1
|
Eiman MN, Kumar S, Serrano Negron YL, Tansey TR, Harbison ST. Genome-wide association in Drosophila identifies a role for Piezo and Proc-R in sleep latency. Sci Rep 2024; 14:260. [PMID: 38168575 PMCID: PMC10761942 DOI: 10.1038/s41598-023-50552-z] [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: 03/28/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024] Open
Abstract
Sleep latency, the amount of time that it takes an individual to fall asleep, is a key indicator of sleep need. Sleep latency varies considerably both among and within species and is heritable, but lacks a comprehensive description of its underlying genetic network. Here we conduct a genome-wide association study of sleep latency. Using previously collected sleep and activity data on a wild-derived population of flies, we calculate sleep latency, confirming significant, heritable genetic variation for this complex trait. We identify 520 polymorphisms in 248 genes contributing to variability in sleep latency. Tests of mutations in 23 candidate genes and additional putative pan-neuronal knockdown of 9 of them implicated CG44153, Piezo, Proc-R and Rbp6 in sleep latency. Two large-effect mutations in the genes Proc-R and Piezo were further confirmed via genetic rescue. This work greatly enhances our understanding of the genetic factors that influence variation in sleep latency.
Collapse
Affiliation(s)
- Matthew N Eiman
- Laboratory of Systems Genetics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Drexel University College of Medicine, Philadelphia, PA, USA
| | - Shailesh Kumar
- Laboratory of Systems Genetics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Division of Neuroscience and Behavior, National Institute On Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Yazmin L Serrano Negron
- Laboratory of Systems Genetics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Terry R Tansey
- Laboratory of Systems Genetics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Susan T Harbison
- Laboratory of Systems Genetics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
2
|
Eicher C, Marty B, Achermann P, Huber R, Landolt HP. Reduced subjective sleep quality in people rating themselves as electro-hypersensitive: An observational study. Sleep Med 2024; 113:165-171. [PMID: 38029625 DOI: 10.1016/j.sleep.2023.11.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/16/2023] [Accepted: 11/16/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Disturbed sleep is among the most frequent health complaints of people exposed to radio frequency electromagnetic fields (RF-EMF) used in mobile telecommunication, particularly in individuals who consider themselves as EMF hypersensitive (EHS). We aimed at investigating whether the EHS status per se is associated with sleep complaints. Because allelic variants of the gene encoding the L-type, voltage-gated calcium channel Cav1.2 (CACNA1C) were previously associated with sleep complaints reminiscent of those reported by EHS individuals, we also explored whether self-rated EHS status and sleep quality associate with these gene variants. METHODS A total of 2'040 participants (1'381 females) aged 18-30 years completed online, validated questionnaires on EMF sensitivity, subjective sleep quality, daytime sleepiness, mentation during sleep, and diurnal preference. They also provided a saliva sample for genotyping three functional variants of CACNA1C (rs7304986, rs16929277 and rs2302729). Eligible participants endorsing the question "Are you electro-hypersensitive?" were considered as "EHS" (n = 105), those denying this question yet believing to develop detrimental health symptoms due to prevailing electromagnetic pollution as "attributers" (n = 254), and the remaining participants as "non-EHS" (n = 1'406). We combined the EHS and attributers into one group for binary analyses. In exploratory analyses, we then tested possible associations between EMF sensitivity, subjective sleep variables and CACNA1C variants using linear and logistic regression. We used age, sex, level of education, presence of sleep disorders and habitual mobile phone use as covariates and corrected with Benjamini-Hochberg False Discovery Rate for multiple comparisons. RESULTS The EHS/attributers consistently reported prolonged sleep latency, reduced sleep quality, higher sleepiness and more nocturnal mentation when compared to non-EHS. Habitual mobile phone use was not associated with self-rated sleep latency and sleep quality scores. While the T-allele of variant rs2302729 of CACNA1C was associated with both, self-reported EMF sensitivity and reduced subjective sleep quality, we found no evidence for the hypothesis that EHS mediates impaired sleep quality via this allelic variant. CONCLUSIONS Irrespective of reported RF-EMF exposure, self-rated EHS/attributers rated subjective sleep quality worse than non-EHS individuals. TRIAL REGISTRATION Swiss National Clinical Trials Portal (SNCTP000002285) and ClinicalTrials.gov (NCT03074617).
Collapse
Affiliation(s)
- Corinne Eicher
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Benjamin Marty
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Peter Achermann
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland; Sleep & Health Zurich, University Center of Competence, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland; Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Reto Huber
- Sleep & Health Zurich, University Center of Competence, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland; Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Zurich, Switzerland; Child Development Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Hans-Peter Landolt
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland; Sleep & Health Zurich, University Center of Competence, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland; Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
3
|
Palagini L, Geoffroy PA, Gehrman PR, Miniati M, Gemignani A, Riemann D. Potential genetic and epigenetic mechanisms in insomnia: A systematic review. J Sleep Res 2023; 32:e13868. [PMID: 36918298 DOI: 10.1111/jsr.13868] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 03/16/2023]
Abstract
Insomnia is a stress-related sleep disorder conceptualised within a diathesis-stress framework, which it is thought to result from predisposing factors interacting with precipitating stressful events that trigger the development of insomnia. Among predisposing factors genetics and epigenetics may play a role. A systematic review of the current evidence for the genetic and epigenetic basis of insomnia was conducted according to Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) system. A total of 24 studies were collected for twins and family heritability, 55 for genome-wide association studies, 26 about candidate genes for insomnia, and eight for epigenetics. Data showed that insomnia is a complex polygenic stress-related disorder, and it is likely to be caused by a synergy of genetic and environmental factors, with stress-related sleep reactivity being the important trait. Even if few studies have been conducted to date on insomnia, epigenetics may be the framework to understand long-lasting consequences of the interaction between genetic and environmental factors and effects of stress on the brain in insomnia. Interestingly, polygenic risk for insomnia has been causally linked to different mental and medical disorders. Probably, by treating insomnia it would be possible to intervene on the effect of stress on the brain and prevent some medical and mental conditions.
Collapse
Affiliation(s)
- Laura Palagini
- Department of Clinical and Experimental Medicine, Unit of Psychiatry, Azienda Ospedaliero Universitaria Pisana AUOP, Pisa, Italy
| | - Pierre A Geoffroy
- Département de Psychiatrie et D'Addictologie, AP-HP, GHU Paris Nord, DMU Neurosciences, Hopital Bichat - Claude Bernard, Paris, France
- GHU Paris - Psychiatry and Neurosciences, Paris, France
- Université de Paris, NeuroDiderot, INSERM, Paris, France
| | - Philip R Gehrman
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Psychiatry, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mario Miniati
- Department of Clinical and Experimental Medicine, Unit of Psychiatry, Azienda Ospedaliero Universitaria Pisana AUOP, Pisa, Italy
| | - Angelo Gemignani
- Unit of Psychology, Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Azienda Ospedaliero Universitaria Pisana AUOP, Pisa, Italy
| | - Dieter Riemann
- Department of Psychiatry and Psychotherapy, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
4
|
Gessner NR, Peiravi M, Zhang F, Yimam S, Springer D, Harbison ST. A conserved role for frizzled in sleep architecture. SLEEP ADVANCES : A JOURNAL OF THE SLEEP RESEARCH SOCIETY 2023; 4:zpad045. [PMID: 38033424 PMCID: PMC10684271 DOI: 10.1093/sleepadvances/zpad045] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/03/2023] [Indexed: 12/02/2023]
Abstract
Previous studies of natural variants in Drosophila melanogaster implicated the Wnt signaling receptor frizzled in sleep. Given that the Wnt signaling pathway is highly conserved across species, we hypothesized that frizzled class receptor 1 (Fzd1), the murine homolog of frizzled, would also have a role in sleep. Using a CRISPR transgenic approach, we removed most of the Fzd1 coding region from C57BL/6N mice. We used a video assay to measure sleep characteristics in Fzd1-deficient mice. As Wnt signaling is known to affect visuospatial memory, we also examined the impact of the deletion on learning and memory using the novel object recognition (NOR) paradigm. Fzd1-deficient mice had altered sleep compared to littermate controls. The mice did not respond differently to the NOR paradigm compared to controls but did display anxiety-like behavior. Our strategy demonstrates that the study of natural variation in Drosophila sleep translates into candidate genes for sleep in vertebrate species such as the mouse.
Collapse
Affiliation(s)
- Nicholas R Gessner
- Laboratory of Systems Genetics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Morteza Peiravi
- Murine Phenotyping Core, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Fan Zhang
- Transgenic Core, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shemsiya Yimam
- Laboratory of Systems Genetics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Danielle Springer
- Murine Phenotyping Core, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Susan T Harbison
- Laboratory of Systems Genetics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
5
|
Austin-Zimmerman I, Levey DF, Giannakopoulou O, Deak JD, Galimberti M, Adhikari K, Zhou H, Denaxas S, Irizar H, Kuchenbaecker K, McQuillin A, Concato J, Buysse DJ, Gaziano JM, Gottlieb DJ, Polimanti R, Stein MB, Bramon E, Gelernter J. Genome-wide association studies and cross-population meta-analyses investigating short and long sleep duration. Nat Commun 2023; 14:6059. [PMID: 37770476 PMCID: PMC10539313 DOI: 10.1038/s41467-023-41249-y] [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: 08/16/2022] [Accepted: 08/28/2023] [Indexed: 09/30/2023] Open
Abstract
Sleep duration has been linked to a wide range of negative health outcomes and to reduced life expectancy. We present genome-wide association studies of short ( ≤ 5 h) and long ( ≥ 10 h) sleep duration in adults of European (N = 445,966), African (N = 27,785), East Asian (N = 3141), and admixed-American (N = 16,250) ancestry from UK Biobank and the Million Veteran Programme. In a cross-population meta-analysis, we identify 84 independent loci for short sleep and 1 for long sleep. We estimate SNP-based heritability for both sleep traits in each ancestry based on population derived linkage disequilibrium (LD) scores using cov-LDSC. We identify positive genetic correlation between short and long sleep traits (rg = 0.16 ± 0.04; p = 0.0002), as well as similar patterns of genetic correlation with other psychiatric and cardiometabolic phenotypes. Mendelian randomisation reveals a directional causal relationship between short sleep and depression, and a bidirectional causal relationship between long sleep and depression.
Collapse
Affiliation(s)
- Isabelle Austin-Zimmerman
- Department of Mental Health Neuroscience, Division of Psychiatry, University College London, London, W1T 7BN, UK
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
| | - Daniel F Levey
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| | - Olga Giannakopoulou
- Department of Mental Health Neuroscience, Division of Psychiatry, University College London, London, W1T 7BN, UK
- UCL Genetics Institute, Division of Biosciences, University College London, London, WC1E 6BT, UK
| | - Joseph D Deak
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| | - Marco Galimberti
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| | - Keyrun Adhikari
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| | - Hang Zhou
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| | - Spiros Denaxas
- Health Data Research UK, Institute of Health Informatics, University College London, London, NW1 2DA, UK
| | - Haritz Irizar
- Department of Mental Health Neuroscience, Division of Psychiatry, University College London, London, W1T 7BN, UK
- Department of Genetics & Genomic Sciences and Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Karoline Kuchenbaecker
- Department of Mental Health Neuroscience, Division of Psychiatry, University College London, London, W1T 7BN, UK
- UCL Genetics Institute, Division of Biosciences, University College London, London, WC1E 6BT, UK
| | - Andrew McQuillin
- Department of Mental Health Neuroscience, Division of Psychiatry, University College London, London, W1T 7BN, UK
| | - John Concato
- School of Medicine, Yale University, New Haven, CT, 06511, USA
- Office of Medical Policy, Center for Drug Evaluation and Research, Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Daniel J Buysse
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - J Michael Gaziano
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, 02130, USA
- Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Daniel J Gottlieb
- VA Boston Healthcare System, 1400 VFW Parkway (111PI), West Roxbury, MA, 02132, USA
- Division of Sleep and Circadian Disorders, Brigham & Women's Hospital, Boston, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Renato Polimanti
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| | - Murray B Stein
- Psychiatry Service, VA San Diego Healthcare System, San Diego, CA, USA
- Departments of Psychiatry and Herbert Wertheim School of Public Health, University of California, San Diego, La Jolla, CA, USA
| | - Elvira Bramon
- Department of Mental Health Neuroscience, Division of Psychiatry, University College London, London, W1T 7BN, UK
- Institute of Cognitive Neuroscience, University College London, London, UK
| | - Joel Gelernter
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA.
| |
Collapse
|
6
|
Lo YJ, Mishra VK, Lo HY, Dubey NK, Lo WC. Clinical Spectrum and Trajectory of Innovative Therapeutic Interventions for Insomnia: A Perspective. Aging Dis 2023; 14:1038-1069. [PMID: 37163444 PMCID: PMC10389812 DOI: 10.14336/ad.2022.1203] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 12/03/2022] [Indexed: 05/12/2023] Open
Abstract
Increasing incidences of insomnia in adults, as well as the aging population, have been reported for their negative impact on the quality of life. Insomnia episodes may be associated with neurocognitive, musculoskeletal, cardiovascular, gastrointestinal, renal, hepatic, and metabolic disorders. Epidemiological evidence also revealed the association of insomnia with oncologic and asthmatic complications, which has been indicated as bidirectional. Two therapeutic approaches including cognitive behavioral therapy (CBT) and drugs-based therapies are being practiced for a long time. However, the adverse events associated with drugs limit their wide and long-term application. Further, Traditional Chinese medicine, acupressure, and pulsed magnetic field therapy may also provide therapeutic relief. Notably, the recently introduced cryotherapy has been demonstrated as a potential candidate for insomnia which could reduce pain, by suppressing oxidative stress and inflammation. It seems that the synergistic therapeutic approach of cryotherapy and the above-mentioned approaches might offer promising prospects to further improve efficacy and safety. Considering these facts, this perspective presents a comprehensive summary of recent advances in pathological aetiologies of insomnia including COVID-19, and its therapeutic management with a greater emphasis on cryotherapy.
Collapse
Affiliation(s)
| | | | | | - Navneet Kumar Dubey
- Victory Biotechnology Co., Ltd., Taipei 114757, Taiwan.
- ShiNeo Technology Co., Ltd., New Taipei City 24262, Taiwan.
| | - Wen-Cheng Lo
- Department of Surgery, Division of Neurosurgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei 11031, Taiwan.
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
7
|
Madrid-Valero JJ, Gregory AM. Behaviour genetics and sleep: A narrative review of the last decade of quantitative and molecular genetic research in humans. Sleep Med Rev 2023; 69:101769. [PMID: 36933344 DOI: 10.1016/j.smrv.2023.101769] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/05/2023]
Abstract
During the last decade quantitative and molecular genetic research on sleep has increased considerably. New behavioural genetics techniques have marked a new era for sleep research. This paper provides a summary of the most important findings from the last ten years, on the genetic and environmental influences on sleep and sleep disorders and their associations with health-related variables (including anxiety and depression) in humans. In this review we present a brief summary of the main methods in behaviour genetic research (such as twin and genome-wide association studies). We then discuss key research findings on: genetic and environmental influences on normal sleep and sleep disorders, as well as on the association between sleep and health variables (highlighting a substantial role for genes in individual differences in sleep and their associations with other variables). We end by discussing future lines of enquiry and drawing conclusions, including those focused on problems and misconceptions associated with research of this type. In this last decade our knowledge about genetic and environmental influences on sleep and its disorders has expanded. Both, twin and genome-wide association studies show that sleep and sleep disorders are substantially influenced by genetic factors and for the very first time multiple specific genetic variants have been associated with sleep traits and disorders.
Collapse
Affiliation(s)
- Juan J Madrid-Valero
- Department of Health Psychology, Faculty of Health Sciences, University of Alicante, Spain.
| | - Alice M Gregory
- Department of Psychology, Goldsmiths, University of London, London, United Kingdom
| |
Collapse
|
8
|
Palermo J, Chesi A, Zimmerman A, Sonti S, Pahl MC, Lasconi C, Brown EB, Pippin JA, Wells AD, Doldur-Balli F, Mazzotti DR, Pack AI, Gehrman PR, Grant SF, Keene AC. Variant-to-gene mapping followed by cross-species genetic screening identifies GPI-anchor biosynthesis as a regulator of sleep. SCIENCE ADVANCES 2023; 9:eabq0844. [PMID: 36608130 PMCID: PMC9821868 DOI: 10.1126/sciadv.abq0844] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 12/05/2022] [Indexed: 05/13/2023]
Abstract
Genome-wide association studies (GWAS) in humans have identified loci robustly associated with several heritable diseases or traits, yet little is known about the functional roles of the underlying causal variants in regulating sleep duration or quality. We applied an ATAC-seq/promoter focused Capture C strategy in human iPSC-derived neural progenitors to carry out a "variant-to-gene" mapping campaign that identified 88 candidate sleep effector genes connected to relevant GWAS signals. To functionally validate the role of the implicated effector genes in sleep regulation, we performed a neuron-specific RNA interference screen in the fruit fly, Drosophila melanogaster, followed by validation in zebrafish. This approach identified a number of genes that regulate sleep including a critical role for glycosylphosphatidylinositol (GPI)-anchor biosynthesis. These results provide the first physical variant-to-gene mapping of human sleep genes followed by a model organism-based prioritization, revealing a conserved role for GPI-anchor biosynthesis in sleep regulation.
Collapse
Affiliation(s)
- Justin Palermo
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| | - Alessandra Chesi
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amber Zimmerman
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA 19104, USA
| | - Shilpa Sonti
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Matthew C. Pahl
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Chiara Lasconi
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Elizabeth B. Brown
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| | - James A. Pippin
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Andrew D. Wells
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA 19104, USA
| | - Fusun Doldur-Balli
- Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA 19104, USA
| | - Diego R. Mazzotti
- Division of Medical Informatics, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66103, USA
- Division of Pulmonary Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Allan I. Pack
- Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA 19104, USA
| | - Phillip R. Gehrman
- Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA 19104, USA
| | - Struan F.A. Grant
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Divisions of Human Genetics and Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alex C. Keene
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
9
|
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
|
10
|
Yang Q, Magnus MC, Kilpi F, Santorelli G, Soares AG, West J, Magnus P, Wright J, Håberg SE, Sanderson E, Lawlor DA, Tilling K, Borges MC. Investigating causal relations between sleep duration and risks of adverse pregnancy and perinatal outcomes: linear and nonlinear Mendelian randomization analyses. BMC Med 2022; 20:295. [PMID: 36089592 PMCID: PMC9465870 DOI: 10.1186/s12916-022-02494-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 07/25/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Observational studies have reported maternal short/long sleep duration to be associated with adverse pregnancy and perinatal outcomes. However, it remains unclear whether there are nonlinear causal effects. Our aim was to use Mendelian randomization (MR) and multivariable regression to examine nonlinear effects of sleep duration on stillbirth (MR only), miscarriage (MR only), gestational diabetes, hypertensive disorders of pregnancy, perinatal depression, preterm birth and low/high offspring birthweight. METHODS We used data from European women in UK Biobank (N=176,897), FinnGen (N=~123,579), Avon Longitudinal Study of Parents and Children (N=6826), Born in Bradford (N=2940) and Norwegian Mother, Father and Child Cohort Study (MoBa, N=14,584). We used 78 previously identified genetic variants as instruments for sleep duration and investigated its effects using two-sample, and one-sample nonlinear (UK Biobank only), MR. We compared MR findings with multivariable regression in MoBa (N=76,669), where maternal sleep duration was measured at 30 weeks. RESULTS In UK Biobank, MR provided evidence of nonlinear effects of sleep duration on stillbirth, perinatal depression and low offspring birthweight. Shorter and longer duration increased stillbirth and low offspring birthweight; shorter duration increased perinatal depression. For example, longer sleep duration was related to lower risk of low offspring birthweight (odds ratio 0.79 per 1 h/day (95% confidence interval: 0.67, 0.93)) in the shortest duration group and higher risk (odds ratio 1.40 (95% confidence interval: 1.06, 1.84)) in the longest duration group, suggesting shorter and longer duration increased the risk. These were supported by the lack of evidence of a linear effect of sleep duration on any outcome using two-sample MR. In multivariable regression, risks of all outcomes were higher in the women reporting <5 and ≥10 h/day sleep compared with the reference category of 8-9 h/day, despite some wide confidence intervals. Nonlinear models fitted the data better than linear models for most outcomes (likelihood ratio P-value=0.02 to 3.2×10-52), except for gestational diabetes. CONCLUSIONS Our results show shorter and longer sleep duration potentially causing higher risks of stillbirth, perinatal depression and low offspring birthweight. Larger studies with more cases are needed to detect potential nonlinear effects on hypertensive disorders of pregnancy, preterm birth and high offspring birthweight.
Collapse
Affiliation(s)
- Qian Yang
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK.
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
| | - Maria C Magnus
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Fanny Kilpi
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Gillian Santorelli
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Ana Gonçalves Soares
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Jane West
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Per Magnus
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - John Wright
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Siri Eldevik Håberg
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Eleanor Sanderson
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- National Institute for Health Research Bristol Biomedical Centre, University Hospitals Bristol NHS Foundation Trust and University of Bristol, Bristol, UK
| | - Kate Tilling
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- National Institute for Health Research Bristol Biomedical Centre, University Hospitals Bristol NHS Foundation Trust and University of Bristol, Bristol, UK
| | - Maria Carolina Borges
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
11
|
Abstract
Genetics is one of the various approaches adopted to understand and control mammalian sleep. Reverse genetics, which is usually applied to analyze sleep in gene-deficient mice, has been the mainstream field of genetic studies on sleep for the past three decades and has revealed that various molecules, including orexin, are involved in sleep regulation. Recently, forward genetic studies in humans and mice have identified gene mutations responsible for heritable sleep abnormalities, such as SIK3, NALCN, DEC2, the neuropeptide S receptor, and β1 adrenergic receptor. Furthermore, the protein kinase A-SIK3 pathway was shown to represent the intracellular neural signaling for sleep need. Large-scale genome-wide analyses of human sleep have been conducted, and many gene loci associated with individual differences in sleep have been found. The development of genome-editing technology and gene transfer by an adeno-associated virus has updated and expanded the genetic studies on mammals. These efforts are expected to elucidate the mechanisms of sleep–wake regulation and develop new therapeutic interventions for sleep disorders.
Collapse
Affiliation(s)
- Hiromasa Funato
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- Department of Anatomy, Faculty of Medicine, Toho University, Ota-ku, Tokyo 951-8585, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas 75390, Texas, USA
- Life Science Center, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan
| |
Collapse
|
12
|
Carnovale C, Perrotta C, Baldelli S, Cattaneo D, Montrasio C, Barbieri SS, Pompilio G, Vantaggiato C, Clementi E, Pozzi M. Antihypertensive drugs and brain function: mechanisms underlying therapeutically beneficial and harmful neuropsychiatric effects. Cardiovasc Res 2022; 119:647-667. [PMID: 35895876 PMCID: PMC10153433 DOI: 10.1093/cvr/cvac110] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 06/15/2022] [Accepted: 06/21/2022] [Indexed: 11/14/2022] Open
Abstract
A bidirectional relationship exists between hypertension and psychiatric disorders, including unipolar and bipolar depression, anxiety, post-traumatic stress disorder (PTSD), psychosis, schizophrenia, mania, and dementia/cognitive decline. Repurposing of antihypertensive drugs to treat mental disorders is thus being explored. A systematic knowledge of the mechanisms of action and clinical consequences of the use of antihypertensive agents on neuropsychiatric functions has not been achieved yet. In this article, we review the putative role of antihypertensive agents in psychiatric disorders, discuss the targets and mechanisms of action, and examine how and to what extent specific drug classes/molecules may trigger, worsen, or mitigate psychiatric symptoms. In addition, we review pharmacokinetics (brain penetration of drugs) and pharmacogenetics data that add important information to assess risks and benefits of antihypertensive drugs in neuropsychiatric settings. The scientific literature shows robust evidence of a positive effect of α1 blockers on PTSD symptoms, nightmares and sleep quality, α2 agonists on core symptoms, executive function and quality of life in Attention-Deficit/Hyperactivity Disorder, PTSD, Tourette's syndrome, and β blockers on anxiety, aggression, working memory, and social communication. Renin-angiotensin system modulators exert protective effects on cognition, depression, and anxiety, and the loop diuretic bumetanide reduced the core symptoms of autism in a subset of patients. There is no evidence of clear benefits of calcium channel blockers in mood disorders in the scientific literature. These findings are mainly from preclinical studies; clinical data are still insufficient or of anecdotal nature, and seldom systematic. The information herewith provided can support a better therapeutic approach to hypertension, tailored to patients with, or with high susceptibility to, psychiatric illness. It may prompt clinical studies exploring the potential benefit of antihypertensive drugs in selected patients with neuropsychiatric comorbidities that include outcomes of neuropsychiatric interest and specifically assess undesirable effects or interactions.
Collapse
Affiliation(s)
- Carla Carnovale
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences (DIBIC), ASST Fatebenefratelli-Sacco University Hospital, Università degli Studi di Milano, 20157 Milano, Italy
| | - Cristiana Perrotta
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences (DIBIC), ASST Fatebenefratelli-Sacco University Hospital, Università degli Studi di Milano, 20157 Milano, Italy
| | - Sara Baldelli
- Unit of Clinical Pharmacology, ASST Fatebenefratelli-Sacco University Hospital, 20157 Milano, Italy
| | - Dario Cattaneo
- Unit of Clinical Pharmacology, ASST Fatebenefratelli-Sacco University Hospital, 20157 Milano, Italy
| | - Cristina Montrasio
- Unit of Clinical Pharmacology, ASST Fatebenefratelli-Sacco University Hospital, 20157 Milano, Italy
| | - Silvia S Barbieri
- Unit of Brain-Heart axis: cellular and molecular mechanisms - Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine - Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy.,Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, Milan, Italy
| | | | - Emilio Clementi
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences (DIBIC), ASST Fatebenefratelli-Sacco University Hospital, Università degli Studi di Milano, 20157 Milano, Italy.,Scientific Institute IRCCS Eugenio Medea, Bosisio Parini (LC), Italy
| | - Marco Pozzi
- Scientific Institute IRCCS Eugenio Medea, Bosisio Parini (LC), Italy
| |
Collapse
|
13
|
Huang J, Cheng Y, Li C, Shang H. Genetic heterogeneity on sleep disorders in Parkinson's disease: a systematic review and meta-analysis. Transl Neurodegener 2022; 11:21. [PMID: 35395825 PMCID: PMC8991652 DOI: 10.1186/s40035-022-00294-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 03/09/2022] [Indexed: 02/08/2023] Open
Abstract
A growing amount of evidence has indicated contributions of variants in causative genes of Parkinson’s disease (PD) to the development of sleep disturbance in PD and prodromal PD stages. In this article, we aimed to investigate the role of genetics in sleep disorders in PD patients and asymptomatic carriers at prodromal stage of PD. A systematic review and meta-analysis of observational studies was conducted based on the MEDLINE, EMBASE and PsychINFO databases. A pooled effect size was calculated by odds ratio (OR) and standard mean difference (SMD). Forty studies were selected for quantitative analysis, including 17 studies on glucocerebrosidase (GBA), 25 studies on Leucine-rich repeat kinase 2 (LRRK2) and 7 on parkin (PRKN) genes, and 3 studies on alpha-synuclein gene (SNCA) were used for qualitative analysis. Patients with PD carrying GBA variants had a significantly higher risk for rapid-eye-movement behavior disorders (RBD) (OR, 1.82) and higher RBD Screening Questionnaire scores (SMD, 0.33). Asymptomatic carriers of GBA variants had higher severity of RBD during follow-up. Patients with PD carrying the LRRK2 G2019S variant had lower risk and severity of RBD compared with those without LRRK2 G2019S. Variants of GBA, LRRK2 and PRKN did not increase or decrease the risk and severity of excessive daytime sleepiness and restless legs syndrome in PD. Our findings suggest that the genetic heterogeneity plays a role in the development of sleep disorders, mainly RBD, in PD and the prodromal stage of PD.
Collapse
Affiliation(s)
- Jingxuan Huang
- Laboratory of Neurodegenerative Disorders, Department of Neurology, Rare Diseases Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yangfan Cheng
- Laboratory of Neurodegenerative Disorders, Department of Neurology, Rare Diseases Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chunyu Li
- Laboratory of Neurodegenerative Disorders, Department of Neurology, Rare Diseases Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Huifang Shang
- Laboratory of Neurodegenerative Disorders, Department of Neurology, Rare Diseases Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
14
|
Abstract
BACKGROUND Shift work is widespread due to 24-h work in many occupations. Understanding differences in individual shift work tolerance (SWT) can help develop coping strategies for shift workers. AIMS This in-depth qualitative review elucidates the architecture of SWT, providing an overview of the research advances in the last decade (2011-2021). METHODS We searched Google Scholar, PubMed and Medline for different word combinations concerning SWT. Genome-wide association studies (GWAS) for the potential genetic basis of SWT were additionally searched in GWAS Central and GWAS Catalogue. RESULTS Eleven new studies were published since 2011, with the proportion of longitudinal studies on SWT having more than doubled in the past decade. They consolidate prior findings (e.g. hardiness most consistently associated with SWT) and discovered additional aspects of SWT like resistance to change and job stress. The 15 large-scale GWAS identified, most of which using UK Biobank (UKB) and 23andMe data, involved mapped genes showing overlap especially within analysis of the same phenotype (e.g. PER2/3 for morningness, PAX8 for sleep duration and LINGO1 for neuroticism). Individual GWAS for additional traits such as resilience have also been published though assessments of gene overlap are not yet possible. CONCLUSIONS Progress regarding longitudinal studies on SWT has been made though a more consistent definition of SWT remains crucial for future research. Non-genetic studies on SWT suggest several important traits and factors; many of which have now also been explored using GWAS. Such evidence could serve as basis for individualized risk prediction and disease prevention approaches for night-shift workers.
Collapse
Affiliation(s)
- J Degenfellner
- Department of Epidemiology, Centre for Public Health, Medical University of Vienna, 1090 Vienna, Austria
| | - E Schernhammer
- Department of Epidemiology, Centre for Public Health, Medical University of Vienna, 1090 Vienna, Austria.,Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
15
|
Faltraco F, Palm D, Coogan A, Simon F, Tucha O, Thome J. Molecular Link between Circadian Rhythmicity and Mood Disorders. Curr Med Chem 2021; 29:5692-5709. [PMID: 34620057 DOI: 10.2174/0929867328666211007113725] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 08/17/2021] [Accepted: 08/26/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The internal clock is driven by circadian genes [e.g., Clock, Bmal1, Per1-3, Cry1-2], hormones [e.g., melatonin, cortisol], as well as zeitgeber ['synchronisers']. Chronic disturbances in the circadian rhythm in patients diagnosed with mood disorders have been recognised for more than 50 years. OBJECTIVES The aim of this review is to summarise the current knowledge and literature regarding circadian rhythms in the context of mood disorders, focussing on the role of circadian genes, hormones, and neurotransmitters. METHOD The review presents the current knowledge and literature regarding circadian rhythms in mood disorders using the Pubmed database. Articles with a focus on circadian rhythms and mood disorders [n=123], particularly from 1973 to 2020, were included. RESULTS The article suggests a molecular link between disruptions in the circadian rhythm and mood disorders. Circadian disturbances, caused by the dysregulation of circadian genes, hormones, and neurotransmitters, often result in a clinical picture resembling depression. CONCLUSION Circadian rhythms are intrinsically linked to affective disorders, such as unipolar depression and bipolar disorder.
Collapse
Affiliation(s)
- Frank Faltraco
- Department of Psychiatry and Psychotherapy, University Medical Center Rostock, Rostock. Germany
| | - Denise Palm
- Department of Psychiatry and Psychotherapy, University Medical Center Rostock, Rostock. Germany
| | - Andrew Coogan
- Department of Psychology, Maynooth University, National University of Ireland, Maynooth. Ireland
| | - Frederick Simon
- Department of Psychiatry and Psychotherapy, University Medical Center Rostock, Rostock. Germany
| | - Oliver Tucha
- Department of Psychiatry and Psychotherapy, University Medical Center Rostock, Rostock. Germany
| | - Johannes Thome
- Department of Psychiatry and Psychotherapy, University Medical Center Rostock, Rostock. Germany
| |
Collapse
|
16
|
Mainieri G, Montini A, Nicotera A, Di Rosa G, Provini F, Loddo G. The Genetics of Sleep Disorders in Children: A Narrative Review. Brain Sci 2021; 11:1259. [PMID: 34679324 PMCID: PMC8534132 DOI: 10.3390/brainsci11101259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 11/16/2022] Open
Abstract
Sleep is a universal, highly preserved process, essential for human and animal life, whose complete functions are yet to be unravelled. Familial recurrence is acknowledged for some sleep disorders, but definite data are lacking for many of them. Genetic studies on sleep disorders have progressed from twin and family studies to candidate gene approaches to culminate in genome-wide association studies (GWAS). Several works disclosed that sleep-wake characteristics, in addition to electroencephalographic (EEG) sleep patterns, have a certain degree of heritability. Notwithstanding, it is rare for sleep disorders to be attributed to single gene defects because of the complexity of the brain network/pathways involved. Besides, the advancing insights in epigenetic gene-environment interactions add further complexity to understanding the genetic control of sleep and its disorders. This narrative review explores the current genetic knowledge in sleep disorders in children, following the International Classification of Sleep Disorders-Third Edition (ICSD-3) categorisation.
Collapse
Affiliation(s)
- Greta Mainieri
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40138 Bologna, Italy; (G.M.); (A.M.)
| | - Angelica Montini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40138 Bologna, Italy; (G.M.); (A.M.)
| | - Antonio Nicotera
- Unit of Child Neurology and Psychiatry, Department of Human Pathology of the Adult and Developmental Age, “Gaetano Barresi” University of Messina, 98124 Messina, Italy; (A.N.); (G.D.R.)
| | - Gabriella Di Rosa
- Unit of Child Neurology and Psychiatry, Department of Human Pathology of the Adult and Developmental Age, “Gaetano Barresi” University of Messina, 98124 Messina, Italy; (A.N.); (G.D.R.)
| | - Federica Provini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40138 Bologna, Italy; (G.M.); (A.M.)
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
| | | |
Collapse
|
17
|
Plante DT, Papale LA, Madrid A, Cook JD, Prairie ML, Alisch RS. PAX8/PAX8-AS1 DNA methylation levels are associated with objective sleep duration in persons with unexplained hypersomnolence using a deep phenotyping approach. Sleep 2021; 44:6305146. [PMID: 34145460 DOI: 10.1093/sleep/zsab108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 12/09/2020] [Indexed: 11/13/2022] Open
Abstract
STUDY OBJECTIVES Patients with unexplained hypersomnolence have significant impairment related to daytime sleepiness and excessive sleep duration, the biological bases of which are poorly understood. This investigation sought to examine relationships between objectively measured hypersomnolence phenotypes and epigenetic modification of candidate hypersomnolence genes to advance this line of inquiry. METHODS Twenty-eight unmedicated clinical patients with unexplained hypersomnolence were evaluated using overnight ad libitum polysomnography, multiple sleep latency testing, infrared pupillometry, and the psychomotor vigilance task. DNA methylation levels on CpG sites annotated to 11 a priori hypersomnolence candidate genes were assessed for statistical association with hypersomnolence measures using independent regression models with adjusted local index of significance (aLIS) P-value threshold of 0.05. RESULTS Nine CpG sites exhibited significant associations between DNA methylation levels and total sleep time measured using ad libitum polysomnography (aLIS p-value < .05). All nine differentially methylated CpG sites were annotated to the paired box 8 (PAX8) gene and its related antisense gene (PAX8-AS1). Among these nine differentially methylated positions was a cluster of five CpG sites located in the body of the PAX8 gene and promoter of PAX8-AS1. CONCLUSIONS This study demonstrates that PAX8/PAX8-AS1 DNA methylation levels are associated with total sleep time in persons with unexplained hypersomnolence. Given prior investigations that have implicated single nucleotide polymorphisms in PAX8/PAX8-AS1 with habitual sleep duration, further research that clarifies the role of DNA methylation levels on these genes in the phenotypic expression of total sleep time is warranted.
Collapse
Affiliation(s)
| | | | - Andy Madrid
- Department of Neurological Surgery, Madison, WI.,Neuroscience Training Program, University of Wisconsin - Madison, Madison, WI
| | | | | | | |
Collapse
|
18
|
Integrative genomics analysis identifies five promising genes implicated in insomnia risk based on multiple omics datasets. Biosci Rep 2021; 40:226183. [PMID: 32830860 PMCID: PMC7468094 DOI: 10.1042/bsr20201084] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 08/15/2020] [Accepted: 08/21/2020] [Indexed: 12/27/2022] Open
Abstract
In recent decades, many genome-wide association studies on insomnia have reported numerous genes harboring multiple risk variants. Nevertheless, the molecular functions of these risk variants conveying risk to insomnia are still ill-studied. In the present study, we integrated GWAS summary statistics (N=386,533) with two independent brain expression quantitative trait loci (eQTL) datasets (N=329) to determine whether expression-associated SNPs convey risk to insomnia. Furthermore, we applied numerous bioinformatics analyses to highlight promising genes associated with insomnia risk. By using Sherlock integrative analysis, we detected 449 significant insomnia-associated genes in the discovery stage. These identified genes were significantly overrepresented in six biological pathways including Huntington’s disease (P=5.58 × 10−5), Alzheimer’s disease (P=5.58 × 10−5), Parkinson’s disease (P=6.34 × 10−5), spliceosome (P=1.17 × 10−4), oxidative phosphorylation (P=1.09 × 10−4), and wnt signaling pathways (P=2.07 × 10−4). Further, five of these identified genes were replicated in an independent brain eQTL dataset. Through a PPI network analysis, we found that there existed highly functional interactions among these five identified genes. Three genes of LDHA (P=0.044), DALRD3 (P=5.0 × 10−5), and HEBP2 (P=0.032) showed significantly lower expression level in brain tissues of insomnic patients than that in controls. In addition, the expression levels of these five genes showed prominently dynamic changes across different time points between behavioral states of sleep and sleep deprivation in mice brain cortex. Together, the evidence of the present study strongly suggested that these five identified genes may represent candidate genes and contributed risk to the etiology of insomnia.
Collapse
|
19
|
Factors Affecting the Quality of Sleep in Children. CHILDREN-BASEL 2021; 8:children8020122. [PMID: 33572155 PMCID: PMC7915148 DOI: 10.3390/children8020122] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/05/2021] [Accepted: 02/06/2021] [Indexed: 12/12/2022]
Abstract
Sleep quality is one of the domains of sleep. Having adequate quality sleep is defined as one’s “feeling fresh” after waking-up. Inadequate sleep quality results in sleep insufficiency producing a variety of symptoms and signs. The central nervous system is affected the most in children, although other system too may be involved. Several factors affect sleep quality in children including genetics, sleep habits, medical problems, parents/caregiver factors, screen time and the child’s environment. These factors are inter-related and dynamic. The outcome of sleep insufficiency is many involving neurocognitive and neurobehavior, mood and emotional issues and specific conditions, like pulmonary hypertension, cor pulmonale and obesity. Management should start with proper history taking to identify the multifaceted nature of the condition. Treatment is planned cognizant of the age of the patient and the associated etiological factors, and should involve both the children and their parents.
Collapse
|
20
|
Awang H, Kankia JD, Daud Z, Roddin R, Rahman AA. Youth attitudes towards an informal roadside metalwork fabrication apprenticeship practice. PROCEEDINGS OF 8TH INTERNATIONAL CONFERENCE ON ADVANCED MATERIALS ENGINEERING & TECHNOLOGY (ICAMET 2020) 2021. [DOI: 10.1063/5.0051686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
21
|
Sleep duration: A review of genome-wide association studies (GWAS) in adults from 2007 to 2020. Sleep Med Rev 2020; 56:101413. [PMID: 33338765 DOI: 10.1016/j.smrv.2020.101413] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 12/20/2022]
Abstract
A modest body of research exists in the area of human sleep genetics, which suggests that specific sleep phenotypes are, like many other complex traits, somewhat heritable. Until 2007 research into sleep genetics relied solely on twin studies, but in the last 13 years with the advent of huge biobanks and very large-scale genome-wide association studies, the field of molecular sleep genetics has seen important advances. To date, the majority have focused on self-reported sleep duration, but in recent years genome-wide association studies of objectively-measured sleep have emerged. These genetic studies have discovered multiple common genetic variants and as such, have provided insight into potential biological pathways, causal relationships between sleep duration and important disease outcomes using Mendelian randomisation. They have also shown that the heritability of these traits may not be as high as previously estimated. This article is the first to provide a detailed review of these recent advances in the genetic epidemiology of sleep duration. Studies were identified using both the GWAS Catalog and PubMed for completeness. Focus is on the genome-wide association studies published to date, including whether and how they have elucidated important biology and advanced knowledge in the area of sleep and health.
Collapse
|
22
|
Li X, Zhao H. Automated feature extraction from population wearable device data identified novel loci associated with sleep and circadian rhythms. PLoS Genet 2020; 16:e1009089. [PMID: 33075057 PMCID: PMC7595622 DOI: 10.1371/journal.pgen.1009089] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 10/29/2020] [Accepted: 08/31/2020] [Indexed: 12/12/2022] Open
Abstract
Wearable devices have been increasingly used in research to provide continuous physical activity monitoring, but how to effectively extract features remains challenging for researchers. To analyze the generated actigraphy data in large-scale population studies, we developed computationally efficient methods to derive sleep and activity features through a Hidden Markov Model-based sleep/wake identification algorithm, and circadian rhythm features through a Penalized Multi-band Learning approach adapted from machine learning. Unsupervised feature extraction is useful when labeled data are unavailable, especially in large-scale population studies. We applied these two methods to the UK Biobank wearable device data and used the derived sleep and circadian features as phenotypes in genome-wide association studies. We identified 53 genetic loci with p<5×10-8 including genes known to be associated with sleep disorders and circadian rhythms as well as novel loci associated with Body Mass Index, mental diseases and neurological disorders, which suggest shared genetic factors of sleep and circadian rhythms with physical and mental health. Further cross-tissue enrichment analysis highlights the important role of the central nervous system and the shared genetic architecture with metabolism-related traits and the metabolic system. Our study demonstrates the effectiveness of our unsupervised methods for wearable device data when additional training data cannot be easily acquired, and our study further expands the application of wearable devices in population studies and genetic studies to provide novel biological insights.
Collapse
Affiliation(s)
- Xinyue Li
- School of Data Science, City University of Hong Kong, Hong Kong, China
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, United States of America
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT, United States of America
- Department of Genetics, Yale University School of Medicine, New Haven, CT, United States of America
| |
Collapse
|
23
|
Li W, Baumbach J, Larsen MJ, Mohammadnejad A, Lund J, Christensen K, Christiansen L, Tan Q. Differential long noncoding RNA profiling of BMI in twins. Epigenomics 2020; 12:1531-1541. [PMID: 32901529 DOI: 10.2217/epi-2020-0033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Many efforts have been deployed to identify genetic variants associated with BMI. Alternatively, we explore epigenetic contribution to BMI variation by focusing on long noncoding RNAs (lncRNAs) which represents a key layer of epigenetic control. Materials & methods: We analyzed lncRNA expression in whole blood of 229 monozygotic twin pairs in association with BMI using generalized estimating equations. Results & conclusion: Six lncRNA probes were identified as significant (false discovery rate <0.05), with BMI showing causal effects on the expression of the significant lncRNAs. Functional annotation of differential profiles identified Gene ontology biological processes including kidney development, regulations of lipid biosynthetic process, circadian rhythm, notch signaling, etc. Whole blood lncRNAs are significantly expressed in response to BMI variation.
Collapse
Affiliation(s)
- Weilong Li
- Unit of Epidemiology, Biostatistics & Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Jan Baumbach
- Computational BioMedicine, Department of Mathematics & Computer Science, University of Southern Denmark, Odense, Denmark.,Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Martin J Larsen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark.,Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Afsaneh Mohammadnejad
- Unit of Epidemiology, Biostatistics & Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Jesper Lund
- Unit of Epidemiology, Biostatistics & Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Kaare Christensen
- Unit of Epidemiology, Biostatistics & Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark.,Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Lene Christiansen
- Unit of Epidemiology, Biostatistics & Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark.,Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Qihua Tan
- Unit of Epidemiology, Biostatistics & Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark.,Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
24
|
Novel candidate genes for ECT response prediction-a pilot study analyzing the DNA methylome of depressed patients receiving electroconvulsive therapy. Clin Epigenetics 2020; 12:114. [PMID: 32727556 PMCID: PMC7388224 DOI: 10.1186/s13148-020-00891-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 06/23/2020] [Indexed: 01/07/2023] Open
Abstract
Background Major depressive disorder (MDD) represents a serious global health concern. The urge for efficient MDD treatment strategies is presently hindered by the incomplete knowledge of its underlying pathomechanism. Despite recent progress (highlighting both genetics and the environment, and thus DNA methylation, to be relevant for its development), 30–50% of MDD patients still fail to reach remission with standard treatment approaches. Electroconvulsive therapy (ECT) is one of the most powerful options for the treatment of pharmacoresistant depression; nevertheless, ECT remission rates barely reach 50% in large-scale naturalistic population-based studies. To optimize MDD treatment strategies and enable personalized medicine in the long- term, prospective indicators of ECT response are thus in great need. Because recent target-driven analyses revealed DNA methylation baseline differences between ECT responder groups, we analyzed the DNA methylome of depressed ECT patients using next-generation sequencing. In this pilot study, we did not only aim to find novel targets for ECT response prediction but also to get a deeper insight into its possible mechanism of action. Results Longitudinal DNA methylation analysis of peripheral blood mononuclear cells isolated from a cohort of treatment-resistant MDD patients (n = 12; time points: before and after 1st and last ECT, respectively) using a TruSeq-Methyl Capture EPIC Kit for library preparation, led to the following results: (1) The global DNA methylation differed neither between the four measured time points nor between ECT responders (n = 8) and non-responders (n = 4). (2) Analyzing the DNA methylation variance for every probe (=1476812 single CpG sites) revealed eight novel candidate genes to be implicated in ECT response (protein-coding genes: RNF175, RNF213, TBC1D14, TMC5, WSCD1; genes encoding for putative long non-coding RNA transcripts: AC018685.2, AC098617.1, CLCN3P1). (3) In addition, DNA methylation of two CpG sites (located within AQP10 and TRERF1) was found to change during the treatment course. Conclusions We suggest ten novel candidate genes to be implicated in either ECT response or its possible mechanism. Because of the small sample size of our pilot study, our findings must be regarded as preliminary.
Collapse
|
25
|
Didikoglu A, Maharani A, Canal MM, Pendleton N, Payton A. Interactions between season of birth, chronological age and genetic polymorphisms in determining later-life chronotype. Mech Ageing Dev 2020; 188:111253. [PMID: 32371234 DOI: 10.1016/j.mad.2020.111253] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/30/2020] [Accepted: 04/17/2020] [Indexed: 01/19/2023]
Abstract
Human chronotype, the temporal pattern of daily behaviors, is influenced by postnatal seasonal programming and ageing. The aim of this study was to investigate genetic variants that are associated with season of birth programming and longitudinal chronotype change. Longitudinal sleep timing and genotype data from 1449 participants were collected for up to 27 years. Gene-environment interaction analysis was performed for 445 candidate single nucleotide polymorphisms that have previously been associated with chronotype. Associations were tested using linear mixed model. We identified 67 suggestively significant genomic loci that have genotype-ageing interaction and 25 genomic loci that may have genotype-season of birth interaction in determining chronotype. We attempted to replicate the results using longitudinal data of the UK Biobank from approximately 30,000 participants. Biological functions of these genes suggest that epigenetic regulation of gene expression and neural development may have roles in these processes. The strongest associated gene for sleep trajectories was ALKBH5, which has functions of DNA repair and epigenetic regulation. A potential candidate gene for postnatal seasonal programming was SIRT1, which has previously been implicated in postnatal programming, ageing and longevity. Genetic diversity may explain the heterogeneity in ageing-related change of sleep timing and postnatal environmental programming of later-life chronotype.
Collapse
Affiliation(s)
- Altug Didikoglu
- Division of Neuroscience & Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK(2).
| | - Asri Maharani
- Division of Nursing, Midwifery & Social Work, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK
| | - Maria Mercè Canal
- Division of Neuroscience & Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK(2)
| | - Neil Pendleton
- Division of Neuroscience & Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK(2)
| | - Antony Payton
- Division of Informatics, Imaging & Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK
| |
Collapse
|
26
|
Genome-wide association analysis of insomnia using data from Partners Biobank. Sci Rep 2020; 10:6928. [PMID: 32332799 PMCID: PMC7181749 DOI: 10.1038/s41598-020-63792-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 03/25/2020] [Indexed: 12/21/2022] Open
Abstract
Insomnia is one of the most prevalent and burdensome mental disorders worldwide, affecting between 10–20% of adults and up to 48% of the geriatric population. It is further associated with substance usage and dependence, as well other psychiatric disorders. In this study, we combined electronic health record (EHR) derived phenotypes and genotype information to conduct a genome wide analysis of insomnia in a 18,055 patient cohort. Diagnostic codes were used to identify 3,135 patients with insomnia. Our genome-wide association study (GWAS) identified one novel genomic risk locus on chromosome 8 (lead SNP rs17052966, p = 4.53 × 10−9, odds ratio = 1.28, se = 0.04). The heritability analysis indicated that common SNPs accounts for 7% (se = 0.02, p = 0.015) of phenotypic variation. We further conducted a large-scale meta-analysis of our results and summary statistics of two recent insomnia GWAS and 13 significant loci were identified. The genetic correlation analysis yielded a strong positive genetic correlation between insomnia and alcohol use (rG = 0.56, se = 0.14, p < 0.001), nicotine use (rG = 0.50, se = 0.12, p < 0.001) and opioid use (rG = 0.43, se = 0.18, p = 0.02) disorders, suggesting a significant common genetic risk factors between insomnia and substance use.
Collapse
|
27
|
Genetics of Circadian and Sleep Measures in Adults: Implications for Sleep Medicine. CURRENT SLEEP MEDICINE REPORTS 2020. [DOI: 10.1007/s40675-020-00165-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
28
|
Bragantini D, Sivertsen B, Gehrman P, Lydersen S, Güzey IC. Genetic polymorphisms associated with sleep-related phenotypes; relationships with individual nocturnal symptoms of insomnia in the HUNT study. BMC MEDICAL GENETICS 2019; 20:179. [PMID: 31718593 PMCID: PMC6852911 DOI: 10.1186/s12881-019-0916-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 10/31/2019] [Indexed: 12/21/2022]
Abstract
Background In recent years, several GWAS (genome wide association studies) of sleep-related traits have identified a number of SNPs (single nucleotides polymorphism) but their relationships with symptoms of insomnia are not known. The aim of this study was to investigate whether SNPs, previously reported in association with sleep-related phenotypes, are associated with individual symptoms of insomnia. Methods We selected participants from the HUNT study (Norway) who reported at least one symptom of insomnia consisting of sleep onset, maintenance or early morning awakening difficulties, (cases, N = 2563) compared to participants who presented no symptoms at all (controls, N = 3665). Cases were further divided in seven subgroups according to different combinations of these three symptoms. We used multinomial logistic regressions to test the association among different patterns of symptoms and 59 SNPs identified in past GWAS studies. Results Although 16 SNPS were significantly associated (p < 0.05) with at least one symptom subgroup, none of the investigated SNPs remained significant after correction for multiple testing using the false discovery rate (FDR) method. Conclusions SNPs associated with sleep-related traits do not replicate on any pattern of insomnia symptoms after multiple tests correction. However, correction in this case may be overly conservative.
Collapse
Affiliation(s)
- Daniela Bragantini
- Department of Research and Development (AFFU), Norwegian University of Science and Technology (NTNU), PO Box 3250 Sluppen, NO-7006, Trondheim, Norway. .,Department of Mental Health, Norwegian University of Science and Technology (NTNU), PO Box 3250 Sluppen, NO-7006, Trondheim, Norway. .,St. Olav's University Hospital, Division of Mental Health Care, Østmarkveien 15, NO-7040, Trondheim, Norway.
| | - Børge Sivertsen
- Department of Mental Health, Norwegian University of Science and Technology (NTNU), PO Box 3250 Sluppen, NO-7006, Trondheim, Norway.,Department of Health Promotion, Norwegian Institute of Public Health, PO Box 973 Sentrum, 5808, Bergen, Norway.,Department of Research and Innovation, Helse-Fonna HF Haugesund Hospital, PO Box 2170, 5504, Haugesund, Norway
| | - Philip Gehrman
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, 3535 Market St., Suite 670, Philadelphia, PA, 19104, USA
| | - Stian Lydersen
- Regional Centre for Child and Youth Mental Health and Child Welfare (RKBU), Norwegian University of Science and Technology (NTNU), P.O. Box 8905, N-7491, Trondheim, Norway
| | - Ismail Cüneyt Güzey
- Department of Research and Development (AFFU), Norwegian University of Science and Technology (NTNU), PO Box 3250 Sluppen, NO-7006, Trondheim, Norway.,Department of Mental Health, Norwegian University of Science and Technology (NTNU), PO Box 3250 Sluppen, NO-7006, Trondheim, Norway.,St. Olav's University Hospital, Division of Mental Health Care, Østmarkveien 15, NO-7040, Trondheim, Norway
| |
Collapse
|
29
|
Wang H, Lane JM, Jones SE, Dashti HS, Ollila HM, Wood AR, van Hees VT, Brumpton B, Winsvold BS, Kantojärvi K, Palviainen T, Cade BE, Sofer T, Song Y, Patel K, Anderson SG, Bechtold DA, Bowden J, Emsley R, Kyle SD, Little MA, Loudon AS, Scheer FAJL, Purcell SM, Richmond RC, Spiegelhalder K, Tyrrell J, Zhu X, Hublin C, Kaprio JA, Kristiansson K, Sulkava S, Paunio T, Hveem K, Nielsen JB, Willer CJ, Zwart JA, Strand LB, Frayling TM, Ray D, Lawlor DA, Rutter MK, Weedon MN, Redline S, Saxena R. Genome-wide association analysis of self-reported daytime sleepiness identifies 42 loci that suggest biological subtypes. Nat Commun 2019; 10:3503. [PMID: 31409809 PMCID: PMC6692391 DOI: 10.1038/s41467-019-11456-7] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 06/27/2019] [Indexed: 01/12/2023] Open
Abstract
Excessive daytime sleepiness (EDS) affects 10-20% of the population and is associated with substantial functional deficits. Here, we identify 42 loci for self-reported daytime sleepiness in GWAS of 452,071 individuals from the UK Biobank, with enrichment for genes expressed in brain tissues and in neuronal transmission pathways. We confirm the aggregate effect of a genetic risk score of 42 SNPs on daytime sleepiness in independent Scandinavian cohorts and on other sleep disorders (restless legs syndrome, insomnia) and sleep traits (duration, chronotype, accelerometer-derived sleep efficiency and daytime naps or inactivity). However, individual daytime sleepiness signals vary in their associations with objective short vs long sleep, and with markers of sleep continuity. The 42 sleepiness variants primarily cluster into two predominant composite biological subtypes - sleep propensity and sleep fragmentation. Shared genetic links are also seen with obesity, coronary heart disease, psychiatric diseases, cognitive traits and reproductive ageing.
Collapse
Affiliation(s)
- Heming Wang
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Jacqueline M Lane
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Samuel E Jones
- Genetics of Complex Traits, University of Exeter Medical School, Exeter, United Kingdom
| | - Hassan S Dashti
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Hanna M Ollila
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, CA, USA
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Andrew R Wood
- Genetics of Complex Traits, University of Exeter Medical School, Exeter, United Kingdom
| | | | - Ben Brumpton
- K.G. Jebsen Centre for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Department of Thoracic and Occupational Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Bendik S Winsvold
- K.G. Jebsen Centre for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
- Division of Clinical Neuroscience, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Katri Kantojärvi
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki, Finland
- Department of Psychiatry and SleepWell Research Program, Faculty of Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Teemu Palviainen
- Institute for Molecular Medicine FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Brian E Cade
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Yanwei Song
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Northeastern University College of Science, Boston, MA, USA
| | - Krunal Patel
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Northeastern University College of Science, Boston, MA, USA
| | - Simon G Anderson
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- The George Alleyne Chronic Disease Research Centre, Caribbean Institute for Health Research, University of the West Indies, Cave Hill, Barbados
| | - David A Bechtold
- Division of Endocrinology, Diabetes & Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jack Bowden
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Richard Emsley
- Division of Endocrinology, Diabetes & Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Simon D Kyle
- Sleep and Circadian Neuroscience Institute, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Max A Little
- Department of Mathematics, Aston University, Birmingham, UK
- Media Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Andrew S Loudon
- Division of Endocrinology, Diabetes & Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Frank A J L Scheer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Shaun M Purcell
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Rebecca C Richmond
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Kai Spiegelhalder
- Clinic for Psychiatry and Psychotherapy, Medical Centre, University of Freiburg, Freiburg, Germany
| | - Jessica Tyrrell
- Genetics of Complex Traits, University of Exeter Medical School, Exeter, United Kingdom
| | - Xiaofeng Zhu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Christer Hublin
- Institute for Molecular Medicine FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Jaakko A Kaprio
- Institute for Molecular Medicine FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Kati Kristiansson
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Sonja Sulkava
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki, Finland
- Department of Psychiatry and SleepWell Research Program, Faculty of Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Tiina Paunio
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki, Finland
- Department of Psychiatry and SleepWell Research Program, Faculty of Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Kristian Hveem
- K.G. Jebsen Centre for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
| | - Jonas B Nielsen
- Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Cristen J Willer
- Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - John-Anker Zwart
- Division of Clinical Neuroscience, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Linn B Strand
- K.G. Jebsen Centre for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| | - Timothy M Frayling
- Genetics of Complex Traits, University of Exeter Medical School, Exeter, United Kingdom
| | - David Ray
- NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, OX39DU, UK
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Martin K Rutter
- Division of Endocrinology, Diabetes & Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester Diabetes Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Michael N Weedon
- Genetics of Complex Traits, University of Exeter Medical School, Exeter, United Kingdom
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
- Department of Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Richa Saxena
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA.
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
30
|
Petersen AS, Barloese MCJ, Snoer A, Soerensen AMS, Jensen RH. Verapamil and Cluster Headache: Still a Mystery. A Narrative Review of Efficacy, Mechanisms and Perspectives. Headache 2019; 59:1198-1211. [DOI: 10.1111/head.13603] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2019] [Indexed: 01/04/2023]
Affiliation(s)
- Anja S. Petersen
- Department of Neurology, Danish Headache Center Rigshospitalet‐Glostrup Glostrup Denmark
| | - Mads C. J. Barloese
- Department of Neurology, Danish Headache Center Rigshospitalet‐Glostrup Glostrup Denmark
- Department of Clinical Physiology and Nuclear Medicine, Center for Functional and Diagnostic Imaging Hvidovre Hospital Hvidovre Denmark
| | - Agneta Snoer
- Department of Neurology, Danish Headache Center Rigshospitalet‐Glostrup Glostrup Denmark
| | - Anne Mette S. Soerensen
- Department of Clinical Pharmacology Bispebjerg and Frederiksberg Hospital Copenhagen Denmark
| | - Rigmor H. Jensen
- Department of Neurology, Danish Headache Center Rigshospitalet‐Glostrup Glostrup Denmark
| |
Collapse
|
31
|
Andrade A, Brennecke A, Mallat S, Brown J, Gomez-Rivadeneira J, Czepiel N, Londrigan L. Genetic Associations between Voltage-Gated Calcium Channels and Psychiatric Disorders. Int J Mol Sci 2019; 20:E3537. [PMID: 31331039 PMCID: PMC6679227 DOI: 10.3390/ijms20143537] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/12/2019] [Accepted: 07/13/2019] [Indexed: 12/23/2022] Open
Abstract
Psychiatric disorders are mental, behavioral or emotional disorders. These conditions are prevalent, one in four adults suffer from any type of psychiatric disorders world-wide. It has always been observed that psychiatric disorders have a genetic component, however, new methods to sequence full genomes of large cohorts have identified with high precision genetic risk loci for these conditions. Psychiatric disorders include, but are not limited to, bipolar disorder, schizophrenia, autism spectrum disorder, anxiety disorders, major depressive disorder, and attention-deficit and hyperactivity disorder. Several risk loci for psychiatric disorders fall within genes that encode for voltage-gated calcium channels (CaVs). Calcium entering through CaVs is crucial for multiple neuronal processes. In this review, we will summarize recent findings that link CaVs and their auxiliary subunits to psychiatric disorders. First, we will provide a general overview of CaVs structure, classification, function, expression and pharmacology. Next, we will summarize tools to study risk loci associated with psychiatric disorders. We will examine functional studies of risk variations in CaV genes when available. Finally, we will review pharmacological evidence of the use of CaV modulators to treat psychiatric disorders. Our review will be of interest for those studying pathophysiological aspects of CaVs.
Collapse
Affiliation(s)
- Arturo Andrade
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA.
| | - Ashton Brennecke
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| | - Shayna Mallat
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| | - Julian Brown
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| | | | - Natalie Czepiel
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| | - Laura Londrigan
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| |
Collapse
|
32
|
Jones SE, van Hees VT, Mazzotti DR, Marques-Vidal P, Sabia S, van der Spek A, Dashti HS, Engmann J, Kocevska D, Tyrrell J, Beaumont RN, Hillsdon M, Ruth KS, Tuke MA, Yaghootkar H, Sharp SA, Ji Y, Harrison JW, Freathy RM, Murray A, Luik AI, Amin N, Lane JM, Saxena R, Rutter MK, Tiemeier H, Kutalik Z, Kumari M, Frayling TM, Weedon MN, Gehrman PR, Wood AR. Genetic studies of accelerometer-based sleep measures yield new insights into human sleep behaviour. Nat Commun 2019; 10:1585. [PMID: 30952852 PMCID: PMC6451011 DOI: 10.1038/s41467-019-09576-1] [Citation(s) in RCA: 173] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 03/14/2019] [Indexed: 01/16/2023] Open
Abstract
Sleep is an essential human function but its regulation is poorly understood. Using accelerometer data from 85,670 UK Biobank participants, we perform a genome-wide association study of 8 derived sleep traits representing sleep quality, quantity and timing, and validate our findings in 5,819 individuals. We identify 47 genetic associations at P < 5 × 10-8, of which 20 reach a stricter threshold of P < 8 × 10-10. These include 26 novel associations with measures of sleep quality and 10 with nocturnal sleep duration. The majority of identified variants associate with a single sleep trait, except for variants previously associated with restless legs syndrome. For sleep duration we identify a missense variant (p.Tyr727Cys) in PDE11A as the likely causal variant. As a group, sleep quality loci are enriched for serotonin processing genes. Although accelerometer-derived measures of sleep are imperfect and may be affected by restless legs syndrome, these findings provide new biological insights into sleep compared to previous efforts based on self-report sleep measures.
Collapse
Affiliation(s)
- Samuel E Jones
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | | | - Diego R Mazzotti
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Perelman School of Medicine of the University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Pedro Marques-Vidal
- Department of Medicine, Internal Medicine, Lausanne University Hospital, Lausanne, 1011, Switzerland
| | - Séverine Sabia
- Research Department of Epidemiology and Public Health, University College London, London, WC1E 6BT, UK
- INSERM, U1153, Epidemiology of Ageing and Neurodegenerative diseases, Université de Paris, Paris, 75010, France
| | - Ashley van der Spek
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3000 CA, The Netherlands
| | - Hassan S Dashti
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Jorgen Engmann
- UCL Institute of Cardiovascular Science, Research department of Population Science and Experimental Medicine, Centre for Translational Genomics, 222 Euston Road, London, NW1 2DA, UK
| | - Desana Kocevska
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3000 CA, The Netherlands
- Department of Child and Adolescent Psychiatry, Erasmus Medical Center, Rotterdam, 3000 CA, The Netherlands
| | - Jessica Tyrrell
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | - Robin N Beaumont
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | - Melvyn Hillsdon
- Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, EX1 2LU, UK
| | - Katherine S Ruth
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | - Marcus A Tuke
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | - Hanieh Yaghootkar
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | - Seth A Sharp
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | - Yingjie Ji
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | - Jamie W Harrison
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | - Rachel M Freathy
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | - Anna Murray
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | - Annemarie I Luik
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3000 CA, The Netherlands
| | - Najaf Amin
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3000 CA, The Netherlands
| | - Jacqueline M Lane
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Richa Saxena
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02111, USA
- Departments of Medicine, Brigham and Women's Hospital and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Martin K Rutter
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, M13 9PL, UK
- Manchester Diabetes Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Oxford Road, 193 Hathersage Road, Manchester, M13 0JE, UK
| | - Henning Tiemeier
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3000 CA, The Netherlands
- Department of Social and Behavioral Science, Harvard TH Chan School of Public Health, Boston, MA, 02115, USA
| | - Zoltán Kutalik
- Institute of Social and Preventive Medicine (IUMSP), Lausanne University Hospital, Lausanne, 1010, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland
| | - Meena Kumari
- ISER, University of Essex, Colchester, Essex, CO4 3SQ, UK
| | - Timothy M Frayling
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK
| | - Michael N Weedon
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK.
| | - Philip R Gehrman
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Perelman School of Medicine of the University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Andrew R Wood
- Genetics of Complex Traits, College of Medicine and Health, University of Exeter, Exeter, EX2 5DW, UK.
| |
Collapse
|
33
|
Dashti HS, Jones SE, Wood AR, Lane JM, van Hees VT, Wang H, Rhodes JA, Song Y, Patel K, Anderson SG, Beaumont RN, Bechtold DA, Bowden J, Cade BE, Garaulet M, Kyle SD, Little MA, Loudon AS, Luik AI, Scheer FAJL, Spiegelhalder K, Tyrrell J, Gottlieb DJ, Tiemeier H, Ray DW, Purcell SM, Frayling TM, Redline S, Lawlor DA, Rutter MK, Weedon MN, Saxena R. Genome-wide association study identifies genetic loci for self-reported habitual sleep duration supported by accelerometer-derived estimates. Nat Commun 2019; 10:1100. [PMID: 30846698 PMCID: PMC6405943 DOI: 10.1038/s41467-019-08917-4] [Citation(s) in RCA: 333] [Impact Index Per Article: 66.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/31/2019] [Indexed: 12/22/2022] Open
Abstract
Sleep is an essential state of decreased activity and alertness but molecular factors regulating sleep duration remain unknown. Through genome-wide association analysis in 446,118 adults of European ancestry from the UK Biobank, we identify 78 loci for self-reported habitual sleep duration (p < 5 × 10−8; 43 loci at p < 6 × 10−9). Replication is observed for PAX8, VRK2, and FBXL12/UBL5/PIN1 loci in the CHARGE study (n = 47,180; p < 6.3 × 10−4), and 55 signals show sign-concordant effects. The 78 loci further associate with accelerometer-derived sleep duration, daytime inactivity, sleep efficiency and number of sleep bouts in secondary analysis (n = 85,499). Loci are enriched for pathways including striatum and subpallium development, mechanosensory response, dopamine binding, synaptic neurotransmission and plasticity, among others. Genetic correlation indicates shared links with anthropometric, cognitive, metabolic, and psychiatric traits and two-sample Mendelian randomization highlights a bidirectional causal link with schizophrenia. This work provides insights into the genetic basis for inter-individual variation in sleep duration implicating multiple biological pathways. Sleep is essential for homeostasis and insufficient or excessive sleep are associated with adverse outcomes. Here, the authors perform GWAS for self-reported habitual sleep duration in adults, supported by accelerometer-derived measures, and identify genetic correlation with psychiatric and metabolic traits
Collapse
Affiliation(s)
- Hassan S Dashti
- Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, MA, USA.,Broad Institute, Cambridge, 02142, MA, USA
| | - Samuel E Jones
- Genetics of Complex Traits, University of Exeter Medical School, Exeter, EX2 5DW, UK
| | - Andrew R Wood
- Genetics of Complex Traits, University of Exeter Medical School, Exeter, EX2 5DW, UK
| | - Jacqueline M Lane
- Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, MA, USA.,Broad Institute, Cambridge, 02142, MA, USA.,Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, MA, USA
| | | | - Heming Wang
- Broad Institute, Cambridge, 02142, MA, USA.,Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, 02115, MA, USA.,Division of Sleep Medicine, Harvard Medical School, Boston, 02115, MA, USA
| | - Jessica A Rhodes
- Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, MA, USA.,Broad Institute, Cambridge, 02142, MA, USA
| | - Yanwei Song
- Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, MA, USA.,Northeastern University College of Science, 176 Mugar Life Sciences, 360 Huntington Avenue, Boston, MA, 02015, USA
| | - Krunal Patel
- Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, MA, USA.,Northeastern University College of Science, 176 Mugar Life Sciences, 360 Huntington Avenue, Boston, MA, 02015, USA
| | - Simon G Anderson
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PL, UK
| | - Robin N Beaumont
- Genetics of Complex Traits, University of Exeter Medical School, Exeter, EX2 5DW, UK
| | - David A Bechtold
- Division of Endocrinology, Diabetes & Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
| | - Jack Bowden
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, BS8 2BN, UK.,Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - Brian E Cade
- Broad Institute, Cambridge, 02142, MA, USA.,Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, 02115, MA, USA.,Division of Sleep Medicine, Harvard Medical School, Boston, 02115, MA, USA
| | - Marta Garaulet
- Department of Physiology, University of Murcia, Murcia, 30100, Spain.,IMIB-Arrixaca, Murcia, 30120, Spain
| | - Simon D Kyle
- Sleep and Circadian Neuroscience Institute, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 7LF, UK
| | - Max A Little
- Department of Mathematics, Aston University, Birmingham, B4 7ET, UK.,Media Lab, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA
| | - Andrew S Loudon
- Division of Endocrinology, Diabetes & Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
| | - Annemarie I Luik
- Sleep and Circadian Neuroscience Institute, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 7LF, UK
| | - Frank A J L Scheer
- Broad Institute, Cambridge, 02142, MA, USA.,Division of Sleep Medicine, Harvard Medical School, Boston, 02115, MA, USA.,Medical Chronobiology Program, Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, 02115, MA, USA
| | - Kai Spiegelhalder
- Clinic for Psychiatry and Psychotherapy, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, 79106, Germany
| | - Jessica Tyrrell
- Genetics of Complex Traits, University of Exeter Medical School, Exeter, EX2 5DW, UK
| | - Daniel J Gottlieb
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, 02115, MA, USA.,Division of Sleep Medicine, Harvard Medical School, Boston, 02115, MA, USA.,VA Boston Healthcare System, Boston, 02132, MA, USA
| | - Henning Tiemeier
- Deprtment of Social and Behavioral Science, Harvard TH Chan School of Public Health, Boston, 02115, MA, USA.,Department of Epidemiology, Erasmus Medical Center, Rotterdam, 3015, The Netherlands
| | - David W Ray
- Division of Endocrinology, Diabetes & Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
| | - Shaun M Purcell
- Department of Psychiatry, Brigham & Women's Hospital, Harvard Medical School, 02115, Boston, MA, USA
| | - Timothy M Frayling
- Genetics of Complex Traits, University of Exeter Medical School, Exeter, EX2 5DW, UK
| | - Susan Redline
- Departments of Medicine, Brigham and Women's Hospital and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, 02115, MA, USA
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, BS8 2BN, UK.,Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - Martin K Rutter
- Division of Endocrinology, Diabetes & Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK.,Manchester Diabetes Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9PL, UK
| | - Michael N Weedon
- Genetics of Complex Traits, University of Exeter Medical School, Exeter, EX2 5DW, UK
| | - Richa Saxena
- Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, MA, USA. .,Broad Institute, Cambridge, 02142, MA, USA. .,Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, MA, USA.
| |
Collapse
|
34
|
Nishiyama T, Nakatochi M, Goto A, Iwasaki M, Hachiya T, Sutoh Y, Shimizu A, Wang C, Tanaka H, Watanabe M, Hosono A, Tamai Y, Yamada T, Yamaji T, Sawada N, Fukumoto K, Otsuka K, Tanno K, Tomita H, Kojima K, Nagasaki M, Hozawa A, Hishida A, Sasakabe T, Nishida Y, Hara M, Ito H, Oze I, Nakamura Y, Mikami H, Ibusuki R, Takezaki T, Koyama T, Kuriyama N, Endoh K, Kuriki K, Turin TC, Naoyuki T, Katsuura-Kamano S, Uemura H, Okada R, Kawai S, Naito M, Momozawa Y, Kubo M, Sasaki M, Yamamoto M, Tsugane S, Wakai K, Suzuki S. Genome-wide association meta-analysis and Mendelian randomization analysis confirm the influence of ALDH2 on sleep durationin the Japanese population. Sleep 2019; 42:5362027. [DOI: 10.1093/sleep/zsz046] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 02/20/2019] [Indexed: 11/14/2022] Open
Affiliation(s)
- Takeshi Nishiyama
- Department of Public Health, Nagoya City University Graduate School of Medicine, Nagoya, Japan
- Department of Public Health, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Masahiro Nakatochi
- Data Science Division, Data Coordinating Center, Department of Advanced Medicine, Nagoya University Hospital, Japan
| | - Atsushi Goto
- Division of Epidemiology, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Motoki Iwasaki
- Division of Epidemiology, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Tsuyoshi Hachiya
- Division of Biomedical Information Analysis, Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate, Japan
| | - Yoichi Sutoh
- Division of Biomedical Information Analysis, Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate, Japan
| | - Atsushi Shimizu
- Division of Biomedical Information Analysis, Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate, Japan
| | - Chaochen Wang
- Department of Public Health, Nagoya City University Graduate School of Medicine, Nagoya, Japan
- Department of Public Health, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Hideo Tanaka
- Osaka Prefectural Kishiwada Public Health Center, Osaka, Japan
| | - Miki Watanabe
- Department of Public Health, Nagoya City University Graduate School of Medicine, Nagoya, Japan
| | - Akihiro Hosono
- Department of Public Health, Nagoya City University Graduate School of Medicine, Nagoya, Japan
| | - Yuya Tamai
- Department of Public Health, Nagoya City University Graduate School of Medicine, Nagoya, Japan
| | | | - Taiki Yamaji
- Division of Epidemiology, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Norie Sawada
- Division of Epidemiology, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Kentaro Fukumoto
- Department of Neuropsychiatry, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Kotaro Otsuka
- Department of Neuropsychiatry, School of Medicine, Iwate Medical University, Iwate, Japan
- Division of Clinical Research and Epidemiology, Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate, Japan
| | - Kozo Tanno
- Division of Clinical Research and Epidemiology, Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate, Japan
- Department of Hygiene and Preventive Medicine, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Hiroaki Tomita
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Kaname Kojima
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Masao Nagasaki
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Atsushi Hozawa
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Asahi Hishida
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tae Sasakabe
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuichiro Nishida
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Megumi Hara
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Hidemi Ito
- Division of Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Isao Oze
- Division of Molecular and Clinical Epidemiology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Yohko Nakamura
- Cancer Prevention Center, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Haruo Mikami
- Cancer Prevention Center, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Rie Ibusuki
- Department of International Islands and Community Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Toshiro Takezaki
- Department of International Islands and Community Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Teruhide Koyama
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Nagato Kuriyama
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Kaori Endoh
- Laboratory of Public Health, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka, Japan
| | - Kiyonori Kuriki
- Laboratory of Public Health, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka, Japan
| | - Tanvir C Turin
- Department of Health Science, Shiga University of Medical Science, Shiga, Japan
| | - Takashima Naoyuki
- Department of Health Science, Shiga University of Medical Science, Shiga, Japan
| | - Sakurako Katsuura-Kamano
- Department of Preventive Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Hirokazu Uemura
- Department of Preventive Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Rieko Okada
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sayo Kawai
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mariko Naito
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Oral Epidemiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
| | - Michiaki Kubo
- Laboratory for Genotyping Development, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
| | - Makoto Sasaki
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate, Japan
- Division of Ultra-High Field MRI and Department of Radiology, Iwate Medical University, Iwate, Japan
| | - Masayuki Yamamoto
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Shoichiro Tsugane
- Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Kenji Wakai
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sadao Suzuki
- Department of Public Health, Nagoya City University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
35
|
Landolt HP, Holst SC, Valomon A. Clinical and Experimental Human Sleep-Wake Pharmacogenetics. Handb Exp Pharmacol 2019; 253:207-241. [PMID: 30443785 DOI: 10.1007/164_2018_175] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Sleep and wakefulness are highly complex processes that are elegantly orchestrated by fine-tuned neurochemical changes among neuronal and non-neuronal ensembles, nuclei, and networks of the brain. Important neurotransmitters and neuromodulators regulating the circadian and homeostatic facets of sleep-wake physiology include melatonin, γ-aminobutyric acid, hypocretin, histamine, norepinephrine, serotonin, dopamine, and adenosine. Dysregulation of these neurochemical systems may cause sleep-wake disorders, which are commonly classified into insomnia disorder, parasomnias, circadian rhythm sleep-wake disorders, central disorders of hypersomnolence, sleep-related movement disorders, and sleep-related breathing disorders. Sleep-wake disorders can have far-reaching consequences on physical, mental, and social well-being and health and, thus, need be treated with effective and rational therapies. Apart from behavioral (e.g., cognitive behavioral therapy for insomnia), physiological (e.g., chronotherapy with bright light), and mechanical (e.g., continuous positive airway pressure treatment of obstructive sleep apnea) interventions, pharmacological treatments often are the first-line clinical option to improve disturbed sleep and wake states. Nevertheless, not all patients respond to pharmacotherapy in uniform and beneficial fashion, partly due to genetic differences. The improved understanding of the neurochemical mechanisms regulating sleep and wakefulness and the mode of action of sleep-wake therapeutics has provided a conceptual framework, to search for functional genetic variants modifying individual drug response phenotypes. This article will summarize the currently known genetic polymorphisms that modulate drug sensitivity and exposure, to partly determine individual responses to sleep-wake pharmacotherapy. In addition, a pharmacogenetic strategy will be outlined how based upon classical and opto-/chemogenetic strategies in animals, as well as human genetic associations, circuit mechanisms regulating sleep-wake functions in humans can be identified. As such, experimental human sleep-wake pharmacogenetics forms a bridge spanning basic research and clinical medicine and constitutes an essential step for the search and development of novel sleep-wake targets and therapeutics.
Collapse
Affiliation(s)
- Hans-Peter Landolt
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland.
- Zürich Center for Interdisciplinary Sleep Research (ZiS), University of Zürich, Zürich, Switzerland.
| | - Sebastian C Holst
- Neurobiology Research Unit and Neuropharm, Department of Neurology, Rigshospitalet, Copenhagen, Denmark
| | - Amandine Valomon
- Wisconsin Institute for Sleep and Consciousness, University of Wisconsin Madison, Madison, WI, USA
| |
Collapse
|
36
|
Mei F, Wu Y, Wu J. The Relationship Between Tryptophan Hydroxylase-2 Gene with Primary Insomnia and Depressive Symptoms in the Han Chinese Population. Balkan Med J 2018; 35:412-416. [PMID: 29952309 PMCID: PMC6251380 DOI: 10.4274/balkanmedj.2017.1406] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background: Insomnia often coexists with depression, and there is compelling evidence for a genetic component in the etiologies of both disorders. Aims: To investigate the relationship between exonic variant (rs4290270) in the tryptophan hydroxylase-2 gene and primary insomnia and symptoms of depression in Han Chinese. Study Design: Case-control study. Methods: This study included 152 patients with primary insomnia and 164 age- and gender-matched normal controls. All patients were investigated by polysomnography for 2 consecutive nights. The depressive symptoms were measured by using a 20-item Zung Self-rating Depression Scale. Sleep quality was assessed with the Pittsburgh Sleep Quality index. The genotypes of the TPH-2 gene polymorphism rs4290270 were determined by the polymerase chain reaction-restriction fragment length polymorphism method. Results: The genotype distributions of the tryptophan hydroxylase-2 gene polymorphism rs4290270 were in Hardy-Weinberg equilibrium in both patients and controls (p>0.05). The allele and genotype distributions of this variant were comparable between patients and controls in all subjects and between genders (all p>0.05). The impact of rs4290270 on self-rating depression scale score changes was statistically significant (p=0.002), with carriers of the A/A genotype having the highest self-rating depression scale score (mean ± standard deviation: 52.73±12.88), followed by the A/T genotype (50.94±11.29, p=0.35) and the T/T genotype (43.48±7.78, p<0.01), and this impact was more obvious in women (p<0.001). Conclusion: The tryptophan hydroxylase-2 gene polymorphism rs4290270 may not be a susceptibility locus for primary insomnia in Han Chinese, but it may be a marker of depressive symptoms.
Collapse
Affiliation(s)
- Feng Mei
- Department of Medical Psychology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yanfeng Wu
- Department of Neurology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jin Wu
- Department of Neurology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
37
|
Abstract
Insomnia is a worldwide problem with substantial deleterious health effects. Twin studies have shown a heritable basis for various sleep-related traits, including insomnia, but robust genetic risk variants have just recently begun to be identified. We conducted genome-wide association studies (GWAS) of soldiers in the Army Study To Assess Risk and Resilience in Servicemembers (STARRS). GWAS were carried out separately for each ancestral group (EUR, AFR, LAT) using logistic regression for each of the STARRS component studies (including 3,237 cases and 14,414 controls), and then meta-analysis was conducted across studies and ancestral groups. Heritability (SNP-based) for lifetime insomnia disorder was significant (h2g = 0.115, p = 1.78 × 10-4 in EUR). A meta-analysis including three ancestral groups and three study cohorts revealed a genome-wide significant locus on Chr 7 (q11.22) (top SNP rs186736700, OR = 0.607, p = 4.88 × 10-9) and a genome-wide significant gene-based association (p = 7.61 × 10-7) in EUR for RFX3 on Chr 9. Polygenic risk for sleeplessness/insomnia severity in UK Biobank was significantly positively associated with likelihood of insomnia disorder in STARRS. Genetic contributions to insomnia disorder in STARRS were significantly positively correlated with major depressive disorder (rg = 0.44, se = 0.22, p = 0.047) and type 2 diabetes (rg = 0.43, se = 0.20, p = 0.037), and negatively with morningness chronotype (rg = -0.34, se = 0.17, p = 0.039) and subjective well being (rg = -0.59, se = 0.23, p = 0.009) in external datasets. Insomnia associated loci may contribute to the genetic risk underlying a range of health conditions including psychiatric disorders and metabolic disease.
Collapse
|
38
|
Zahari Z, Ibrahim MA, Musa N, Tan SC, Mohamad N, Ismail R. Sleep quality and OPRM1 polymorphisms: a cross-sectional study among opioid-naive individuals. BRAZ J PHARM SCI 2018. [DOI: 10.1590/s2175-97902018000117217] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Zalina Zahari
- Universiti Sultan Zainal Abidin, Malaysia; Universiti Sains Malaysia, Malaysia
| | | | | | | | - Nasir Mohamad
- Universiti Sains Malaysia, Malaysia; Universiti Sultan Zainal Abidin, Malaysia
| | - Rusli Ismail
- Universiti Sains Malaysia, Malaysia; Universiti Sultan Zainal Abidin, Malaysia
| |
Collapse
|
39
|
Gan-Or Z, Alcalay RN, Rouleau GA, Postuma RB. Sleep disorders and Parkinson disease; lessons from genetics. Sleep Med Rev 2018; 41:101-112. [PMID: 29449121 DOI: 10.1016/j.smrv.2018.01.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/04/2017] [Accepted: 01/15/2018] [Indexed: 02/08/2023]
Abstract
Parkinson disease is a common, age-related neurodegenerative disorder, projected to afflict millions of individuals in the near future. Understanding its etiology and identifying clinical, genetic or biological markers for Parkinson disease onset and progression is therefore of major importance. Various sleep-related disorders are the most common group of non-motor symptoms in advanced Parkinson disease, but they can also occur during its prodromal phase. However, with the exception of REM sleep behavior disorder, it is unclear whether they are part of the early pathological process of Parkinson disease, or if they develop as Parkinson disease advances because of treatments and neurodegeneration progression. The advancements in genetic studies in the past two decades have generated a wealth of information, and recent genetic studies offer new insight on the association of sleep-related disorders with Parkinson disease. More specifically, comparing genetic data between Parkinson disease and sleep-related disorders can clarify their association, which may assist in determining whether they can serve as clinical markers for Parkinson disease risk or progression. In this review, we discuss the current knowledge on the genetics of sleep-related disorders in Parkinson disease context, and the potential implications on research, diagnosis, counseling and treatment.
Collapse
Affiliation(s)
- Ziv Gan-Or
- Montreal Neurological Institute, McGill University, Montréal, QC, Canada; Department of Human Genetics, McGill University, Montréal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada.
| | - Roy N Alcalay
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Guy A Rouleau
- Montreal Neurological Institute, McGill University, Montréal, QC, Canada; Department of Human Genetics, McGill University, Montréal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Ronald B Postuma
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| |
Collapse
|
40
|
Melroy-Greif WE, Gizer IR, Wilhelmsen KC, Ehlers CL. Genetic Influences on Evening Preference Overlap with Those for Bipolar Disorder in a Sample of Mexican Americans and American Indians. Twin Res Hum Genet 2017; 20:499-510. [PMID: 29192581 PMCID: PMC6013261 DOI: 10.1017/thg.2017.62] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Diurnal preference (e.g., being an owl or lark) has been associated with several psychiatric disorders including bipolar disorder (BP), major depressive disorder, and substance use disorders. Previous large-scale genome-wide association studies (GWAS) aimed at identifying genetic influences on diurnal preference have exclusively included subjects of European ancestry. This study examined the genetic architecture of diurnal preference in two minority samples: a young adult sample of Mexican Americans (MAs), and a family-based sample of American Indians (AIs). Typed or imputed variants from exome chip data from the MA sample and low pass whole-genome sequencing from the AI cohort were analyzed using a mixed linear model approach for association with being an owl, as defined by a usual bedtime after 23:00 hrs. Genetic risk score (GRS) profiling detected shared genetic risk between evening preference and related disorders. Four variants in KIAA1549 like (KIAA1549L), a gene previously associated with attempted suicide in bipolar patients, were suggestively associated with being an owl at p < 1.82E-05; post hoc analyses showed the top variant trending in both the MA and AI cohorts at p = 2.50E-05 and p = .030, respectively. Variants associated with BP at p < .03 from the Psychiatric Genomics Consortium nominally predicted being an owl in the MA/AI cohort at p = .012. This study provides some additional evidence that genetic risk factors for BP also confer risk for being an owl in MAs/AIs and that evening preference may be a useful endophenotype for future studies of BP.
Collapse
Affiliation(s)
| | - Ian R. Gizer
- Department of Psychological Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Kirk C. Wilhelmsen
- Renaissance Computing Institute (RENCI), Chapel Hill, NC 27517, USA
- Departments of Genetics and Neurology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Cindy L. Ehlers
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
41
|
Ahmad Z, Moustafa YW, Stiller JW, Pavlovich MA, Raheja UK, Gragnoli C, Snitker S, Nazem S, Dagdag A, Fang B, Fuchs D, Lowry CA, Postolache TT. Sleep onset insomnia, daytime sleepiness and sleep duration in relationship to Toxoplasma gondii IgG seropositivity and serointensity. Pteridines 2017; 28:195-204. [PMID: 29657364 PMCID: PMC5894504 DOI: 10.1515/pterid-2017-0010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Toxoplasma gondii (T. gondii) infects central nervous tissue and is kept in relative dormancy by a healthy immune system. Sleep disturbances have been found to precipitate mental illness, suicidal behavior and car accidents, which have been previously linked to T. gondii as well. We speculated that if sleep disruption, particularly insomnia, would mediate, at least partly, the link between T. gondii infection and related behavioral dysregulation, then we would be able to identify significant associations between sleep disruption and T. gondii. The mechanisms for such an association may involve dopamine (DA) production by T. gondii, or collateral effects of immune activation necessary to keep T. gondii in check. Sleep questionnaires from 2031 Old Order Amish were analyzed in relationship to T. gondii-IgG antibodies measured by enzyme-linked immunosorbent assay (ELISA). Toxoplasma gondii seropositivity and serointensity were not associated with any of the sleep latency variables or Epworth Sleepiness Scale (ESS). A secondary analysis identified, after adjustment for age group, a statistical trend toward shorter sleep duration in seropositive men (p = 0.07). In conclusion, it is unlikely that sleep disruption mediates links between T. gondii and mental illness or behavioral dysregulation. Trending gender differences in associations between T. gondii and shorter sleep need further investigation.
Collapse
Affiliation(s)
- Zaki Ahmad
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yara W. Moustafa
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA; and Saint Elizabeths’ Hospital, Psychiatry Residency Training Program, Washington, DC, USA
| | - John W. Stiller
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA; Saint Elizabeths’ Hospital, Department of Neurology, Washington, DC, USA; and Maryland State Athletic Commission, Baltimore, MD, USA
| | - Mary A. Pavlovich
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA; and Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Uttam K. Raheja
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA; and Child and Adolescent Psychiatry Residency Program, Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Claudia Gragnoli
- Division of Endocrinology, Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA; Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA; and Molecular Biology Laboratory, Bios Biotech Multi Diagnostic Health Center, Rome, Italy
| | - Soren Snitker
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sarra Nazem
- Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Denver, CO, USA; Department of Psychiatry, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA; and Department of Physical Medicine and Rehabilitation, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Aline Dagdag
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Beverly Fang
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Christopher A. Lowry
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA; Department of Physical Medicine and Rehabilitation and Center for Neuroscience, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA; and Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Denver, CO, USA
| | - Teodor T. Postolache
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA; Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Denver, CO, USA; and Mental Illness Research, Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 5, VA Capitol Health Care Network, Baltimore, MD, USA
| |
Collapse
|
42
|
Hammerschlag AR, Stringer S, de Leeuw CA, Sniekers S, Taskesen E, Watanabe K, Blanken TF, Dekker K, te Lindert BHW, Wassing R, Jonsdottir I, Thorleifsson G, Stefansson H, Gislason T, Berger K, Schormair B, Wellmann J, Winkelmann J, Stefansson K, Oexle K, Van Someren EJW, Posthuma D. Genome-wide association analysis of insomnia complaints identifies risk genes and genetic overlap with psychiatric and metabolic traits. Nat Genet 2017; 49:1584-1592. [PMID: 28604731 PMCID: PMC5600256 DOI: 10.1038/ng.3888] [Citation(s) in RCA: 182] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 05/03/2017] [Indexed: 12/31/2022]
Abstract
Persistent insomnia is among the most frequent complaints in general practice. To identify genetic factors for insomnia complaints, we performed a genome-wide association study (GWAS) and a genome-wide gene-based association study (GWGAS) in 113,006 individuals. We identify three loci and seven genes associated with insomnia complaints, with the associations for one locus and five genes supported by joint analysis with an independent sample (n = 7,565). Our top association (MEIS1, P < 5 × 10-8) has previously been implicated in restless legs syndrome (RLS). Additional analyses favor the hypothesis that MEIS1 exhibits pleiotropy for insomnia and RLS and show that the observed association with insomnia complaints cannot be explained only by the presence of an RLS subgroup within the cases. Sex-specific analyses suggest that there are different genetic architectures between the sexes in addition to shared genetic factors. We show substantial positive genetic correlation of insomnia complaints with internalizing personality traits and metabolic traits and negative correlation with subjective well-being and educational attainment. These findings provide new insight into the genetic architecture of insomnia.
Collapse
Affiliation(s)
- Anke R Hammerschlag
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, The Netherlands
| | - Sven Stringer
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, The Netherlands
| | - Christiaan A de Leeuw
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, The Netherlands
| | - Suzanne Sniekers
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, The Netherlands
| | - Erdogan Taskesen
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, The Netherlands
- Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands
| | - Kyoko Watanabe
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, The Netherlands
| | - Tessa F Blanken
- Department of Sleep and Cognition, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
- Departments of Integrative Neurophysiology and Psychiatry, Amsterdam Neuroscience, VU University and Medical Center, Amsterdam, The Netherlands
| | - Kim Dekker
- Department of Sleep and Cognition, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Bart HW te Lindert
- Department of Sleep and Cognition, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Rick Wassing
- Department of Sleep and Cognition, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Ingileif Jonsdottir
- deCODE genetics / Amgen Inc., Reykjavík, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | - Thorarinn Gislason
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
- Department of Respiratory Medicine and Sleep, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
| | - Klaus Berger
- Institute of Epidemiology and Social Medicine, University of Muenster, Muenster, Germany
| | - Barbara Schormair
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
- Institute of Human Genetics, Technische Universität München, Munich, Germany
| | - Juergen Wellmann
- Institute of Epidemiology and Social Medicine, University of Muenster, Muenster, Germany
| | - Juliane Winkelmann
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
- Institute of Human Genetics, Technische Universität München, Munich, Germany
- Neurologische Klinik und Poliklinik, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Kari Stefansson
- deCODE genetics / Amgen Inc., Reykjavík, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Konrad Oexle
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
| | - Eus JW Van Someren
- Department of Sleep and Cognition, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
- Departments of Integrative Neurophysiology and Psychiatry, Amsterdam Neuroscience, VU University and Medical Center, Amsterdam, The Netherlands
| | - Danielle Posthuma
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, The Netherlands
- Department of Clinical Genetics, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
43
|
Kantojärvi K, Liuhanen J, Saarenpää-Heikkilä O, Satomaa AL, Kylliäinen A, Pölkki P, Jaatela J, Toivola A, Milani L, Himanen SL, Porkka-Heiskanen T, Paavonen J, Paunio T. Variants in calcium voltage-gated channel subunit Alpha1 C-gene (CACNA1C) are associated with sleep latency in infants. PLoS One 2017; 12:e0180652. [PMID: 28792954 PMCID: PMC5549883 DOI: 10.1371/journal.pone.0180652] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 06/19/2017] [Indexed: 12/12/2022] Open
Abstract
Genetic variants in CACNA1C (calcium voltage-gated channel subunit alpha1 C) are associated with bipolar disorder and schizophrenia where sleep disturbances are common. In an experimental model, Cacna1c has been found to modulate the electrophysiological architecture of sleep. There are strong genetic influences for consolidation of sleep in infancy, but only a few studies have thus far researched the genetic factors underlying the process. We hypothesized that genetic variants in CACNA1C affect the regulation of sleep in early development. Seven variants that were earlier associated (genome-wide significantly) with psychiatric disorders at CACNA1C were selected for analyses. The study sample consists of 1086 infants (520 girls and 566 boys) from the Finnish CHILD-SLEEP birth cohort (genotyped by Illumina Infinium PsychArray BeadChip). Sleep length, latency, and nightly awakenings were reported by the parents of the infants with a home-delivered questionnaire at 8 months of age. The genetic influence of CACNA1C variants on sleep in infants was examined by using PLINK software. Three of the examined CACNA1C variants, rs4765913, rs4765914, and rs2239063, were associated with sleep latency (permuted P<0.05). There was no significant association between studied variants and night awakenings or sleep duration. CACNA1C variants for psychiatric disorders were found to be associated with long sleep latency among 8-month-old infants. It remains to be clarified whether the findings refer to defective regulation of sleep, or to distractibility of sleep under external influences.
Collapse
Affiliation(s)
- Katri Kantojärvi
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki, Finland
- Department of Psychiatry, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Johanna Liuhanen
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki, Finland
- Department of Psychiatry, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | | | - Anna-Liisa Satomaa
- Department of Clinical Neurophysiology, Tampere University Hospital, Medical Imaging Centre and Hospital Pharmacy, Pirkanmaa Hospital District, Tampere, Finland
| | - Anneli Kylliäinen
- School of Social Sciences and Humanities/Psychology, University of Tampere, Tampere, Finland
| | - Pirjo Pölkki
- Department of Social Sciences, University of Eastern Finland, Kuopio, Finland
| | - Julia Jaatela
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki, Finland
- Department of Psychiatry, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Auli Toivola
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki, Finland
- Department of Psychiatry, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Lili Milani
- The Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Sari-Leena Himanen
- Department of Clinical Neurophysiology, Tampere University Hospital, Medical Imaging Centre and Hospital Pharmacy, Pirkanmaa Hospital District, Tampere, Finland
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | | | - Juulia Paavonen
- Child and Adolescent Mental Health, National Institute for Health and Welfare, Helsinki, Finland
| | - Tiina Paunio
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki, Finland
- Department of Psychiatry, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| |
Collapse
|
44
|
Rohde PD, Gaertner B, Ward K, Sørensen P, Mackay TFC. Genomic Analysis of Genotype-by-Social Environment Interaction for Drosophila melanogaster Aggressive Behavior. Genetics 2017; 206:1969-1984. [PMID: 28550016 PMCID: PMC5560801 DOI: 10.1534/genetics.117.200642] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 05/22/2017] [Indexed: 02/06/2023] Open
Abstract
Human psychiatric disorders such as schizophrenia, bipolar disorder, and attention-deficit/hyperactivity disorder often include adverse behaviors including increased aggressiveness. Individuals with psychiatric disorders often exhibit social withdrawal, which can further increase the probability of conducting a violent act. Here, we used the inbred, sequenced lines of the Drosophila Genetic Reference Panel (DGRP) to investigate the genetic basis of variation in male aggressive behavior for flies reared in a socialized and socially isolated environment. We identified genetic variation for aggressive behavior, as well as significant genotype-by-social environmental interaction (GSEI); i.e., variation among DGRP genotypes in the degree to which social isolation affected aggression. We performed genome-wide association (GWA) analyses to identify genetic variants associated with aggression within each environment. We used genomic prediction to partition genetic variants into gene ontology (GO) terms and constituent genes, and identified GO terms and genes with high prediction accuracies in both social environments and for GSEI. The top predictive GO terms significantly increased the proportion of variance explained, compared to prediction models based on all segregating variants. We performed genomic prediction across environments, and identified genes in common between the social environments that turned out to be enriched for genome-wide associated variants. A large proportion of the associated genes have previously been associated with aggressive behavior in Drosophila and mice. Further, many of these genes have human orthologs that have been associated with neurological disorders, indicating partially shared genetic mechanisms underlying aggression in animal models and human psychiatric disorders.
Collapse
Affiliation(s)
- Palle Duun Rohde
- Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, Aarhus University, 8830 Tjele, Denmark
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, 8000 Aarhus, Denmark
- ISEQ, Center for Integrative Sequencing, Aarhus University, 8000 Aarhus, Denmark
| | - Bryn Gaertner
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27695
- Program in Genetics, North Carolina State University, Raleigh, North Carolina 27695
- W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina 27695
| | - Kirsty Ward
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27695
- Program in Genetics, North Carolina State University, Raleigh, North Carolina 27695
- W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina 27695
| | - Peter Sørensen
- Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, Aarhus University, 8830 Tjele, Denmark
| | - Trudy F C Mackay
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27695
- Program in Genetics, North Carolina State University, Raleigh, North Carolina 27695
- W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina 27695
| |
Collapse
|
45
|
Harrison PJ, Cipriani A, Harmer CJ, Nobre AC, Saunders K, Goodwin GM, Geddes JR. Innovative approaches to bipolar disorder and its treatment. Ann N Y Acad Sci 2017; 1366:76-89. [PMID: 27111134 PMCID: PMC4850752 DOI: 10.1111/nyas.13048] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 02/25/2016] [Accepted: 03/01/2016] [Indexed: 12/29/2022]
Abstract
All psychiatric disorders have suffered from a dearth of truly novel pharmacological interventions. In bipolar disorder, lithium remains a mainstay of treatment, six decades since its effects were serendipitously discovered. The lack of progress reflects several factors, including ignorance of the disorder's pathophysiology and the complexities of the clinical phenotype. After reviewing the current status, we discuss some ways forward. First, we highlight the need for a richer characterization of the clinical profile, facilitated by novel devices and new forms of data capture and analysis; such data are already promoting a reevaluation of the phenotype, with an emphasis on mood instability rather than on discrete clinical episodes. Second, experimental medicine can provide early indications of target engagement and therapeutic response, reducing the time, cost, and risk involved in evaluating potential mood stabilizers. Third, genomic data can inform target identification and validation, such as the increasing evidence for involvement of calcium channel genes in bipolar disorder. Finally, new methods and models relevant to bipolar disorder, including stem cells and genetically modified mice, are being used to study key pathways and drug effects. A combination of these approaches has real potential to break the impasse and deliver genuinely new treatments.
Collapse
Affiliation(s)
- Paul J Harrison
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom.,Oxford Health NHS Foundation Trust, Oxford, United Kingdom
| | - Andrea Cipriani
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom.,Oxford Health NHS Foundation Trust, Oxford, United Kingdom
| | - Catherine J Harmer
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom.,Oxford Health NHS Foundation Trust, Oxford, United Kingdom
| | - Anna C Nobre
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom.,Oxford Health NHS Foundation Trust, Oxford, United Kingdom.,Oxford Centre for Human Brain Activity, Warneford Hospital, Oxford, United Kingdom
| | - Kate Saunders
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom.,Oxford Health NHS Foundation Trust, Oxford, United Kingdom
| | - Guy M Goodwin
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom.,Oxford Health NHS Foundation Trust, Oxford, United Kingdom
| | - John R Geddes
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom.,Oxford Health NHS Foundation Trust, Oxford, United Kingdom
| |
Collapse
|
46
|
Veatch OJ, Keenan BT, Gehrman PR, Malow BA, Pack AI. Pleiotropic genetic effects influencing sleep and neurological disorders. Lancet Neurol 2017; 16:158-170. [PMID: 28102151 DOI: 10.1016/s1474-4422(16)30339-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 10/04/2016] [Accepted: 11/09/2016] [Indexed: 10/20/2022]
Abstract
Research evidence increasingly points to the large impact of sleep disturbances on public health. Many aspects of sleep are heritable and genes influencing traits such as timing, EEG characteristics, sleep duration, and response to sleep loss have been identified. Notably, large-scale genome-wide analyses have implicated numerous genes with small effects on sleep timing. Additionally, there has been considerable progress in the identification of genes influencing risk for some neurological sleep disorders. For restless legs syndrome, implicated variants are typically in genes associated with neuronal development. By contrast, genes conferring risk for narcolepsy function in the immune system. Many genetic variants associated with sleep disorders are also implicated in neurological disorders in which sleep abnormalities are common; for example, variation in genes involved in synaptic homoeostasis are implicated in autism spectrum disorder and sleep-wake control. Further investigation into pleiotropic roles of genes influencing both sleep and neurological disorders could lead to new treatment strategies for a variety of sleep disturbances.
Collapse
Affiliation(s)
- Olivia J Veatch
- Department of Neurology, Vanderbilt University, Nashville, TN, USA; Center for Sleep and Circadian Neurobiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Brendan T Keenan
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Philip R Gehrman
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Beth A Malow
- Department of Neurology, Vanderbilt University, Nashville, TN, USA
| | - Allan I Pack
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Division of Sleep Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
47
|
Abstract
The circadian clock interacts with the sleep homeostatic drive in humans. Chronotype and sleep parameters show substantial heritability, underscoring a genetic component to these measures. This article reviews the genetic underpinnings of chronotype and of sleep, including sleepiness, sleep quality and latency, and sleep timing and duration in healthy adult sleepers, drawing on candidate gene and genome-wide association studies. Notably, both circadian and noncircadian genes associate with individual differences in chronotype and in sleep parameters. The article concludes with a brief discussion of future research directions.
Collapse
Affiliation(s)
- Namni Goel
- Division of Sleep and Chronobiology, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, 1017 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104-6021, USA.
| |
Collapse
|
48
|
Higgins GA, Allyn-Feuer A, Georgoff P, Nikolian V, Alam HB, Athey BD. Mining the topography and dynamics of the 4D Nucleome to identify novel CNS drug pathways. Methods 2017; 123:102-118. [PMID: 28385536 DOI: 10.1016/j.ymeth.2017.03.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 03/10/2017] [Indexed: 12/16/2022] Open
Abstract
The pharmacoepigenome can be defined as the active, noncoding province of the genome including canonical spatial and temporal regulatory mechanisms of gene regulation that respond to xenobiotic stimuli. Many psychotropic drugs that have been in clinical use for decades have ill-defined mechanisms of action that are beginning to be resolved as we understand the transcriptional hierarchy and dynamics of the nucleus. In this review, we describe spatial, temporal and biomechanical mechanisms mediated by psychotropic medications. Focus is placed on a bioinformatics pipeline that can be used both for detection of pharmacoepigenomic variants that discretize drug response and adverse events to improve pharmacogenomic testing, and for the discovery of novel CNS therapeutics. This approach integrates the functional topology and dynamics of the transcriptional hierarchy of the pharmacoepigenome, gene variant-driven identification of pharmacogenomic regulatory domains, and mesoscale mapping for the discovery of novel CNS pharmacodynamic pathways in human brain. Examples of the application of this pipeline are provided, including the discovery of valproic acid (VPA) mediated transcriptional reprogramming of neuronal cell fate following injury, and mapping of a CNS pathway glutamatergic pathway for the mood stabilizer lithium. These examples in regulatory pharmacoepigenomics illustrate how ongoing research using the 4D nucleome provides a foundation to further insight into previously unrecognized psychotropic drug pharmacodynamic pathways in the human CNS.
Collapse
Affiliation(s)
- Gerald A Higgins
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, USA
| | - Ari Allyn-Feuer
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, USA
| | - Patrick Georgoff
- Department of Surgery, University of Michigan Medical School, USA
| | - Vahagn Nikolian
- Department of Surgery, University of Michigan Medical School, USA
| | - Hasan B Alam
- Department of Surgery, University of Michigan Medical School, USA
| | - Brian D Athey
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, USA; Michigan Institute for Data Science (MIDAS), USA.
| |
Collapse
|
49
|
Houghton KT, Forrest A, Awad A, Atkinson LZ, Stockton S, Harrison PJ, Geddes JR, Cipriani A. Biological rationale and potential clinical use of gabapentin and pregabalin in bipolar disorder, insomnia and anxiety: protocol for a systematic review and meta-analysis. BMJ Open 2017; 7:e013433. [PMID: 28348186 PMCID: PMC5372050 DOI: 10.1136/bmjopen-2016-013433] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Gabapentin has been extensively prescribed off-label for psychiatric indications, with little established evidence of efficacy. Gabapentin and pregabalin, a very similar drug with the same mechanism of action, bind to a subunit of voltage-dependent calcium channels which are implicated in the aetiopathogenesis of bipolar disorder, anxiety and insomnia. This systematic review and meta-analysis aims to collect and critically appraise all the available evidence about the efficacy and tolerability of gabapentin and pregabalin in the treatment of bipolar disorder, insomnia and anxiety. METHODS AND ANALYSIS We will include all randomised controlled trials (RCTs) reported as double-blind and comparing gabapentin or pregabalin with placebo or any other active pharmacological treatment (any preparation, dose, frequency, route of delivery or setting) in patients with bipolar disorder, anxiety or insomnia. For consideration of adverse effects (tolerability), single-blind or open-label RCTs and non-randomised evidence will also be summarised. The main outcomes will be efficacy (measured as dichotomous and continuous outcome) and acceptability (proportion of patients who dropped out of the allocated treatment). Published and unpublished studies will be sought through relevant database searches, trial registries and websites; all reference selection and data extraction will be conducted by at least 2 independent reviewers. We will conduct a random-effects meta-analysis to synthesise all evidence for each outcome. Heterogeneity between studies will be investigated by the I2 statistic. Data from included studies will be entered into a funnel plot for investigation of small-study effects. No subgroup analysis will be undertaken, but we will carry out sensitivity analyses about combination treatment, psychiatric comorbidity, use of rescue medications and fixed versus random-effects model. ETHICS AND DISSEMINATION This review does not require ethical approval. This protocol has been registered on PROSPERO (CRD42016041802). The results of the systematic review will be disseminated via publication in a peer-reviewed journal.
Collapse
Affiliation(s)
- Kerensa T Houghton
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, UK
| | - Alexandra Forrest
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford OX3 7JX, UK
| | - Amine Awad
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, UK
| | - Lauren Z Atkinson
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, UK
| | - Sarah Stockton
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, UK
| | - Paul J Harrison
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford OX3 7JX, UK
| | - John R Geddes
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford OX3 7JX, UK
| | - Andrea Cipriani
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford OX3 7JX, UK
| |
Collapse
|
50
|
Lane JM, Liang J, Vlasac I, Anderson SG, Bechtold DA, Bowden J, Emsley R, Gill S, Little MA, Luik AI, Loudon A, Scheer FAJL, Purcell SM, Kyle SD, Lawlor DA, Zhu X, Redline S, Ray DW, Rutter MK, Saxena R. Genome-wide association analyses of sleep disturbance traits identify new loci and highlight shared genetics with neuropsychiatric and metabolic traits. Nat Genet 2017; 49:274-281. [PMID: 27992416 PMCID: PMC5491693 DOI: 10.1038/ng.3749] [Citation(s) in RCA: 238] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 11/21/2016] [Indexed: 12/16/2022]
Abstract
Chronic sleep disturbances, associated with cardiometabolic diseases, psychiatric disorders and all-cause mortality, affect 25-30% of adults worldwide. Although environmental factors contribute substantially to self-reported habitual sleep duration and disruption, these traits are heritable and identification of the genes involved should improve understanding of sleep, mechanisms linking sleep to disease and development of new therapies. We report single- and multiple-trait genome-wide association analyses of self-reported sleep duration, insomnia symptoms and excessive daytime sleepiness in the UK Biobank (n = 112,586). We discover loci associated with insomnia symptoms (near MEIS1, TMEM132E, CYCL1 and TGFBI in females and WDR27 in males), excessive daytime sleepiness (near AR-OPHN1) and a composite sleep trait (near PATJ (INADL) and HCRTR2) and replicate a locus associated with sleep duration (at PAX8). We also observe genetic correlation between longer sleep duration and schizophrenia risk (rg = 0.29, P = 1.90 × 10-13) and between increased levels of excessive daytime sleepiness and increased measures for adiposity traits (body mass index (BMI): rg = 0.20, P = 3.12 × 10-9; waist circumference: rg = 0.20, P = 2.12 × 10-7).
Collapse
Affiliation(s)
- Jacqueline M Lane
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Jingjing Liang
- Department of Epidemiology and Biostatistics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Irma Vlasac
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Simon G Anderson
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- George Institute for Global Health, University of Oxford, Oxford Martin School, Oxford, UK
| | - David A Bechtold
- Division of Endocrinology, Diabetes and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jack Bowden
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Richard Emsley
- Division of Population Health, Health Services Research and Primary Care, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | | | - Max A Little
- Engineering and Applied Science, Aston University, Birmingham, UK
- Media Laboratory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Annemarie I Luik
- Sleep and Circadian Neuroscience Institute, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Andrew Loudon
- Division of Endocrinology, Diabetes and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Frank A J L Scheer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital and Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Shaun M Purcell
- Broad Institute, Cambridge, Massachusetts, USA
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Simon D Kyle
- Sleep and Circadian Neuroscience Institute, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Xiaofeng Zhu
- Department of Epidemiology and Biostatistics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Susan Redline
- Department of Medicine, Brigham and Women's Hospital and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - David W Ray
- Division of Endocrinology, Diabetes and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Martin K Rutter
- Division of Endocrinology, Diabetes and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester Diabetes Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Richa Saxena
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital and Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|