1
|
Zenge C, Ordureau A. Ubiquitin system mutations in neurological diseases. Trends Biochem Sci 2024; 49:875-887. [PMID: 38972780 PMCID: PMC11455613 DOI: 10.1016/j.tibs.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/31/2024] [Accepted: 06/12/2024] [Indexed: 07/09/2024]
Abstract
Neuronal ubiquitin balance impacts the fate of countless cellular proteins, and its disruption is associated with various neurological disorders. The ubiquitin system is critical for proper neuronal cell state transitions and the clearance of misfolded or aggregated proteins that threaten cellular integrity. This article reviews the state of and recent advancements in our understanding of the disruptions to components of the ubiquitin system, in particular E3 ligases and deubiquitylases, in neurodevelopmental and neurodegenerative diseases. Specific focus is on enzymes with recent progress in their characterization, including identifying enzyme-substrate pairs, the use of stem cell and animal models, and the development of therapeutics for ubiquitin-related diseases.
Collapse
Affiliation(s)
- Colin Zenge
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Alban Ordureau
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
2
|
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
|
3
|
Malik DM, Rhoades SD, Kain P, Sengupta A, Sehgal A, Weljie AM. Altered Metabolism during the Dark Period in Drosophila Short Sleep Mutants. J Proteome Res 2024; 23:3823-3836. [PMID: 38836855 DOI: 10.1021/acs.jproteome.4c00106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Sleep is regulated via circadian mechanisms, but effects of sleep disruption on physiological rhythms, in particular metabolic cycling, remain unclear. To examine this question, we probed diurnal metabolic alterations of two Drosophila short sleep mutants, fumin and sleepless. Samples were collected with high temporal sampling (every 2 h) over 24 h under a 12:12 light:dark cycle, and profiling was done using an ion-switching LCMS/MS method. Fewer metabolites with 24 h oscillations were noted with short sleep (50 and 46 in fumin and sleepless, BH. Q < 0.2 by RAIN analysis) compared to a wild-type control (iso31, 63 with BH. Q < 0.2), and peak phases of the sleep mutants were consolidated into two major phase peaks at mid-day and middle of night. Overall, altered nicotinate/nicotinamide, alanine/aspartate/glutamate, acetylcholine, glyoxylate/dicarboxylate, and TCA cycle metabolism were observed in the short sleep mutants, indicative of increased energetic demand and oxidative stress compared to wild type. Both changes in cycling and discriminant models suggest unique alterations in the dark period indicative of constrained metabolic networks. Thus, we conclude that sleep loss alters metabolic function uniquely throughout the day, and further examination of specific mechanisms is warranted.
Collapse
Affiliation(s)
- Dania M Malik
- Pharmacology Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Seth D Rhoades
- Pharmacology Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Fulgens Consulting, LLC, Cambridge, Massachusetts 02142, United States
| | - Pinky Kain
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Arjun Sengupta
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Amita Sehgal
- Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Aalim M Weljie
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
4
|
Chowdhury B, Shafer OT. Drosophila sleep homeostasis in sickness and in health. Sleep 2024; 47:zsae128. [PMID: 38899406 DOI: 10.1093/sleep/zsae128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Indexed: 06/21/2024] Open
Affiliation(s)
- Budhaditya Chowdhury
- The Advanced Science Research Center, The City University of New York; The Graduate Center at the City University of New York, New York, NY, USA
| | - Orie T Shafer
- The Advanced Science Research Center, The City University of New York; The Graduate Center at the City University of New York, New York, NY, USA
| |
Collapse
|
5
|
ElGrawani W, Sun G, Kliem FP, Sennhauser S, Pierre-Ferrer S, Rosi-Andersen A, Boccalaro I, Bethge P, Heo WD, Helmchen F, Adamantidis AR, Forger DB, Robles MS, Brown SA. BDNF-TrkB signaling orchestrates the buildup process of local sleep. Cell Rep 2024; 43:114500. [PMID: 39046880 DOI: 10.1016/j.celrep.2024.114500] [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: 12/11/2023] [Revised: 05/15/2024] [Accepted: 06/27/2024] [Indexed: 07/27/2024] Open
Abstract
Sleep debt accumulates during wakefulness, leading to increased slow wave activity (SWA) during sleep, an encephalographic marker for sleep need. The use-dependent demands of prior wakefulness increase sleep SWA locally. However, the circuitry and molecular identity of this "local sleep" remain unclear. Using pharmacology and optogenetic perturbations together with transcriptomics, we find that cortical brain-derived neurotrophic factor (BDNF) regulates SWA via the activation of tyrosine kinase B (TrkB) receptor and cAMP-response element-binding protein (CREB). We map BDNF/TrkB-induced sleep SWA to layer 5 (L5) pyramidal neurons of the cortex, independent of neuronal firing per se. Using mathematical modeling, we here propose a model of how BDNF's effects on synaptic strength can increase SWA in ways not achieved through increased firing alone. Proteomic analysis further reveals that TrkB activation enriches ubiquitin and proteasome subunits. Together, our study reveals that local SWA control is mediated by BDNF-TrkB-CREB signaling in L5 excitatory cortical neurons.
Collapse
Affiliation(s)
- Waleed ElGrawani
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland.
| | - Guanhua Sun
- Department of Mathematics, University of Michigan, Ann Arbor, MI, USA
| | - Fabian P Kliem
- Institute of Medical Psychology and Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Germany
| | - Simon Sennhauser
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Sara Pierre-Ferrer
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland
| | - Alex Rosi-Andersen
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland
| | - Ida Boccalaro
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, Bern, Switzerland
| | - Philipp Bethge
- Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland; Brain Research Institute, University of Zurich, Zurich, Switzerland
| | - Won Do Heo
- Department of Biological Science, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Fritjof Helmchen
- Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland; Brain Research Institute, University of Zurich, Zurich, Switzerland; University Research Priority Program (URPP), Adaptive Brain Circuits in Development and Learning, University of Zurich, Zurich, Switzerland
| | - Antoine R Adamantidis
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, Bern, Switzerland.
| | - Daniel B Forger
- Department of Mathematics, University of Michigan, Ann Arbor, MI, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA.
| | - Maria S Robles
- Institute of Medical Psychology and Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Germany.
| | - Steven A Brown
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
6
|
Weiss JT, Blundell MZ, Singh P, Donlea JM. Sleep deprivation drives brain-wide changes in cholinergic presynapse abundance in Drosophila melanogaster. Proc Natl Acad Sci U S A 2024; 121:e2312664121. [PMID: 38498719 PMCID: PMC10990117 DOI: 10.1073/pnas.2312664121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 02/19/2024] [Indexed: 03/20/2024] Open
Abstract
Sleep is an evolutionarily conserved state that supports brain functions, including synaptic plasticity, in species across the animal kingdom. Here, we examine the neuroanatomical and cell-type distribution of presynaptic scaling in the fly brain after sleep loss. We previously found that sleep loss drives accumulation of the active zone scaffolding protein Bruchpilot (BRP) within cholinergic Kenyon cells of the Drosophila melanogaster mushroom body (MB), but not in other classes of MB neurons. To test whether similar cell type-specific trends in plasticity occur broadly across the brain, we used a flp-based genetic reporter to label presynaptic BRP in cholinergic, dopaminergic, GABAergic, or glutamatergic neurons. We then collected whole-brain confocal image stacks of BRP intensity to systematically quantify BRP, a marker of presynapse abundance, across 37 neuropil regions of the central fly brain. Our results indicate that sleep loss, either by overnight (12-h) mechanical stimulation or chronic sleep disruption in insomniac mutants, broadly elevates cholinergic synapse abundance across the brain, while synapse abundance in neurons that produce other neurotransmitters undergoes weaker, if any, changes. Extending sleep deprivation to 24 h drives brain-wide upscaling in glutamatergic, but not other, synapses. Finally, overnight male-male social pairings induce increased BRP in excitatory synapses despite male-female pairings eliciting more waking activity, suggesting experience-specific plasticity. Within neurotransmitter class and waking context, BRP changes are similar across the 37 neuropil domains, indicating that similar synaptic scaling rules may apply across the brain during acute sleep loss and that sleep need may broadly alter excitatory-inhibitory balance in the central brain.
Collapse
Affiliation(s)
- Jacqueline T. Weiss
- Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, CA90095
- Neuroscience Interdepartmental Program, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Mei Z. Blundell
- Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, CA90095
| | - Prabhjit Singh
- Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, CA90095
| | - Jeffrey M. Donlea
- Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, CA90095
| |
Collapse
|
7
|
Tener SJ, Lin Z, Park SJ, Oraedu K, Ulgherait M, Van Beek E, Martínez-Muñiz A, Pantalia M, Gatto JA, Volpi J, Stavropoulos N, Ja WW, Canman JC, Shirasu-Hiza M. Neuronal knockdown of Cullin3 as a Drosophila model of autism spectrum disorder. Sci Rep 2024; 14:1541. [PMID: 38233464 PMCID: PMC10794434 DOI: 10.1038/s41598-024-51657-9] [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: 06/01/2023] [Accepted: 01/06/2024] [Indexed: 01/19/2024] Open
Abstract
Mutations in Cullin-3 (Cul3), a conserved gene encoding a ubiquitin ligase, are strongly associated with autism spectrum disorder (ASD). Here, we characterize ASD-related pathologies caused by neuron-specific Cul3 knockdown in Drosophila. We confirmed that neuronal Cul3 knockdown causes short sleep, paralleling sleep disturbances in ASD. Because sleep defects and ASD are linked to metabolic dysregulation, we tested the starvation response of neuronal Cul3 knockdown flies; they starved faster and had lower triacylglyceride levels than controls, suggesting defects in metabolic homeostasis. ASD is also characterized by increased biomarkers of oxidative stress; we found that neuronal Cul3 knockdown increased sensitivity to hyperoxia, an exogenous oxidative stress. Additional hallmarks of ASD are deficits in social interactions and learning. Using a courtship suppression assay that measures social interactions and memory of prior courtship, we found that neuronal Cul3 knockdown reduced courtship and learning compared to controls. Finally, we found that neuronal Cul3 depletion alters the anatomy of the mushroom body, a brain region required for memory and sleep. Taken together, the ASD-related phenotypes of neuronal Cul3 knockdown flies establish these flies as a genetic model to study molecular and cellular mechanisms underlying ASD pathology, including metabolic and oxidative stress dysregulation and neurodevelopment.
Collapse
Affiliation(s)
- Samantha J Tener
- 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
| | - Scarlet J Park
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, 33458, USA
| | - Kairaluchi Oraedu
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Matthew Ulgherait
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Emily Van Beek
- Waksman Institute, Rutgers University, Piscataway, NJ, 08854, USA
| | - Andrés Martínez-Muñiz
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Meghan Pantalia
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Jared A Gatto
- 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
| | | | - William W Ja
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, 33458, USA
| | - Julie C Canman
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Mimi Shirasu-Hiza
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
8
|
Negrutskii BS, Porubleva LV, Malinowska A, Novosylna OV, Dadlez M, Knudsen CR. Understanding functions of eEF1 translation elongation factors beyond translation. A proteomic approach. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 138:67-99. [PMID: 38220433 DOI: 10.1016/bs.apcsb.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Mammalian translation elongation factors eEF1A1 and eEF1A2 are 92% homologous isoforms whose mutually exclusive tissue-specific expression is regulated during development. The isoforms have similar translation functionality, but show differences in spatial organization and participation in various processes, such as oncogenesis and virus reproduction. The differences may be due to their ability to interact with isoform-specific partner proteins. We used the identified sets of eEF1A1 or eEF1A2 partner proteins to identify cell complexes and/or processes specific to one particular isoform. As a result, we found isoform-specific interactions reflecting the involvement of different eEF1A isoforms in different cellular processes, including actin-related, chromatin-remodeling, ribonuclease H2, adenylyl cyclase, and Cul3-RING ubiquitin ligase complexes as well as initiation of mitochondrial transcription. An essential by-product of our analysis is the elucidation of a number of cellular processes beyond protein biosynthesis, where both isoforms appear to participate such as large ribosomal subunit biogenesis, mRNA splicing, DNA mismatch repair, 26S proteasome activity, P-body and exosomes formation, protein targeting to the membrane. This information suggests that a relatively high content of eEF1A in the cell may be necessary not only to maintain efficient translation, but also to ensure its participation in various cellular processes, where some roles of eEF1A have not yet been described. We believe that the data presented here will be useful for deciphering new auxiliary functions of eEF1A and its isoforms, and provide a new look at the known non-canonical functions of this main component of the human translation-elongation machinery.
Collapse
Affiliation(s)
- Boris S Negrutskii
- Institute of Molecular Biology and Genetics, Kyiv, Ukraine; Aarhus Institute of Advanced Sciences, Høegh-Guldbergs, Aarhus C, Denmark; Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen, Aarhus C, Denmark.
| | | | - Agata Malinowska
- Institute of Biochemistry and Biophysics, PAN, Pawinskiego, Warsaw, Poland
| | | | - Michal Dadlez
- Institute of Biochemistry and Biophysics, PAN, Pawinskiego, Warsaw, Poland
| | - Charlotte R Knudsen
- Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen, Aarhus C, Denmark
| |
Collapse
|
9
|
Brown EB, Zhang J, Lloyd E, Lanzon E, Botero V, Tomchik S, Keene AC. Neurofibromin 1 mediates sleep depth in Drosophila. PLoS Genet 2023; 19:e1011049. [PMID: 38091360 PMCID: PMC10763969 DOI: 10.1371/journal.pgen.1011049] [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: 09/11/2023] [Revised: 01/03/2024] [Accepted: 11/03/2023] [Indexed: 01/04/2024] Open
Abstract
Neural regulation of sleep and metabolic homeostasis are critical in many aspects of human health. Despite extensive epidemiological evidence linking sleep dysregulation with obesity, diabetes, and metabolic syndrome, little is known about the neural and molecular basis for the integration of sleep and metabolic function. The RAS GTPase-activating gene Neurofibromin (Nf1) has been implicated in the regulation of sleep and metabolic rate, raising the possibility that it serves to integrate these processes, but the effects on sleep consolidation and physiology remain poorly understood. A key hallmark of sleep depth in mammals and flies is a reduction in metabolic rate during sleep. Here, we examine multiple measures of sleep quality to determine the effects of Nf1 on sleep-dependent changes in arousal threshold and metabolic rate. Flies lacking Nf1 fail to suppress metabolic rate during sleep, raising the possibility that loss of Nf1 prevents flies from integrating sleep and metabolic state. Sleep of Nf1 mutant flies is fragmented with a reduced arousal threshold in Nf1 mutants, suggesting Nf1 flies fail to enter deep sleep. The effects of Nf1 on sleep can be localized to a subset of neurons expressing the GABAA receptor Rdl. Sleep loss has been associated with changes in gut homeostasis in flies and mammals. Selective knockdown of Nf1 in Rdl-expressing neurons within the nervous system increases gut permeability and reactive oxygen species (ROS) in the gut, raising the possibility that loss of sleep quality contributes to gut dysregulation. Together, these findings suggest Nf1 acts in GABA-sensitive neurons to modulate sleep depth in Drosophila.
Collapse
Affiliation(s)
- Elizabeth B. Brown
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
- Department of Biological Sciences, Florida State University, Tallahassee, Florida, United States of America
| | - Jiwei Zhang
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| | - Evan Lloyd
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| | - Elizabeth Lanzon
- Jupiter Life Science Initiative, Florida Atlantic University, Jupiter, Florida, United States of America
| | - Valentina Botero
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Seth Tomchik
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Alex C. Keene
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
10
|
Malik DM, Sengupta A, Sehgal A, Weljie AM. Altered Metabolism During the Dark Period in Drosophila Short Sleep Mutants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.30.564858. [PMID: 37961245 PMCID: PMC10634958 DOI: 10.1101/2023.10.30.564858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Sleep is an almost universally required state in biology. Disrupted sleep has been associated with adverse health risks including metabolic perturbations. Sleep is in part regulated via circadian mechanisms, however, metabolic dysfunction at different times of day arising from sleep disruption is unclear. We used targeted liquid chromatography-mass spectrometry to probe metabolic alterations using high-resolution temporal sampling of two Drosophila short sleep mutants, fumin and sleepless, across a circadian day. Discriminant analyses revealed overall distinct metabolic profiles for mutants when compared to a wild type dataset. Altered levels of metabolites involved in nicotinate/nicotinamide, alanine, aspartate, and glutamate, glyoxylate and dicarboxylate metabolism, and the TCA cycle were observed in mutants suggesting increased energetic demands. Furthermore, rhythmicity analyses revealed fewer 24 hr rhythmic metabolites in both mutants. Interestingly, mutants displayed two major peaks in phases while wild type displayed phases that were less concerted. In contrast to 24 hr rhythmic metabolites, an increase in the number of 12 hr rhythmic metabolites was observed in fumin while sleepless displayed a decrease. These results support that decreased sleep alters the overall metabolic profile with short sleep mutants displaying altered metabolite levels associated with a number of pathways in addition to altered neurotransmitter levels.
Collapse
Affiliation(s)
- Dania M. Malik
- Pharmacology Graduate Group
- Department of Systems Pharmacology and Translational Therapeutics
- Institute for Translational Medicine and Therapeutics
| | - Arjun Sengupta
- Department of Systems Pharmacology and Translational Therapeutics
- Institute for Translational Medicine and Therapeutics
| | - Amita Sehgal
- Chronobiology and Sleep Institute
- Howard Hughes Medical Institute
| | - Aalim M. Weljie
- Department of Systems Pharmacology and Translational Therapeutics
- Institute for Translational Medicine and Therapeutics
- Chronobiology and Sleep Institute
| |
Collapse
|
11
|
Durkin J, Poe AR, Belfer SJ, Rodriguez A, Tang SH, Walker JA, Kayser MS. Neurofibromin 1 regulates early developmental sleep in Drosophila. Neurobiol Sleep Circadian Rhythms 2023; 15:100101. [PMID: 37593040 PMCID: PMC10428071 DOI: 10.1016/j.nbscr.2023.100101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/30/2023] [Accepted: 08/06/2023] [Indexed: 08/19/2023] Open
Abstract
Sleep disturbances are common in neurodevelopmental disorders, but knowledge of molecular factors that govern sleep in young animals is lacking. Evidence across species, including Drosophila, suggests that juvenile sleep has distinct functions and regulatory mechanisms in comparison to sleep in maturity. In flies, manipulation of most known adult sleep regulatory genes is not associated with sleep phenotypes during early developmental (larval) stages. Here, we examine the role of the neurodevelopmental disorder-associated gene Neurofibromin 1 (Nf1) in sleep during numerous developmental periods. Mutations in Neurofibromin 1 (Nf1) are associated with sleep and circadian disorders in humans and adult flies. We find in flies that Nf1 acts to regulate sleep across the lifespan, beginning during larval stages. Nf1 is required in neurons for this function, as is signaling via the Alk pathway. These findings identify Nf1 as one of a small number of genes positioned to regulate sleep across developmental periods.
Collapse
Affiliation(s)
- Jaclyn Durkin
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Amy R. Poe
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Samuel J. Belfer
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Anyara Rodriguez
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Si Hao Tang
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - James A. Walker
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Matthew S. Kayser
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
12
|
Axelrod S, Li X, Sun Y, Lincoln S, Terceros A, O’Neil J, Wang Z, Nguyen A, Vora A, Spicer C, Shapiro B, Young MW. The Drosophila blood-brain barrier regulates sleep via Moody G protein-coupled receptor signaling. Proc Natl Acad Sci U S A 2023; 120:e2309331120. [PMID: 37831742 PMCID: PMC10589661 DOI: 10.1073/pnas.2309331120] [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: 06/02/2023] [Accepted: 08/28/2023] [Indexed: 10/15/2023] Open
Abstract
Sleep is vital for most animals, yet its mechanism and function remain unclear. We found that permeability of the BBB (blood-brain barrier)-the organ required for the maintenance of homeostatic levels of nutrients, ions, and other molecules in the brain-is modulated by sleep deprivation (SD) and can cell-autonomously effect sleep changes. We observed increased BBB permeability in known sleep mutants as well as in acutely sleep-deprived animals. In addition to molecular tracers, SD-induced BBB changes also increased the penetration of drugs used in the treatment of brain pathologies. After chronic/genetic or acute SD, rebound sleep or administration of the sleeping aid gaboxadol normalized BBB permeability, showing that SD effects on the BBB are reversible. Along with BBB permeability, RNA levels of the BBB master regulator moody are modulated by sleep. Conversely, altering BBB permeability alone through glia-specific modulation of moody, gαo, loco, lachesin, or neuroglian-each a well-studied regulator of BBB function-was sufficient to induce robust sleep phenotypes. These studies demonstrate a tight link between BBB permeability and sleep and indicate a unique role for the BBB in the regulation of sleep.
Collapse
Affiliation(s)
- Sofia Axelrod
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Xiaoling Li
- International Personalized Cancer Center, Tianjin Cancer Hospital Airport Hospital, Tianjin300308, China
| | - Yingwo Sun
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Samantha Lincoln
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Andrea Terceros
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Jenna O’Neil
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Zikun Wang
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Andrew Nguyen
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Aabha Vora
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Carmen Spicer
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Benjamin Shapiro
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Michael W. Young
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| |
Collapse
|
13
|
Shah R, Shah VK, Emin M, Gao S, Sampogna RV, Aggarwal B, Chang A, St-Onge MP, Malik V, Wang J, Wei Y, Jelic S. Mild sleep restriction increases endothelial oxidative stress in female persons. Sci Rep 2023; 13:15360. [PMID: 37717072 PMCID: PMC10505226 DOI: 10.1038/s41598-023-42758-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 09/14/2023] [Indexed: 09/18/2023] Open
Abstract
Sleep restriction is associated with increased cardiovascular risk, which is more pronounced in female than male persons. We reported recently first causal evidence that mild, prolonged sleep restriction mimicking "real-life" conditions impairs endothelial function, a key step in the development and progression of cardiovascular disease, in healthy female persons. However, the underlying mechanisms are unclear. In model organisms, sleep restriction increases oxidative stress and upregulates antioxidant response via induction of the antioxidant regulator nuclear factor (erythroid-derived 2)-like 2 (Nrf2). Here, we assessed directly endothelial cell oxidative stress and antioxidant responses in healthy female persons (n = 35) after 6 weeks of mild sleep restriction (1.5 h less than habitual sleep) using randomized crossover design. Sleep restriction markedly increased endothelial oxidative stress without upregulating antioxidant response. Using RNA-seq and a predicted protein-protein interaction database, we identified reduced expression of endothelial Defective in Cullin Neddylation-1 Domain Containing 3 (DCUN1D3), a protein that licenses Nrf2 antioxidant responses, as a mediator of impaired endothelial antioxidant response in sleep restriction. Thus, sleep restriction impairs clearance of endothelial oxidative stress that over time increases cardiovascular risk.Trial Registration: NCT02835261 .
Collapse
Affiliation(s)
- Riddhi Shah
- Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Vikash Kumar Shah
- Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Memet Emin
- Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Su Gao
- Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Rosemary V Sampogna
- Division of Nephrology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Brooke Aggarwal
- Division of Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Audrey Chang
- NewYork-Presbyterian Morgan Stanley Children's Hospital, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Marie-Pierre St-Onge
- Division of General Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Vikas Malik
- Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Columbia Center for Human Development and Columbia Stem Cell Initiative, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Jianlong Wang
- Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Columbia Center for Human Development and Columbia Stem Cell Initiative, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Ying Wei
- Division of Biostatistics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Sanja Jelic
- Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
14
|
Jiang W, Wang W, Kong Y, Zheng S. Structural basis for the ubiquitination of G protein βγ subunits by KCTD5/Cullin3 E3 ligase. SCIENCE ADVANCES 2023; 9:eadg8369. [PMID: 37450587 PMCID: PMC10348674 DOI: 10.1126/sciadv.adg8369] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 06/14/2023] [Indexed: 07/18/2023]
Abstract
G protein-coupled receptor (GPCR) signaling is precisely controlled to avoid overstimulation that results in detrimental consequences. Gβγ signaling is negatively regulated by a Cullin3 (Cul3)-dependent E3 ligase, KCTD5, which triggers ubiquitination and degradation of free Gβγ. Here, we report the cryo-electron microscopy structures of the KCTD5-Gβγ fusion complex and the KCTD7-Cul3 complex. KCTD5 in pentameric form engages symmetrically with five copies of Gβγ through its C-terminal domain. The unique pentameric assembly of the KCTD5/Cul3 E3 ligase places the ubiquitin-conjugating enzyme (E2) and the modification sites of Gβγ in close proximity and allows simultaneous transfer of ubiquitin from E2 to five Gβγ subunits. Moreover, we show that ubiquitination of Gβγ by KCTD5 is important for fine-tuning cyclic adenosine 3´,5´-monophosphate signaling of GPCRs. Our studies provide unprecedented insights into mechanisms of substrate recognition by unusual pentameric E3 ligases and highlight the KCTD family as emerging regulators of GPCR signaling.
Collapse
Affiliation(s)
- Wentong Jiang
- Graduate School of Peking Union Medical College, Beijing 100730, China
- National Institute of Biological Sciences, Beijing 102206, China
| | - Wei Wang
- National Institute of Biological Sciences, Beijing 102206, China
- School of Life Sciences, Peking University, Beijing 100871, China
| | - Yinfei Kong
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Sanduo Zheng
- Graduate School of Peking Union Medical College, Beijing 100730, China
- National Institute of Biological Sciences, Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 100084, China
| |
Collapse
|
15
|
Li Y, Zhou X, Cheng C, Ding G, Zhao P, Tan K, Chen L, Perrimon N, Veenstra JA, Zhang L, Song W. Gut AstA mediates sleep deprivation-induced energy wasting in Drosophila. Cell Discov 2023; 9:49. [PMID: 37221172 DOI: 10.1038/s41421-023-00541-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 03/13/2023] [Indexed: 05/25/2023] Open
Abstract
Severe sleep deprivation (SD) has been highly associated with systemic energy wasting, such as lipid loss and glycogen depletion. Despite immune dysregulation and neurotoxicity observed in SD animals, whether and how the gut-secreted hormones participate in SD-induced disruption of energy homeostasis remains largely unknown. Using Drosophila as a conserved model organism, we characterize that production of intestinal Allatostatin A (AstA), a major gut-peptide hormone, is robustly increased in adult flies bearing severe SD. Interestingly, the removal of AstA production in the gut using specific drivers significantly improves lipid loss and glycogen depletion in SD flies without affecting sleep homeostasis. We reveal the molecular mechanisms whereby gut AstA promotes the release of an adipokinetic hormone (Akh), an insulin counter-regulatory hormone functionally equivalent to mammalian glucagon, to mobilize systemic energy reserves by remotely targeting its receptor AstA-R2 in Akh-producing cells. Similar regulation of glucagon secretion and energy wasting by AstA/galanin is also observed in SD mice. Further, integrating single-cell RNA sequencing and genetic validation, we uncover that severe SD results in ROS accumulation in the gut to augment AstA production via TrpA1. Altogether, our results demonstrate the essential roles of the gut-peptide hormone AstA in mediating SD-associated energy wasting.
Collapse
Affiliation(s)
- Yingge Li
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Xiaoya Zhou
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Chen Cheng
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Guangming Ding
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Peng Zhao
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Kai Tan
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
| | - Lixia Chen
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Norbert Perrimon
- Department of Genetics, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - Jan A Veenstra
- INCIA, UMR 5287 CNRS, University of Bordeaux, Talence, France
| | - Luoying Zhang
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Song
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China.
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
16
|
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: 1.0] [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
|
17
|
Wang Z, Lincoln S, Nguyen AD, Li W, Young MW. Chronic sleep loss disrupts rhythmic gene expression in Drosophila. Front Physiol 2022; 13:1048751. [PMID: 36467698 PMCID: PMC9716074 DOI: 10.3389/fphys.2022.1048751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/01/2022] [Indexed: 11/19/2022] Open
Abstract
Genome-wide profiling of rhythmic gene expression has offered new avenues for studying the contribution of circadian clock to diverse biological processes. Sleep has been considered one of the most important physiological processes that are regulated by the circadian clock, however, the effects of chronic sleep loss on rhythmic gene expression remain poorly understood. In the present study, we exploited Drosophila sleep mutants insomniac 1 (inc 1 ) and wide awake D2 (wake D2 ) as models for chronic sleep loss. We profiled the transcriptomes of head tissues collected from 4-week-old wild type flies, inc 1 and wake D2 at timepoints around the clock. Analysis of gene oscillation revealed a substantial loss of rhythmicity in inc 1 and wake D2 compared to wild type flies, with most of the affected genes common to both mutants. The disruption of gene oscillation was not due to changes in average gene expression levels. We also identified a subset of genes whose loss of rhythmicity was shared among animals with chronic sleep loss and old flies, suggesting a contribution of aging to chronic, sleep-loss-induced disruption of gene oscillation.
Collapse
Affiliation(s)
- Zikun Wang
- Laboratory of Genetics, The Rockefeller University, New York, NY, United States
| | - Samantha Lincoln
- Laboratory of Genetics, The Rockefeller University, New York, NY, United States
| | - Andrew D. Nguyen
- Laboratory of Genetics, The Rockefeller University, New York, NY, United States
| | - Wanhe Li
- Laboratory of Genetics, The Rockefeller University, New York, NY, United States
- Department of Biology, Center for Biological Clocks Research, Texas A&M University, College Station, United States
| | - Michael W. Young
- Laboratory of Genetics, The Rockefeller University, New York, NY, United States
| |
Collapse
|
18
|
Harbison ST. What have we learned about sleep from selective breeding strategies? Sleep 2022; 45:zsac147. [PMID: 36111812 PMCID: PMC9644121 DOI: 10.1093/sleep/zsac147] [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: 04/28/2022] [Revised: 06/19/2022] [Indexed: 09/18/2023] Open
Abstract
Selective breeding is a classic technique that enables an experimenter to modify a heritable target trait as desired. Direct selective breeding for extreme sleep and circadian phenotypes in flies successfully alters these behaviors, and sleep and circadian perturbations emerge as correlated responses to selection for other traits in mice, rats, and dogs. The application of sequencing technologies to the process of selective breeding identifies the genetic network impacting the selected trait in a holistic way. Breeding techniques preserve the extreme phenotypes generated during selective breeding, generating community resources for further functional testing. Selective breeding is thus a unique strategy that can explore the phenotypic limits of sleep and circadian behavior, discover correlated responses of traits having shared genetic architecture with the target trait, identify naturally-occurring genomic variants and gene expression changes that affect trait variability, and pinpoint genes with conserved roles.
Collapse
Affiliation(s)
- Susan T Harbison
- Laboratory of Systems Genetics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD,USA
| |
Collapse
|
19
|
Sleep Modulates Alcohol Toxicity in Drosophila. Int J Mol Sci 2022; 23:ijms232012091. [PMID: 36292943 PMCID: PMC9603330 DOI: 10.3390/ijms232012091] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 11/19/2022] Open
Abstract
Alcohol abuse is a significant public health problem. While considerable research has shown that alcohol use affects sleep, little is known about the role of sleep deprivation in alcohol toxicity. We investigated sleep as a factor modulating alcohol toxicity using Drosophila melanogaster, a model for studies of sleep, alcohol, and aging. Following 24 h of sleep deprivation using a paradigm that similarly affects males and females and induces rebound sleep, flies were given binge-like alcohol exposures. Sleep deprivation increased mortality, with no sex-dependent differences. Sleep deprivation also abolished functional tolerance measured at 24 h after the initial alcohol exposure, although there was no effect on alcohol absorbance or clearance. We investigated the effect of chronic sleep deprivation using mutants with decreased sleep, insomniac and insulin-like peptide 2, finding increased alcohol mortality. Furthermore, we investigated whether pharmacologically inducing sleep prior to alcohol exposure using the GABAA-receptor agonist 4,5,6,7-tetrahydroisoxazolo(5,4-c)pyridin-3-ol (THIP) mitigated the effects of alcohol toxicity on middle-aged flies, flies with environmentally disrupted circadian clocks, and flies with short sleep. Pharmacologically increasing sleep prior to alcohol exposure decreased alcohol-induced mortality. Thus, sleep prior to binge-like alcohol exposure affects alcohol-induced mortality, even in vulnerable groups such as aging flies and those with circadian dysfunction.
Collapse
|
20
|
Vaughen JP, Theisen E, Rivas-Serna IM, Berger AB, Kalakuntla P, Anreiter I, Mazurak VC, Rodriguez TP, Mast JD, Hartl T, Perlstein EO, Reimer RJ, Clandinin MT, Clandinin TR. Glial control of sphingolipid levels sculpts diurnal remodeling in a circadian circuit. Neuron 2022; 110:3186-3205.e7. [PMID: 35961319 PMCID: PMC10868424 DOI: 10.1016/j.neuron.2022.07.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/21/2022] [Accepted: 07/14/2022] [Indexed: 11/19/2022]
Abstract
Structural plasticity in the brain often necessitates dramatic remodeling of neuronal processes, with attendant reorganization of the cytoskeleton and membranes. Although cytoskeletal restructuring has been studied extensively, how lipids might orchestrate structural plasticity remains unclear. We show that specific glial cells in Drosophila produce glucocerebrosidase (GBA) to locally catabolize sphingolipids. Sphingolipid accumulation drives lysosomal dysfunction, causing gba1b mutants to harbor protein aggregates that cycle across circadian time and are regulated by neural activity, the circadian clock, and sleep. Although the vast majority of membrane lipids are stable across the day, a specific subset that is highly enriched in sphingolipids cycles daily in a gba1b-dependent fashion. Remarkably, both sphingolipid biosynthesis and degradation are required for the diurnal remodeling of circadian clock neurites, which grow and shrink across the day. Thus, dynamic sphingolipid regulation by glia enables diurnal circuit remodeling and proper circadian behavior.
Collapse
Affiliation(s)
- John P Vaughen
- Department of Neurobiology, Stanford University, Stanford, CA 94305, USA; Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA
| | - Emma Theisen
- Department of Neurobiology, Stanford University, Stanford, CA 94305, USA
| | - Irma Magaly Rivas-Serna
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Andrew B Berger
- Department of Neurobiology, Stanford University, Stanford, CA 94305, USA
| | - Prateek Kalakuntla
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA
| | - Ina Anreiter
- Department of Neurobiology, Stanford University, Stanford, CA 94305, USA
| | - Vera C Mazurak
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | | | - Joshua D Mast
- Perlara PBC, 2625 Alcatraz Ave #435, Berkeley, CA 94705, USA
| | - Tom Hartl
- Perlara PBC, 2625 Alcatraz Ave #435, Berkeley, CA 94705, USA
| | | | - Richard J Reimer
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - M Thomas Clandinin
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Thomas R Clandinin
- Department of Neurobiology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
21
|
Huang G, Dierick HA. The need for unbiased genetic screens to dissect aggression in Drosophila melanogaster. Front Behav Neurosci 2022; 16:901453. [PMID: 35979224 PMCID: PMC9377312 DOI: 10.3389/fnbeh.2022.901453] [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: 03/22/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Aggression is an evolutionarily conserved behavior present in most animals and is necessary for survival when competing for limited resources and mating partners. Studies have shown that aggression is modulated both genetically and epigenetically, but details of how the molecular and cellular mechanisms interact to determine aggressive behavior remain to be elucidated. In recent decades, Drosophila melanogaster has emerged as a powerful model system to understand the mechanisms that regulate aggression. Surprisingly most of the findings discovered to date have not come from genetic screens despite the fly's long and successful history of using screens to unravel its biology. Here, we highlight the tools and techniques used to successfully screen for aggression-linked behavioral elements in Drosophila and discuss the potential impact future screens have in advancing our knowledge of the underlying genetic and neural circuits governing aggression.
Collapse
Affiliation(s)
- Gary Huang
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Herman A Dierick
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
22
|
Ukita Y, Okumura M, Chihara T. Ubiquitin proteasome system in circadian rhythm and sleep homeostasis: Lessons from Drosophila. Genes Cells 2022; 27:381-391. [PMID: 35438236 PMCID: PMC9322287 DOI: 10.1111/gtc.12935] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/27/2022] [Accepted: 03/28/2022] [Indexed: 11/30/2022]
Abstract
Sleep is regulated by two main processes: the circadian clock and sleep homeostasis. Circadian rhythms have been well studied at the molecular level. In the Drosophila circadian clock neurons, the core clock proteins are precisely regulated by post-translational modifications and degraded via the ubiquitin-proteasome system (UPS). Sleep homeostasis, however, is less understood; nevertheless, recent reports suggest that proteasome-mediated degradation of core clock proteins or synaptic proteins contributes to the regulation of sleep amount. Here, we review the molecular mechanism of the UPS and summarize the role of protein degradation in the regulation of circadian clock and homeostatic sleep in Drosophila. Moreover, we discuss the potential interaction between circadian clock and homeostatic sleep regulation with a prime focus on E3 ubiquitin ligases.
Collapse
Affiliation(s)
- Yumiko Ukita
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Misako Okumura
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan.,Program of Basic Biology, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Takahiro Chihara
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan.,Program of Basic Biology, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
23
|
Gao Z, Wang A, Zhao Y, Zhang X, Yuan X, Li N, Xu C, Wang S, Zhu Y, Zhu J, Guan J, Liu F, Yin S. Integrative Proteome and Ubiquitinome Analyses Reveal the Substrates of BTBD9 and Its Underlying Mechanism in Sleep Regulation. ACS OMEGA 2022; 7:11839-11852. [PMID: 35449961 PMCID: PMC9016840 DOI: 10.1021/acsomega.1c07262] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/24/2022] [Indexed: 06/14/2023]
Abstract
Ubiquitination is a major posttranslational modification of proteins that affects their stability, and E3 ligases play a key role in ubiquitination by specifically recognizing their substrates. BTBD9, an adaptor of the Cullin-RING ligase complex, is responsible for substrate recognition and is associated with sleep homeostasis. However, the substrates of BTBD9-mediated ubiquitination remain unknown. Here, we generated an SH-SY5Y cell line stably expressing BTBD9 and performed proteomic analysis combined with ubiquitinome analysis to identify the downstream targets of BTBD9. Through this approach, we identified four potential BTBD9-mediated ubiquitination substrates that are targeted for degradation. Among these candidate substrates, inosine monophosphate dehydrogenase (IMPDH2), a novel target of BTBD9-mediated degradation, is a potential risk gene for sleep dysregulation. In conclusion, these findings not only demonstrate that proteomic analysis can be a useful general approach for the systematic identification of E3 ligase substrates but also identify novel substrates of BTBD9, providing a resource for future studies of sleep regulation mechanisms.
Collapse
Affiliation(s)
- Zhenfei Gao
- Department
of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Otolaryngology Institute of Shanghai Jiao Tong University,
Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai 200233, China
- Shanghai
Key Laboratory of Sleep Disordered Breathing, Yishan Road 600, Shanghai 200233, China
| | - Anzhao Wang
- Department
of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Otolaryngology Institute of Shanghai Jiao Tong University,
Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai 200233, China
- Shanghai
Key Laboratory of Sleep Disordered Breathing, Yishan Road 600, Shanghai 200233, China
| | - Yongxu Zhao
- CAS
Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai
Institute of Nutrition and Health, Shanghai Institutes for Biological
Sciences, University of Chinese Academy of Sciences, Chinese Academy
of Sciences, Shanghai 200231, China
| | - Xiaoxu Zhang
- Department
of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Otolaryngology Institute of Shanghai Jiao Tong University,
Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai 200233, China
- Shanghai
Key Laboratory of Sleep Disordered Breathing, Yishan Road 600, Shanghai 200233, China
| | - Xiangshan Yuan
- Department
of Anatomy and Histoembryology, School of Basic Medical Sciences,
State Key Laboratory of Medical Neurobiology and MOE Frontiers Center
for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200231, China
| | - Niannian Li
- Department
of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Otolaryngology Institute of Shanghai Jiao Tong University,
Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai 200233, China
- Shanghai
Key Laboratory of Sleep Disordered Breathing, Yishan Road 600, Shanghai 200233, China
| | - Chong Xu
- Department
of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Otolaryngology Institute of Shanghai Jiao Tong University,
Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai 200233, China
- Shanghai
Key Laboratory of Sleep Disordered Breathing, Yishan Road 600, Shanghai 200233, China
| | - Shenming Wang
- Department
of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Otolaryngology Institute of Shanghai Jiao Tong University,
Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai 200233, China
- Shanghai
Key Laboratory of Sleep Disordered Breathing, Yishan Road 600, Shanghai 200233, China
| | - Yaxin Zhu
- Department
of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Otolaryngology Institute of Shanghai Jiao Tong University,
Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai 200233, China
- Shanghai
Key Laboratory of Sleep Disordered Breathing, Yishan Road 600, Shanghai 200233, China
| | - Jingyu Zhu
- Department
of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Otolaryngology Institute of Shanghai Jiao Tong University,
Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai 200233, China
- Shanghai
Key Laboratory of Sleep Disordered Breathing, Yishan Road 600, Shanghai 200233, China
| | - Jian Guan
- Department
of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Otolaryngology Institute of Shanghai Jiao Tong University,
Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai 200233, China
- Shanghai
Key Laboratory of Sleep Disordered Breathing, Yishan Road 600, Shanghai 200233, China
| | - Feng Liu
- Department
of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Otolaryngology Institute of Shanghai Jiao Tong University,
Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai 200233, China
- Shanghai
Key Laboratory of Sleep Disordered Breathing, Yishan Road 600, Shanghai 200233, China
| | - Shankai Yin
- Department
of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Otolaryngology Institute of Shanghai Jiao Tong University,
Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai 200233, China
- Shanghai
Key Laboratory of Sleep Disordered Breathing, Yishan Road 600, Shanghai 200233, China
| |
Collapse
|
24
|
Wu J, Fang S, Lu KT, Kumar G, Reho JJ, Brozoski DT, Otanwa AJ, Hu C, Nair AR, Wackman KK, Agbor LN, Grobe JL, Sigmund CD. Endothelial Cullin3 Mutation Impairs Nitric Oxide-Mediated Vasodilation and Promotes Salt-Induced Hypertension. FUNCTION (OXFORD, ENGLAND) 2022; 3:zqac017. [PMID: 35493997 PMCID: PMC9045850 DOI: 10.1093/function/zqac017] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 01/13/2023]
Abstract
Human hypertension caused by in-frame deletion of CULLIN3 exon-9 (Cul3∆9) is driven by renal and vascular mechanisms. We bred conditionally activatable Cul3∆9 transgenic mice with tamoxifen-inducible Tie2-CREERT2 mice to test the importance of endothelial Cul3. The resultant mice (E-Cul3∆9) trended towards elevated nighttime blood pressure (BP) correlated with increased nighttime activity, but displayed no difference in daytime BP or activity. Male and female E-Cul3∆9 mice together exhibited a decline in endothelial-dependent relaxation in carotid artery. Male but not female E-Cul3∆9 mice displayed severe endothelial dysfunction in cerebral basilar artery. There was no impairment in mesenteric artery and no difference in smooth muscle function, suggesting the effects of Cul3∆9 are arterial bed-specific and sex-dependent. Expression of Cul3∆9 in primary mouse aortic endothelial cells decreased endogenous Cul3 protein, phosphorylated (S1177) endothelial nitric oxide synthase (eNOS) and nitric oxide (NO) production. Protein phosphatase (PP) 2A, a known Cul3 substrate, dephosphorylates eNOS. Cul3∆9-induced impairment of eNOS activity was rescued by a selective PP2A inhibitor okadaic acid, but not by a PP1 inhibitor tautomycetin. Because NO deficiency contributes to salt-induced hypertension, we tested the salt-sensitivity of E-Cul3∆9 mice. While both male and female E-Cul3∆9 mice developed salt-induced hypertension and renal injury, the pressor effect of salt was greater in female mutants. The increased salt-sensitivity in female E-Cul3∆9 mice was associated with decreased renovascular relaxation and impaired natriuresis in response to a sodium load. Thus, CUL3 mutations in the endothelium may contribute to human hypertension in part through decreased endothelial NO bioavailability, renovascular dysfunction, and increased salt-sensitivity of BP.
Collapse
Affiliation(s)
- Jing Wu
- Deparment of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, 53226 Wisconsin, USA
| | - Shi Fang
- Deparment of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, 53226 Wisconsin, USA,Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, 52242 Iowa, USA
| | - Ko-Ting Lu
- Deparment of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, 53226 Wisconsin, USA
| | - Gaurav Kumar
- Deparment of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, 53226 Wisconsin, USA
| | - John J Reho
- Deparment of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, 53226 Wisconsin, USA
| | - Daniel T Brozoski
- Deparment of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, 53226 Wisconsin, USA
| | - Adokole J Otanwa
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, 52242 Iowa, USA
| | - Chunyan Hu
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, 52242 Iowa, USA
| | - Anand R Nair
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, 52242 Iowa, USA
| | - Kelsey K Wackman
- Deparment of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, 53226 Wisconsin, USA
| | - Larry N Agbor
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, 52242 Iowa, USA
| | - Justin L Grobe
- Deparment of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, 53226 Wisconsin, USA
| | | |
Collapse
|
25
|
Doldur-Balli F, Imamura T, Veatch OJ, Gong NN, Lim DC, Hart MP, Abel T, Kayser MS, Brodkin ES, Pack AI. Synaptic dysfunction connects autism spectrum disorder and sleep disturbances: A perspective from studies in model organisms. Sleep Med Rev 2022; 62:101595. [PMID: 35158305 PMCID: PMC9064929 DOI: 10.1016/j.smrv.2022.101595] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/24/2021] [Accepted: 01/19/2022] [Indexed: 01/03/2023]
Abstract
Sleep disturbances (SD) accompany many neurodevelopmental disorders, suggesting SD is a transdiagnostic process that can account for behavioral deficits and influence underlying neuropathogenesis. Autism Spectrum Disorder (ASD) comprises a complex set of neurodevelopmental conditions characterized by challenges in social interaction, communication, and restricted, repetitive behaviors. Diagnosis of ASD is based primarily on behavioral criteria, and there are no drugs that target core symptoms. Among the co-occurring conditions associated with ASD, SD are one of the most prevalent. SD often arises before the onset of other ASD symptoms. Sleep interventions improve not only sleep but also daytime behaviors in children with ASD. Here, we examine sleep phenotypes in multiple model systems relevant to ASD, e.g., mice, zebrafish, fruit flies and worms. Given the functions of sleep in promoting brain connectivity, neural plasticity, emotional regulation and social behavior, all of which are of critical importance in ASD pathogenesis, we propose that synaptic dysfunction is a major mechanism that connects ASD and SD. Common molecular targets in this interplay that are involved in synaptic function might be a novel avenue for therapy of individuals with ASD experiencing SD. Such therapy would be expected to improve not only sleep but also other ASD symptoms.
Collapse
Affiliation(s)
- Fusun Doldur-Balli
- Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA.
| | - Toshihiro Imamura
- Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA; Division of Pulmonary and Sleep Medicine, Children's Hospital of Philadelphia, Philadelphia, USA
| | - Olivia J Veatch
- Department of Psychiatry and Behavioral Sciences, School of Medicine, The University of Kansas Medical Center, Kansas City, USA
| | - Naihua N Gong
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Diane C Lim
- Pulmonary, Allergy, Critical Care and Sleep Medicine Division, Department of Medicine, Miller School of Medicine, University of Miami, Miami, USA
| | - Michael P Hart
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Ted Abel
- Iowa Neuroscience Institute and Department of Neuroscience & Pharmacology, University of Iowa, Iowa City, USA
| | - Matthew S Kayser
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA; Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA; Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Edward S Brodkin
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Allan I Pack
- Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
26
|
Li Q, Jang H, Lim KY, Lessing A, Stavropoulos N. insomniac links the development and function of a sleep-regulatory circuit. eLife 2021; 10:65437. [PMID: 34908527 PMCID: PMC8758140 DOI: 10.7554/elife.65437] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 10/15/2021] [Indexed: 11/17/2022] Open
Abstract
Although many genes are known to influence sleep, when and how they impact sleep-regulatory circuits remain ill-defined. Here, we show that insomniac (inc), a conserved adaptor for the autism-associated Cul3 ubiquitin ligase, acts in a restricted period of neuronal development to impact sleep in adult Drosophila. The loss of inc causes structural and functional alterations within the mushroom body (MB), a center for sensory integration, associative learning, and sleep regulation. In inc mutants, MB neurons are produced in excess, develop anatomical defects that impede circuit assembly, and are unable to promote sleep when activated in adulthood. Our findings link neurogenesis and postmitotic development of sleep-regulatory neurons to their adult function and suggest that developmental perturbations of circuits that couple sensory inputs and sleep may underlie sleep dysfunction in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Qiuling Li
- Neuroscience Institute, Department of Neuroscience and Physiology, New York University School of MedicineNew YorkUnited States
| | - Hyunsoo Jang
- Neuroscience Institute, Department of Neuroscience and Physiology, New York University School of MedicineNew YorkUnited States
| | - Kayla Y Lim
- Neuroscience Institute, Department of Neuroscience and Physiology, New York University School of MedicineNew YorkUnited States
| | - Alexie Lessing
- Neuroscience Institute, Department of Neuroscience and Physiology, New York University School of MedicineNew YorkUnited States
| | - Nicholas Stavropoulos
- Neuroscience Institute, Department of Neuroscience and Physiology, New York University School of MedicineNew YorkUnited States
- Waksman Institute, Rutgers UniversityPiscatawayUnited States
| |
Collapse
|
27
|
Li W, Wang Z, Syed S, Lyu C, Lincoln S, O'Neil J, Nguyen AD, Feng I, Young MW. Chronic social isolation signals starvation and reduces sleep in Drosophila. Nature 2021; 597:239-244. [PMID: 34408325 PMCID: PMC8429171 DOI: 10.1038/s41586-021-03837-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/20/2021] [Indexed: 12/11/2022]
Abstract
Social isolation and loneliness have potent effects on public health1-4. Research in social psychology suggests that compromised sleep quality is a key factor that links persistent loneliness to adverse health conditions5,6. Although experimental manipulations have been widely applied to studying the control of sleep and wakefulness in animal models, how normal sleep is perturbed by social isolation is unknown. Here we report that chronic, but not acute, social isolation reduces sleep in Drosophila. We use quantitative behavioural analysis and transcriptome profiling to differentiate between brain states associated with acute and chronic social isolation. Although the flies had uninterrupted access to food, chronic social isolation altered the expression of metabolic genes and induced a brain state that signals starvation. Chronically isolated animals exhibit sleep loss accompanied by overconsumption of food, which resonates with anecdotal findings of loneliness-associated hyperphagia in humans. Chronic social isolation reduces sleep and promotes feeding through neural activities in the peptidergic fan-shaped body columnar neurons of the fly. Artificial activation of these neurons causes misperception of acute social isolation as chronic social isolation and thereby results in sleep loss and increased feeding. These results present a mechanistic link between chronic social isolation, metabolism, and sleep, addressing a long-standing call for animal models focused on loneliness7.
Collapse
Affiliation(s)
- Wanhe Li
- Laboratory of Genetics, The Rockefeller University, New York, NY, USA.
| | - Zikun Wang
- Laboratory of Genetics, The Rockefeller University, New York, NY, USA
| | - Sheyum Syed
- Department of Physics, University of Miami, Coral Gables, FL, USA
| | - Cheng Lyu
- Laboratory of Integrative Brain Function, The Rockefeller University, New York, NY, USA
| | - Samantha Lincoln
- Laboratory of Genetics, The Rockefeller University, New York, NY, USA
| | - Jenna O'Neil
- Laboratory of Genetics, The Rockefeller University, New York, NY, USA
| | - Andrew D Nguyen
- Laboratory of Genetics, The Rockefeller University, New York, NY, USA
| | - Irena Feng
- Laboratory of Genetics, The Rockefeller University, New York, NY, USA
| | - Michael W Young
- Laboratory of Genetics, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
28
|
Dai X, Zhou E, Yang W, Mao R, Zhang W, Rao Y. Molecular resolution of a behavioral paradox: sleep and arousal are regulated by distinct acetylcholine receptors in different neuronal types in Drosophila. Sleep 2021; 44:6119684. [PMID: 33493349 DOI: 10.1093/sleep/zsab017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 12/21/2020] [Indexed: 02/02/2023] Open
Abstract
Sleep and arousal are both important for animals. The neurotransmitter acetylcholine (ACh) has long been found to promote both sleep and arousal in mammals, an apparent paradox which has also been found to exist in flies, causing much confusion in understanding sleep and arousal. Here, we have systematically studied all 13 ACh receptors (AChRs) in Drosophila to understand mechanisms underlying ACh function in sleep and arousal. We found that exogenous stimuli-induced arousal was decreased in nAChRα3 mutants, whereas sleep was decreased in nAChRα2 and nAChRβ2 mutants. nAChRα3 functions in dopaminergic neurons to promote exogenous stimuli-induced arousal, whereas nAChRα2 and β2 function in octopaminergic neurons to promote sleep. Our studies have revealed that a single transmitter can promote endogenous sleep and exogenous stimuli-induced arousal through distinct receptors in different types of downstream neurons.
Collapse
Affiliation(s)
- Xihuimin Dai
- PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Beijing Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Beijing, China.,Chinese Institute for Brain Research, Beijing, China.,Howard Hughes Medical Institute, Department of Biology, Brandeis University, Waltham, MA
| | - Enxing Zhou
- PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Beijing Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Beijing, China
| | - Wei Yang
- PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Beijing Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Beijing, China.,Chinese Institute for Brain Research, Beijing, China
| | - Renbo Mao
- Graduate School of Peking Union Medical College, Beijing, China.,National Institute of Biological Sciences, Beijing, China
| | - Wenxia Zhang
- PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Beijing Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Beijing, China
| | - Yi Rao
- PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Beijing Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Beijing, China.,Chinese Institute for Brain Research, Beijing, China
| |
Collapse
|
29
|
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: 1.7] [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]
|
30
|
Pattinson CL, Guedes VA, Edwards K, Mithani S, Yun S, Taylor P, Dunbar K, Kim HS, Lai C, Roy MJ, Gill JM. Excessive daytime sleepiness is associated with altered gene expression in military personnel and veterans with posttraumatic stress disorder: an RNA sequencing study. Sleep 2021; 43:5802516. [PMID: 32191323 DOI: 10.1093/sleep/zsaa036] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 02/11/2020] [Indexed: 11/13/2022] Open
Abstract
STUDY OBJECTIVES Posttraumatic stress disorder (PTSD) is a common condition for military personnel and veterans. PTSD has been shown to impact gene expression, however, to date no study has examined comorbid conditions which may also impact gene expression, for example, excessive daytime sleepiness (EDS). As such, this study sought to examine gene expression using RNA sequencing across three group comparisons of military personnel and veterans: (1) PTSD with EDS (PTSDwEDS) versus PTSD without EDS (PTSDw/outEDS), (2) Controls (no PTSD or EDS) versus PTSDwEDS, and (3) Controls versus PTSDw/outEDS. METHODS We performed experimental RNA-seq using Illumina's HiSeq 2500 Sequencing System. We also used Ingenuity Pathway Analysis (IPA), a bioinformatics application, to identify gene pathways and networks which may be disrupted. RESULTS There were only two genes that were significantly dysregulated between the Controls and PTSDw/outEDS, therefore IPA analysis was not conducted. However, comparisons revealed that there was significant gene dysregulation between Controls and the PTSDwEDS (251 genes), and the PTSDwEDS versus the PTSDw/outEDS (1,873 genes) groups. Four candidate networks were identified via the IPA software for analysis. Significantly dysregulated genes across the four candidate networks were associated with sleep and circadian function, metabolism, mitochondrial production and function, ubiquitination, and the glutamate system. CONCLUSIONS These results suggest that PTSD with concurrent EDS is associated with gene dysregulation. This dysregulation may present additional biological and health consequences for these military personnel and veterans. Further research, to track these gene changes over time and to determine the cause of the EDS reported, is vital.
Collapse
Affiliation(s)
- Cassandra L Pattinson
- National Institutes of Nursing Research, National Institutes of Health, Bethesda, MD.,Institute for Social Science Research, University of Queensland, Indooroopilly, Queensland, Australia
| | - Vivian A Guedes
- National Institutes of Nursing Research, National Institutes of Health, Bethesda, MD
| | - Katie Edwards
- National Institutes of Nursing Research, National Institutes of Health, Bethesda, MD.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD
| | - Sara Mithani
- National Institutes of Nursing Research, National Institutes of Health, Bethesda, MD
| | - Sijung Yun
- National Institutes of Nursing Research, National Institutes of Health, Bethesda, MD.,Yotta Biomed, LLC, Bethesda, MD
| | - Patricia Taylor
- Institute for Social Science Research, University of Queensland, Indooroopilly, Queensland, Australia.,Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD
| | - Kerri Dunbar
- Institute for Social Science Research, University of Queensland, Indooroopilly, Queensland, Australia.,Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD
| | - Hyung-Suk Kim
- National Institutes of Nursing Research, National Institutes of Health, Bethesda, MD
| | - Chen Lai
- National Institutes of Nursing Research, National Institutes of Health, Bethesda, MD
| | - Michael J Roy
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD.,Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD
| | - Jessica M Gill
- National Institutes of Nursing Research, National Institutes of Health, Bethesda, MD
| |
Collapse
|
31
|
Belfer SJ, Bashaw AG, Perlis ML, Kayser MS. A Drosophila model of sleep restriction therapy for insomnia. Mol Psychiatry 2021; 26:492-507. [PMID: 30824866 PMCID: PMC6717687 DOI: 10.1038/s41380-019-0376-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 01/23/2019] [Accepted: 02/11/2019] [Indexed: 01/03/2023]
Abstract
Insomnia is the most common sleep disorder among adults, especially affecting individuals of advanced age or with neurodegenerative disease. Insomnia is also a common comorbidity across psychiatric disorders. Cognitive behavioral therapy for insomnia (CBT-I) is the first-line treatment for insomnia; a key component of this intervention is restriction of sleep opportunity, which optimizes matching of sleep ability and opportunity, leading to enhanced sleep drive. Despite the well-documented efficacy of CBT-I, little is known regarding how CBT-I works at a cellular and molecular level to improve sleep, due in large part to an absence of experimentally-tractable animals models of this intervention. Here, guided by human behavioral sleep therapies, we developed a Drosophila model for sleep restriction therapy (SRT) of insomnia. We demonstrate that restriction of sleep opportunity through manipulation of environmental cues improves sleep efficiency in multiple short-sleeping Drosophila mutants. The response to sleep opportunity restriction requires ongoing environmental inputs, but is independent of the molecular circadian clock. We apply this sleep opportunity restriction paradigm to aging and Alzheimer's disease fly models, and find that sleep impairments in these models are reversible with sleep restriction, with associated improvement in reproductive fitness and extended lifespan. This work establishes a model to investigate the neurobiological basis of CBT-I, and provides a platform that can be exploited toward novel treatment targets for insomnia.
Collapse
Affiliation(s)
- Samuel J. Belfer
- Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States,Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States
| | - Alexander G. Bashaw
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States
| | - Michael L. Perlis
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States,Behavioral Sleep Medicine Program, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States,Center for Sleep and Circadian Neurobiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States
| | - Matthew S. Kayser
- Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States,Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States,Center for Sleep and Circadian Neurobiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States,Chronobiology Program, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, United States,Lead Contact and Correspondence: Dr. Matthew S. Kayser, Clinical Research Building, Room 320, 415 Curie Boulevard, Philadelphia, PA 19143, , Telephone: 215 898 8268, Fax: 215 898 0509
| |
Collapse
|
32
|
Sehgal A. The 2020 Pittendrigh/Aschoff Lecture: My Circadian Journey. J Biol Rhythms 2021; 36:84-96. [PMID: 33428509 PMCID: PMC8815313 DOI: 10.1177/0748730420982398] [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] [Indexed: 11/16/2022]
Abstract
The circadian field has come a long way since I started as a postdoctoral fellow ~30 years ago. At the time, the only known animal clock gene was period, so I had the privilege of witnessing, and participating in, the molecular revolution that took us from the discovery of the circadian clock mechanism to the identification of pathways that link clocks to behavior and physiology. This lecture highlights my role and perspective in these developments, and also demonstrates how the successful use of Drosophila for studies of circadian rhythms inspired us to develop a fly model for sleep. I also touch upon my experiences as a non-white immigrant woman navigating my way through the US science and education system, and hope my story will be of interest to some.
Collapse
Affiliation(s)
- Amita Sehgal
- Howard Hughes Medical Institute, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
33
|
Whole blood transcriptome analysis using RNA sequencing in individuals with insomnia disorder and good sleepers: a pilot study. Sleep Med 2021; 80:1-8. [PMID: 33530007 DOI: 10.1016/j.sleep.2021.01.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 01/10/2023]
Abstract
BACKGROUND Insomnia is a highly prevalent condition that is associated with negative health outcomes, yet little is known about the underlying molecular mechanisms. METHOD RNA sequencing was conducted using blood samples from 15 individuals with primary insomnia and 15 age- and gender-matched good sleeper controls. The RNA library was sequenced with 150 base pair paired-ends on the Illumina NovaSeq-6000 platform. Alignment was performed using human reference genome hg38. Differential gene expression analysis was performed using DESeq2 following alignment, using log fold change ±0.50, and had a false discovery rate p-value <0.05. Pathway analysis was performed using Ingenuity Pathway Analysis. RESULTS We found 288 differentially expressed genes in insomnia patients when compared to controls. Upregulated genes included LINC02224 (Long Intergenic Non-Protein Coding RNA 2224), DUX4L9 (Double Homeobox 4 Like 9), and TUSC3 (Tumor Suppressor Candidate 3) and down regulated genes included CTXN2 (Cortexin 2), CSMD1 (CUB And Sushi Multiple Domains 1), and SLC12A1 (Solute Carrier Family 12 Member 1). Ingenuity® Pathway Analysis (IPA) revealed 3 associated networks (score>40) with genes and hubs related to inflammation (nuclear factor-kB), oxidative stress (Mitochondrial complex 1) and ubiquitination. CONCLUSION Differentially expressed genes in this analysis are functionally associated with inflammation and immune response, mitochondrial and metabolic processes. Further research into the transcriptomic changes in insomnia is needed to understand related pathways to the disorder and provide new avenues for diagnostics and therapeutics.
Collapse
|
34
|
Kumar S, Tunc I, Tansey TR, Pirooznia M, Harbison ST. Identification of Genes Contributing to a Long Circadian Period in Drosophila Melanogaster. J Biol Rhythms 2020; 36:239-253. [PMID: 33274675 DOI: 10.1177/0748730420975946] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The endogenous circadian period of animals and humans is typically very close to 24 h. Individuals with much longer circadian periods have been observed, however, and in the case of humans, these deviations have health implications. Previously, we observed a line of Drosophila with a very long average period of 31.3 h for locomotor activity behavior. Preliminary mapping indicated that the long period did not map to known canonical clock genes but instead mapped to multiple chromosomes. Using RNA-Seq, we surveyed the whole transcriptome of fly heads from this line across time and compared it with a wild-type control. A three-way generalized linear model revealed that approximately two-thirds of the genes were expressed differentially among the two genotypes, while only one quarter of the genes varied across time. Using these results, we applied algorithms to search for genes that oscillated over 24 h, identifying genes not previously known to cycle. We identified 166 differentially expressed genes that overlapped with a previous Genome-wide Association Study (GWAS) of circadian behavior, strongly implicating them in the long-period phenotype. We tested mutations in 45 of these genes for their effect on the circadian period. Mutations in Alk, alph, CG10089, CG42540, CG6034, Kairos (CG6123), CG8768, klg, Lar, sick, and tinc had significant effects on the circadian period, with seven of these mutations increasing the circadian period of locomotor activity behavior. Genetic rescue of mutant Kairos restored the circadian period to wild-type levels, suggesting it has a critical role in determining period length in constant darkness.
Collapse
Affiliation(s)
- Shailesh Kumar
- Laboratory of Systems Genetics, Systems Biology Center, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Ilker Tunc
- Bioinformatics and Computational Biology Core, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Terry R Tansey
- Laboratory of Systems Genetics, Systems Biology Center, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Mehdi Pirooznia
- Bioinformatics and Computational Biology Core, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Susan T Harbison
- Laboratory of Systems Genetics, Systems Biology Center, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| |
Collapse
|
35
|
K + Channel Tetramerization Domain 5 (KCTD5) Protein Regulates Cell Migration, Focal Adhesion Dynamics and Spreading through Modulation of Ca 2+ Signaling and Rac1 Activity. Cells 2020; 9:cells9102273. [PMID: 33053687 PMCID: PMC7600296 DOI: 10.3390/cells9102273] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/03/2020] [Accepted: 10/05/2020] [Indexed: 01/08/2023] Open
Abstract
Cell migration is critical for several physiological and pathophysiological processes. It depends on the coordinated action of kinases, phosphatases, Rho-GTPases proteins, and Ca2+ signaling. Interestingly, ubiquitination events have emerged as regulatory elements of migration. Thus, the role of proteins involved in ubiquitination processes could be relevant to a complete understanding of pro-migratory mechanisms. KCTD5 is a member of Potassium Channel Tetramerization Domain (KCTD) proteins that have been proposed as a putative adaptor for Cullin3-E3 ubiquitin ligase and a novel regulatory protein of TRPM4 channels. Here, we study whether KCTD5 participates in cell migration-associated mechanisms, such as focal adhesion dynamics and cellular spreading. Our results show that KCTD5 CRISPR/Cas9- and shRNA-based depletion in B16-F10 cells promoted an increase in cell migration and cell spreading, and a decrease in the focal adhesion area, consistent with an increased focal adhesion disassembly rate. The expression of a dominant-negative mutant of Rho-GTPases Rac1 precluded the KCTD5 depletion-induced increase in cell spreading. Additionally, KCTD5 silencing decreased the serum-induced Ca2+ response, and the reversion of this with ionomycin abolished the KCTD5 knockdown-induced decrease in focal adhesion size. Together, these data suggest that KCTD5 acts as a regulator of cell migration by modulating cell spreading and focal adhesion dynamics through Rac1 activity and Ca2+ signaling, respectively.
Collapse
|
36
|
Abstract
Sleep is a fundamental property conserved across species. The homeostatic induction of sleep indicates the presence of a mechanism that is progressively activated by the awake state and that induces sleep. Several lines of evidence support that such function, namely, sleep need, lies in the neuronal assemblies rather than specific brain regions and circuits. However, the molecular mechanism underlying the dynamics of sleep need is still unclear. This review aims to summarize recent studies mainly in rodents indicating that protein phosphorylation, especially at the synapses, could be the molecular entity associated with sleep need. Genetic studies in rodents have identified a set of kinases that promote sleep. The activity of sleep-promoting kinases appears to be elevated during the awake phase and in sleep deprivation. Furthermore, the proteomic analysis demonstrated that the phosphorylation status of synaptic protein is controlled by the sleep-wake cycle. Therefore, a plausible scenario may be that the awake-dependent activation of kinases modifies the phosphorylation status of synaptic proteins to promote sleep. We also discuss the possible importance of multisite phosphorylation on macromolecular protein complexes to achieve the slow dynamics and physiological functions of sleep in mammals.
Collapse
Affiliation(s)
- Koji L Ode
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroki R Ueda
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics, Osaka, Japan
| |
Collapse
|
37
|
Hypnotic effect of thalidomide is independent of teratogenic ubiquitin/proteasome pathway. Proc Natl Acad Sci U S A 2020; 117:23106-23112. [PMID: 32848052 DOI: 10.1073/pnas.1917701117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Thalidomide exerts its teratogenic and immunomodulatory effects by binding to cereblon (CRBN) and thereby inhibiting/modifying the CRBN-mediated ubiquitination pathway consisting of the Cullin4-DDB1-ROC1 E3 ligase complex. The mechanism of thalidomide's classical hypnotic effect remains largely unexplored, however. Here we examined whether CRBN is involved in the hypnotic effect of thalidomide by generating mice harboring a thalidomide-resistant mutant allele of Crbn (Crbn YW/AA knock-in mice). Thalidomide increased non-REM sleep time in Crbn YW/AA knock-in homozygotes and heterozygotes to a similar degree as seen in wild-type littermates. Thalidomide similarly depressed excitatory synaptic transmission in the cortical slices obtained from wild-type and Crbn YW/AA homozygous knock-in mice without affecting GABAergic inhibition. Thalidomide induced Fos expression in vasopressin-containing neurons of the supraoptic nucleus and reduced Fos expression in the tuberomammillary nuclei. Thus, thalidomide's hypnotic effect seems to share some downstream mechanisms with general anesthetics and GABAA-activating sedatives but does not involve the teratogenic CRBN-mediated ubiquitin/proteasome pathway.
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW The goal of this review is to evaluate recent advances in understanding the pivotal roles of Cullin-3 (CUL3) in blood pressure regulation with a focus on its actions in the kidney and blood vessels. RECENT FINDINGS Cul3-based ubiquitin ligase regulates renal electrolyte transport, vascular tone, and redox homeostasis by facilitating the normal turnover of (1) with-no-lysine kinases in the distal nephron, (2) RhoA and phosphodiesterase 5 in the vascular smooth muscle, and (3) nuclear factor E2-related factor 2 in antioxidant responses. CUL3 mutations identified in familial hyperkalemic hypertension (FHHt) yield a mutant protein lacking exon 9 (CUL3∆9) which displays dual gain and loss of function. CUL3∆9 acts in a dominant manner to impair CUL3-mediated substrate ubiquitylation and degradation. The consequent accumulation of substrates and overactivation of downstream signaling cause FHHt through increased sodium reabsorption, enhanced vasoconstriction, and decreased vasodilation. CUL3 ubiquitin ligase maintains normal cardiovascular and renal physiology through posttranslational modification of key substrates which regulate blood pressure. Interference with CUL3 disturbs these key downstream pathways. Further understanding the spatial and temporal specificity of how CUL3 functions in these pathways is necessary to identify novel therapeutic targets for hypertension.
Collapse
|
39
|
Vaccaro A, Kaplan Dor Y, Nambara K, Pollina EA, Lin C, Greenberg ME, Rogulja D. Sleep Loss Can Cause Death through Accumulation of Reactive Oxygen Species in the Gut. Cell 2020; 181:1307-1328.e15. [PMID: 32502393 DOI: 10.1016/j.cell.2020.04.049] [Citation(s) in RCA: 233] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 01/15/2020] [Accepted: 04/24/2020] [Indexed: 02/06/2023]
Abstract
The view that sleep is essential for survival is supported by the ubiquity of this behavior, the apparent existence of sleep-like states in the earliest animals, and the fact that severe sleep loss can be lethal. The cause of this lethality is unknown. Here we show, using flies and mice, that sleep deprivation leads to accumulation of reactive oxygen species (ROS) and consequent oxidative stress, specifically in the gut. ROS are not just correlates of sleep deprivation but drivers of death: their neutralization prevents oxidative stress and allows flies to have a normal lifespan with little to no sleep. The rescue can be achieved with oral antioxidant compounds or with gut-targeted transgenic expression of antioxidant enzymes. We conclude that death upon severe sleep restriction can be caused by oxidative stress, that the gut is central in this process, and that survival without sleep is possible when ROS accumulation is prevented. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Alexandra Vaccaro
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Yosef Kaplan Dor
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Keishi Nambara
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Cindy Lin
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Dragana Rogulja
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
40
|
Malik DM, Paschos GK, Sehgal A, Weljie AM. Circadian and Sleep Metabolomics Across Species. J Mol Biol 2020; 432:3578-3610. [PMID: 32376454 PMCID: PMC7781158 DOI: 10.1016/j.jmb.2020.04.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/28/2020] [Accepted: 04/28/2020] [Indexed: 02/06/2023]
Abstract
Under normal circadian function, metabolic control is temporally coordinated across tissues and behaviors with a 24-h period. However, circadian disruption results in negative consequences for metabolic homeostasis including energy or redox imbalances. Yet, circadian disruption has become increasingly prevalent within today's society due to many factors including sleep loss. Metabolic consequences of both have been revealed by metabolomics analyses of circadian biology and sleep. Specifically, two primary analytical platforms, mass spectrometry and nuclear magnetic resonance spectroscopy, have been used to study molecular clock and sleep influences on overall metabolic rhythmicity. For example, human studies have demonstrated the prevalence of metabolic rhythms in human biology, as well as pan-metabolome consequences of sleep disruption. However, human studies are limited to peripheral metabolic readouts primarily through minimally invasive procedures. For further tissue- and organism-specific investigations, a number of model systems have been studied, based upon the conserved nature of both the molecular clock and sleep across species. Here we summarize human studies as well as key findings from metabolomics studies using mice, Drosophila, and zebrafish. While informative, a limitation in existing literature is a lack of interpretation regarding dynamic synthesis or catabolism within metabolite pools. To this extent, future work incorporating isotope tracers, specific metabolite reporters, and single-cell metabolomics may provide a means of exploring dynamic activity in pathways of interest.
Collapse
Affiliation(s)
- Dania M Malik
- Pharmacology Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Georgios K Paschos
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amita Sehgal
- Penn Chronobiology, University of Pennsylvania, Philadelphia, PA 19104, USA; Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Aalim M Weljie
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
41
|
Maurer GW, Malita A, Nagy S, Koyama T, Werge TM, Halberg KA, Texada MJ, Rewitz K. Analysis of genes within the schizophrenia-linked 22q11.2 deletion identifies interaction of night owl/LZTR1 and NF1 in GABAergic sleep control. PLoS Genet 2020; 16:e1008727. [PMID: 32339168 PMCID: PMC7205319 DOI: 10.1371/journal.pgen.1008727] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 05/07/2020] [Accepted: 03/20/2020] [Indexed: 12/14/2022] Open
Abstract
The human 22q11.2 chromosomal deletion is one of the strongest identified genetic risk factors for schizophrenia. Although the deletion spans a number of known genes, the contribution of each of these to the 22q11.2 deletion syndrome (DS) is not known. To investigate the effect of individual genes within this interval on the pathophysiology associated with the deletion, we analyzed their role in sleep, a behavior affected in virtually all psychiatric disorders, including the 22q11.2 DS. We identified the gene LZTR1 (night owl, nowl) as a regulator of night-time sleep in Drosophila. In humans, LZTR1 has been associated with Ras-dependent neurological diseases also caused by Neurofibromin-1 (Nf1) deficiency. We show that Nf1 loss leads to a night-time sleep phenotype nearly identical to that of nowl loss and that nowl negatively regulates Ras and interacts with Nf1 in sleep regulation. Furthermore, nowl is required for metabolic homeostasis, suggesting that LZTR1 may contribute to the genetic susceptibility to obesity associated with the 22q11.2 DS. Knockdown of nowl or Nf1 in GABA-responsive sleep-promoting neurons elicits the sleep phenotype, and this defect can be rescued by increased GABAA receptor signaling, indicating that Nowl regulates sleep through modulation of GABA signaling. Our results suggest that nowl/LZTR1 may be a conserved regulator of GABA signaling important for normal sleep that contributes to the 22q11.2 DS. Schizophrenia is a devastating mental disorder with a large genetic component to disease predisposition. One of the strongest genetic risk factors for this disorder is a relatively small genetic deletion of 43 genes on the 22nd chromosome, called 22q11.2, which confers about a 25% risk of schizophrenia development. However, it is likely that only some of these deleted genes affect disease risk, so we tested most of them individually. One of the main symptoms of schizophrenia is disturbed sleep. Sleep is an evolutionarily conserved behavior that can be easily studied in the fruit fly Drosophila melanogaster, so we investigated the effect on sleep of blocking expression of the fly homologs of most of the 22q11.2 genes and identified the gene LZTR1 (night owl, nowl) as an important sleep regulator. We found that Nowl/LZTR1 is required for inhibition of the Ras pathway and interacts genetically with the Ras inhibitor NF1. Nowl/LZTR1 appears to function in sleep by modulating inhibitory GABA signaling, which is affected in schizophrenia. Thus, this gene may underlie some of the phenotypes of the human schizophrenia-risk deletion.
Collapse
Affiliation(s)
- Gianna W. Maurer
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Alina Malita
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Stanislav Nagy
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Takashi Koyama
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Thomas M. Werge
- Institute for Biological Psychiatry, Mental Health Centre Sct. Hans, Roskilde, Denmark
| | | | - Michael J. Texada
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kim Rewitz
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
42
|
Covert sleep-related biological processes are revealed by probabilistic analysis in Drosophila. Proc Natl Acad Sci U S A 2020; 117:10024-10034. [PMID: 32303656 PMCID: PMC7211995 DOI: 10.1073/pnas.1917573117] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Reduced sleep duration and disrupted sleep quality are correlated with adverse mental and physical health outcomes. Better tools for measuring the internal drives for sleep and wake in model organisms would facilitate understanding the role of sleep quality in health. We defined two conditional probabilities, P(Wake) and P(Doze), that can be calculated from recordings of Drosophila activity without disturbing the animal. We demonstrated that P(Wake) is a measure of sleep depth and that P(Doze) is a measure of sleep pressure. In parallel, we developed an automatic classifier for state-based analysis of Drosophila behavior. These analysis tools will improve our understanding of the pharmacology and neuronal regulation of behavioral drives in the Drosophila brain. Sleep pressure and sleep depth are key regulators of wake and sleep. Current methods of measuring these parameters in Drosophila melanogaster have low temporal resolution and/or require disrupting sleep. Here we report analysis tools for high-resolution, noninvasive measurement of sleep pressure and depth from movement data. Probability of initiating activity, P(Wake), measures sleep depth while probability of ceasing activity, P(Doze), measures sleep pressure. In vivo and computational analyses show that P(Wake) and P(Doze) are largely independent and control the amount of total sleep. We also develop a Hidden Markov Model that allows visualization of distinct sleep/wake substates. These hidden states have a predictable relationship with P(Doze) and P(Wake), suggesting that the methods capture the same behaviors. Importantly, we demonstrate that both the Doze/Wake probabilities and the sleep/wake substates are tied to specific biological processes. These metrics provide greater mechanistic insight into behavior than measuring the amount of sleep alone.
Collapse
|
43
|
Rivas J, Díaz N, Silva I, Morales D, Lavanderos B, Álvarez A, Saldías MP, Pulgar E, Cruz P, Maureira D, Flores G, Colombo A, Blanco C, Contreras HR, Jaña F, Gallegos I, Concha ML, Vergara-Jaque A, Poblete H, González W, Varela D, Trimmer JS, Cáceres M, Cerda O. KCTD5, a novel TRPM4-regulatory protein required for cell migration as a new predictor for breast cancer prognosis. FASEB J 2020; 34:7847-7865. [PMID: 32301552 DOI: 10.1096/fj.201901195rrr] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 03/03/2020] [Accepted: 03/26/2020] [Indexed: 12/23/2022]
Abstract
Transient receptor potential melastatin 4 (TRPM4) is a Ca2+ -activated nonselective cationic channel that regulates cell migration and contractility. Increased TRPM4 expression has been related to pathologies, in which cytoskeletal rearrangement and cell migration are altered, such as metastatic cancer. Here, we identify the K+ channel tetramerization domain 5 (KCTD5) protein, a putative adaptor of cullin3 E3 ubiquitin ligase, as a novel TRPM4-interacting protein. We demonstrate that KCTD5 is a positive regulator of TRPM4 activity by enhancing its Ca2+ sensitivity. We show that through its effects on TRPM4 that KCTD5 promotes cell migration and contractility. Finally, we observed that both TRPM4 and KCTD5 expression are increased in distinct patterns in different classes of breast cancer tumor samples. Together, these data support that TRPM4 activity can be regulated through expression levels of either TRPM4 or KCTD5, not only contributing to increased understanding of the molecular mechanisms involved on the regulation of these important ion channels, but also providing information that could inform treatments based on targeting these distinct molecules that define TRPM4 activity.
Collapse
Affiliation(s)
- José Rivas
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,Departamento de Ciencias de la Salud, Universidad de Aysén, Coyhaique, Chile
| | - Nicolás Díaz
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Ian Silva
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Danna Morales
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,Program of Physiology and Biophysics, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Boris Lavanderos
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Alhejandra Álvarez
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - María Paz Saldías
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Eduardo Pulgar
- Program of Anatomy and Developmental Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Pablo Cruz
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Diego Maureira
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Guillermo Flores
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Alicia Colombo
- Departamento de Oncología Básico Clínica, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Departamento de Anatomía Patológica, Hospital Clínico Universidad de Chile, Universidad de Chile, Santiago, Chile
| | - Constanza Blanco
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Héctor R Contreras
- Departamento de Oncología Básico Clínica, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Fabián Jaña
- Departamento de Ciencias de la Salud, Universidad de Aysén, Coyhaique, Chile.,The Wound Repair, Treatment and Health (WoRTH) Initiative, Santiago, Chile
| | - Ivan Gallegos
- Departamento de Oncología Básico Clínica, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Departamento de Anatomía Patológica, Hospital Clínico Universidad de Chile, Universidad de Chile, Santiago, Chile
| | - Miguel L Concha
- Program of Anatomy and Developmental Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile.,Millennium Nucleus on Physics of Active Matter, Santiago, Chile
| | - Ariela Vergara-Jaque
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,Multidisciplinary Scientific Nucleus, Universidad de Talca, Talca, Chile.,Center for Bioinformatics and Molecular Simulations (CBSM), Faculty of Engineering, Universidad de Talca, Talca, Chile
| | - Horacio Poblete
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,Multidisciplinary Scientific Nucleus, Universidad de Talca, Talca, Chile.,Center for Bioinformatics and Molecular Simulations (CBSM), Faculty of Engineering, Universidad de Talca, Talca, Chile
| | - Wendy González
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,Center for Bioinformatics and Molecular Simulations (CBSM), Faculty of Engineering, Universidad de Talca, Talca, Chile
| | - Diego Varela
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,Program of Physiology and Biophysics, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - James S Trimmer
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA, USA
| | - Mónica Cáceres
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,The Wound Repair, Treatment and Health (WoRTH) Initiative, Santiago, Chile
| | - Oscar Cerda
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,The Wound Repair, Treatment and Health (WoRTH) Initiative, Santiago, Chile
| |
Collapse
|
44
|
Singh P, Donlea JM. Bidirectional Regulation of Sleep and Synapse Pruning after Neural Injury. Curr Biol 2020; 30:1063-1076.e3. [PMID: 32142703 DOI: 10.1016/j.cub.2019.12.065] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 09/10/2019] [Accepted: 12/19/2019] [Indexed: 12/14/2022]
Abstract
Following acute neural injury, severed axons undergo programmed Wallerian degeneration over several following days. While sleep has been linked with synaptic reorganization under other conditions, the role of sleep in responses to neural injuries remains poorly understood. To study the relationship between sleep and neural injury responses, we examined Drosophila melanogaster following the removal of antennae or other sensory tissues. Daytime sleep is elevated after antennal or wing injury, but sleep returns to baseline levels within 24 h after injury. Similar increases in sleep are not observed when olfactory receptor neurons are silenced or when other sensory organs are severed, suggesting that increased sleep after injury is not attributed to sensory deprivation, nociception, or generalized inflammatory responses. Neuroprotective disruptions of the E3 ubiquitin ligase highwire and c-Jun N-terminal kinase basket in olfactory receptor neurons weaken the sleep-promoting effects of antennal injury, suggesting that post-injury sleep may be influenced by the clearance of damaged neurons. Finally, we show that pre-synaptic active zones are preferentially removed from severed axons within hours after injury and that depriving recently injured flies of sleep slows the removal of both active zones and damaged axons. These data support a bidirectional interaction between sleep and synapse pruning after antennal injury: locally increasing the need to clear neural debris is associated with increased sleep, which is required for efficient active zone removal after injury.
Collapse
Affiliation(s)
- Prabhjit Singh
- Department of Neurobiology, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA 90095-1763, USA
| | - Jeffrey M Donlea
- Department of Neurobiology, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA 90095-1763, USA.
| |
Collapse
|
45
|
Huang S, Piao C, Beuschel CB, Götz T, Sigrist SJ. Presynaptic Active Zone Plasticity Encodes Sleep Need in Drosophila. Curr Biol 2020; 30:1077-1091.e5. [PMID: 32142702 DOI: 10.1016/j.cub.2020.01.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 09/26/2019] [Accepted: 01/07/2020] [Indexed: 01/04/2023]
Abstract
Sleep is universal across species and essential for quality of life and health, as evidenced by the consequences of sleep loss. Sleep might homeostatically normalize synaptic gains made over wake states in order to reset information processing and storage and support learning, and sleep-associated synaptic (ultra)structural changes have been demonstrated recently. However, causal relationships between the molecular and (ultra)structural status of synapses, sleep homeostatic regulation, and learning processes have yet to be established. We show here that the status of the presynaptic active zone can directly control sleep in Drosophila. Short sleep mutants showed a brain-wide upregulation of core presynaptic scaffold proteins and release factors. Increasing the gene copy number of ELKS-family scaffold master organizer Bruchpilot (BRP) not only mimicked changes in the active zone scaffold and release proteins but importantly provoked sleep in a dosage-dependent manner, qualitatively and quantitatively reminiscent of sleep deprivation effects. Conversely, reducing the brp copy number decreased sleep in short sleep mutant backgrounds, suggesting a specific role of the active zone plasticity in homeostatic sleep regulation. Finally, elimination of BRP specifically in the sleep-promoting R2 neurons of 4xBRP animals partially restored sleep patterns and rescued learning deficits. Our results suggest that the presynaptic active zone plasticity driven by BRP operates as a sleep homeostatic actuator that also restricts periods of effective learning.
Collapse
Affiliation(s)
- Sheng Huang
- Institute for Biology/Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany; NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Charitéplatz 1, 10117 Berlin, Germany
| | - Chengji Piao
- Institute for Biology/Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
| | - Christine B Beuschel
- Institute for Biology/Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
| | - Torsten Götz
- Institute for Biology/Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
| | - Stephan J Sigrist
- Institute for Biology/Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany; NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
46
|
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: 2.8] [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
|
47
|
FUNATO H. Forward genetic approach for behavioral neuroscience using animal models. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2020; 96:10-31. [PMID: 31932526 PMCID: PMC6974404 DOI: 10.2183/pjab.96.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 10/18/2019] [Indexed: 06/10/2023]
Abstract
Forward genetics is a powerful approach to understand the molecular basis of animal behaviors. Fruit flies were the first animal to which this genetic approach was applied systematically and have provided major discoveries on behaviors including sexual, learning, circadian, and sleep-like behaviors. The development of different classes of model organism such as nematodes, zebrafish, and mice has enabled genetic research to be conducted using more-suitable organisms. The unprecedented success of forward genetic approaches was the identification of the transcription-translation negative feedback loop composed of clock genes as a fundamental and conserved mechanism of circadian rhythm. This approach has now expanded to sleep/wakefulness in mice. A conventional strategy such as dominant and recessive screenings can be modified with advances in DNA sequencing and genome editing technologies.
Collapse
Affiliation(s)
- Hiromasa FUNATO
- Department of Anatomy, Faculty of Medicine, Toho University, Tokyo, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
48
|
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: 604] [Impact Index Per Article: 120.8] [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
|
49
|
Toda H, Williams JA, Gulledge M, Sehgal A. A sleep-inducing gene, nemuri, links sleep and immune function in Drosophila. Science 2019; 363:509-515. [PMID: 30705188 PMCID: PMC6505470 DOI: 10.1126/science.aat1650] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 06/29/2018] [Accepted: 11/26/2018] [Indexed: 12/19/2022]
Abstract
Sleep remains a major mystery of biology. In particular, little is known about the mechanisms that account for the drive to sleep. In an unbiased screen of more than 12,000 Drosophila lines, we identified a single gene, nemuri, that induces sleep. The NEMURI protein is an antimicrobial peptide that can be secreted ectopically to drive prolonged sleep (with resistance to arousal) and to promote survival after infection. Loss of nemuri increased arousability during daily sleep and attenuated the acute increase in sleep induced by sleep deprivation or bacterial infection. Conditions that increase sleep drive induced expression of nemuri in a small number of fly brain neurons and targeted it to the sleep-promoting, dorsal fan-shaped body. We propose that NEMURI is a bona fide sleep homeostasis factor that is particularly important under conditions of high sleep need; because these conditions include sickness, our findings provide a link between sleep and immune function.
Collapse
Affiliation(s)
| | | | - Michael Gulledge
- Howard Hughes Medical Institute, Chronobiology Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|
50
|
Kikuma K, Li X, Perry S, Li Q, Goel P, Chen C, Kim D, Stavropoulos N, Dickman D. Cul3 and insomniac are required for rapid ubiquitination of postsynaptic targets and retrograde homeostatic signaling. Nat Commun 2019; 10:2998. [PMID: 31278365 PMCID: PMC6611771 DOI: 10.1038/s41467-019-10992-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 06/14/2019] [Indexed: 01/05/2023] Open
Abstract
At the Drosophila neuromuscular junction, inhibition of postsynaptic glutamate receptors activates retrograde signaling that precisely increases presynaptic neurotransmitter release to restore baseline synaptic strength. However, the nature of the underlying postsynaptic induction process remains enigmatic. Here, we design a forward genetic screen to discover factors in the postsynaptic compartment necessary to generate retrograde homeostatic signaling. This approach identified insomniac (inc), a putative adaptor for the Cullin-3 (Cul3) ubiquitin ligase complex, which together with Cul3 is essential for normal sleep regulation. Interestingly, we find that Inc and Cul3 rapidly accumulate at postsynaptic compartments following acute receptor inhibition and are required for a local increase in mono-ubiquitination. Finally, we show that Peflin, a Ca2+-regulated Cul3 co-adaptor, is necessary for homeostatic communication, suggesting a relationship between Ca2+ signaling and control of Cul3/Inc activity in the postsynaptic compartment. Our study suggests that Cul3/Inc-dependent mono-ubiquitination, compartmentalized at postsynaptic densities, gates retrograde signaling and provides an intriguing molecular link between the control of sleep and homeostatic plasticity at synapses. The authors use a forward genetic screen to discover postsynaptic factors required for homeostatic synaptic plasticity at the Drosophila neuromuscular junction. They identify insomniac and the ubiquitin ligase Cul3, genes involved in sleep regulation, to be necessary for retrograde homeostatic signalling at this synapse.
Collapse
Affiliation(s)
- Koto Kikuma
- Department of Neurobiology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Xiling Li
- Department of Neurobiology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Sarah Perry
- Department of Neurobiology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Qiuling Li
- Neuroscience Institute, Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY, 10016, USA
| | - Pragya Goel
- Department of Neurobiology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Catherine Chen
- Department of Neurobiology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Daniel Kim
- Department of Neurobiology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Nicholas Stavropoulos
- Neuroscience Institute, Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY, 10016, USA
| | - Dion Dickman
- Department of Neurobiology, University of Southern California, Los Angeles, CA, 90089, USA.
| |
Collapse
|