespan and causes structural abnormalities in neuronal dendritic structures, a phenotype which has been linked to schizophrenia. Furthermore, cognitive impairment and altered sleep patterning are some of the most common symptoms of schizophrenia. Therefore, to understand the potential contribution of PRKAB2 to the 1q21.1 syndrome, we tested whether AMPK alterations might cause defects in learning and sleep. Our studies show that lack of PRKAB2 and AMPK-complex activity in the nervous system leads to reduced learning and to dramatic sleep disturbances. Thus, our data links a single 1q21.1-related gene with phenotypes that resemble common symptoms of neuropsychiatric disorders, suggesting that this gene, PRKAB2, may contribute to the risk of developing schizophrenia.
Collapse
Affiliation(s)
- Stanislav Nagy
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Gianna W Maurer
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Julie L Hentze
- Department of Biology, University of Copenhagen, Copenhagen, Denmark.,Institute for Biological Psychiatry, Mental Health Centre Sct. Hans, Roskilde, Denmark.,Department of Pathology, Herlev Hospital, Herlev, Denmark
| | - Morten Rose
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Thomas M Werge
- Institute for Biological Psychiatry, Mental Health Centre Sct. Hans, Roskilde, Denmark
| | - Kim Rewitz
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
23
|
Graze RM, Tzeng RY, Howard TS, Arbeitman MN. Perturbation of IIS/TOR signaling alters the landscape of sex-differential gene expression in Drosophila. BMC Genomics 2018; 19:893. [PMID: 30526477 PMCID: PMC6288939 DOI: 10.1186/s12864-018-5308-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 11/23/2018] [Indexed: 12/15/2022] Open
Abstract
Background The core functions of the insulin/insulin-like signaling and target of rapamycin (IIS/TOR) pathway are nutrient sensing, energy homeostasis, growth, and regulation of stress responses. This pathway is also known to interact directly and indirectly with the sex determination regulatory hierarchy. The IIS/TOR pathway plays a role in directing sexually dimorphic traits, including dimorphism of growth, metabolism, stress and behavior. Previous studies of sexually dimorphic gene expression in the adult head, which includes both nervous system and endocrine tissues, have revealed variation in sex-differential expression, depending in part on genotype and environment. To understand the degree to which the environmentally responsive insulin signaling pathway contributes to sexual dimorphism of gene expression, we examined the effect of perturbation of the pathway on gene expression in male and female Drosophila heads. Results Our data reveal a large effect of insulin signaling on gene expression, with greater than 50% of genes examined changing expression. Males and females have a shared gene expression response to knock-down of InR function, with significant enrichment for pathways involved in metabolism. Perturbation of insulin signaling has a greater impact on gene expression in males, with more genes changing expression and with gene expression differences of larger magnitude. Primarily as a consequence of the response in males, we find that reduced insulin signaling results in a striking increase in sex-differential expression. This includes sex-differences in expression of immune, defense and stress response genes, genes involved in modulating reproductive behavior, genes linking insulin signaling and ageing, and in the insulin signaling pathway itself. Conclusions Our results demonstrate that perturbation of insulin signaling results in thousands of genes displaying sex differences in expression that are not differentially expressed in control conditions. Thus, insulin signaling may play a role in variability of somatic, sex-differential expression. The finding that perturbation of the IIS/TOR pathway results in an altered landscape of sex-differential expression suggests a role of insulin signaling in the physiological underpinnings of trade-offs, sexual conflict and sex differences in expression variability. Electronic supplementary material The online version of this article (10.1186/s12864-018-5308-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rita M Graze
- Department of Biological Sciences, Auburn University, 101 Rouse Life Sciences building, Auburn, AL, 36849-5407, USA.
| | - Ruei-Ying Tzeng
- Biomedical Sciences Department, Florida State University, College of Medicine, 1115 West Call Street, Tallahassee, FL, 32306, USA
| | - Tiffany S Howard
- Department of Biological Sciences, Auburn University, 101 Rouse Life Sciences building, Auburn, AL, 36849-5407, USA
| | - Michelle N Arbeitman
- Biomedical Sciences Department, Florida State University, College of Medicine, 1115 West Call Street, Tallahassee, FL, 32306, USA.
| |
Collapse
|
24
|
Serine metabolism in the brain regulates starvation-induced sleep suppression in Drosophila melanogaster. Proc Natl Acad Sci U S A 2018; 115:7129-7134. [PMID: 29915051 PMCID: PMC6142195 DOI: 10.1073/pnas.1719033115] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Foraging and sleep are two conflicting behaviors in starved animals; however, it remains elusive how metabolic status governs sleep drive. In this study, we show that a biosynthetic pathway for the amino acid serine is transcriptionally up-regulated by starvation in adult fly brains. The behavioral response to genetic manipulation of key enzymes involved in serine metabolism supports the sleep-suppressing effect of serine in response to starvation. In a society where daily diet is becoming increasingly important to the sleep quality of individuals, our study defines an amino acid metabolic pathway that underlies adaptive sleep behaviors upon dietary stress. Sleep and metabolism are physiologically and behaviorally intertwined; however, the molecular basis for their interaction remains poorly understood. Here, we identified a serine metabolic pathway as a key mediator for starvation-induced sleep suppression. Transcriptome analyses revealed that enzymes involved in serine biosynthesis were induced upon starvation in Drosophila melanogaster brains. Genetic mutants of astray (aay), a fly homolog of the rate-limiting phosphoserine phosphatase in serine biosynthesis, displayed reduced starvation-induced sleep suppression. In contrast, a hypomorphic mutation in a serine/threonine-metabolizing enzyme, serine/threonine dehydratase (stdh), exaggerated starvation-induced sleep suppression. Analyses of double mutants indicated that aay and stdh act on the same genetic pathway to titrate serine levels in the head as well as to adjust starvation-induced sleep behaviors. RNA interference-mediated depletion of aay expression in neurons, using cholinergic Gal4 drivers, phenocopied aay mutants, while a nicotinic acetylcholine receptor antagonist selectively rescued the exaggerated starvation-induced sleep suppression in stdh mutants. Taken together, these data demonstrate that neural serine metabolism controls sleep during starvation, possibly via cholinergic signaling. We propose that animals have evolved a sleep-regulatory mechanism that reprograms amino acid metabolism for adaptive sleep behaviors in response to metabolic needs.
Collapse
|
25
|
Ly S, Pack AI, Naidoo N. The neurobiological basis of sleep: Insights from Drosophila. Neurosci Biobehav Rev 2018; 87:67-86. [PMID: 29391183 PMCID: PMC5845852 DOI: 10.1016/j.neubiorev.2018.01.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 01/22/2018] [Accepted: 01/24/2018] [Indexed: 12/12/2022]
Abstract
Sleep is a biological enigma that has raised numerous questions about the inner workings of the brain. The fundamental question of why our nervous systems have evolved to require sleep remains a topic of ongoing scientific deliberation. This question is largely being addressed by research using animal models of sleep. Drosophila melanogaster, also known as the common fruit fly, exhibits a sleep state that shares common features with many other species. Drosophila sleep studies have unearthed an immense wealth of knowledge about the neuroscience of sleep. Given the breadth of findings published on Drosophila sleep, it is important to consider how all of this information might come together to generate a more holistic understanding of sleep. This review provides a comprehensive summary of the neurobiology of Drosophila sleep and explores the broader insights and implications of how sleep is regulated across species and why it is necessary for the brain.
Collapse
Affiliation(s)
- Sarah Ly
- Center for Sleep and Circadian Neurobiology, 125 South 31st St., Philadelphia, PA, 19104-3403, United States.
| | - Allan I Pack
- Center for Sleep and Circadian Neurobiology, 125 South 31st St., Philadelphia, PA, 19104-3403, United States; Division of Sleep Medicine/Department of Medicine, University of Pennsylvania Perelman School of Medicine, 125 South 31st St., Philadelphia, PA, 19104-3403, United States
| | - Nirinjini Naidoo
- Center for Sleep and Circadian Neurobiology, 125 South 31st St., Philadelphia, PA, 19104-3403, United States; Division of Sleep Medicine/Department of Medicine, University of Pennsylvania Perelman School of Medicine, 125 South 31st St., Philadelphia, PA, 19104-3403, United States.
| |
Collapse
|
26
|
Moskalev A, Shaposhnikov M, Zemskaya N, Belyi A, Dobrovolskaya E, Patova A, Guvatova Z, Lukyanova E, Snezhkina A, Kudryavtseva A. Transcriptome analysis reveals mechanisms of geroprotective effects of fucoxanthin in Drosophila. BMC Genomics 2018; 19:77. [PMID: 29504896 PMCID: PMC5836829 DOI: 10.1186/s12864-018-4471-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Background We have previously showed that the carotenoid fucoxanthin can increase the lifespan in Drosophila melanogaster and Caenorhabditis elegans. However, the molecular mechanisms of the geroprotective effect of fucoxanthin have not been studied so far. Results Here, we studied the effects of fucoxanthin on the Drosophila aging process at the molecular and the whole organism levels. At the organismal level, fucoxanthin increased the median lifespan and had a positive effect on fecundity, fertility, intestinal barrier function, and nighttime sleep. Transcriptome analysis revealed 57 differentially expressed genes involved in 17 KEGG (Kyoto Encyclopedia of Genes and Genomes) pathways. Among the most represented molecular pathways induced by fucoxanthin, a significant portion is related to longevity, including MAPK, mTOR, Wnt, Notch, and Hippo signaling pathways, autophagy, translation, glycolysis, oxidative phosphorylation, apoptosis, immune response, neurogenesis, sleep, and response to DNA damage. Conclusions Life-extending effects of fucoxanthin are associated with differential expression of longevity-associated genes. Electronic supplementary material The online version of this article (10.1186/s12864-018-4471-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alexey Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia. .,Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of Sciences, Syktyvkar, Russia.
| | - Mikhail Shaposhnikov
- Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of Sciences, Syktyvkar, Russia
| | - Nadezhda Zemskaya
- Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of Sciences, Syktyvkar, Russia
| | - Alexey Belyi
- Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of Sciences, Syktyvkar, Russia
| | - Eugenia Dobrovolskaya
- Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of Sciences, Syktyvkar, Russia
| | - Anna Patova
- Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of Sciences, Syktyvkar, Russia
| | - Zulfiya Guvatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Elena Lukyanova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Anastasiya Snezhkina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Anna Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
27
|
Abstract
Sleep is essential for proper brain function in mammals and insects. During sleep, animals are disconnected from the external world; they show high arousal thresholds and changed brain activity. Sleep deprivation results in a sleep rebound. Research using the fruit fly, Drosophila melanogaster, has helped us understand the genetic and neuronal control of sleep. Genes involved in sleep control code for ion channels, factors influencing neurotransmission and neuromodulation, and proteins involved in the circadian clock. The neurotransmitters/neuromodulators involved in sleep control are GABA, dopamine, acetylcholine, serotonin, and several neuropeptides. Sleep is controlled by the interplay between sleep homeostasis and the circadian clock. Putative sleep-wake centers are located in higher-order brain centers that are indirectly connected to the circadian clock network. The primary function of sleep appears to be the downscaling of synapses that have been built up during wakefulness. Thus, brain homeostasis is maintained and learning and memory are assured.
Collapse
Affiliation(s)
- Charlotte Helfrich-Förster
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, University of Würzburg, 97074 Würzburg, Germany;
| |
Collapse
|
28
|
RNA-Sequencing of Drosophila melanogaster Head Tissue on High-Sugar and High-Fat Diets. G3-GENES GENOMES GENETICS 2018; 8:279-290. [PMID: 29141990 PMCID: PMC5765356 DOI: 10.1534/g3.117.300397] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Obesity has been shown to increase risk for cardiovascular disease and type-2 diabetes. In addition, it has been implicated in aggravation of neurological conditions such as Alzheimer’s. In the model organism Drosophila melanogaster, a physiological state mimicking diet-induced obesity can be induced by subjecting fruit flies to a solid medium disproportionately higher in sugar than protein, or that has been supplemented with a rich source of saturated fat. These flies can exhibit increased circulating glucose levels, increased triglyceride content, insulin-like peptide resistance, and behavior indicative of neurological decline. We subjected flies to variants of the high-sugar diet, high-fat diet, or normal (control) diet, followed by a total RNA extraction from fly heads of each diet group for the purpose of Poly-A selected RNA-Sequencing. Our objective was to identify the effects of obesogenic diets on transcriptome patterns, how they differed between obesogenic diets, and identify genes that may relate to pathogenesis accompanying an obesity-like state. Gene ontology analysis indicated an overrepresentation of affected genes associated with immunity, metabolism, and hemocyanin in the high-fat diet group, and CHK, cell cycle activity, and DNA binding and transcription in the high-sugar diet group. Our results also indicate differences in the effects of the high-fat diet and high-sugar diet on expression profiles in head tissue of flies, despite the reportedly similar phenotypic impacts of the diets. The impacted genes, and how they may relate to pathogenesis in the Drosophila obesity-like state, warrant further experimental investigation.
Collapse
|
29
|
Thimgan MS, Kress N, Lisse J, Fiebelman C, Hilderbrand T. The acyl-CoA Synthetase, pudgy, Promotes Sleep and Is Required for the Homeostatic Response to Sleep Deprivation. Front Endocrinol (Lausanne) 2018; 9:464. [PMID: 30186232 PMCID: PMC6110854 DOI: 10.3389/fendo.2018.00464] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/27/2018] [Indexed: 12/12/2022] Open
Abstract
The regulation of sleep and the response to sleep deprivation rely on multiple biochemical pathways. A critical connection is the link between sleep and metabolism. Metabolic changes can disrupt sleep, and conversely decreased sleep can alter the metabolic environment. There is building evidence that lipid metabolism, in particular, is a critical part of mounting the homeostatic response to sleep deprivation. We have evaluated an acyl-CoA synthetase, pudgy (pdgy), for its role in sleep and response to sleep deprivation. When pdgy transcript levels are decreased through transposable element disruption of the gene, mutant flies showed lower total sleep times and increased sleep fragmentation at night compared to genetic controls. Consistent with disrupted sleep, mutant flies had a decreased lifespan compared to controls. pdgy disrupted fatty acid handling as pdgy mutants showed increased sensitivity to starvation and exhibited lower fat stores. Moreover, the response to sleep deprivation is reduced when compared to a control flies. When we decreased the transcript levels for pdgy using RNAi, the response to sleep deprivation was decreased compared to background controls. In addition, when the pdgy transcription is rescued throughout the fly, the response to sleep deprivation is restored. These data demonstrate that the regulation and function of acyl-CoA synthetase plays a critical role in regulating sleep and the response to sleep deprivation. Endocrine and metabolic signals that alter transcript levels of pdgy impact sleep regulation or interfere with the homeostatic response to sleep deprivation.
Collapse
Affiliation(s)
- Matthew S. Thimgan
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, United States
- *Correspondence: Matthew S. Thimgan
| | - Natalie Kress
- Department of Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, MO, United States
| | - Josh Lisse
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, United States
| | - Courtney Fiebelman
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, United States
| | - Thomas Hilderbrand
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, United States
| |
Collapse
|
30
|
O'Callaghan EK, Green EW, Franken P, Mongrain V. Omics Approaches in Sleep-Wake Regulation. Handb Exp Pharmacol 2018; 253:59-81. [PMID: 29796779 DOI: 10.1007/164_2018_125] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Although sleep seems an obvious and simple behaviour, it is extremely complex involving numerous interactions both at the neuronal and the molecular levels. While we have gained detailed insight into the molecules and neuronal networks responsible for the circadian organization of sleep and wakefulness, the molecular underpinnings of the homeostatic aspect of sleep regulation are still unknown and the focus of a considerable research effort. In the last 20 years, the development of techniques allowing the simultaneous measurement of hundreds to thousands of molecular targets (i.e. 'omics' approaches) has enabled the unbiased study of the molecular pathways regulated by and regulating sleep. In this chapter, we will review how the different omics approaches, including transcriptomics, epigenomics, proteomics, and metabolomics, have advanced sleep research. We present relevant data in the framework of the two-process model in which circadian and homeostatic processes interact to regulate sleep. The integration of the different omics levels, known as 'systems genetics', will eventually lead to a better understanding of how information flows from the genome, to molecules, to networks, and finally to sleep both in health and disease.
Collapse
Affiliation(s)
- Emma K O'Callaghan
- Center for Advanced Research in Sleep Medicine and Research Center, Hôpital du Sacré-Coeur de Montréal, Montreal, QC, Canada.,Department of Neuroscience, Université de Montréal, Montreal, QC, Canada
| | - Edward W Green
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Paul Franken
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Valérie Mongrain
- Center for Advanced Research in Sleep Medicine and Research Center, Hôpital du Sacré-Coeur de Montréal, Montreal, QC, Canada. .,Department of Neuroscience, Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
31
|
Selection for long and short sleep duration in Drosophila melanogaster reveals the complex genetic network underlying natural variation in sleep. PLoS Genet 2017; 13:e1007098. [PMID: 29240764 PMCID: PMC5730107 DOI: 10.1371/journal.pgen.1007098] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 11/01/2017] [Indexed: 12/16/2022] Open
Abstract
Why do some individuals need more sleep than others? Forward mutagenesis screens in flies using engineered mutations have established a clear genetic component to sleep duration, revealing mutants that convey very long or short sleep. Whether such extreme long or short sleep could exist in natural populations was unknown. We applied artificial selection for high and low night sleep duration to an outbred population of Drosophila melanogaster for 13 generations. At the end of the selection procedure, night sleep duration diverged by 9.97 hours in the long and short sleeper populations, and 24-hour sleep was reduced to 3.3 hours in the short sleepers. Neither long nor short sleeper lifespan differed appreciably from controls, suggesting little physiological consequences to being an extreme long or short sleeper. Whole genome sequence data from seven generations of selection revealed several hundred thousand changes in allele frequencies at polymorphic loci across the genome. Combining the data from long and short sleeper populations across generations in a logistic regression implicated 126 polymorphisms in 80 candidate genes, and we confirmed three of these genes and a larger genomic region with mutant and chromosomal deficiency tests, respectively. Many of these genes could be connected in a single network based on previously known physical and genetic interactions. Candidate genes have known roles in several classic, highly conserved developmental and signaling pathways—EGFR, Wnt, Hippo, and MAPK. The involvement of highly pleiotropic pathway genes suggests that sleep duration in natural populations can be influenced by a wide variety of biological processes, which may be why the purpose of sleep has been so elusive. One of the biggest mysteries in biology is the need to sleep. Sleep duration has an underlying genetic basis, suggesting that very long and short sleep times could be bred for experimentally. How far can sleep duration be driven up or down? Here we achieved extremely long and short night sleep duration by subjecting a wild-derived population of Drosophila melanogaster to an experimental breeding program. At the end of the breeding program, long sleepers averaged 9.97 hours more nightly sleep than short sleepers. We analyzed whole-genome sequences from seven generations of the experimental breeding to identify allele frequencies that diverged between long and short sleepers, and verified genes and genomic regions with mutation and deficiency testing. These alleles map to classic developmental and signaling pathways, implicating many diverse processes that potentially affect sleep duration.
Collapse
|
32
|
Donlea JM. Neuronal and molecular mechanisms of sleep homeostasis. CURRENT OPINION IN INSECT SCIENCE 2017; 24:51-57. [PMID: 29208223 DOI: 10.1016/j.cois.2017.09.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 09/13/2017] [Accepted: 09/13/2017] [Indexed: 06/07/2023]
Abstract
Sleep is necessary for survival, and prolonged waking causes a homeostatic increase in the need for recovery sleep. Homeostasis is a core component of sleep regulation and has been tightly conserved across evolution from invertebrates to man. Homeostatic sleep regulation was first identified among insects in cockroaches several decades ago, but the characterization of sleep rebound in Drosophila melanogaster opened the use of insect model species to understand homeostatic functions and regulation of sleep. This review describes circuits in two neuropil structures, the central complex and mushroom bodies, that influence sleep homeostasis and neuromodulatory systems that influence the accrual of homeostatic sleep need.
Collapse
Affiliation(s)
- Jeffrey M Donlea
- Department of Neurobiology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA 90095-1763, USA.
| |
Collapse
|
33
|
Seugnet L, Dissel S, Thimgan M, Cao L, Shaw PJ. Identification of Genes that Maintain Behavioral and Structural Plasticity during Sleep Loss. Front Neural Circuits 2017; 11:79. [PMID: 29109678 PMCID: PMC5660066 DOI: 10.3389/fncir.2017.00079] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/05/2017] [Indexed: 11/23/2022] Open
Abstract
Although patients with primary insomnia experience sleep disruption, they are able to maintain normal performance on a variety of cognitive tasks. This observation suggests that insomnia may be a condition where predisposing factors simultaneously increase the risk for insomnia and also mitigate against the deleterious consequences of waking. To gain insight into processes that might regulate sleep and buffer neuronal circuits during sleep loss, we manipulated three genes, fat facet (faf), highwire (hiw) and the GABA receptor Resistance to dieldrin (Rdl), that were differentially modulated in a Drosophila model of insomnia. Our results indicate that increasing faf and decreasing hiw or Rdl within wake-promoting large ventral lateral clock neurons (lLNvs) induces sleep loss. As expected, sleep loss induced by decreasing hiw in the lLNvs results in deficits in short-term memory and increases of synaptic growth. However, sleep loss induced by knocking down Rdl in the lLNvs protects flies from sleep-loss induced deficits in short-term memory and increases in synaptic markers. Surprisingly, decreasing hiw and Rdl within the Mushroom Bodies (MBs) protects against the negative effects of sleep deprivation (SD) as indicated by the absence of a subsequent homeostatic response, or deficits in short-term memory. Together these results indicate that specific genes are able to disrupt sleep and protect against the negative consequences of waking in a circuit dependent manner.
Collapse
Affiliation(s)
- Laurent Seugnet
- Centre de Recherche en Neurosciences de Lyon, U1028/UMR 5292, Team WAKING, Université Claude Bernard Lyon 1, INSERM U1028, CNRS UMR 5292, Lyon, France
| | - Stephane Dissel
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, United States
| | - Matthew Thimgan
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, United States
| | - Lijuan Cao
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, United States
| | - Paul J Shaw
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
34
|
Rohde PD, Gaertner B, Ward K, Sørensen P, Mackay TFC. Genomic Analysis of Genotype-by-Social Environment Interaction for Drosophila melanogaster Aggressive Behavior. Genetics 2017; 206:1969-1984. [PMID: 28550016 PMCID: PMC5560801 DOI: 10.1534/genetics.117.200642] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 05/22/2017] [Indexed: 02/06/2023] Open
Abstract
Human psychiatric disorders such as schizophrenia, bipolar disorder, and attention-deficit/hyperactivity disorder often include adverse behaviors including increased aggressiveness. Individuals with psychiatric disorders often exhibit social withdrawal, which can further increase the probability of conducting a violent act. Here, we used the inbred, sequenced lines of the Drosophila Genetic Reference Panel (DGRP) to investigate the genetic basis of variation in male aggressive behavior for flies reared in a socialized and socially isolated environment. We identified genetic variation for aggressive behavior, as well as significant genotype-by-social environmental interaction (GSEI); i.e., variation among DGRP genotypes in the degree to which social isolation affected aggression. We performed genome-wide association (GWA) analyses to identify genetic variants associated with aggression within each environment. We used genomic prediction to partition genetic variants into gene ontology (GO) terms and constituent genes, and identified GO terms and genes with high prediction accuracies in both social environments and for GSEI. The top predictive GO terms significantly increased the proportion of variance explained, compared to prediction models based on all segregating variants. We performed genomic prediction across environments, and identified genes in common between the social environments that turned out to be enriched for genome-wide associated variants. A large proportion of the associated genes have previously been associated with aggressive behavior in Drosophila and mice. Further, many of these genes have human orthologs that have been associated with neurological disorders, indicating partially shared genetic mechanisms underlying aggression in animal models and human psychiatric disorders.
Collapse
Affiliation(s)
- Palle Duun Rohde
- Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, Aarhus University, 8830 Tjele, Denmark
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, 8000 Aarhus, Denmark
- ISEQ, Center for Integrative Sequencing, Aarhus University, 8000 Aarhus, Denmark
| | - Bryn Gaertner
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27695
- Program in Genetics, North Carolina State University, Raleigh, North Carolina 27695
- W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina 27695
| | - Kirsty Ward
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27695
- Program in Genetics, North Carolina State University, Raleigh, North Carolina 27695
- W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina 27695
| | - Peter Sørensen
- Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, Aarhus University, 8830 Tjele, Denmark
| | - Trudy F C Mackay
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27695
- Program in Genetics, North Carolina State University, Raleigh, North Carolina 27695
- W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina 27695
| |
Collapse
|
35
|
Identifying pathways modulating sleep duration: from genomics to transcriptomics. Sci Rep 2017; 7:4555. [PMID: 28676676 PMCID: PMC5496883 DOI: 10.1038/s41598-017-04027-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 05/10/2017] [Indexed: 01/02/2023] Open
Abstract
Recognizing that insights into the modulation of sleep duration can emerge by exploring the functional relationships among genes, we used this strategy to explore the genome-wide association results for this trait. We detected two major signalling pathways (ion channels and the ERBB signalling family of tyrosine kinases) that could be replicated across independent GWA studies meta-analyses. To investigate the significance of these pathways for sleep modulation, we performed transcriptome analyses of short sleeping flies’ heads (knockdown for the ABCC9 gene homolog; dSur). We found significant alterations in gene-expression in the short sleeping knockdowns versus controls flies, which correspond to pathways associated with sleep duration in our human studies. Most notably, the expression of Rho and EGFR (members of the ERBB signalling pathway) genes was down- and up-regulated, respectively, consistently with the established role of these genes for sleep consolidation in Drosophila. Using a disease multifactorial interaction network, we showed that many of the genes of the pathways indicated to be relevant for sleep duration had functional evidence of their involvement with sleep regulation, circadian rhythms, insulin secretion, gluconeogenesis and lipogenesis.
Collapse
|
36
|
Nikonova EV, Gilliland JDA, Tanis KQ, Podtelezhnikov AA, Rigby AM, Galante RJ, Finney EM, Stone DJ, Renger JJ, Pack AI, Winrow CJ. Transcriptional Profiling of Cholinergic Neurons From Basal Forebrain Identifies Changes in Expression of Genes Between Sleep and Wake. Sleep 2017; 40:3608773. [PMID: 28419375 PMCID: PMC6075396 DOI: 10.1093/sleep/zsx059] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Study objective To assess differences in gene expression in cholinergic basal forebrain cells between sleeping and sleep-deprived mice sacrificed at the same time of day. Methods Tg(ChAT-eGFP)86Gsat mice expressing enhanced green fluorescent protein (eGFP) under control of the choline acetyltransferase (Chat) promoter were utilized to guide laser capture of cholinergic cells in basal forebrain. Messenger RNA expression levels in these cells were profiled using microarrays. Gene expression in eGFP(+) neurons was compared (1) to that in eGFP(-) neurons and to adjacent white matter, (2) between 7:00 am (lights on) and 7:00 pm (lights off), (3) between sleep-deprived and sleeping animals at 0, 3, 6, and 9 hours from lights on. Results There was a marked enrichment of ChAT and other markers of cholinergic neurons in eGFP(+) cells. Comparison of gene expression in these eGFP(+) neurons between 7:00 am and 7:00 pm revealed expected differences in the expression of clock genes (Arntl2, Per1, Per2, Dbp, Nr1d1) as well as mGluR3. Comparison of expression between spontaneous sleep and sleep-deprived groups sacrificed at the same time of day revealed a number of transcripts (n = 55) that had higher expression in sleep deprivation compared to sleep. Genes upregulated in sleep deprivation predominantly were from the protein folding pathway (25 transcripts, including chaperones). Among 42 transcripts upregulated in sleep was the cold-inducible RNA-binding protein. Conclusions Cholinergic cell signatures were characterized. Whether the identified genes are changing as a consequence of differences in behavioral state or as part of the molecular regulatory mechanism remains to be determined.
Collapse
Affiliation(s)
- Elena V Nikonova
- Genetics and Pharmacogenomics, Merck Research Laboratories, Merck & Co., Inc., West Point, PA
| | - Jason DA Gilliland
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, PA
| | - Keith Q Tanis
- Genetics and Pharmacogenomics, Merck Research Laboratories, Merck & Co., Inc., West Point, PA
| | - Alexei A Podtelezhnikov
- Genetics and Pharmacogenomics, Merck Research Laboratories, Merck & Co., Inc., West Point, PA
| | - Alison M Rigby
- Department of Neuroscience, Merck & Co., Inc., West Point, PA
| | - Raymond J Galante
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, PA
| | - Eva M Finney
- Genetics and Pharmacogenomics, Merck Research Laboratories, Merck & Co., Inc., West Point, PA
| | - David J Stone
- Genetics and Pharmacogenomics, Merck Research Laboratories, Merck & Co., Inc., West Point, PA
| | - John J Renger
- Department of Neuroscience, Merck & Co., Inc., West Point, PA
| | - Allan I Pack
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, PA
| | | |
Collapse
|
37
|
Circadian Rhythms and Sleep in Drosophila melanogaster. Genetics 2017; 205:1373-1397. [PMID: 28360128 DOI: 10.1534/genetics.115.185157] [Citation(s) in RCA: 236] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 11/17/2016] [Indexed: 02/07/2023] Open
Abstract
The advantages of the model organism Drosophila melanogaster, including low genetic redundancy, functional simplicity, and the ability to conduct large-scale genetic screens, have been essential for understanding the molecular nature of circadian (∼24 hr) rhythms, and continue to be valuable in discovering novel regulators of circadian rhythms and sleep. In this review, we discuss the current understanding of these interrelated biological processes in Drosophila and the wider implications of this research. Clock genes period and timeless were first discovered in large-scale Drosophila genetic screens developed in the 1970s. Feedback of period and timeless on their own transcription forms the core of the molecular clock, and accurately timed expression, localization, post-transcriptional modification, and function of these genes is thought to be critical for maintaining the circadian cycle. Regulators, including several phosphatases and kinases, act on different steps of this feedback loop to ensure strong and accurately timed rhythms. Approximately 150 neurons in the fly brain that contain the core components of the molecular clock act together to translate this intracellular cycling into rhythmic behavior. We discuss how different groups of clock neurons serve different functions in allowing clocks to entrain to environmental cues, driving behavioral outputs at different times of day, and allowing flexible behavioral responses in different environmental conditions. The neuropeptide PDF provides an important signal thought to synchronize clock neurons, although the details of how PDF accomplishes this function are still being explored. Secreted signals from clock neurons also influence rhythms in other tissues. SLEEP is, in part, regulated by the circadian clock, which ensures appropriate timing of sleep, but the amount and quality of sleep are also determined by other mechanisms that ensure a homeostatic balance between sleep and wake. Flies have been useful for identifying a large set of genes, molecules, and neuroanatomic loci important for regulating sleep amount. Conserved aspects of sleep regulation in flies and mammals include wake-promoting roles for catecholamine neurotransmitters and involvement of hypothalamus-like regions, although other neuroanatomic regions implicated in sleep in flies have less clear parallels. Sleep is also subject to regulation by factors such as food availability, stress, and social environment. We are beginning to understand how the identified molecules and neurons interact with each other, and with the environment, to regulate sleep. Drosophila researchers can also take advantage of increasing mechanistic understanding of other behaviors, such as learning and memory, courtship, and aggression, to understand how sleep loss impacts these behaviors. Flies thus remain a valuable tool for both discovery of novel molecules and deep mechanistic understanding of sleep and circadian rhythms.
Collapse
|
38
|
Wang JB, Lu HL, St. Leger RJ. The genetic basis for variation in resistance to infection in the Drosophila melanogaster genetic reference panel. PLoS Pathog 2017; 13:e1006260. [PMID: 28257468 PMCID: PMC5352145 DOI: 10.1371/journal.ppat.1006260] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 03/15/2017] [Accepted: 02/24/2017] [Indexed: 01/01/2023] Open
Abstract
Individuals vary extensively in the way they respond to disease but the genetic basis of this variation is not fully understood. We found substantial individual variation in resistance and tolerance to the fungal pathogen Metarhizium anisopliae Ma549 using the Drosophila melanogaster Genetic Reference Panel (DGRP). In addition, we found that host defense to Ma549 was correlated with defense to the bacterium Pseudomonas aeruginosa Pa14, and several previously published DGRP phenotypes including oxidative stress sensitivity, starvation stress resistance, hemolymph glucose levels, and sleep indices. We identified polymorphisms associated with differences between lines in both their mean survival times and microenvironmental plasticity, suggesting that lines differ in their ability to adapt to variable pathogen exposures. The majority of polymorphisms increasing resistance to Ma549 were sex biased, located in non-coding regions, had moderately large effect and were rare, suggesting that there is a general cost to defense. Nevertheless, host defense was not negatively correlated with overall longevity and fecundity. In contrast to Ma549, minor alleles were concentrated in the most Pa14-susceptible as well as the most Pa14-resistant lines. A pathway based analysis revealed a network of Pa14 and Ma549-resistance genes that are functionally connected through processes that encompass phagocytosis and engulfment, cell mobility, intermediary metabolism, protein phosphorylation, axon guidance, response to DNA damage, and drug metabolism. Functional testing with insertional mutagenesis lines indicates that 12/13 candidate genes tested influence susceptibility to Ma549. Many candidate genes have homologs identified in studies of human disease, suggesting that genes affecting variation in susceptibility are conserved across species.
Collapse
Affiliation(s)
- Jonathan B. Wang
- Department of Entomology, University of Maryland College Park, College Park, Maryland, United States of America
| | - Hsiao-Ling Lu
- Department of Entomology, University of Maryland College Park, College Park, Maryland, United States of America
| | - Raymond J. St. Leger
- Department of Entomology, University of Maryland College Park, College Park, Maryland, United States of America
- * E-mail:
| |
Collapse
|
39
|
Nitta Y, Yamazaki D, Sugie A, Hiroi M, Tabata T. DISCO Interacting Protein 2 regulates axonal bifurcation and guidance of Drosophila mushroom body neurons. Dev Biol 2016; 421:233-244. [PMID: 27908785 DOI: 10.1016/j.ydbio.2016.11.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 11/16/2016] [Accepted: 11/16/2016] [Indexed: 10/20/2022]
Abstract
Axonal branching is one of the key processes within the enormous complexity of the nervous system to enable a single neuron to send information to multiple targets. However, the molecular mechanisms that control branch formation are poorly understood. In particular, previous studies have rarely addressed the mechanisms underlying axonal bifurcation, in which axons form new branches via splitting of the growth cone. We demonstrate that DISCO Interacting Protein 2 (DIP2) is required for precise axonal bifurcation in Drosophila mushroom body (MB) neurons by suppressing ectopic bifurcation and regulating the guidance of sister axons. We also found that DIP2 localize to the plasma membrane. Domain function analysis revealed that the AMP-synthetase domains of DIP2 are essential for its function, which may involve exerting a catalytic activity that modifies fatty acids. Genetic analysis and subsequent biochemical analysis suggested that DIP2 is involved in the fatty acid metabolization of acyl-CoA. Taken together, our results reveal a function of DIP2 in the developing nervous system and provide a potential functional relationship between fatty acid metabolism and axon morphogenesis.
Collapse
Affiliation(s)
- Yohei Nitta
- Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Daisuke Yamazaki
- Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Atsushi Sugie
- Department of Neuroscience Disease, Center for Transdisciplinary Research, Niigata University, 757, Ichibancho, Asahimachidori, Chuo-ku, Niigata-shi, Niigata 951-8585, Japan
| | - Makoto Hiroi
- Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Tetsuya Tabata
- Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan.
| |
Collapse
|
40
|
Dissel S, Klose M, Donlea J, Cao L, English D, Winsky-Sommerer R, van Swinderen B, Shaw PJ. Enhanced sleep reverses memory deficits and underlying pathology in Drosophila models of Alzheimer's disease. Neurobiol Sleep Circadian Rhythms 2016; 2:15-26. [PMID: 29094110 PMCID: PMC5662006 DOI: 10.1016/j.nbscr.2016.09.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
To test the hypothesis that sleep can reverse cognitive impairment during Alzheimer's disease, we enhanced sleep in flies either co-expressing human amyloid precursor protein and Beta-secretase (APP:BACE), or in flies expressing human tau. The ubiquitous expression of APP:BACE or human tau disrupted sleep. The sleep deficits could be reversed and sleep could be enhanced when flies were administered the GABA-A agonist 4,5,6,7-tetrahydroisoxazolo-[5,4-c]pyridine-3-ol (THIP). Expressing APP:BACE disrupted both Short-term memory (STM) and Long-term memory (LTM) as assessed using Aversive Phototaxic Suppression (APS) and courtship conditioning. Flies expressing APP:BACE also showed reduced levels of the synaptic protein discs large (DLG). Enhancing sleep in memory-impaired APP:BACE flies fully restored both STM and LTM and restored DLG levels. Sleep also restored STM to flies expressing human tau. Using live-brain imaging of individual clock neurons expressing both tau and the cAMP sensor Epac1-camps, we found that tau disrupted cAMP signaling. Importantly, enhancing sleep in flies expressing human tau restored proper cAMP signaling. Thus, we demonstrate that sleep can be used as a therapeutic to reverse deficits that accrue during the expression of toxic peptides associated with Alzheimer's disease. THIP can be used to enhance sleep in two Drosophila models of Alzheimer's disease. Enhanced sleep reverses memory deficits in fly's expressing human APP:BACE and tau. Enhanced sleep restores cAMP levels in clock neurons expressing tau. Sleep can be used as a therapeutic to reverse Alzheimer's disease related deficits.
Collapse
Affiliation(s)
- Stephane Dissel
- Department of Neuroscience, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, U.S.A
| | - Markus Klose
- Department of Neuroscience, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, U.S.A
| | - Jeff Donlea
- Department of Neurobiology, University of California: Los Angeles Los Angeles, California, U.S.A
| | - Lijuan Cao
- Department of Neuroscience, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, U.S.A
| | - Denis English
- Department of Neuroscience, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, U.S.A
| | - Raphaelle Winsky-Sommerer
- Surrey Sleep Research Centre, Faculty of Health and Medical Sciences University of Surrey Guildford Surrey, GU2 7XH, United Kingdom
| | - Bruno van Swinderen
- Queensland Brain Institute, The University of Queensland, Brisbane Qld 4072 Australia
| | - Paul J Shaw
- Department of Neuroscience, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, U.S.A
| |
Collapse
|
41
|
Abstract
Drosophila has proven to be a powerful model to identify genes and circuits that impact sleep. While the majority of studies have primarily been interested in identifying manipulations that alter sleep time, a growing body of work has begun to focus on how changing sleep influences functional outcomes such as cognitive performance, structural plasticity, and metabolism to name a few. Evaluating sleep time provides an appropriate entry point into elucidating sleep function. However, it is not possible to fully understand how a manipulation has impacted sleep regulation without first establishing how it has affected the animals’ well-being. Synaptic plasticity and memory are important functional outcomes that can be used to asses an animal’s status. In this manuscript, we review recent advances in studies examining sleep, memory, and performance. We conclude that as Drosophila sleep researchers expand their analysis beyond sleep time, the opportunities to discover the function of sleep will be enhanced.
Collapse
|