The zebrafish mutant dreammist implicates sodium homeostasis in sleep regulation

Sleep is a nearly universal feature of animal behaviour, yet many of the molecular, genetic, and neuronal substrates that orchestrate sleep/wake transitions lie undiscovered. Employing a viral insertion sleep screen in larval zebrafish, we identified a novel gene, dreammist (dmist), whose loss results in behavioural hyperactivity and reduced sleep at night. The neuronally expressed dmist gene is conserved across vertebrates and encodes a small single-pass transmembrane protein that is structurally similar to the Na+,K+-ATPase regulator, FXYD1/Phospholemman. Disruption of either fxyd1 or atp1a3a, a Na+,K+-ATPase alpha-3 subunit associated with several heritable movement disorders in humans, led to decreased night-time sleep. Since atpa1a3a and dmist mutants have elevated intracellular Na+ levels and non-additive effects on sleep amount at night, we propose that Dmist-dependent enhancement of Na+ pump function modulates neuronal excitability to maintain normal sleep behaviour.

[1]  Justus M. Kebschull,et al.  Hyperexcitable arousal circuits drive sleep instability during aging , 2022, Science.

[2]  Oriol Vinyals,et al.  Highly accurate protein structure prediction with AlphaFold , 2021, Nature.

[3]  Joseph M. Fernandez,et al.  A simple and effective F0 knockout method for rapid screening of behaviour and other complex phenotypes , 2020, bioRxiv.

[4]  D. Prober,et al.  The Serotonergic Raphe Promote Sleep in Zebrafish and Mice , 2019, Neuron.

[5]  Meena Kumari,et al.  Genetic studies of accelerometer-based sleep measures yield new insights into human sleep behaviour , 2019, Nature Communications.

[6]  Gero Miesenböck,et al.  A potassium channel β-subunit couples mitochondrial electron transport to sleep , 2019, Nature.

[7]  Max A. Little,et al.  Genome-wide association study identifies genetic loci for self-reported habitual sleep duration supported by accelerometer-derived estimates , 2019, Nature Communications.

[8]  V. Vacic,et al.  Genome-wide analysis of insomnia in 1,331,010 individuals identifies new risk loci and functional pathways , 2019, Nature Genetics.

[9]  A. Sehgal,et al.  A sleep-inducing gene, nemuri, links sleep and immune function in Drosophila , 2019, Science.

[10]  Max A. Little,et al.  Biological and clinical insights from genetics of insomnia symptoms , 2019, Nature Genetics.

[11]  Sujay M Kansagra,et al.  Polysomnography Findings and Sleep Disorders in Children With Alternating Hemiplegia of Childhood. , 2019, Journal of clinical sleep medicine : JCSM : official publication of the American Academy of Sleep Medicine.

[12]  K. Domschke,et al.  Developmental pathways towards mood disorders in adult life: Is there a role for sleep disturbances? , 2019, Journal of affective disorders.

[13]  Oriol Pavón Arocas,et al.  The Neuropeptide Galanin Is Required for Homeostatic Rebound Sleep following Increased Neuronal Activity , 2019, Neuron.

[14]  A. Nakashima,et al.  Dec1 and CLOCK Regulate Na+/K+-ATPase &bgr;1 Subunit Expression and Blood Pressure , 2018, Hypertension.

[15]  Gunnar H. D. Poplawski,et al.  Super-Mendelian inheritance mediated by CRISPR/Cas9 in the female mouse germline , 2018, Nature.

[16]  R. Deo,et al.  A Rapid Method for Directed Gene Knockout for Screening in G0 Zebrafish. , 2018, Developmental cell.

[17]  D. Prober,et al.  Neuropeptide Y Regulates Sleep by Modulating Noradrenergic Signaling , 2017, Current Biology.

[18]  D. Prober,et al.  Author response: Genetic and neuronal regulation of sleep by neuropeptide VF , 2017 .

[19]  H. Yost,et al.  CRISPR/Cas9‐Directed Gene Editing for the Generation of Loss‐of‐Function Mutants in High‐Throughput Zebrafish F0 Screens , 2017, Current protocols in molecular biology.

[20]  H. Poulsen,et al.  The Structure and Function of the Na,K-ATPase Isoforms in Health and Disease , 2017, Front. Physiol..

[21]  J. Rihel,et al.  Zebrafish sleep: from geneZZZ to neuronZZZ , 2017, Current Opinion in Neurobiology.

[22]  D. Raizen,et al.  The RFamide receptor DMSR-1 regulates stress-induced sleep in C. elegans , 2017, eLife.

[23]  Tomohiro Suzuki,et al.  Forward-genetics analysis of sleep in randomly mutagenized mice , 2016, Nature.

[24]  W. Joiner,et al.  Unraveling the Evolutionary Determinants of Sleep , 2016, Current Biology.

[25]  Maiken Nedergaard,et al.  Changes in the composition of brain interstitial ions control the sleep-wake cycle , 2016, Science.

[26]  Joseph M. Fernandez,et al.  Estrogens Suppress a Behavioral Phenotype in Zebrafish Mutants of the Autism Risk Gene, CNTNAP2 , 2016, Neuron.

[27]  A. Schier,et al.  A Zebrafish Genetic Screen Identifies Neuromedin U as a Regulator of Sleep/Wake States , 2016, Neuron.

[28]  James Y. Zou Analysis of protein-coding genetic variation in 60,706 humans , 2015, Nature.

[29]  D. Prober,et al.  Norepinephrine is required to promote wakefulness and for hypocretin-induced arousal in zebrafish , 2015, eLife.

[30]  D. Raizen,et al.  FMRFamide signaling promotes stress-induced sleep in Drosophila , 2015, Brain, Behavior, and Immunity.

[31]  C. Moens,et al.  Rapid reverse genetic screening using CRISPR in zebrafish , 2015, Nature Methods.

[32]  D. Prober,et al.  Melatonin Is Required for the Circadian Regulation of Sleep , 2015, Neuron.

[33]  Syed M. Adil,et al.  Knock‐in mouse model of alternating hemiplegia of childhood: Behavioral and electrophysiologic characterization , 2015, Epilepsia.

[34]  W. Huber,et al.  Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 , 2014, Genome Biology.

[35]  K. Ikeda,et al.  Heterozygous mice deficient in Atp1a3 exhibit motor deficits by chronic restraint stress , 2014, Behavioural Brain Research.

[36]  T. Maniatis,et al.  An RNA-Sequencing Transcriptome and Splicing Database of Glia, Neurons, and Vascular Cells of the Cerebral Cortex , 2014, The Journal of Neuroscience.

[37]  Tessa G. Montague,et al.  Efficient Mutagenesis by Cas9 Protein-Mediated Oligonucleotide Insertion and Large-Scale Assessment of Single-Guide RNAs , 2014, PloS one.

[38]  George M. Church,et al.  CHOPCHOP: a CRISPR/Cas9 and TALEN web tool for genome editing , 2014, Nucleic Acids Res..

[39]  Mohamad A Mikati,et al.  Distinct neurological disorders with ATP1A3 mutations , 2014, The Lancet Neurology.

[40]  W. Joiner,et al.  SLEEPLESS Is a Bifunctional Regulator of Excitability and Cholinergic Synaptic Transmission , 2014, Current Biology.

[41]  P. Ingham,et al.  A method for high-throughput PCR-based genotyping of larval zebrafish tail biopsies. , 2013, BioTechniques.

[42]  A. Schier,et al.  Sites of action of sleep and wake drugs: insights from model organisms , 2013, Current Opinion in Neurobiology.

[43]  T. Sakurai Orexin deficiency and narcolepsy , 2013, Current Opinion in Neurobiology.

[44]  E. Rosato,et al.  Circadian Regulation of the Na+/K+-Atpase Alpha Subunit in the Visual System Is Mediated by the Pacemaker and by Retina Photoreceptors in Drosophila Melanogaster , 2013, PloS one.

[45]  Susan R. Wente,et al.  Efficient multiplex biallelic zebrafish genome editing using a CRISPR nuclease system , 2013, Proceedings of the National Academy of Sciences.

[46]  M. Shattock,et al.  Novel regulation of cardiac Na pump via phospholemman. , 2013, Journal of molecular and cellular cardiology.

[47]  S. Satake,et al.  Enhanced inhibitory neurotransmission in the cerebellar cortex of Atp1a3‐deficient heterozygous mice , 2013, The Journal of physiology.

[48]  H. C. Beck,et al.  α3Na+/K+-ATPase Deficiency Causes Brain Ventricle Dilation and Abrupt Embryonic Motility in Zebrafish* , 2013, The Journal of Biological Chemistry.

[49]  Matteo Pellegrini,et al.  A large-scale zebrafish gene knockout resource for the genome-wide study of gene function , 2013, Genome research.

[50]  H. Brismar,et al.  A Specific and Essential Role for Na,K-ATPase α3 in Neurons Co-expressing α1 and α3* , 2012, The Journal of Biological Chemistry.

[51]  David B. Goldstein,et al.  De novo mutations in ATP1A3 cause alternating hemiplegia of childhood , 2012, Nature Genetics.

[52]  L. A. Lowery,et al.  Multiple roles for the Na,K-ATPase subunits, Atp1a1 and Fxyd1, during brain ventricle development. , 2012, Developmental biology.

[53]  T. Meitinger,et al.  A KATP channel gene effect on sleep duration: from genome-wide association studies to function in Drosophila , 2011, Molecular Psychiatry.

[54]  J. Roder,et al.  Mania-like behavior induced by genetic dysfunction of the neuron-specific Na+,K+-ATPase α3 sodium pump , 2011, Proceedings of the National Academy of Sciences.

[55]  U. Gether,et al.  SLC6 Neurotransmitter Transporters: Structure, Function, and Regulation , 2011, Pharmacological Reviews.

[56]  F. Gebauer,et al.  Cytoplasmic polyadenylation and translational control. , 2011, Current opinion in genetics & development.

[57]  A. Sehgal,et al.  Genetics of Sleep and Sleep Disorders , 2011, Cell.

[58]  Sean R. Eddy,et al.  Hidden Markov model speed heuristic and iterative HMM search procedure , 2010, BMC Bioinformatics.

[59]  Kazutaka Katoh,et al.  Parallelization of the MAFFT multiple sequence alignment program , 2010, Bioinform..

[60]  S. Haggarty,et al.  Zebrafish Behavioral Profiling Links Drugs to Biological Targets and Rest/Wake Regulation , 2010, Science.

[61]  C. Ackerley,et al.  Mutation I810N in the α3 isoform of Na+,K+-ATPase causes impairments in the sodium pump and hyperexcitability in the CNS , 2009, Proceedings of the National Academy of Sciences.

[62]  Chiara Cirelli,et al.  The genetic and molecular regulation of sleep: from fruit flies to humans , 2009, Nature Reviews Neuroscience.

[63]  Steffen Schmidt,et al.  Nonsense-Mediated mRNA Decay Effectors Are Essential for Zebrafish Embryonic Development and Survival , 2009, Molecular and Cellular Biology.

[64]  A. Sehgal,et al.  Identification of SLEEPLESS, a Sleep-Promoting Factor , 2008, Science.

[65]  A. Tucker,et al.  Phospholemman-Mediated Activation of Na/K-ATPase Limits [Na]i and Inotropic State During &bgr;-Adrenergic Stimulation in Mouse Ventricular Myocytes , 2008, Circulation.

[66]  Melissa Hardy,et al.  The Tol2kit: A multisite gateway‐based construction kit for Tol2 transposon transgenesis constructs , 2007, Developmental dynamics : an official publication of the American Association of Anatomists.

[67]  S. Sivasubbu,et al.  Insertional mutagenesis strategies in zebrafish , 2007, Genome Biology.

[68]  Giulio Tononi,et al.  Sleep in Kcna2 knockout mice , 2007, BMC Biology.

[69]  Giulio Tononi,et al.  Drosophila Hyperkinetic Mutants Have Reduced Sleep and Impaired Memory , 2007, The Journal of Neuroscience.

[70]  Alexander F. Schier,et al.  Hypocretin/Orexin Overexpression Induces An Insomnia-Like Phenotype in Zebrafish , 2006, The Journal of Neuroscience.

[71]  Anton J. Enright,et al.  Zebrafish MiR-430 Promotes Deadenylation and Clearance of Maternal mRNAs , 2006, Science.

[72]  Giulio Tononi,et al.  Reduced sleep in Drosophila Shaker mutants , 2005, Nature.

[73]  Elizabeth Schroeder Activity , 2005, Encyclopedic Dictionary of Archaeology.

[74]  J. Lingrel,et al.  The α1 Isoform of Na,K-ATPase Regulates Cardiac Contractility and Functionally Interacts and Co-localizes with the Na/Ca Exchanger in Heart* , 2004, Journal of Biological Chemistry.

[75]  William B Dobyns,et al.  Mutations in the Na+/K+-ATPase α3 Gene ATP1A3 Are Associated with Rapid-Onset Dystonia Parkinsonism , 2004, Neuron.

[76]  K. Geering,et al.  FXYD Proteins: New Tissue‐ and Isoform‐Specific Regulators of Na,K‐ATPase , 2003, Annals of the New York Academy of Sciences.

[77]  L. Jones,et al.  Phospholemman, a Single-Span Membrane Protein, Is an Accessory Protein of Na,K-ATPase in Cerebellum and Choroid Plexus , 2003, The Journal of Neuroscience.

[78]  K. Geering,et al.  Phospholemman (FXYD1) associates with Na,K-ATPase and regulates its transport properties , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[79]  C. Clarke,et al.  A gene expression screen in zebrafish embryogenesis. , 2001, Genome research.

[80]  A. Therien,et al.  Mechanisms of sodium pump regulation. , 2000, American journal of physiology. Cell physiology.

[81]  Sebastiaan Overeem,et al.  A mutation in a case of early onset narcolepsy and a generalized absence of hypocretin peptides in human narcoleptic brains , 2000, Nature Medicine.

[82]  E. Rael,et al.  The FXYD gene family of small ion transport regulators or channels: cDNA sequence, protein signature sequence, and expression. , 2000, Genomics.

[83]  Emmanuel Mignot,et al.  The Sleep Disorder Canine Narcolepsy Is Caused by a Mutation in the Hypocretin (Orexin) Receptor 2 Gene , 1999, Cell.

[84]  Jon T. Willie,et al.  Narcolepsy in orexin Knockout Mice Molecular Genetics of Sleep Regulation , 1999, Cell.

[85]  A. Klip,et al.  Hormonal regulation of the Na(+)-K(+)-ATPase: mechanisms underlying rapid and sustained changes in pump activity. , 1995, The American journal of physiology.

[86]  C. Kimmel,et al.  Stages of embryonic development of the zebrafish , 1995, Developmental dynamics : an official publication of the American Association of Anatomists.

[87]  KM McGrail,et al.  Immunofluorescent localization of three Na,K-ATPase isozymes in the rat central nervous system: both neurons and glia can express more than one Na,K-ATPase , 1991, The Journal of neuroscience : the official journal of the Society for Neuroscience.

[88]  J. Lingrel,et al.  Structure-function relationships in the sodium-potassium ATPase .alpha. subunit: site-directed mutagenesis of glutamine-111 to arginine and asparagine-122 to aspartic acid generates a ouabain-resistant enzyme , 1988 .

[89]  Joseph M. Fernandez,et al.  Estrogens Suppress a Behavioral Phenotype in Zebrafish Mutants of the Autism Risk Gene , CNTNAP 2 Highlights , 2016 .

[90]  B. Thisse,et al.  High-resolution in situ hybridization to whole-mount zebrafish embryos , 2007, Nature Protocols.