1
|
Czajewski I, Swain B, Xu J, McDowall L, Ferenbach AT, van Aalten DMF. Rescuable sleep and synaptogenesis phenotypes in a Drosophila model of O-GlcNAc transferase intellectual disability. eLife 2024; 13:e90376. [PMID: 39535175 PMCID: PMC11623933 DOI: 10.7554/elife.90376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
O-GlcNAcylation is an essential intracellular protein modification mediated by O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA). Recently, missense mutations in OGT have been linked to intellectual disability, indicating that this modification is important for the development and functioning of the nervous system. However, the processes that are most sensitive to perturbations in O-GlcNAcylation remain to be identified. Here, we uncover quantifiable phenotypes in the fruit fly Drosophila melanogaster carrying a patient-derived OGT mutation in the catalytic domain. Hypo-O-GlcNAcylation leads to defects in synaptogenesis and reduced sleep stability. Both these phenotypes can be partially rescued by genetically or chemically targeting OGA, suggesting that a balance of OGT/OGA activity is required for normal neuronal development and function.
Collapse
Affiliation(s)
- Ignacy Czajewski
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Bijayalaxmi Swain
- Section of Neurobiology and DANDRITE, Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
| | - Jiawei Xu
- Section of Neurobiology and DANDRITE, Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
| | - Laurin McDowall
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Andrew T Ferenbach
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
- Section of Neurobiology and DANDRITE, Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
| | - Daan MF van Aalten
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
- Section of Neurobiology and DANDRITE, Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
| |
Collapse
|
2
|
Ortiz-Vega N, Lobato AG, Canic T, Zhu Y, Lazopulo S, Syed S, Zhai RG. Regulation of proteostasis by sleep through autophagy in Drosophila models of Alzheimer's disease. Life Sci Alliance 2024; 7:e202402681. [PMID: 39237365 PMCID: PMC11377308 DOI: 10.26508/lsa.202402681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024] Open
Abstract
Sleep and circadian rhythm dysfunctions are common clinical features of Alzheimer's disease (AD). Increasing evidence suggests that in addition to being a symptom, sleep disturbances can also drive the progression of neurodegeneration. Protein aggregation is a pathological hallmark of AD; however, the molecular pathways behind how sleep affects protein homeostasis remain elusive. Here we demonstrate that sleep modulation influences proteostasis and the progression of neurodegeneration in Drosophila models of tauopathy. We show that sleep deprivation enhanced Tau aggregational toxicity resulting in exacerbated synaptic degeneration. In contrast, sleep induction using gaboxadol led to reduced toxic Tau accumulation in neurons as a result of modulated autophagic flux and enhanced clearance of ubiquitinated Tau, suggesting altered protein processing and clearance that resulted in improved synaptic integrity and function. These findings highlight the complex relationship between sleep and regulation of protein homeostasis and the neuroprotective potential of sleep-enhancing therapeutics to slow the progression or delay the onset of neurodegeneration.
Collapse
Affiliation(s)
- Natalie Ortiz-Vega
- Department of Neurology, University of Chicago, Chicago, IL, USA
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
- Graduate Program in Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Amanda G Lobato
- Department of Neurology, University of Chicago, Chicago, IL, USA
| | - Tijana Canic
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Physics, University of Miami, Coral Gables, FL, USA
| | - Yi Zhu
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | - Sheyum Syed
- Department of Physics, University of Miami, Coral Gables, FL, USA
| | - R Grace Zhai
- Department of Neurology, University of Chicago, Chicago, IL, USA
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
3
|
Sitaraman D, Vecsey CG, Koochagian C. Activity Monitoring for Analysis of Sleep in Drosophila melanogaster. Cold Spring Harb Protoc 2024; 2024:pdb.top108095. [PMID: 38336390 DOI: 10.1101/pdb.top108095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
Sleep is important for survival, and the need for sleep is conserved across species. In the past two decades, the fruit fly Drosophila melanogaster has emerged as a promising system in which to study the genetic, neural, and physiological bases of sleep. Through significant advances in our understanding of the regulation of sleep in flies, the field is poised to address several open questions about sleep, such as how the need for sleep is encoded, how molecular regulators of sleep are situated within brain networks, and what the functions of sleep are. Here, we describe key findings, open questions, and commonly used methods that have been used to inform existing theories and develop new ways of thinking about the function, regulation, and adaptability of sleep behavior.
Collapse
Affiliation(s)
- Divya Sitaraman
- Department of Psychology, College of Science, California State University, Hayward, California 94542, USA
| | | | - Casey Koochagian
- Neuroscience Program, Skidmore College, Saratoga Springs, New York 12866, USA
| |
Collapse
|
4
|
Shih MFM, Zhang J, Brown EB, Dubnau J, Keene AC. Targeted single cell expression profiling identifies integrators of sleep and metabolic state. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.614841. [PMID: 39386468 PMCID: PMC11463630 DOI: 10.1101/2024.09.25.614841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Animals modulate sleep in accordance with their internal and external environments. Metabolic cues are particularly potent regulators of sleep, allowing animals to alter their sleep timing and amount depending on food availability and foraging duration. The fruit fly, Drosophila melanogaster, suppresses sleep in response to acute food deprivation, presumably to forage for food. This process is dependent on a single pair of Lateral Horn Leucokinin (LHLK) neurons, that secrete the neuropeptide Leucokinin. These neurons signal to insulin producing cells and suppress sleep under periods of starvation. The identification of individual neurons that modulate sleep-metabolism interactions provides the opportunity to examine the cellular changes associated with sleep modulation. Here, we use single-cell sequencing of LHLK neurons to examine the transcriptional responses to starvation. We validate that a Patch-seq approach selectively isolates RNA from individual LHLK neurons. Single-cell CEL-Seq comparisons of LHLK neurons between fed and 24-hr starved flies identified 24 genes that are differentially expressed in accordance with starvation state. In total, 12 upregulated genes and 12 downregulated genes were identified. Gene-ontology analysis showed an enrichment for Attacins, a family of anti-microbial peptides, along with several transcripts with diverse roles in regulating cellular function. Targeted knockdown of differentially expressed genes identified multiple genes that function within LHLK neurons to regulate sleep-metabolism interactions. Functionally validated genes include an essential role for the E3 ubiquitin Ligase insomniac, the sorbitol dehydrogenase Sodh1, as well as AttacinC and AttacinB in starvation-induced sleep suppression. Taken together, these findings provide a pipeline for identifying novel regulators of sleep-metabolism interactions within individual neurons.
Collapse
Affiliation(s)
| | - Jiwei Zhang
- Department of Biology, Texas A&M University, College Station, TX 77840
| | | | - Joshua Dubnau
- Dept of Anesthesiology, Stony Brook School of Medicine, Stony Brook NY, 11794
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook NY, 11794
| | - Alex C. Keene
- Department of Biology, Texas A&M University, College Station, TX 77840
| |
Collapse
|
5
|
Van De Poll M, Tainton-Heap L, Troup M, van Swinderen B. Whole-Brain Electrophysiology and Calcium Imaging in Drosophila during Sleep and Wake. Cold Spring Harb Protoc 2024; 2024:pdb.top108394. [PMID: 38148172 DOI: 10.1101/pdb.top108394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
Sleep is likely a whole-brain phenomenon, with most of the brain probably benefiting from this state of decreased arousal. Recent advances in our understanding of some potential sleep functions, such as metabolite clearance and synaptic homeostasis, make it evident why the whole brain is likely impacted by sleep: All neurons have synapses, and all neurons produce waste metabolites. Sleep experiments in the fly Drosophila melanogaster suggest that diverse sleep functions appear to be conserved across all animals. Studies of brain activity during sleep in humans typically involve multidimensional data sets, such as those acquired by electroencephalograms (EEGs) or functional magnetic resonance imaging (fMRI), and these whole-brain read-outs often reveal important qualities of different sleep stages, such as changes in frequency dynamics or connectivity. Recently, various techniques have been developed that allow for the recording of neural activity simultaneously across multiple regions of the fly brain. These whole-brain-recording approaches will be important for better understanding sleep physiology and function, as they provide a more comprehensive view of neural dynamics during sleep and wake in a relevant model system. Here, we present a brief summary of some of the findings derived from sleep activity recording studies in sleeping Drosophila flies and discuss the value of electrophysiological versus calcium imaging techniques. Although these involve very different preparations, they both highlight the value of multidimensional data for studying sleep in this model system, like the use of both EEG and fMRI in humans.
Collapse
Affiliation(s)
- Matthew Van De Poll
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Lucy Tainton-Heap
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Michael Troup
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Bruno van Swinderen
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
6
|
Murakami H, Ko T, Ouchi H, Namba T, Ebihara S, Kobayashi S. Bifidobacterium adolescentis SBT2786 Improves Sleep Quality in Japanese Adults with Relatively High Levels of Stress: A Randomized, Double-Blind, Placebo-Controlled Study. Nutrients 2024; 16:1702. [PMID: 38892634 PMCID: PMC11174696 DOI: 10.3390/nu16111702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Sleep disorders associated with lifestyle changes and unhealthy habits are major public health concerns. Our previous study showed that Bifidobacterium adolescentis SBT2786 has a potent sleep-promoting effect on fruit flies. Fruit flies share many similarities with mammals, making them suitable model organisms for studying sleep. Thus, in the present study, we conducted a randomized, double-blind, placebo-controlled clinical trial to test whether SBT2786 has sleep-enhancing effects in humans. In this study, 61 participants in the SBT2786 group and 65 participants in the placebo group were analyzed. The results showed that SBT2786 increased sleep time; however, it predominantly increased light sleep and did not improve subjective sleep quality. Interestingly, mood improvement was observed. A subgroup analysis was conducted on participants with high stress levels, and results showed that these participants experienced an increase in sleep duration and an improvement in sleepiness upon waking up and reported feeling well-rested during the day. We concluded that SBT2786 may improve sleep quality, particularly in individuals experiencing high levels of stress, and that SBT2786 can be used as a dietary supplement to improve sleep and mood.
Collapse
Affiliation(s)
- Hiroki Murakami
- Milk Science Research Institute, MEGMILK SNOW BRAND Co., Ltd., 1-1-2 Minamidai, Kawagoe, Saitama 350-1165, Japan; (H.M.); (T.K.); (H.O.)
| | - Taro Ko
- Milk Science Research Institute, MEGMILK SNOW BRAND Co., Ltd., 1-1-2 Minamidai, Kawagoe, Saitama 350-1165, Japan; (H.M.); (T.K.); (H.O.)
| | - Haruka Ouchi
- Milk Science Research Institute, MEGMILK SNOW BRAND Co., Ltd., 1-1-2 Minamidai, Kawagoe, Saitama 350-1165, Japan; (H.M.); (T.K.); (H.O.)
| | - Toshiharu Namba
- CPCC Company Limited, 3-3-10 Nihonbashi Hongokucho, Chuo-ku, Tokyo 103-0021, Japan;
| | - Shukuko Ebihara
- Chiyoda Paramedical Care Clinic, 3-3-10 Nihonbashi Hongokucho, Chuo-ku, Tokyo 103-0021, Japan
| | - Shunjiro Kobayashi
- Milk Science Research Institute, MEGMILK SNOW BRAND Co., Ltd., 1-1-2 Minamidai, Kawagoe, Saitama 350-1165, Japan; (H.M.); (T.K.); (H.O.)
| |
Collapse
|
7
|
Frank MG. Molecular clues to the evolution of sleep. Sleep 2024; 47:zsae049. [PMID: 38367018 DOI: 10.1093/sleep/zsae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Indexed: 02/19/2024] Open
Affiliation(s)
- Marcos G Frank
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
- Gleason Institute for Neuroscience, Washington State University, Spokane, WA, USA
- Sleep Performance and Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| |
Collapse
|
8
|
Ko K, Ahn Y, Cheon GY, Suh HJ, Cho YJ, Park SS, Hong KB. Effects of Dendropanax morbiferus Leaf Extract on Sleep Parameters in Invertebrate and Vertebrate Models. Antioxidants (Basel) 2023; 12:1890. [PMID: 37891970 PMCID: PMC10604594 DOI: 10.3390/antiox12101890] [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: 09/15/2023] [Revised: 10/19/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Dendropanax morbiferus is highly valued in traditional medicine and has been used to alleviate the symptoms of numerous diseases owing to its excellent antioxidant activity. This study aimed to evaluate the sleep promotion and related signaling pathways of D. morbiferus extract (DE) via behavioral analysis, molecular biological techniques, and electrophysiological measurements in invertebrate and vertebrate models. In Drosophila, the group treated with 4% DE experienced decreased subjective nighttime movement and sleep bout and increased total sleeping time. Moreover, substantial changes in locomotor activity, including distance moved, velocity, and movement, were confirmed in the 4% DE-treated group. Compared to Drosophila in which insomnia and oxidative stress were induced by exposure to 0.1% caffeine, the DE-treated group improved sleep-related parameters to the level of the normal group. In the Drosophila model, exposure to 4% DE upregulated the expression of gamma-aminobutyric acid (GABA)-related receptors and serotonin receptor (5-HT1A), along with the expression of antioxidant-related factors, glutathione, and catalase. In the pentobarbital-induced sleep test using ICR mice, the duration of sleep was markedly increased by high concentration of DE. In addition, through the electroencephalography analysis of SD-rats, a significant increase in non-rapid-eye-movement sleep and delta waves was confirmed with high concentrations of DE administration. The increase in sleep time and improvement in sleep quality were confirmed to be related to the expression of altered GABA receptors and the enhancement of the contents of the neurotransmitters GABA and serotonin (5-HT) because of high DE administration. High-dose administration of DE also increased the expression of antioxidant-related factors in the brain and significantly decreased malondialdehyde content. Taken together, DE induced improvements in sleep quantity and quality by regulating neurotransmitter content and related receptor expression, along with high antioxidant activity, and may have a therapeutic effect on sleep disorders.
Collapse
Affiliation(s)
- Kayoung Ko
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Republic of Korea
| | - Yejin Ahn
- Research Group of Functional Food Materials, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea
| | - Ga Yeong Cheon
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Republic of Korea
| | - Hyung Joo Suh
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Yun Jae Cho
- R&D Center, JEJUPANATEK Inc., Jeju 63243, Republic of Korea
| | - Sung-Soo Park
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Republic of Korea
| | - Ki-Bae Hong
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Republic of Korea
| |
Collapse
|
9
|
Cattabriga G, Giordani G, Gargiulo G, Cavaliere V. Effect of aminergic signaling on the humoral innate immunity response of Drosophila. Front Physiol 2023; 14:1249205. [PMID: 37693001 PMCID: PMC10483126 DOI: 10.3389/fphys.2023.1249205] [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: 06/28/2023] [Accepted: 08/10/2023] [Indexed: 09/12/2023] Open
Abstract
Biogenic amines are crucial signaling molecules that modulate various physiological life functions both in vertebrates and invertebrates. In humans, these neurotransmitters influence the innate and adaptive immunity systems. In this work, we analyzed whether the aminergic neurotransmission of dopamine, serotonin, and octopamine could have an impact on the humoral innate immune response of Drosophila melanogaster. This is a powerful model system widely used to uncover the insect innate immunity mechanisms which are also conserved in mammals. We found that the neurotransmission of all these amines positively modulates the Toll-responsive antimicrobial peptide (AMP) drosomycin (drs) gene in adult flies infected with the Micrococcus luteus bacterium. Indeed, we showed that either blocking the neurotransmission in their specific aminergic neurons by expressing shibirets (Shits) or silencing the vesicular monoamine transporter gene (dVMAT) by RNAi caused a significantly reduced expression of the Toll-responsive drs gene. However, upon M. luteus infection, the block of aminergic transmission did not alter the expression of AMP attacin genes responding to the immune deficiency (Imd) and Toll pathways. Overall, our results not only reveal a neuroimmune function for biogenic amines in humoral immunity but also further highlight the complexity of the network controlling AMP gene regulation.
Collapse
Affiliation(s)
- Giulia Cattabriga
- Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum Università di Bologna, Bologna, Italy
| | - Giorgia Giordani
- Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum Università di Bologna, Bologna, Italy
| | - Giuseppe Gargiulo
- Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum Università di Bologna, Bologna, Italy
| | - Valeria Cavaliere
- Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum Università di Bologna, Bologna, Italy
- Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology (BAT Center), University of Napoli “Federico II”, Naples, Italy
| |
Collapse
|
10
|
Souto-Maior C, Serrano Negron YL, Harbison ST. Nonlinear expression patterns and multiple shifts in gene network interactions underlie robust phenotypic change in Drosophila melanogaster selected for night sleep duration. PLoS Comput Biol 2023; 19:e1011389. [PMID: 37561813 PMCID: PMC10443883 DOI: 10.1371/journal.pcbi.1011389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 08/22/2023] [Accepted: 07/25/2023] [Indexed: 08/12/2023] Open
Abstract
All but the simplest phenotypes are believed to result from interactions between two or more genes forming complex networks of gene regulation. Sleep is a complex trait known to depend on the system of feedback loops of the circadian clock, and on many other genes; however, the main components regulating the phenotype and how they interact remain an unsolved puzzle. Genomic and transcriptomic data may well provide part of the answer, but a full account requires a suitable quantitative framework. Here we conducted an artificial selection experiment for sleep duration with RNA-seq data acquired each generation. The phenotypic results are robust across replicates and previous experiments, and the transcription data provides a high-resolution, time-course data set for the evolution of sleep-related gene expression. In addition to a Hierarchical Generalized Linear Model analysis of differential expression that accounts for experimental replicates we develop a flexible Gaussian Process model that estimates interactions between genes. 145 gene pairs are found to have interactions that are different from controls. Our method appears to be not only more specific than standard correlation metrics but also more sensitive, finding correlations not significant by other methods. Statistical predictions were compared to experimental data from public databases on gene interactions. Mutations of candidate genes implicated by our results affected night sleep, and gene expression profiles largely met predicted gene-gene interactions.
Collapse
Affiliation(s)
- Caetano Souto-Maior
- Laboratory of Systems Genetics, Systems Biology Center, National Heart Lung and Blood Institute, Bethesda, Maryland, United States of America
| | - Yazmin L. Serrano Negron
- Laboratory of Systems Genetics, Systems Biology Center, National Heart Lung and Blood Institute, Bethesda, Maryland, United States of America
| | - Susan T. Harbison
- Laboratory of Systems Genetics, Systems Biology Center, National Heart Lung and Blood Institute, Bethesda, Maryland, United States of America
| |
Collapse
|
11
|
Goda T, Umezaki Y, Hamada FN. Molecular and Neural Mechanisms of Temperature Preference Rhythm in Drosophila melanogaster. J Biol Rhythms 2023; 38:326-340. [PMID: 37222551 PMCID: PMC10330063 DOI: 10.1177/07487304231171624] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Temperature influences animal physiology and behavior. Animals must set an appropriate body temperature to maintain homeostasis and maximize survival. Mammals set their body temperatures using metabolic and behavioral strategies. The daily fluctuation in body temperature is called the body temperature rhythm (BTR). For example, human body temperature increases during wakefulness and decreases during sleep. BTR is controlled by the circadian clock, is closely linked with metabolism and sleep, and entrains peripheral clocks located in the liver and lungs. However, the underlying mechanisms of BTR are largely unclear. In contrast to mammals, small ectotherms, such as Drosophila, control their body temperatures by choosing appropriate environmental temperatures. The preferred temperature of Drosophila increases during the day and decreases at night; this pattern is referred to as the temperature preference rhythm (TPR). As flies are small ectotherms, their body temperature is close to that of the surrounding environment. Thus, Drosophila TPR produces BTR, which exhibits a pattern similar to that of human BTR. In this review, we summarize the regulatory mechanisms of TPR, including recent studies that describe neuronal circuits relaying ambient temperature information to dorsal neurons (DNs). The neuropeptide diuretic hormone 31 (DH31) and its receptor (DH31R) regulate TPR, and a mammalian homolog of DH31R, the calcitonin receptor (CALCR), also plays an important role in mouse BTR regulation. In addition, both fly TPR and mammalian BTR are separately regulated from another clock output, locomotor activity rhythms. These findings suggest that the fundamental mechanisms of BTR regulation may be conserved between mammals and flies. Furthermore, we discuss the relationships between TPR and other physiological functions, such as sleep. The dissection of the regulatory mechanisms of Drosophila TPR could facilitate an understanding of mammalian BTR and the interaction between BTR and sleep regulation.
Collapse
Affiliation(s)
- Tadahiro Goda
- Department of Neurobiology, Physiology & Behavior, University of California, Davis, Davis, California
| | - Yujiro Umezaki
- Department of Neurobiology, Physiology & Behavior, University of California, Davis, Davis, California
| | - Fumika N. Hamada
- Department of Neurobiology, Physiology & Behavior, University of California, Davis, Davis, California
| |
Collapse
|
12
|
Pantalia M, Lin Z, Tener SJ, Qiao B, Tang G, Ulgherait M, O'Connor R, Delventhal R, Volpi J, Syed S, Itzhak N, Canman JC, Fernández MP, Shirasu-Hiza M. Drosophila mutants lacking the glial neurotransmitter-modifying enzyme Ebony exhibit low neurotransmitter levels and altered behavior. Sci Rep 2023; 13:10411. [PMID: 37369755 PMCID: PMC10300103 DOI: 10.1038/s41598-023-36558-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Inhibitors of enzymes that inactivate amine neurotransmitters (dopamine, serotonin), such as catechol-O-methyltransferase (COMT) and monoamine oxidase (MAO), are thought to increase neurotransmitter levels and are widely used to treat Parkinson's disease and psychiatric disorders, yet the role of these enzymes in regulating behavior remains unclear. Here, we investigated the genetic loss of a similar enzyme in the model organism Drosophila melanogaster. Because the enzyme Ebony modifies and inactivates amine neurotransmitters, its loss is assumed to increase neurotransmitter levels, increasing behaviors such as aggression and courtship and decreasing sleep. Indeed, ebony mutants have been described since 1960 as "aggressive mutants," though this behavior has not been quantified. Using automated machine learning-based analyses, we quantitatively confirmed that ebony mutants exhibited increased aggressive behaviors such as boxing but also decreased courtship behaviors and increased sleep. Through tissue-specific knockdown, we found that ebony's role in these behaviors was specific to glia. Unexpectedly, direct measurement of amine neurotransmitters in ebony brains revealed that their levels were not increased but reduced. Thus, increased aggression is the anomalous behavior for this neurotransmitter profile. We further found that ebony mutants exhibited increased aggression only when fighting each other, not when fighting wild-type controls. Moreover, fights between ebony mutants were less likely to end with a clear winner than fights between controls or fights between ebony mutants and controls. In ebony vs. control fights, ebony mutants were more likely to win. Together, these results suggest that ebony mutants exhibit prolonged aggressive behavior only in a specific context, with an equally dominant opponent.
Collapse
Affiliation(s)
- Meghan Pantalia
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Zhi Lin
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Samantha J Tener
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Bing Qiao
- Department of Physics, University of Miami, Coral Gables, FL, 33146, USA
| | - Grace Tang
- Department of Neuroscience and Behavior, Barnard College, New York, NY, 10027, USA
| | - Matthew Ulgherait
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Reed O'Connor
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | | | - Julia Volpi
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Sheyum Syed
- Department of Physics, University of Miami, Coral Gables, FL, 33146, USA
| | - Nissim Itzhak
- Division of Human Genetics and Metabolic Disease, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pediatrics, Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Julie C Canman
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - María Paz Fernández
- Department of Neuroscience and Behavior, Barnard College, New York, NY, 10027, USA
| | - Mimi Shirasu-Hiza
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
13
|
Bhattacharya D, Górska-Andrzejak J, Abaquita TAL, Pyza E. Effects of adenosine receptor overexpression and silencing in neurons and glial cells on lifespan, fitness, and sleep of Drosophila melanogaster. Exp Brain Res 2023:10.1007/s00221-023-06649-y. [PMID: 37335362 DOI: 10.1007/s00221-023-06649-y] [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: 10/03/2022] [Accepted: 05/28/2023] [Indexed: 06/21/2023]
Abstract
A single adenosine receptor gene (dAdoR) has been detected in Drosophila melanogaster. However, its function in different cell types of the nervous system is mostly unknown. Therefore, we overexpressed or silenced the dAdoR gene in eye photoreceptors, all neurons, or glial cells and examined the fitness of flies, the amount and daily pattern of sleep, and the influence of dAdoR silencing on Bruchpilot (BRP) presynaptic protein. Furthermore, we examined the dAdoR and brp gene expression in young and old flies. We found that a higher level of dAdoR in the retina photoreceptors, all neurons, and glial cells negatively influenced the survival rate and lifespan of male and female Drosophila in a cell-dependent manner and to a different extent depending on the age of the flies. In old flies, expression of both dAdoR and brp was higher than in young ones. An excess of dAdoR in neurons improved climbing in older individuals. It also influenced sleep by lengthening nighttime sleep and siesta. In turn, silencing of dAdoR decreased the lifespan of flies, although it increased the survival rate of young flies. It hindered the climbing of older males and females, but did not change sleep. Silencing also affected the daily pattern of BRP abundance, especially when dAdoR expression was decreased in glial cells. The obtained results indicate the role of adenosine and dAdoR in the regulation of fitness in flies that is based on communication between neurons and glial cells, and the effect of glial cells on synapses.
Collapse
Affiliation(s)
| | | | | | - Elżbieta Pyza
- Department of Cell Biology and Imaging, Jagellonian University, Kraków, Poland.
| |
Collapse
|
14
|
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
|
15
|
Landis JE, Sungu K, Sipe H, Copeland JM. RNAi of Complex I and V of the electron transport chain in glutamate neurons extends life span, increases sleep, and decreases locomotor activity in Drosophila melanogaster. PLoS One 2023; 18:e0286828. [PMID: 37319260 PMCID: PMC10270625 DOI: 10.1371/journal.pone.0286828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 05/24/2023] [Indexed: 06/17/2023] Open
Abstract
RNAi targeting the electron transport chain has been proven to prolong life span in many different species, and experiments specifically with Drosophila melanogaster and Caenorhabditis elegans have shown a distinct role for neurons. To determine which subset of neurons is implicated in this life span extension, we used the GAL4/UAS system to activate RNAi against genes of Complex I and Complex V. We found life span extension of 18-24% with two glutamate neuron (D42 and VGlut) GAL4 lines. We used the GAL80 system to determine if the overlapping set of glutamate neurons in these two GAL4 lines imparts the life span extension. Limiting GAL4 activity to non-VGlut glutamate neurons in the D42 background failed to extend life span, suggesting that glutamate neurons have an important role in aging. Interestingly, RNAi of the electron transport chain in D42 glutamate neurons also caused an increase in daytime and nighttime sleep and a decrease in nighttime locomotor activity. Changes to sleep patterns and prolonged life span were not accompanied by any changes in female fertility or response to starvation. Our findings demonstrate that a small subset of neurons can control life span, and further studies can look into the contributions made by glutamate neurons.
Collapse
Affiliation(s)
- Jessie E. Landis
- Department of Biology, Eastern Mennonite University, Harrisonburg, VA, United States of America
| | - Kevin Sungu
- Department of Biology, Eastern Mennonite University, Harrisonburg, VA, United States of America
| | - Hannah Sipe
- Department of Biology, Eastern Mennonite University, Harrisonburg, VA, United States of America
| | - Jeffrey M. Copeland
- Department of Biology, Eastern Mennonite University, Harrisonburg, VA, United States of America
| |
Collapse
|
16
|
Ko T, Murakami H, Kobayashi S, Kamikouchi A, Ishimoto H. Behavioral screening of sleep-promoting effects of human intestinal and food-associated bacteria on Drosophila melanogaster. Genes Cells 2023; 28:433-446. [PMID: 36914986 PMCID: PMC11447928 DOI: 10.1111/gtc.13025] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023]
Abstract
Commensal microbes influence various aspects of vertebrate and invertebrate brain function. We previously reported that Lactiplantibacillus plantarum SBT2227 promotes sleep in the fruit fly, Drosophila melanogaster. However, how widely the sleep-promoting effects are conserved in gut bacterial species remains unknown. In this study, we orally administered human intestinal and food-associated bacterial species (39 in total) to flies and investigated their effects on sleep. Six species of bacteria were found to have significant sleep-promoting effects. Of these, we further investigated Bifidobacterium adolescentis, which had the greatest sleep-promoting effect, and found that the strength of the sleep effect varied among strains of the same bacterial species. The B. adolescentis strains BA2786 and BA003 showed strong and weak effects on sleep, respectively. Transcriptome characteristics compared between the heads of flies treated with BA2786 or BA003 revealed that the gene expression of the insulin-like receptor (InR) was increased in BA2786-fed flies. Furthermore, a heterozygous mutation in InR suppressed the sleep-promoting effect of BA2786. These results suggest that orally administered sleep-promoting bacteria (at least BA2786), may act on insulin signaling to modulate brain function for sleep.
Collapse
Affiliation(s)
- Taro Ko
- Graduate School of Science, Nagoya University, Nagoya, Aichi, Japan
- Milk Science Research Institute, Megmilk Snow Brand Co., Ltd, Kawagoe, Saitama, Japan
| | - Hiroki Murakami
- Graduate School of Science, Nagoya University, Nagoya, Aichi, Japan
- Milk Science Research Institute, Megmilk Snow Brand Co., Ltd, Kawagoe, Saitama, Japan
| | - Shunjiro Kobayashi
- Graduate School of Science, Nagoya University, Nagoya, Aichi, Japan
- Milk Science Research Institute, Megmilk Snow Brand Co., Ltd, Kawagoe, Saitama, Japan
| | - Azusa Kamikouchi
- Graduate School of Science, Nagoya University, Nagoya, Aichi, Japan
| | - Hiroshi Ishimoto
- Graduate School of Science, Nagoya University, Nagoya, Aichi, Japan
| |
Collapse
|
17
|
Bologa AM, Stoica I, Constantin ND, Ecovoiu AA. The Landscape of the DNA Transposons in the Genome of the Horezu_LaPeri Strain of Drosophila melanogaster. INSECTS 2023; 14:494. [PMID: 37367310 DOI: 10.3390/insects14060494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023]
Abstract
Natural transposons (NTs) represent mobile DNA sequences found in both prokaryotic and eukaryotic genomes. Drosophila melanogaster (the fruit fly) is a eukaryotic model organism with NTs standing for about 20% of its genome and has contributed significantly to the understanding of various aspects of transposon biology. Our study describes an accurate approach designed to map class II transposons (DNA transposons) in the genome of the Horezu_LaPeri fruit fly strain, consecutive to Oxford Nanopore Technology sequencing. A whole genome bioinformatics analysis was conducted using Genome ARTIST_v2, LoRTE and RepeatMasker tools to identify DNA transposons insertions. Then, a gene ontology enrichment analysis was performed in order to evaluate the potential adaptive role of some DNA transposons insertions. Herein, we describe DNA transposon insertions specific for the Horezu_LaPeri genome and a predictive functional analysis of some insertional alleles. The PCR validation of P-element insertions specific for this fruit fly strain, along with a putative consensus sequence for the KP element, is also reported. Overall, the genome of the Horezu_LaPeri strain contains several insertions of DNA transposons associated with genes known to be involved in adaptive processes. For some of these genes, insertional alleles obtained via mobilization of the artificial transposons were previously reported. This is a very alluring aspect, as it suggests that insertional mutagenesis experiments conducting adaptive predictions for laboratory strains may be confirmed by mirroring insertions which are expected to be found at least in some natural fruit fly strains.
Collapse
Affiliation(s)
- Alexandru Marian Bologa
- Department of Genetics, Faculty of Biology, University of Bucharest, 060101 Bucharest, Romania
| | - Ileana Stoica
- Department of Genetics, Faculty of Biology, University of Bucharest, 060101 Bucharest, Romania
| | | | - Alexandru Al Ecovoiu
- Department of Genetics, Faculty of Biology, University of Bucharest, 060101 Bucharest, Romania
| |
Collapse
|
18
|
Palermo J, Chesi A, Zimmerman A, Sonti S, Pahl MC, Lasconi C, Brown EB, Pippin JA, Wells AD, Doldur-Balli F, Mazzotti DR, Pack AI, Gehrman PR, Grant SF, Keene AC. Variant-to-gene mapping followed by cross-species genetic screening identifies GPI-anchor biosynthesis as a regulator of sleep. SCIENCE ADVANCES 2023; 9:eabq0844. [PMID: 36608130 PMCID: PMC9821868 DOI: 10.1126/sciadv.abq0844] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 12/05/2022] [Indexed: 05/13/2023]
Abstract
Genome-wide association studies (GWAS) in humans have identified loci robustly associated with several heritable diseases or traits, yet little is known about the functional roles of the underlying causal variants in regulating sleep duration or quality. We applied an ATAC-seq/promoter focused Capture C strategy in human iPSC-derived neural progenitors to carry out a "variant-to-gene" mapping campaign that identified 88 candidate sleep effector genes connected to relevant GWAS signals. To functionally validate the role of the implicated effector genes in sleep regulation, we performed a neuron-specific RNA interference screen in the fruit fly, Drosophila melanogaster, followed by validation in zebrafish. This approach identified a number of genes that regulate sleep including a critical role for glycosylphosphatidylinositol (GPI)-anchor biosynthesis. These results provide the first physical variant-to-gene mapping of human sleep genes followed by a model organism-based prioritization, revealing a conserved role for GPI-anchor biosynthesis in sleep regulation.
Collapse
Affiliation(s)
- Justin Palermo
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| | - Alessandra Chesi
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amber Zimmerman
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA 19104, USA
| | - Shilpa Sonti
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Matthew C. Pahl
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Chiara Lasconi
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Elizabeth B. Brown
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| | - James A. Pippin
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Andrew D. Wells
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA 19104, USA
| | - Fusun Doldur-Balli
- Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA 19104, USA
| | - Diego R. Mazzotti
- Division of Medical Informatics, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66103, USA
- Division of Pulmonary Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Allan I. Pack
- Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA 19104, USA
| | - Phillip R. Gehrman
- Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA 19104, USA
| | - Struan F.A. Grant
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Divisions of Human Genetics and Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alex C. Keene
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
19
|
Cabana-Domínguez J, Antón-Galindo E, Fernàndez-Castillo N, Singgih EL, O'Leary A, Norton WH, Strekalova T, Schenck A, Reif A, Lesch KP, Slattery D, Cormand B. The translational genetics of ADHD and related phenotypes in model organisms. Neurosci Biobehav Rev 2023; 144:104949. [PMID: 36368527 DOI: 10.1016/j.neubiorev.2022.104949] [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: 07/01/2022] [Revised: 11/02/2022] [Accepted: 11/05/2022] [Indexed: 11/10/2022]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a highly prevalent neurodevelopmental disorder resulting from the interaction between genetic and environmental risk factors. It is well known that ADHD co-occurs frequently with other psychiatric disorders due, in part, to shared genetics factors. Although many studies have contributed to delineate the genetic landscape of psychiatric disorders, their specific molecular underpinnings are still not fully understood. The use of animal models can help us to understand the role of specific genes and environmental stimuli-induced epigenetic modifications in the pathogenesis of ADHD and its comorbidities. The aim of this review is to provide an overview on the functional work performed in rodents, zebrafish and fruit fly and highlight the generated insights into the biology of ADHD, with a special focus on genetics and epigenetics. We also describe the behavioral tests that are available to study ADHD-relevant phenotypes and comorbid traits in these models. Furthermore, we have searched for new models to study ADHD and its comorbidities, which can be useful to test potential pharmacological treatments.
Collapse
Affiliation(s)
- Judit Cabana-Domínguez
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia, Spain.
| | - Ester Antón-Galindo
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia, Spain
| | - Noèlia Fernàndez-Castillo
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia, Spain
| | - Euginia L Singgih
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Aet O'Leary
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany; Division of Neuropsychopharmacology, Department of Psychology, University of Tartu, Tartu, Estonia
| | - William Hg Norton
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Tatyana Strekalova
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany, and Department of Neuropsychology and Psychiatry, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, the Netherlands
| | - Annette Schenck
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany, and Department of Neuropsychology and Psychiatry, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, the Netherlands
| | - David Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Bru Cormand
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia, Spain.
| |
Collapse
|
20
|
Huang S, Piao C, Beuschel CB, Zhao Z, Sigrist SJ. A brain-wide form of presynaptic active zone plasticity orchestrates resilience to brain aging in Drosophila. PLoS Biol 2022; 20:e3001730. [PMID: 36469518 PMCID: PMC9721493 DOI: 10.1371/journal.pbio.3001730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 11/07/2022] [Indexed: 12/10/2022] Open
Abstract
The brain as a central regulator of stress integration determines what is threatening, stores memories, and regulates physiological adaptations across the aging trajectory. While sleep homeostasis seems to be linked to brain resilience, how age-associated changes intersect to adapt brain resilience to life history remains enigmatic. We here provide evidence that a brain-wide form of presynaptic active zone plasticity ("PreScale"), characterized by increases of active zone scaffold proteins and synaptic vesicle release factors, integrates resilience by coupling sleep, longevity, and memory during early aging of Drosophila. PreScale increased over the brain until mid-age, to then decreased again, and promoted the age-typical adaption of sleep patterns as well as extended longevity, while at the same time it reduced the ability of forming new memories. Genetic induction of PreScale also mimicked early aging-associated adaption of sleep patterns and the neuronal activity/excitability of sleep control neurons. Spermidine supplementation, previously shown to suppress early aging-associated PreScale, also attenuated the age-typical sleep pattern changes. Pharmacological induction of sleep for 2 days in mid-age flies also reset PreScale, restored memory formation, and rejuvenated sleep patterns. Our data suggest that early along the aging trajectory, PreScale acts as an acute, brain-wide form of presynaptic plasticity to steer trade-offs between longevity, sleep, and memory formation in a still plastic phase of early brain aging.
Collapse
Affiliation(s)
- Sheng Huang
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| | - Chengji Piao
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| | - Christine B. Beuschel
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| | - Zhiying Zhao
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - Stephan J. Sigrist
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| |
Collapse
|
21
|
Knapp EM, Kaiser A, Arnold RC, Sampson MM, Ruppert M, Xu L, Anderson MI, Bonanno SL, Scholz H, Donlea JM, Krantz DE. Mutation of the Drosophila melanogaster serotonin transporter dSERT impacts sleep, courtship, and feeding behaviors. PLoS Genet 2022; 18:e1010289. [PMID: 36409783 PMCID: PMC9721485 DOI: 10.1371/journal.pgen.1010289] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 12/05/2022] [Accepted: 11/08/2022] [Indexed: 11/22/2022] Open
Abstract
The Serotonin Transporter (SERT) regulates extracellular serotonin levels and is the target of most current drugs used to treat depression. The mechanisms by which inhibition of SERT activity influences behavior are poorly understood. To address this question in the model organism Drosophila melanogaster, we developed new loss of function mutations in Drosophila SERT (dSERT). Previous studies in both flies and mammals have implicated serotonin as an important neuromodulator of sleep, and our newly generated dSERT mutants show an increase in total sleep and altered sleep architecture that is mimicked by feeding the SSRI citalopram. Differences in daytime versus nighttime sleep architecture as well as genetic rescue experiments unexpectedly suggest that distinct serotonergic circuits may modulate daytime versus nighttime sleep. dSERT mutants also show defects in copulation and food intake, akin to the clinical side effects of SSRIs and consistent with the pleomorphic influence of serotonin on the behavior of D. melanogaster. Starvation did not overcome the sleep drive in the mutants and in male dSERT mutants, the drive to mate also failed to overcome sleep drive. dSERT may be used to further explore the mechanisms by which serotonin regulates sleep and its interplay with other complex behaviors.
Collapse
Affiliation(s)
- Elizabeth M. Knapp
- Department of Psychiatry, University of California, Los Angeles, California, United States of America
| | - Andrea Kaiser
- Department of Biology, Institute of Zoology, Albertus-Magnus University of Cologne, Cologne, Germany
| | - Rebecca C. Arnold
- Department of Psychiatry, University of California, Los Angeles, California, United States of America
| | - Maureen M. Sampson
- Department of Psychiatry, University of California, Los Angeles, California, United States of America
| | - Manuela Ruppert
- Department of Biology, Institute of Zoology, Albertus-Magnus University of Cologne, Cologne, Germany
| | - Li Xu
- Department of Biology, Institute of Zoology, Albertus-Magnus University of Cologne, Cologne, Germany
| | | | - Shivan L. Bonanno
- Department of Psychiatry, University of California, Los Angeles, California, United States of America
| | - Henrike Scholz
- Department of Biology, Institute of Zoology, Albertus-Magnus University of Cologne, Cologne, Germany
| | - Jeffrey M. Donlea
- Department of Neurobiology, University of California, Los Angeles, California, United States of America
| | - David E. Krantz
- Department of Psychiatry, University of California, Los Angeles, California, United States of America
| |
Collapse
|
22
|
Pandey P, Kaur G, Babu K. Crosstalk between neurons and glia through G-protein coupled receptors: Insights from Caenorhabditis elegans. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 193:119-144. [PMID: 36357074 DOI: 10.1016/bs.pmbts.2022.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The past decades have witnessed a dogmatic shift from glia as supporting cells in the nervous system to their active roles in neurocentric functions. Neurons and glia communicate and show bidirectional responses through tripartite synapses. Studies across species indicate that neurotransmitters released by neurons are perceived by glial receptors, which allow for gliotransmitter release. These gliotransmitters can result in activation of neurons via neuronal GPCR receptors. However, studies of these molecular interactions are in their infancy. Caenorhabditis elegans has a conserved neuron-glia architectural repertoire with molecular and functional resemblance to mammals. Further, glia in C. elegans can be manipulated through ablation and mutations allowing for deciphering of glial dependent processes in vivo at single glial resolutions. Here, we will review recent findings from vertebrate and invertebrate organisms with a focus on how C. elegans can be used to advance our understanding of neuron-glia interactions through GPCRs.
Collapse
Affiliation(s)
- Pratima Pandey
- Indian Institute of Science Education and Research, Mohali, Punjab, India.
| | - Gazaldeep Kaur
- National Agri-Food Biotechnology Institute, Mohali, Punjab, India
| | - Kavita Babu
- Indian Institute of Science, Bangalore, Karnataka, India.
| |
Collapse
|
23
|
Brown EB, Klok J, Keene AC. Measuring metabolic rate in single flies during sleep and waking states via indirect calorimetry. J Neurosci Methods 2022; 376:109606. [PMID: 35483506 PMCID: PMC9310448 DOI: 10.1016/j.jneumeth.2022.109606] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 04/13/2022] [Accepted: 04/18/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND Drosophila melanogaster is a leading genetic model for studying the neural regulation of sleep. Sleep is associated with changes in behavior and physiological state that are largely conserved across species. The investigation of sleep in flies has predominantly focused on behavioral readouts of sleep because physiological measurements, including changes in brain activity and metabolic rate, are less accessible. We have previously used stop-flow indirect calorimetry to measure whole body metabolic rate in single flies and have shown that in flies, like mammals, metabolic rate is reduced during sleep. NEW METHOD Here, we describe a modified version of this system that allows for efficient and highly sensitive acquisition of CO2 output from single flies. RESULTS In this modified system, we show that sleep-dependent changes in metabolic rate are diminished in aging flies, supporting the notion that sleep quality is reduced as flies age. We also describe a modification that allows for simultaneous acquisition of CO2 and O2 levels, providing a respiratory quotient that quantifies how metabolic stores are utilized. We find that the respiratory quotient identified in flies on an all-sugar diet is suggestive of lipogenesis, where the dietary sugar provided to the flies is being converted to fat. COMPARISON WITH EXISTING METHODS AND CONCLUSIONS Taken together, the measurement of metabolic rate via indirect calorimetry not only provides a physiological readout of sleep depth, but also provides insight the metabolic regulation of nutrient utilization, with broad applications to genetic studies in flies.
Collapse
Affiliation(s)
- Elizabeth B Brown
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| | - Jaco Klok
- Sable Systems International, Las Vegas, NV 89032, USA
| | - Alex C Keene
- Department of Biology, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
24
|
Ko T, Murakami H, Kamikouchi A, Ishimoto H. Biogenic action of Lactobacillus plantarum SBT2227 promotes sleep in Drosophila melanogaster. iScience 2022; 25:104626. [PMID: 35811846 PMCID: PMC9257349 DOI: 10.1016/j.isci.2022.104626] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/25/2022] [Accepted: 06/13/2022] [Indexed: 11/25/2022] Open
Abstract
Lactic acid bacteria (LAB) influence multiple aspects of host brain function via the production of active metabolites in the gut, which is known as the pre/probiotic action. However, little is known about the biogenic effects of LAB on host brain function. Here, we reported that the Lactobacillus plantarum SBT2227 promoted sleep in Drosophila melanogaster. Administration of SBT2227 primarily increased the amount of sleep and decreased sleep latency at the beginning of night-time. The sleep-promoting effects of SBT2227 were independent of the existing gut flora. Furthermore, heat treatment or mechanical crushing of SBT2227 did not suppress the sleep-promoting effects, indicative of biogenic action. Transcriptome analysis and RNAi mini-screening for gut-derived peptide hormones revealed the requirement of neuropeptide F, a homolog of the mammalian neuropeptide Y, for the action of SBT2227. These biogenic effects of SBT2227 on the host sleep provide new insights into the interaction between the brain and gut bacteria. Lactobacillus plantarum SBT2227 promotes sleep at the onset of nighttime Existing intestinal microbes do not affect the SBT2227 sleep effect Heat-stable intracellular/intramembrane components are candidates for active substances Neuropeptide F is required for the sleep-promoting effect of SBT2227
Collapse
|
25
|
Chaturvedi R, Stork T, Yuan C, Freeman MR, Emery P. Astrocytic GABA transporter controls sleep by modulating GABAergic signaling in Drosophila circadian neurons. Curr Biol 2022; 32:1895-1908.e5. [PMID: 35303417 PMCID: PMC9090989 DOI: 10.1016/j.cub.2022.02.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 01/11/2022] [Accepted: 02/23/2022] [Indexed: 11/16/2022]
Abstract
A precise balance between sleep and wakefulness is essential to sustain a good quality of life and optimal brain function. GABA is known to play a key and conserved role in sleep control, and GABAergic tone should, therefore, be tightly controlled in sleep circuits. Here, we examined the role of the astrocytic GABA transporter (GAT) in sleep regulation using Drosophila melanogaster. We found that a hypomorphic gat mutation (gat33-1) increased sleep amount, decreased sleep latency, and increased sleep consolidation at night. Interestingly, sleep defects were suppressed when gat33-1 was combined with a mutation disrupting wide-awake (wake), a gene that regulates the cell-surface levels of the GABAA receptor resistance to dieldrin (RDL) in the wake-promoting large ventral lateral neurons (l-LNvs). Moreover, RNAi knockdown of rdl and its modulators dnlg4 and wake in these circadian neurons also suppressed gat33-1 sleep phenotypes. Brain immunohistochemistry showed that GAT-expressing astrocytes were located near RDL-positive l-LNv cell bodies and dendritic processes. We concluded that astrocytic GAT decreases GABAergic tone and RDL activation in arousal-promoting LNvs, thus determining proper sleep amount and quality in Drosophila.
Collapse
Affiliation(s)
- Ratna Chaturvedi
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Tobias Stork
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Chunyan Yuan
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Marc R Freeman
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Patrick Emery
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
26
|
Karam CS, Williams BL, Jones SK, Javitch JA. The Role of the Dopamine Transporter in the Effects of Amphetamine on Sleep and Sleep Architecture in Drosophila. Neurochem Res 2022; 47:177-189. [PMID: 33630236 PMCID: PMC8384956 DOI: 10.1007/s11064-021-03275-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 01/12/2021] [Accepted: 02/10/2021] [Indexed: 12/26/2022]
Abstract
The dopamine transporter (DAT) mediates the inactivation of released dopamine (DA) through its reuptake, and thereby plays an important homeostatic role in dopaminergic neurotransmission. Amphetamines exert their stimulant effects by targeting DAT and inducing the reverse transport of DA, leading to a dramatic increase of extracellular DA. Animal models have proven critical to investigating the molecular and cellular mechanisms underlying transporter function and its modulation by psychostimulants such as amphetamine. Here we establish a behavioral model for amphetamine action using adult Drosophila melanogaster. We use it to characterize the effects of amphetamine on sleep and sleep architecture. Our data show that amphetamine induces hyperactivity and disrupts sleep in a DA-dependent manner. Flies that do not express a functional DAT (dDAT null mutants) have been shown to be hyperactive and to exhibit significantly reduced sleep at baseline. Our data show that, in contrast to its action in control flies, amphetamine decreases the locomotor activity of dDAT null mutants and restores their sleep by modulating distinct aspects of sleep structure. To begin to explore the circuitry involved in the actions of amphetamine on sleep, we also describe the localization of dDAT throughout the fly brain, particularly in neuropils known to regulate sleep. Together, our data establish Drosophila as a robust model for studying the regulatory mechanisms that govern DAT function and psychostimulant action.
Collapse
Affiliation(s)
- Caline S Karam
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Brenna L Williams
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Sandra K Jones
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Jonathan A Javitch
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA.
- Department of Pharmacology, Columbia University Vagelos College of Physicians and Surgeons, 1051 Riverside Dr, Unit 19, New York, NY, 10032, USA.
| |
Collapse
|
27
|
Parmar S, Tadavarty R, Sastry BR. G-protein coupled receptors and synaptic plasticity in sleep deprivation. World J Psychiatry 2021; 11:954-980. [PMID: 34888167 PMCID: PMC8613756 DOI: 10.5498/wjp.v11.i11.954] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/05/2021] [Accepted: 09/19/2021] [Indexed: 02/06/2023] Open
Abstract
Insufficient sleep has been correlated to many physiological and psychoneurological disorders. Over the years, our understanding of the state of sleep has transcended from an inactive period of rest to a more active state involving important cellular and molecular processes. In addition, during sleep, electrophysiological changes also occur in pathways in specific regions of the mammalian central nervous system (CNS). Activity mediated synaptic plasticity in the CNS can lead to long-term and sometimes permanent strengthening and/or weakening synaptic strength affecting neuronal network behaviour. Memory consolidation and learning that take place during sleep cycles, can be affected by changes in synaptic plasticity during sleep disturbances. G-protein coupled receptors (GPCRs), with their versatile structural and functional attributes, can regulate synaptic plasticity in CNS and hence, may be potentially affected in sleep deprived conditions. In this review, we aim to discuss important functional changes that can take place in the CNS during sleep and sleep deprivation and how changes in GPCRs can lead to potential problems with therapeutics with pharmacological interventions.
Collapse
Affiliation(s)
- Shweta Parmar
- Department of Anesthesiology, Pharmacology and Therapeutics, The University of British Columbia, Vancouver V6T 1Z3, British Columbia, Canada
| | - Ramakrishna Tadavarty
- Department of Anesthesiology, Pharmacology and Therapeutics, The University of British Columbia, Vancouver V6T 1Z3, British Columbia, Canada
| | - Bhagavatula R Sastry
- Department of Anesthesiology, Pharmacology and Therapeutics, The University of British Columbia, Vancouver V6T 1Z3, British Columbia, Canada
| |
Collapse
|
28
|
Van De Poll MN, van Swinderen B. Balancing Prediction and Surprise: A Role for Active Sleep at the Dawn of Consciousness? Front Syst Neurosci 2021; 15:768762. [PMID: 34803618 PMCID: PMC8602873 DOI: 10.3389/fnsys.2021.768762] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/08/2021] [Indexed: 11/14/2022] Open
Abstract
The brain is a prediction machine. Yet the world is never entirely predictable, for any animal. Unexpected events are surprising, and this typically evokes prediction error signatures in mammalian brains. In humans such mismatched expectations are often associated with an emotional response as well, and emotional dysregulation can lead to cognitive disorders such as depression or schizophrenia. Emotional responses are understood to be important for memory consolidation, suggesting that positive or negative 'valence' cues more generally constitute an ancient mechanism designed to potently refine and generalize internal models of the world and thereby minimize prediction errors. On the other hand, abolishing error detection and surprise entirely (as could happen by generalization or habituation) is probably maladaptive, as this might undermine the very mechanism that brains use to become better prediction machines. This paradoxical view of brain function as an ongoing balance between prediction and surprise suggests a compelling approach to study and understand the evolution of consciousness in animals. In particular, this view may provide insight into the function and evolution of 'active' sleep. Here, we propose that active sleep - when animals are behaviorally asleep but their brain seems awake - is widespread beyond mammals and birds, and may have evolved as a mechanism for optimizing predictive processing in motile creatures confronted with constantly changing environments. To explore our hypothesis, we progress from humans to invertebrates, investigating how a potential role for rapid eye movement (REM) sleep in emotional regulation in humans could be re-examined as a conserved sleep function that co-evolved alongside selective attention to maintain an adaptive balance between prediction and surprise. This view of active sleep has some interesting implications for the evolution of subjective awareness and consciousness in animals.
Collapse
Affiliation(s)
| | - Bruno van Swinderen
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
29
|
Murakami K, Palermo J, Stanhope BA, Gibbs AG, Keene AC. A screen for sleep and starvation resistance identifies a wake-promoting role for the auxiliary channel unc79. G3 (BETHESDA, MD.) 2021; 11:6300522. [PMID: 34849820 PMCID: PMC8496288 DOI: 10.1093/g3journal/jkab199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/25/2021] [Indexed: 11/22/2022]
Abstract
The regulation of sleep and metabolism are highly interconnected, and dysregulation of sleep is linked to metabolic diseases that include obesity, diabetes, and heart disease. Furthermore, both acute and long-term changes in diet potently impact sleep duration and quality. To identify novel factors that modulate interactions between sleep and metabolic state, we performed a genetic screen for their roles in regulating sleep duration, starvation resistance, and starvation-dependent modulation of sleep. This screen identified a number of genes with potential roles in regulating sleep, metabolism, or both processes. One such gene encodes the auxiliary ion channel UNC79, which was implicated in both the regulation of sleep and starvation resistance. Genetic knockdown or mutation of unc79 results in flies with increased sleep duration, as well as increased starvation resistance. Previous findings have shown that unc79 is required in pacemaker for 24-hours circadian rhythms. Here, we find that unc79 functions in the mushroom body, but not pacemaker neurons, to regulate sleep duration and starvation resistance. Together, these findings reveal spatially localized separable functions of unc79 in the regulation of circadian behavior, sleep, and metabolic function.
Collapse
Affiliation(s)
- Kazuma Murakami
- Department of Biological Sciences, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Justin Palermo
- Department of Biological Sciences, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Bethany A Stanhope
- Department of Biological Sciences, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Allen G Gibbs
- Department of Biological Sciences, University of Nevada, Las Vegas, Las Vegas, NV 89154, USA
| | - Alex C Keene
- Department of Biological Sciences, Florida Atlantic University, Jupiter, FL 33458, USA
| |
Collapse
|
30
|
Huang S, Sigrist SJ. Presynaptic and postsynaptic long-term plasticity in sleep homeostasis. Curr Opin Neurobiol 2021; 69:1-10. [DOI: 10.1016/j.conb.2020.11.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/03/2020] [Accepted: 11/15/2020] [Indexed: 12/25/2022]
|
31
|
Sleep contributes to preference for novel food odours in Drosophila melanogaster. Sci Rep 2021; 11:9395. [PMID: 33931708 PMCID: PMC8087676 DOI: 10.1038/s41598-021-88967-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 04/20/2021] [Indexed: 11/21/2022] Open
Abstract
The importance of sleep in maintaining cognitive functions such as learning and memory has been reported in both vertebrates and invertebrates. Previous studies demonstrated that sleep deprivation impaired the olfactory memory retention of fruit flies as described in the classical conditioning paradigm. Here, we show that sleep deprivation leads to a preference for the odours of the rearing environment in Drosophila melanogaster. Flies whose sleep had been disturbed with periodic rotation stimuli during night-time preferred apple cider vinegar (ACV) to broth, while this preference was lower in flies without sleep deprivation and those rotated during daytime. Experiments using single odours showed an increase in responses to ACV due to sleep deprivation. These results suggest that sleep functions in food odour preference. Flies grown on medium supplemented with ACV showed greater preference for ACV, and those grown with broth supplementation showed a greater preference for broth under sleep-deprived conditions. These results suggest that flies with night-time sleep deprivation become attached to the environment on which they have developed, and that sleep contributes to preference for novel food odours. This study offers an approach to investigating the interaction between sleep and neural disorders concerning cognitive deficits towards novel stimuli.
Collapse
|
32
|
Tasman K, Rands SA, Hodge JJL. The Power of Drosophila melanogaster for Modeling Neonicotinoid Effects on Pollinators and Identifying Novel Mechanisms. Front Physiol 2021; 12:659440. [PMID: 33967830 PMCID: PMC8096932 DOI: 10.3389/fphys.2021.659440] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/29/2021] [Indexed: 02/06/2023] Open
Abstract
Neonicotinoids are the most widely used insecticides in the world and are implicated in the widespread population declines of insects including pollinators. Neonicotinoids target nicotinic acetylcholine receptors which are expressed throughout the insect central nervous system, causing a wide range of sub-lethal effects on non-target insects. Here, we review the potential of the fruit fly Drosophila melanogaster to model the sub-lethal effects of neonicotinoids on pollinators, by utilizing its well-established assays that allow rapid identification and mechanistic characterization of these effects. We compare studies on the effects of neonicotinoids on lethality, reproduction, locomotion, immunity, learning, circadian rhythms and sleep in D. melanogaster and a range of pollinators. We also highlight how the genetic tools available in D. melanogaster, such as GAL4/UAS targeted transgene expression system combined with RNAi lines to any gene in the genome including the different nicotinic acetylcholine receptor subunit genes, are set to elucidate the mechanisms that underlie the sub-lethal effects of these common pesticides. We argue that studying pollinators and D. melanogaster in tandem allows rapid elucidation of mechanisms of action, which translate well from D. melanogaster to pollinators. We focus on the recent identification of novel and important sublethal effects of neonicotinoids on circadian rhythms and sleep. The comparison of effects between D. melanogaster and pollinators and the use of genetic tools to identify mechanisms make a powerful partnership for the future discovery and testing of more specific insecticides.
Collapse
Affiliation(s)
- Kiah Tasman
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Sean A. Rands
- School of Biological Sciences, University of Bristol, Bristol, United Kingdom
| | - James J. L. Hodge
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
33
|
Panchin Y, Kovalzon VM. Total Wake: Natural, Pathological, and Experimental Limits to Sleep Reduction. Front Neurosci 2021; 15:643496. [PMID: 33897357 PMCID: PMC8058214 DOI: 10.3389/fnins.2021.643496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/26/2021] [Indexed: 11/16/2022] Open
Abstract
Sleep is not considered a pathological state, but it consumes a third of conscious human life. This share is much more than most optimistic life extension forecasts that biotechnologies or experimental and medical interventions can offer. Are there insurmountable physical or biological limitations to reducing the duration of sleep? How far can it be avoided without fatal consequences? What means can reduce the length of sleep? It is widely accepted that sleep is necessary for long-term survival. Here we review the limited yet intriguing evidence that is not consistent with this notion. We concentrate on clinical cases of complete and partial loss of sleep and on human mutations that result in a short sleep phenotype. These observations are supported by new animal studies and are discussed from the perspective of sleep evolution. Two separate hypotheses suggest distinct approaches for remodeling our sleep machinery. If sleep serves an unidentified vital physiological function, this indispensable function has to be identified before "sleep prosthesis" (technical, biological, or chemical) can be developed. If sleep has no vital function, but rather represents a timing mechanism for adaptive inactivity, sleep could be reduced by forging the sleep generation system itself, with no adverse effects.
Collapse
Affiliation(s)
- Yuri Panchin
- Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia
- Department of Mathematical Methods in Biology, Belozersky Institute, Lomonosov Moscow State University, Moscow, Russia
| | - Vladimir M. Kovalzon
- Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia
- Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
34
|
Neonicotinoids disrupt memory, circadian behaviour and sleep. Sci Rep 2021; 11:2061. [PMID: 33479461 PMCID: PMC7820356 DOI: 10.1038/s41598-021-81548-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 12/28/2020] [Indexed: 02/06/2023] Open
Abstract
Globally, neonicotinoids are the most used insecticides, despite their well-documented sub-lethal effects on beneficial insects. Neonicotinoids are nicotinic acetylcholine receptor agonists. Memory, circadian rhythmicity and sleep are essential for efficient foraging and pollination and require nicotinic acetylcholine receptor signalling. The effect of field-relevant concentrations of the European Union-banned neonicotinoids: imidacloprid, clothianidin, thiamethoxam and thiacloprid were tested on Drosophila memory, circadian rhythms and sleep. Field-relevant concentrations of imidacloprid, clothianidin and thiamethoxam disrupted learning, behavioural rhythmicity and sleep whilst thiacloprid exposure only affected sleep. Exposure to imidacloprid and clothianidin prevented the day/night remodelling and accumulation of pigment dispersing factor (PDF) neuropeptide in the dorsal terminals of clock neurons. Knockdown of the neonicotinoid susceptible Dα1 and Dβ2 nicotinic acetylcholine receptor subunits in the mushroom bodies or clock neurons recapitulated the neonicotinoid like deficits in memory or sleep/circadian behaviour respectively. Disruption of learning, circadian rhythmicity and sleep are likely to have far-reaching detrimental effects on beneficial insects in the field.
Collapse
|
35
|
Souto-Maior C, Serrano Negron YL, Harbison ST. Natural selection on sleep duration in Drosophila melanogaster. Sci Rep 2020; 10:20652. [PMID: 33244154 PMCID: PMC7691507 DOI: 10.1038/s41598-020-77680-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/10/2020] [Indexed: 11/30/2022] Open
Abstract
Sleep is ubiquitous across animal species, but why it persists is not well understood. Here we observe natural selection act on Drosophila sleep by relaxing bi-directional artificial selection for extreme sleep duration for 62 generations. When artificial selection was suspended, sleep increased in populations previously selected for short sleep. Likewise, sleep decreased in populations previously selected for long sleep when artificial selection was relaxed. We measured the corresponding changes in the allele frequencies of genomic variants responding to artificial selection. The allele frequencies of these variants reversed course in response to relaxed selection, and for short sleepers, the changes exceeded allele frequency changes that would be expected under random genetic drift. These observations suggest that the variants are causal polymorphisms for sleep duration responding to natural selection pressure. These polymorphisms may therefore pinpoint the most important regions of the genome maintaining variation in sleep duration.
Collapse
Affiliation(s)
- Caetano Souto-Maior
- Laboratory of Systems Genetics, Systems Biology Center, National Heart Lung and Blood Institute, Bethesda, MD, USA
| | - Yazmin L Serrano Negron
- Laboratory of Systems Genetics, Systems Biology Center, National Heart Lung and Blood Institute, Bethesda, MD, USA
| | - Susan T Harbison
- Laboratory of Systems Genetics, Systems Biology Center, National Heart Lung and Blood Institute, Bethesda, MD, USA.
| |
Collapse
|
36
|
Zhao X, Li Y, Zhao Z, Du J. Extra sex combs buffers sleep-related stresses through regulating Heat shock proteins. FASEB J 2020; 35:e21190. [PMID: 33220007 DOI: 10.1096/fj.202001303rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 11/11/2022]
Abstract
The impact of global warming on the life of the earth is increasingly concerned. Previous studies indicated that temperature changes have a serious impact on insect sleep. Sleep is critical for animals as it has many important physiological functions. It is of great significance to study the regulation mechanism of temperature-induced sleep changes for understanding the impact of global warming on insects. More importantly, understanding how these pressures regulate sleep can provide insights into improving sleep. In this study, we found that extra sex combs (ESC) are a regulatory factor in this process. Our data showed that ESC was an upstream negative regulatory factor of Heat shock proteins (Hsps), and it could regulate sleep in mushroom and ellipsoid of Drosophila. ESC mutation exaggerates the sleep change caused by temperature, while buffering the shortening of life caused by sleep deprivation. These phenotypes can be rescued by Hsps mutants. Therefore, we concluded that the ESC buffers sleep-related stresses through regulating Hsps.
Collapse
Affiliation(s)
- Xianguo Zhao
- Department of Entomology, MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Yahong Li
- Department of Entomology, MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Zhangwu Zhao
- Department of Entomology, MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Juan Du
- Department of Entomology, MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| |
Collapse
|
37
|
Ajayi OM, Eilerts DF, Bailey ST, Vinauger C, Benoit JB. Do Mosquitoes Sleep? Trends Parasitol 2020; 36:888-897. [PMID: 32952061 PMCID: PMC8094063 DOI: 10.1016/j.pt.2020.08.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/17/2020] [Accepted: 08/20/2020] [Indexed: 10/23/2022]
Abstract
Sleep is a phenomenon conserved across the animal kingdom, where studies on Drosophila melanogaster have revealed that sleep phenotypes and molecular underpinnings are similar to those in mammals. However, little is known about sleep in blood-feeding arthropods, which have a critical role in public health as disease vectors. Specifically, sleep studies in mosquitoes are lacking despite considerable focus on how circadian processes, which have a central role in regulating sleep/wake cycles, impact activity, feeding, and immunity. Here, we review observations which suggest that sleep-like states likely occur in mosquitoes and discuss the potential role of sleep in relation to mosquito biology and their ability to function as disease vectors.
Collapse
Affiliation(s)
- Oluwaseun M Ajayi
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45221, USA. @mail.uc.edu
| | - Diane F Eilerts
- Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Samuel T Bailey
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Clément Vinauger
- Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Joshua B Benoit
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45221, USA. @uc.edu
| |
Collapse
|
38
|
Mazzotta GM, Damulewicz M, Cusumano P. Better Sleep at Night: How Light Influences Sleep in Drosophila. Front Physiol 2020; 11:997. [PMID: 33013437 PMCID: PMC7498665 DOI: 10.3389/fphys.2020.00997] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/22/2020] [Indexed: 01/25/2023] Open
Abstract
Sleep-like states have been described in Drosophila and the mechanisms and factors that generate and define sleep-wake profiles in this model organism are being thoroughly investigated. Sleep is controlled by both circadian and homeostatic mechanisms, and environmental factors such as light, temperature, and social stimuli are fundamental in shaping and confining sleep episodes into the correct time of the day. Among environmental cues, light seems to have a prominent function in modulating the timing of sleep during the 24 h and, in this review, we will discuss the role of light inputs in modulating the distribution of the fly sleep-wake cycles. This phenomenon is of growing interest in the modern society, where artificial light exposure during the night is a common trait, opening the possibility to study Drosophila as a model organism for investigating shift-work disorders.
Collapse
Affiliation(s)
| | - Milena Damulewicz
- Department of Cell Biology and Imaging, Jagiellonian University, Kraków, Poland
| | - Paola Cusumano
- Department of Biology, University of Padova, Padua, Italy
| |
Collapse
|
39
|
Kang YY, Wachi Y, Engdorf E, Fumagalli E, Wang Y, Myers J, Massey S, Greiss A, Xu S, Roman G. Normal Ethanol Sensitivity and Rapid Tolerance Require the G Protein Receptor Kinase 2 in Ellipsoid Body Neurons in Drosophila. Alcohol Clin Exp Res 2020; 44:1686-1699. [PMID: 32573992 PMCID: PMC7485117 DOI: 10.1111/acer.14396] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 06/12/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND G protein signaling pathways are key neuromodulatory mechanisms for behaviors and neurological functions that affect the impact of ethanol (EtOH) on locomotion, arousal, and synaptic plasticity. Here, we report a novel role for the Drosophila G protein-coupled receptor kinase 2 (GPRK2) as a member of the GRK4/5/6 subfamily in modulating EtOH-induced behaviors. METHODS We studied the requirement of Drosophila Gprk2 for naïve sensitivity to EtOH sedation and ability of the fly to develop rapid tolerance after a single exposure to EtOH, using the loss of righting reflex (LORR) and fly group activity monitor (FlyGrAM) assays. RESULTS Loss-of-function Gprk2 mutants demonstrate an increase in alcohol-induced hyperactivity, reduced sensitivity to the sedative effects of EtOH, and diminished rapid tolerance after a single intoxicating exposure. The requirement for Gprk2 in EtOH sedation and rapid tolerance maps to ellipsoid body neurons within the Drosophila brain, suggesting that wild-type Gprk2 is required for modulation of locomotion and alertness. However, even though Gprk2 loss of function leads to decreased and fragmented sleep, this change in the sleep state does not depend on Gprk2 expression in the ellipsoid body. CONCLUSION Our work on GPRK2 has established a role for this GRK4/5/6 subfamily member in EtOH sensitivity and rapid tolerance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Shiyu Xu
- University of HoustonHoustonTexas
| | - Gregg Roman
- University of MississippiUniversityMississippi
| |
Collapse
|
40
|
Van der Auwera P, Frooninckx L, Buscemi K, Vance RT, Watteyne J, Mirabeau O, Temmerman L, De Haes W, Fancsalszky L, Gottschalk A, Raizen DM, Nelson MD, Schoofs L, Beets I. RPamide neuropeptides NLP-22 and NLP-2 act through GnRH-like receptors to promote sleep and wakefulness in C. elegans. Sci Rep 2020; 10:9929. [PMID: 32555288 PMCID: PMC7303124 DOI: 10.1038/s41598-020-66536-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 05/22/2020] [Indexed: 01/26/2023] Open
Abstract
Sleep and wakefulness are fundamental behavioral states of which the underlying molecular principles are becoming slowly elucidated. Transitions between these states require the coordination of multiple neurochemical and modulatory systems. In Caenorhabditis elegans sleep occurs during a larval transition stage called lethargus and is induced by somnogenic neuropeptides. Here, we identify two opposing neuropeptide/receptor signaling pathways: NLP-22 promotes behavioral quiescence, whereas NLP-2 promotes movement during lethargus, by signaling through gonadotropin-releasing hormone (GnRH) related receptors. Both NLP-2 and NLP-22 belong to the RPamide neuropeptide family and share sequence similarities with neuropeptides of the bilaterian GnRH, adipokinetic hormone (AKH) and corazonin family. RPamide neuropeptides dose-dependently activate the GnRH/AKH-like receptors GNRR-3 and GNRR-6 in a cellular receptor activation assay. In addition, nlp-22-induced locomotion quiescence requires the receptor gnrr-6. By contrast, wakefulness induced by nlp-2 overexpression is diminished by deletion of either gnrr-3 or gnrr-6. nlp-2 is expressed in a pair of olfactory AWA neurons and cycles with larval periodicity, as reported for nlp-22, which is expressed in RIA. Our data suggest that the somnogenic NLP-22 neuropeptide signals through GNRR-6, and that both GNRR-3 and GNRR-6 are required for the wake-promoting action of NLP-2 neuropeptides.
Collapse
Affiliation(s)
- Petrus Van der Auwera
- Department of Biology, KU Leuven, Naamsestraat 59, 3000, Leuven, Belgium
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University, Max-von-Laue-Strasse 15, D-60438, Frankfurt, Germany
| | - Lotte Frooninckx
- Department of Biology, KU Leuven, Naamsestraat 59, 3000, Leuven, Belgium
| | - Kristen Buscemi
- Department of Biology, Saint Joseph's University, 5600 City Ave, Philadelphia, PA, 19131, USA
| | - Ryan T Vance
- Department of Biology, Saint Joseph's University, 5600 City Ave, Philadelphia, PA, 19131, USA
| | - Jan Watteyne
- Department of Biology, KU Leuven, Naamsestraat 59, 3000, Leuven, Belgium
| | | | - Liesbet Temmerman
- Department of Biology, KU Leuven, Naamsestraat 59, 3000, Leuven, Belgium
| | - Wouter De Haes
- Department of Biology, KU Leuven, Naamsestraat 59, 3000, Leuven, Belgium
| | - Luca Fancsalszky
- Department of Biology, KU Leuven, Naamsestraat 59, 3000, Leuven, Belgium
| | - Alexander Gottschalk
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University, Max-von-Laue-Strasse 15, D-60438, Frankfurt, Germany
| | - David M Raizen
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 415 Curie Blvd, Philadelphia, PA, 19104, USA
| | - Matthew D Nelson
- Department of Biology, Saint Joseph's University, 5600 City Ave, Philadelphia, PA, 19131, USA
| | - Liliane Schoofs
- Department of Biology, KU Leuven, Naamsestraat 59, 3000, Leuven, Belgium.
| | - Isabel Beets
- Department of Biology, KU Leuven, Naamsestraat 59, 3000, Leuven, Belgium.
| |
Collapse
|
41
|
Dissel S. Drosophila as a Model to Study the Relationship Between Sleep, Plasticity, and Memory. Front Physiol 2020; 11:533. [PMID: 32547415 PMCID: PMC7270326 DOI: 10.3389/fphys.2020.00533] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 04/30/2020] [Indexed: 12/28/2022] Open
Abstract
Humans spend nearly a third of their life sleeping, yet, despite decades of research the function of sleep still remains a mystery. Sleep has been linked with various biological systems and functions, including metabolism, immunity, the cardiovascular system, and cognitive functions. Importantly, sleep appears to be present throughout the animal kingdom suggesting that it must provide an evolutionary advantage. Among the many possible functions of sleep, the relationship between sleep, and cognition has received a lot of support. We have all experienced the negative cognitive effects associated with a night of sleep deprivation. These can include increased emotional reactivity, poor judgment, deficit in attention, impairment in learning and memory, and obviously increase in daytime sleepiness. Furthermore, many neurological diseases like Alzheimer’s disease often have a sleep disorder component. In some cases, the sleep disorder can exacerbate the progression of the neurological disease. Thus, it is clear that sleep plays an important role for many brain functions. In particular, sleep has been shown to play a positive role in the consolidation of long-term memory while sleep deprivation negatively impacts learning and memory. Importantly, sleep is a behavior that is adapted to an individual’s need and influenced by many external and internal stimuli. In addition to being an adaptive behavior, sleep can also modulate plasticity in the brain at the level of synaptic connections between neurons and neuronal plasticity influences sleep. Understanding how sleep is modulated by internal and external stimuli and how sleep can modulate memory and plasticity is a key question in neuroscience. In order to address this question, several animal models have been developed. Among them, the fruit fly Drosophila melanogaster with its unparalleled genetics has proved to be extremely valuable. In addition to sleep, Drosophila has been shown to be an excellent model to study many complex behaviors, including learning, and memory. This review describes our current knowledge of the relationship between sleep, plasticity, and memory using the fly model.
Collapse
Affiliation(s)
- Stephane Dissel
- Department of Molecular Biology and Biochemistry, School of Biological and Chemical Sciences, University of Missouri-Kansas City, Kansas City, MO, United States
| |
Collapse
|
42
|
A Pilot Study on Sleep Quality, Forgiveness, Religion, Spirituality, and General Health of Women Living in a Homeless Mission. Holist Nurs Pract 2020; 34:49-56. [DOI: 10.1097/hnp.0000000000000362] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
43
|
Hill VM, O’Connor RM, Shirasu-Hiza M. Tired and stressed: Examining the need for sleep. Eur J Neurosci 2020; 51:494-508. [PMID: 30295966 PMCID: PMC6453762 DOI: 10.1111/ejn.14197] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 09/04/2018] [Accepted: 09/18/2018] [Indexed: 12/31/2022]
Abstract
A key feature of circadian rhythms is the sleep/wake cycle. Sleep causes reduced responsiveness to the environment, which puts animals in a particularly vulnerable state; yet sleep has been conserved throughout evolution, indicating that it fulfils a vital purpose. A core function of sleep across species has not been identified, but substantial advances in sleep research have been made in recent years using the genetically tractable model organism, Drosophila melanogaster. This review describes the universality of sleep, the regulation of sleep, and current theories on the function of sleep, highlighting a historical and often overlooked theory called the Free Radical Flux Theory of Sleep. Additionally, we summarize our recent work with short-sleeping Drosophila mutants and other genetic and pharmacological tools for manipulating sleep which supports an antioxidant theory of sleep and demonstrates a bi-directional relationship between sleep and oxidative stress.
Collapse
Affiliation(s)
- Vanessa M. Hill
- Department of Genetics and Development; Columbia University Medical Center; NY, NY, 10032; USA
| | - Reed M. O’Connor
- Department of Genetics and Development; Columbia University Medical Center; NY, NY, 10032; USA
| | - Mimi Shirasu-Hiza
- Department of Genetics and Development; Columbia University Medical Center; NY, NY, 10032; USA
| |
Collapse
|
44
|
De Nobrega AK, Lyons LC. Aging and the clock: Perspective from flies to humans. Eur J Neurosci 2020; 51:454-481. [PMID: 30269400 PMCID: PMC6441388 DOI: 10.1111/ejn.14176] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 09/10/2018] [Accepted: 09/17/2018] [Indexed: 12/15/2022]
Abstract
Endogenous circadian oscillators regulate molecular, cellular and physiological rhythms, synchronizing tissues and organ function to coordinate activity and metabolism with environmental cycles. The technological nature of modern society with round-the-clock work schedules and heavy reliance on personal electronics has precipitated a striking increase in the incidence of circadian and sleep disorders. Circadian dysfunction contributes to an increased risk for many diseases and appears to have adverse effects on aging and longevity in animal models. From invertebrate organisms to humans, the function and synchronization of the circadian system weakens with age aggravating the age-related disorders and pathologies. In this review, we highlight the impacts of circadian dysfunction on aging and longevity and the reciprocal effects of aging on circadian function with examples from Drosophila to humans underscoring the highly conserved nature of these interactions. Additionally, we review the potential for using reinforcement of the circadian system to promote healthy aging and mitigate age-related pathologies. Advancements in medicine and public health have significantly increased human life span in the past century. With the demographics of countries worldwide shifting to an older population, there is a critical need to understand the factors that shape healthy aging. Drosophila melanogaster, as a model for aging and circadian interactions, has the capacity to facilitate the rapid advancement of research in this area and provide mechanistic insights for targeted investigations in mammals.
Collapse
Affiliation(s)
- Aliza K De Nobrega
- Program in Neuroscience, Department of Biological Science, Florida State University, Tallahassee, Florida
| | - Lisa C Lyons
- Program in Neuroscience, Department of Biological Science, Florida State University, Tallahassee, Florida
| |
Collapse
|
45
|
Xie J, Wang D, Ling S, Yang G, Yang Y, Chen W. High-Salt Diet Causes Sleep Fragmentation in Young Drosophila Through Circadian Rhythm and Dopaminergic Systems. Front Neurosci 2019; 13:1271. [PMID: 31849585 PMCID: PMC6895215 DOI: 10.3389/fnins.2019.01271] [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: 08/21/2019] [Accepted: 11/08/2019] [Indexed: 12/21/2022] Open
Abstract
Salt (sodium chloride) is an essential dietary requirement, but excessive consumption has long-term adverse consequences. A high-salt diet (HSD) increases the risk of chronic diseases such as cardiovascular conditions and diabetes and is also associated with poor sleep quality. Little is known, however, about the neural circuit mechanisms that mediate HSD-induced sleep changes. In this study, we sought to identify the effects of HSD on the sleep and related neural circuit mechanisms of Drosophila. Strikingly, we found that HSD causes young Drosophila to exhibit a fragmented sleep phenotype similar to that of normal aging individuals. Importantly, we further showed that HSD slightly impairs circadian rhythms and that the HSD-induced sleep changes are dependent on the circadian rhythm system. In addition, we demonstrated that HSD-induced sleep changes are dopaminergic-system dependent. Together, these results provide insight into how elevated salt in the diet can affect sleep quality.
Collapse
Affiliation(s)
- Jiayu Xie
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, China
| | - Danfeng Wang
- Institute of Applied Ecology, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Shengan Ling
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, China
| | - Guang Yang
- Institute of Applied Ecology, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yufeng Yang
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, China
| | - Wenfeng Chen
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, China
| |
Collapse
|
46
|
Molecular mechanisms and physiological importance of circadian rhythms. Nat Rev Mol Cell Biol 2019; 21:67-84. [PMID: 31768006 DOI: 10.1038/s41580-019-0179-2] [Citation(s) in RCA: 631] [Impact Index Per Article: 105.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2019] [Indexed: 12/12/2022]
Abstract
To accommodate daily recurring environmental changes, animals show cyclic variations in behaviour and physiology, which include prominent behavioural states such as sleep-wake cycles but also a host of less conspicuous oscillations in neurological, metabolic, endocrine, cardiovascular and immune functions. Circadian rhythmicity is created endogenously by genetically encoded molecular clocks, whose components cooperate to generate cyclic changes in their own abundance and activity, with a periodicity of about a day. Throughout the body, such molecular clocks convey temporal control to the function of organs and tissues by regulating pertinent downstream programmes. Synchrony between the different circadian oscillators and resonance with the solar day is largely enabled by a neural pacemaker, which is directly responsive to certain environmental cues and able to transmit internal time-of-day representations to the entire body. In this Review, we discuss aspects of the circadian clock in Drosophila melanogaster and mammals, including the components of these molecular oscillators, the function and mechanisms of action of central and peripheral clocks, their synchronization and their relevance to human health.
Collapse
|
47
|
Suo S, Harada K, Matsuda S, Kyo K, Wang M, Maruyama K, Awaji T, Tsuboi T. Sexually Dimorphic Regulation of Behavioral States by Dopamine in Caenorhabditis elegans. J Neurosci 2019; 39:4668-4683. [PMID: 30988167 PMCID: PMC6561698 DOI: 10.1523/jneurosci.2985-18.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 11/21/2022] Open
Abstract
Sex differences in behavior allow animals to effectively mate and reproduce. However, the mechanism by which biological sex regulates behavioral states, which underlie the regulation of sex-shared behaviors, such as locomotion, is largely unknown. In this study, we studied sex differences in the behavioral states of Caenorhabditis elegans and found that males spend less time in a low locomotor activity state than hermaphrodites and that dopamine generates this sex difference. In males, dopamine reduces the low activity state by acting in the same pathway as polycystic kidney disease-related genes that function in male-specific neurons. In hermaphrodites, dopamine increases the low activity state by suppression of octopamine signaling in the sex-shared SIA neurons, which have reduced responsiveness to octopamine in males. Furthermore, dopamine promotes exploration both inside and outside of bacterial lawn (the food source) in males and suppresses it in hermaphrodites. These results demonstrate that sexually dimorphic signaling allows the same neuromodulator to promote adaptive behavior for each sex.SIGNIFICANCE STATEMENT The mechanisms that generate sex differences in sex-shared behaviors, including locomotion, are not well understood. We show that there are sex differences in the regulation of behavioral states in the model animal Caenorhabditis elegans Dopamine promotes the high locomotor activity state in males, which must search for mates to reproduce, and suppresses it in self-fertilizing hermaphrodites through distinct molecular mechanisms. This study demonstrates that sex-specific signaling generates sex differences in the regulation of behavioral states, which in turn modulates the locomotor activity to suit reproduction for each sex.
Collapse
Affiliation(s)
- Satoshi Suo
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, Saitama, 350-0495, Japan,
| | - Kazuki Harada
- Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Tokyo, 153-8902, Japan
| | - Shogo Matsuda
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, 113-0033, Japan, and
| | - Koki Kyo
- Department of Human Sciences, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, 080-8555, Japan
| | - Min Wang
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, Saitama, 350-0495, Japan
| | - Kei Maruyama
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, Saitama, 350-0495, Japan
| | - Takeo Awaji
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, Saitama, 350-0495, Japan
| | - Takashi Tsuboi
- Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Tokyo, 153-8902, Japan
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, 113-0033, Japan, and
| |
Collapse
|
48
|
Daywake, an Anti-siesta Gene Linked to a Splicing-Based Thermostat from an Adjoining Clock Gene. Curr Biol 2019; 29:1728-1734.e4. [PMID: 31080079 DOI: 10.1016/j.cub.2019.04.039] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/26/2019] [Accepted: 04/12/2019] [Indexed: 01/25/2023]
Abstract
Sleep is fundamental to animal survival but is a vulnerable state that also limits how much time can be devoted to critical wake-dependent activities [1]. Although many animals are day-active and sleep at night, they exhibit a midday nap, or "siesta," that can vary in intensity and is usually more prominent on warm days. In humans, the balance between maintaining the wake state or sleeping during the day has important health implications [2], but the mechanisms underlying this dynamic regulation are poorly understood. Using the well-established Drosophila melanogaster animal model to study sleep [3], we identify a new wake-sleep regulator that we term daywake (dyw). dyw encodes a juvenile hormone-binding protein [4] that functions in neurons as a day-specific anti-siesta gene, with little effect on sleep levels during the nighttime or in the absence of light. Remarkably, dyw expression is stimulated in trans via cold-enhanced splicing of the dmpi8 intron [5] from the reverse-oriented but slightly overlapping period (per) clock gene [6]. The functionally integrated dmpi8-dyw genetic unit operates as a "behavioral temperate acclimator" by increasingly counterbalancing siesta-promoting pathways as daily temperatures become cooler and carry reduced risks from daytime heat exposure. While daily patterns of when animals are awake and when they sleep are largely scheduled by the circadian timing system, dyw implicates a less recognized class of modulatory wake-sleep regulators that primarily function to enhance flexibility in wake-sleep preference, a behavioral plasticity that is commonly observed in animals during the midday, raising the possibility of shared mechanisms.
Collapse
|