1
|
Sitaraman D, Vecsey CG, Koochagian C. Activity Monitoring for Analysis of Sleep in Drosophila melanogaster. Cold Spring Harb Protoc 2024; 2024:pdb.top108095. [PMID: 38336390 DOI: 10.1101/pdb.top108095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
Sleep is important for survival, and the need for sleep is conserved across species. In the past two decades, the fruit fly Drosophila melanogaster has emerged as a promising system in which to study the genetic, neural, and physiological bases of sleep. Through significant advances in our understanding of the regulation of sleep in flies, the field is poised to address several open questions about sleep, such as how the need for sleep is encoded, how molecular regulators of sleep are situated within brain networks, and what the functions of sleep are. Here, we describe key findings, open questions, and commonly used methods that have been used to inform existing theories and develop new ways of thinking about the function, regulation, and adaptability of sleep behavior.
Collapse
Affiliation(s)
- Divya Sitaraman
- Department of Psychology, College of Science, California State University, Hayward, California 94542, USA
| | | | - Casey Koochagian
- Neuroscience Program, Skidmore College, Saratoga Springs, New York 12866, USA
| |
Collapse
|
2
|
Flores CC, Pasetto NA, Wang H, Dimitrov AG, Davis JF, Jiang Z, Davis CJ, Gerstner JR. Sleep and diurnal alternative polyadenylation sites associated with human APA-linked brain disorders. NPJ BIOLOGICAL TIMING AND SLEEP 2024; 1:11. [PMID: 39493890 PMCID: PMC11530375 DOI: 10.1038/s44323-024-00012-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/23/2024] [Indexed: 11/05/2024]
Abstract
Disruption of sleep and circadian rhythms are a comorbid feature of many pathologies, and can negatively influence many health conditions, including neurodegenerative disease, metabolic illness, cancer, and various neurological disorders. Genetic association studies linking sleep and circadian disturbances with disease susceptibility have mainly focused on changes in gene expression due to mutations, such as single-nucleotide polymorphisms. The interaction between sleep and/or circadian rhythms with the use of Alternative Polyadenylation (APA) has been largely undescribed, particularly in the context of other disorders. APA generates transcript isoforms by utilizing various polyadenylation sites (PASs) from the same gene affecting its mRNA translation, stability, localization, and subsequent function. Here we identified unique APAs expressed in rat brain over time-of-day, immediately following sleep deprivation, and the subsequent recovery period. From these data, we performed a secondary analysis of these sleep- or time-of-day associated PASs with recently described APA-linked human brain disorder susceptibility genes.
Collapse
Affiliation(s)
- Carlos C. Flores
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA USA
| | - Nickolas A. Pasetto
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA USA
| | - Hongyang Wang
- Department of Animal Sciences, College of Agricultural, Human, and Natural Resource Sciences, Washington State University, Pullman, WA USA
- Institute of Animal Husbandry & Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, China
| | - Alexander G. Dimitrov
- Department of Mathematics and Statistics, College of Arts and Sciences, Washington State University, Vancouver, WA USA
| | - Jon F. Davis
- Department of Integrative Physiology and Neuroscience, Pullman, WA USA
- Integrated Physiology Research, Novo Nordisk, Lexington, MA USA
| | - Zhihua Jiang
- Department of Animal Sciences, College of Agricultural, Human, and Natural Resource Sciences, Washington State University, Pullman, WA USA
| | - Christopher J. Davis
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA USA
- Department of Integrative Physiology and Neuroscience, Pullman, WA USA
- Sleep and Performance Research Center, Washington State University, Spokane, WA USA
- Steve Gleason Institute for Neuroscience, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA USA
| | - Jason R. Gerstner
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA USA
- Department of Integrative Physiology and Neuroscience, Pullman, WA USA
- Sleep and Performance Research Center, Washington State University, Spokane, WA USA
- Steve Gleason Institute for Neuroscience, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA USA
| |
Collapse
|
3
|
Lloyd E, Xia F, Moore K, Zertuche C, Rastogi A, Kozol R, Kenzior O, Warren W, Appelbaum L, Moran RL, Zhao C, Duboue E, Rohner N, Keene AC. Elevated DNA Damage without signs of aging in the short-sleeping Mexican Cavefish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.18.590174. [PMID: 38659770 PMCID: PMC11042282 DOI: 10.1101/2024.04.18.590174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Dysregulation of sleep has widespread health consequences and represents an enormous health burden. Short-sleeping individuals are predisposed to the effects of neurodegeneration, suggesting a critical role for sleep in the maintenance of neuronal health. While the effects of sleep on cellular function are not completely understood, growing evidence has identified an association between sleep loss and DNA damage, raising the possibility that sleep facilitates efficient DNA repair. The Mexican tetra fish, Astyanax mexicanus provides a model to investigate the evolutionary basis for changes in sleep and the consequences of sleep loss. Multiple cave-adapted populations of these fish have evolved to sleep for substantially less time compared to surface populations of the same species without identifiable impacts on healthspan or longevity. To investigate whether the evolved sleep loss is associated with DNA damage and cellular stress, we compared the DNA Damage Response (DDR) and oxidative stress levels between A. mexicanus populations. We measured markers of chronic sleep loss and discovered elevated levels of the DNA damage marker γH2AX in the brain, and increased oxidative stress in the gut of cavefish, consistent with chronic sleep deprivation. Notably, we found that acute UV-induced DNA damage elicited an increase in sleep in surface fish but not in cavefish. On a transcriptional level, only the surface fish activated the photoreactivation repair pathway following UV damage. These findings suggest a reduction of the DDR in cavefish compared to surface fish that coincides with elevated DNA damage in cavefish. To examine DDR pathways at a cellular level, we created an embryonic fibroblast cell line from the two populations of A. mexicanus . We observed that both the DDR and DNA repair were diminished in the cavefish cells, corroborating the in vivo findings and suggesting that the acute response to DNA damage is lost in cavefish. To investigate the long-term impact of these changes, we compared the transcriptome in the brain and gut of aged surface fish and cavefish. Strikingly, many genes that are differentially expressed between young and old surface fish do not transcriptionally vary by age in cavefish. Taken together, these findings suggest that cavefish have developed resilience to sleep loss, despite possessing cellular hallmarks of chronic sleep deprivation.
Collapse
|
4
|
Liu AC, Shen Y, Serbinski CR, He H, Roman D, Endale M, Aschbacher-Smith L, King KA, Granadillo JL, López I, Krueger DA, Dye TJ, Smith DF, Hogenesch JB, Prada CE. Clinical and functional studies of MTOR variants in Smith-Kingsmore syndrome reveal deficits of circadian rhythm and sleep-wake behavior. HGG ADVANCES 2024; 5:100333. [PMID: 39030910 PMCID: PMC11342114 DOI: 10.1016/j.xhgg.2024.100333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/13/2024] [Accepted: 07/15/2024] [Indexed: 07/22/2024] Open
Abstract
Heterozygous de novo or inherited gain-of-function mutations in the MTOR gene cause Smith-Kingsmore syndrome (SKS). SKS is a rare autosomal dominant condition, and individuals with SKS display macrocephaly/megalencephaly, developmental delay, intellectual disability, and seizures. A few dozen individuals are reported in the literature. Here, we report a cohort of 28 individuals with SKS that represent nine MTOR pathogenic variants. We conducted a detailed natural history study and found pathophysiological deficits among individuals with SKS in addition to the common neurodevelopmental symptoms. These symptoms include sleep-wake disturbance, hyperphagia, and hyperactivity, indicative of homeostatic imbalance. To characterize these variants, we developed cell models and characterized their functional consequences. We showed that these SKS variants display a range of mechanistic target of rapamycin (mTOR) activities and respond to the mTOR inhibitor, rapamycin, differently. For example, the R1480_C1483del variant we identified here and the previously known C1483F are more active than wild-type controls and less responsive to rapamycin. Further, we showed that SKS mutations dampened circadian rhythms and low-dose rapamycin improved the rhythm amplitude, suggesting that optimal mTOR activity is required for normal circadian function. As SKS is caused by gain-of-function mutations in MTOR, rapamycin was used to treat several patients. While higher doses of rapamycin caused delayed sleep-wake phase disorder in a subset of patients, optimized lower doses improved sleep. Our study expands the clinical and molecular spectrum of SKS and supports further studies for mechanism-guided treatment options to improve sleep-wake behavior and overall health.
Collapse
Affiliation(s)
- Andrew C Liu
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA.
| | - Yang Shen
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Carolyn R Serbinski
- Divisions of Human Genetics, Neurology, Immunobiology, Pediatric Otolaryngology, and Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Division of Genetics, Genomics & Metabolism, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Hongzhi He
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Destino Roman
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Mehari Endale
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Lindsey Aschbacher-Smith
- Divisions of Human Genetics, Neurology, Immunobiology, Pediatric Otolaryngology, and Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Katherine A King
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Jorge L Granadillo
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Isabel López
- Pediatric Neurology Unit, Department of Neurology, Clínica Las Condes, Santiago, Chile
| | - Darcy A Krueger
- Divisions of Human Genetics, Neurology, Immunobiology, Pediatric Otolaryngology, and Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Thomas J Dye
- Divisions of Human Genetics, Neurology, Immunobiology, Pediatric Otolaryngology, and Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - David F Smith
- Divisions of Pediatric Otolaryngology and Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; The Sleep Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; The Center for Circadian Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Otolaryngology Head and Neck Surgery, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - John B Hogenesch
- Divisions of Human Genetics, Neurology, Immunobiology, Pediatric Otolaryngology, and Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Carlos E Prada
- Divisions of Human Genetics, Neurology, Immunobiology, Pediatric Otolaryngology, and Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Division of Genetics, Genomics & Metabolism, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA; Department of Pediatrics, Feinberg School of Medicine of Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
5
|
Flores CC, Pasetto NA, Wang H, Dimitrov AG, Davis JF, Jiang Z, Davis CJ, Gerstner JR. Sleep and diurnal alternative polyadenylation sites associated with human APA-linked brain disorders. RESEARCH SQUARE 2024:rs.3.rs-4707772. [PMID: 39149473 PMCID: PMC11326403 DOI: 10.21203/rs.3.rs-4707772/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Disruption of sleep and circadian rhythms are a comorbid feature of many pathologies, and can negatively influence many health conditions, including neurodegenerative disease, metabolic illness, cancer, and various neurological disorders. Genetic association studies linking sleep and circadian disturbances with disease susceptibility have mainly focused on changes in gene expression due to mutations, such as single-nucleotide polymorphisms. The interaction between sleep and/or circadian rhythms with the use of Alternative Polyadenylation (APA) has been largely undescribed, particularly in the context of other disorders. APA is a process that generates various transcript isoforms of the same gene affecting its mRNA translation, stability, localization, and subsequent function. Here we identified unique APAs expressed in rat brain over time-of-day, immediately following sleep deprivation, and the subsequent recovery period. From these data, we performed a secondary analysis of these sleep- or time-of-day associated PASs with recently described APA-linked human brain disorder susceptibility genes.
Collapse
|
6
|
Rößler DC, Klein BA. More sleep for behavioral ecologists. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART A, ECOLOGICAL AND INTEGRATIVE PHYSIOLOGY 2024. [PMID: 39034483 DOI: 10.1002/jez.2856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/23/2024]
Abstract
From jellyfish to parrot fish and roundworms to homeotherms, all animals are thought to sleep. Despite its presumed universality, sleep is a poorly understood behavior, varying significantly in its expression across, and even within, animal lineages. There is still no consensus about the origin, architecture, ecology of sleep, or even its defining characters. The field of behavioral ecology has the potential to extend our knowledge of sleep behavior to nontraditional models and in ecologically relevant settings. Here, we highlight current efforts in diversifying the field to generate stronger synergies between historically human-focused sleep research and behavioral ecology. Our primary aim is for behavioral ecology to enhance sleep research by contributing crucial observations as well as by creating novel comparative and evolutionary frameworks. At the same time, sleep research can enhance behavioral ecology by exposing the relevance of sleep to wakeful behaviors. Nikolaas Tinbergen's four levels of analysis have served as a foundation for comprehensively addressing questions in behavior, and we introduce some Tinbergian approaches to examine the interplay between sleep and wake under ecologically meaningful conditions.
Collapse
Affiliation(s)
- Daniela C Rößler
- Department of Biology, University of Konstanz, Konstanz, Germany
- Zukunftskolleg, University of Konstanz, Konstanz, Germany
- Department of Ecology of Animal Societies, Max Planck Institute of Animal Behavior, Konstanz, Germany
| | - Barrett A Klein
- Biology Department, University of Wisconsin-La Crosse, La Crosse, USA
| |
Collapse
|
7
|
Pandi-Perumal SR, Saravanan KM, Paul S, Namasivayam GP, Chidambaram SB. Waking Up the Sleep Field: An Overview on the Implications of Genetics and Bioinformatics of Sleep. Mol Biotechnol 2024; 66:919-931. [PMID: 38198051 DOI: 10.1007/s12033-023-01009-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 11/28/2023] [Indexed: 01/11/2024]
Abstract
Sleep genetics is an intriguing, as yet less understood, understudied, emerging area of biological and medical discipline. A generalist may not be aware of the current status of the field given the variety of journals that have published studies on the genetics of sleep and the circadian clock over the years. For researchers venturing into this fascinating area, this review thus includes fundamental features of circadian rhythm and genetic variables impacting sleep-wake cycles. Sleep/wake pathway medication exposure and susceptibility are influenced by genetic variations, and the responsiveness of sleep-related medicines is influenced by several functional polymorphisms. This review highlights the features of the circadian timing system and then a genetic perspective on wakefulness and sleep, as well as the relationship between sleep genetics and sleep disorders. Neurotransmission genes, as well as circadian and sleep/wake receptors, exhibit functional variability. Experiments on animals and humans have shown that these genetic variants impact clock systems, signaling pathways, nature, amount, duration, type, intensity, quality, and quantity of sleep. In this regard, the overview covers research on sleep genetics, the genomic properties of several popular model species used in sleep studies, homologs of mammalian genes, sleep disorders, and related genes. In addition, the study includes a brief discussion of sleep, narcolepsy, and restless legs syndrome from the viewpoint of a model organism. It is suggested that the understanding of genetic clues on sleep function and sleep disorders may, in future, result in an evidence-based, personalized treatment of sleep disorders.
Collapse
Affiliation(s)
- Seithikurippu R Pandi-Perumal
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education and Research, Mysuru, Karnataka, 570015, India
- Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, 602105, India
- Division of Research and Development, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Konda Mani Saravanan
- Department of Biotechnology, Bharath Institute of Higher Education and Research, Chennai, Tamil Nadu, 600073, India
| | - Sayan Paul
- Department of Biochemistry & Molecular Biology, The University of Texas Medical Branch at Galveston, Galveston, TX, 77555, USA
| | - Ganesh Pandian Namasivayam
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), A210, Kyoto University Institute for Advanced Study, Yoshida Ushinomiya-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Saravana Babu Chidambaram
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education and Research, Mysuru, Karnataka, 570015, India.
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, Karnataka, 570015, India.
- Special Interest Group - Brain, Behaviour and Cognitive Neurosciences, JSS Academy of Higher Education & Research, Mysuru, Karnataka, 570015, India.
| |
Collapse
|
8
|
Uy G, Farrell LN, Faheem SF, Kinne LE, Adore MG, Im SH, Fairman R. The Effects of poly-GA and poly-PR C9orf72 Dipeptide Repeats on Sleep Patterns in Drosophila melanogaster. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.000973. [PMID: 38495583 PMCID: PMC10943360 DOI: 10.17912/micropub.biology.000973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 01/31/2024] [Accepted: 03/01/2024] [Indexed: 03/19/2024]
Abstract
C9orf72 is the most common familial gene associated with amyotrophic lateral sclerosis (ALS). Dipeptide repeats (DPRs) encoded by an expanded nucleotide repeat sequence in the C9orf72 gene were found in the sleep-related neurons of patients, indicating a role of DPRs in ALS-associated sleep disruptions. Poly-GA or poly-PR DPRs were expressed in male Drosophila melanogaster to study their effect on sleep . Poly-PR expression caused sleep disruptions while poly-GA expression did not. This study validates the use of Drosophila as an in vivo model system for exploring the roles of DPRs in perturbing the underlying molecular mechanisms in sleep regulation.
Collapse
Affiliation(s)
- Genevieve Uy
- Chemistry, Haverford College, Philadelphia, Pennsylvania, United States
| | - Laura N. Farrell
- Neuroscience, Haverford College, Philadelphia, Pennsylvania, United States
| | - Syeda F. Faheem
- Biology, Haverford College, Philadelphia, Pennsylvania, United States
| | - Lauren E. Kinne
- Biology, Haverford College, Philadelphia, Pennsylvania, United States
| | - Madison G. Adore
- Biology, Haverford College, Philadelphia, Pennsylvania, United States
| | - Seol Hee Im
- Biology, Haverford College, Philadelphia, Pennsylvania, United States
| | - Robert Fairman
- Biology, Haverford College, Philadelphia, Pennsylvania, United States
| |
Collapse
|
9
|
Flores CC, Pasetto NA, Wang H, Dimitrov A, Davis JF, Jiang Z, Davis CJ, Gerstner JR. Identification of sleep and circadian alternative polyadenylation sites associated with APA-linked human brain disorders. RESEARCH SQUARE 2024:rs.3.rs-3867797. [PMID: 38313253 PMCID: PMC10836116 DOI: 10.21203/rs.3.rs-3867797/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
Sleep and circadian rhythm disruptions are comorbid features of many pathologies and can negatively influence numerous health conditions, including degenerative diseases, metabolic illnesses, cancer, and various neurological disorders. Genetic association studies linking sleep and circadian disturbances with disease susceptibility have mainly focused on changes in gene expression due to mutations, such as single-nucleotide polymorphisms. Thus, associations between sleep and/or circadian rhythm and alternative polyadenylation (APA), particularly in the context of other health challenges, are largely undescribed. APA is a process that generates various transcript isoforms from the same gene, resulting in effects on mRNA translation, stability, localization, and subsequent function. Here, we have identified unique APAs in rat brain that exhibit time-of-day-dependent oscillations in expression as well as APAs that are altered by sleep deprivation and the subsequent recovery period. Genes affected by APA usage include Mapt/Tau, Ntrk2, Homer1A, Sin3band Sorl. Sorl1 has two APAs which cycle with a 24 h period, one additional APA cycles with a 12 h period and one more that is reduced during recovery sleep. Finally, we compared sleep- or circadian-associated APAs with recently described APA-linked brain disorder susceptibility genes and found 46 genes in common.
Collapse
|
10
|
Kompotis K, Mang GM, Hubbard J, Jimenez S, Emmenegger Y, Polysopoulos C, Hor CN, Wigger L, Hébert SS, Mongrain V, Franken P. Cortical miR-709 links glutamatergic signaling to NREM sleep EEG slow waves in an activity-dependent manner. Proc Natl Acad Sci U S A 2024; 121:e2220532121. [PMID: 38207077 PMCID: PMC10801902 DOI: 10.1073/pnas.2220532121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 11/29/2023] [Indexed: 01/13/2024] Open
Abstract
MicroRNAs (miRNAs) are key post-transcriptional regulators of gene expression that have been implicated in a plethora of neuronal processes. Nevertheless, their role in regulating brain activity in the context of sleep has so far received little attention. To test their involvement, we deleted mature miRNAs in post-mitotic neurons at two developmental ages, i.e., in early adulthood using conditional Dicer knockout (cKO) mice and in adult mice using an inducible conditional Dicer cKO (icKO) line. In both models, electroencephalographic (EEG) activity was affected and the response to sleep deprivation (SD) altered; while the rapid-eye-movement sleep (REMS) rebound was compromised in both, the increase in EEG delta (1 to 4 Hz) power during non-REMS (NREMS) was smaller in cKO mice and larger in icKO mice compared to controls. We subsequently investigated the effects of SD on the forebrain miRNA transcriptome and found that the expression of 48 miRNAs was affected, and in particular that of the activity-dependent miR-709. In vivo inhibition of miR-709 in the brain increased EEG power during NREMS in the slow-delta (0.75 to 1.75 Hz) range, particularly after periods of prolonged wakefulness. Transcriptome analysis of primary cortical neurons in vitro revealed that miR-709 regulates genes involved in glutamatergic neurotransmission. A subset of these genes was also affected in the cortices of sleep-deprived, miR-709-inhibited mice. Our data implicate miRNAs in the regulation of EEG activity and indicate that miR-709 links neuronal activity during wakefulness to brain synchrony during sleep through the regulation of glutamatergic signaling.
Collapse
Affiliation(s)
- Konstantinos Kompotis
- Center for Integrative Genomics, University of Lausanne, LausanneCH-1015, Switzerland
- Institute of Pharmacology and Toxicology, University of Zurich, ZurichCH-8057, Switzerland
| | - Géraldine M. Mang
- Center for Integrative Genomics, University of Lausanne, LausanneCH-1015, Switzerland
| | - Jeffrey Hubbard
- Center for Integrative Genomics, University of Lausanne, LausanneCH-1015, Switzerland
| | - Sonia Jimenez
- Center for Integrative Genomics, University of Lausanne, LausanneCH-1015, Switzerland
| | - Yann Emmenegger
- Center for Integrative Genomics, University of Lausanne, LausanneCH-1015, Switzerland
| | - Christos Polysopoulos
- Department of Biostatistics, Epidemiology, Biostatistics and Prevention Institute, University of Zurich, ZurichCH-8057, Switzerland
| | - Charlotte N. Hor
- Center for Integrative Genomics, University of Lausanne, LausanneCH-1015, Switzerland
| | - Leonore Wigger
- Genomic Technologies Facility, Center for Integrative Genomics, University of Lausanne, LausanneCH-1015, Switzerland
| | - Sébastien S. Hébert
- Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Axe Neurosciences, Québec, QCG1V 4G2, Canada
- Département de psychiatrie et de neurosciences, Faculté de médecine, Université Laval, Québec, QCG1V 0A6, Canada
| | - Valérie Mongrain
- Department of Neuroscience, Université de Montréal, Montréal, QCH3T 1J4, Canada
- Centre de recherche, Centre hospitalier de l’Université de Montréal, Montréal, QCH2X 0A9, Canada
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Montréal, QCH4J 1C5, Canada
| | - Paul Franken
- Center for Integrative Genomics, University of Lausanne, LausanneCH-1015, Switzerland
| |
Collapse
|
11
|
Bae GY, Ko K, Yang E, Park SS, Suh HJ, Hong KB. Combined Effects of Ziziphus jujuba, Dimocarpus longan, and Lactuca sativa on Sleep-Related Behaviors through GABAergic Signaling. Foods 2023; 13:1. [PMID: 38201029 PMCID: PMC10778002 DOI: 10.3390/foods13010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/11/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
We aimed to analyze the increase in the sleep-promoting effects based on the mixed ratio of botanical extracts, Ziziphus jujuba seeds, Dimocarpus longan fruits, and Lactuca sativa leaves, using animal models. Behavioral analyses, including an analysis of the total sleep time of Drosophila melanogaster, were conducted to select the optimal mixed ratio of the three botanical extracts. The effects were verified in a caffeine-induced sleepless model, specific neurotransmitter receptor antagonists, and ICR mice. In D. melanogaster exposed to 2.0% of each extract, group behavior was significantly reduced, and the mixed extracts of Z. jujuba, D. longan, and L. sativa (4:1:1 and 1:4:1) significantly increased the total sleep time with individual fruit flies. In the caffeine-induced insomnia model, mixed extracts (4:1:1 and 1:4:1) led to the highest increase in total sleep time. An analysis of locomotor ability revealed a significant reduction in the mobility percentage in the mixed extract groups (0:0:1, 1:0:1, 1:1:1, 4:1:1, and 1:4:1). The administration of Z. jujuba extract and mixed extracts (4:1:1) significantly increased the expression of GABAA-R, whereas the administration of the mixed extracts (4:1:1) and (1:4:1) significantly increased the expression of GABAB-R1 and GABAB-R2, respectively. D. longan extract and the mixed ratio (1:4:1) reduced the subjective nighttime movement and increased the total sleep time in the presence of flumazenil. An analysis of ICR mice indicated that the administration of mixed extracts (4:1:1) significantly increased sleep duration in a dose-dependent manner. These results indicated that the mixed ratio of Z. jujuba, D. longan, and L. sativa extracts, particularly the mixed ratio of 4:1:1, may have sleep-enhancing effects in fruit flies and mice. The study also identified changes in gene expression related to GABA receptors, indicating the potential mechanism for the observed sleep-promoting effects.
Collapse
Affiliation(s)
- Gi Yeon Bae
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea; (G.Y.B.); (H.J.S.)
| | - Kayoung Ko
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Republic of Korea; (K.K.); (E.Y.); (S.-S.P.)
| | - Eunseon Yang
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Republic of Korea; (K.K.); (E.Y.); (S.-S.P.)
| | - Sung-Soo Park
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Republic of Korea; (K.K.); (E.Y.); (S.-S.P.)
| | - Hyung Joo Suh
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea; (G.Y.B.); (H.J.S.)
- BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul 02841, Republic of Korea
| | - Ki-Bae Hong
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Republic of Korea; (K.K.); (E.Y.); (S.-S.P.)
| |
Collapse
|
12
|
Wu Z, Liu L, Li L, Cao X, Jia W, Liao X, Zhao Z, Qi H, Fan G, Lu H, Shu C, Zhen M, Wang C, Bai C. Oral nano-antioxidants improve sleep by restoring intestinal barrier integrity and preventing systemic inflammation. Natl Sci Rev 2023; 10:nwad309. [PMID: 38204453 PMCID: PMC10781441 DOI: 10.1093/nsr/nwad309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/23/2023] [Accepted: 11/28/2023] [Indexed: 01/12/2024] Open
Abstract
Sleep deprivation (SD) is a severe public health threat that can cause systemic inflammation and nerve damage. Few effective and side-effect-free drugs are available to address SD. However, the bidirectional communications between the brain and gut provide new strategies for anti-SD therapeutics. Here we explored oral delivery of fullerene nano-antioxidants (FNAO) in the SD model to improve sleep by regulating abnormal intestinal barrier and systemic inflammation via the brain-gut axis. SD caused excessive reactive oxygen species (ROS) production and hyperactive inflammatory responses in the intestines of zebrafish and mouse models, leading to disturbed sleep patterns and reduced brain nerve activity. Of note, based on the property of the conjugated π bond of the C60 structure to absorb unpaired electrons, oral FNAO efficiently reduced the excessive ROS in the intestines, maintained redox homeostasis and intestinal barrier integrity, and ameliorated intestinal and systemic inflammation, resulting in superior sleep improvement. Our findings suggest that maintaining intestinal homeostasis may be a promising avenue for SD-related nerve injury therapy.
Collapse
Affiliation(s)
- Zhanfeng Wu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lei Liu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lei Li
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinran Cao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wang Jia
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaodan Liao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhongpu Zhao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hedong Qi
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guoqiang Fan
- School of Pharmacy, Wenzhou Medical University, Wenzhou 325000, China
| | - Huiqiang Lu
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou 341000, China
| | - Chunying Shu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mingming Zhen
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunru Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunli Bai
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
13
|
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
|
14
|
Talwar P, Deantoni M, Van Egroo M, Muto V, Chylinski D, Koshmanova E, Jaspar M, Meyer C, Degueldre C, Berthomier C, Luxen A, Salmon E, Collette F, Dijk DJ, Schmidt C, Phillips C, Maquet P, Sherif S, Vandewalle G. In vivo marker of brainstem myelin is associated to quantitative sleep parameters in healthy young men. Sci Rep 2023; 13:20873. [PMID: 38012207 PMCID: PMC10682495 DOI: 10.1038/s41598-023-47753-x] [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: 08/22/2023] [Accepted: 11/17/2023] [Indexed: 11/29/2023] Open
Abstract
The regional integrity of brain subcortical structures has been implicated in sleep-wake regulation, however, their associations with sleep parameters remain largely unexplored. Here, we assessed association between quantitative Magnetic Resonance Imaging (qMRI)-derived marker of the myelin content of the brainstem and the variability in the sleep electrophysiology in a large sample of 18-to-31 years healthy young men (N = 321; ~ 22 years). Separate Generalized Additive Model for Location, Scale and Shape (GAMLSS) revealed that sleep onset latency and slow wave energy were significantly associated with MTsat estimates in the brainstem (pcorrected ≤ 0.03), with overall higher MTsat value associated with values reflecting better sleep quality. The association changed with age, however (MTsat-by-age interaction-pcorrected ≤ 0.03), with higher MTsat value linked to better values in the two sleep metrics in the younger individuals of our sample aged ~ 18 to 20 years. Similar associations were detected across different parts of the brainstem (pcorrected ≤ 0.03), suggesting that the overall maturation and integrity of the brainstem was associated with both sleep metrics. Our results suggest that myelination of the brainstem nuclei essential to regulation of sleep is associated with inter-individual differences in sleep characteristics during early adulthood. They may have implications for sleep disorders or neurological diseases related to myelin.
Collapse
Affiliation(s)
- Puneet Talwar
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
| | - Michele Deantoni
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
| | - Maxime Van Egroo
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
| | - Vincenzo Muto
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium
- Psychology and Cognitive Neuroscience Research Unit, University of Liège, Liège, Belgium
| | - Daphne Chylinski
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
| | - Ekaterina Koshmanova
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
| | - Mathieu Jaspar
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium
| | - Christelle Meyer
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium
| | - Christian Degueldre
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
| | | | - André Luxen
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
| | - Eric Salmon
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
- Psychology and Cognitive Neuroscience Research Unit, University of Liège, Liège, Belgium
- Department of Neurology, CHU of Liège, Liège, Belgium
| | - Fabienne Collette
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
- Psychology and Cognitive Neuroscience Research Unit, University of Liège, Liège, Belgium
| | - D-J Dijk
- Sleep Research Centre, University of Surrey, Guildford, UK
- UK Dementia Research Institute, University of Surrey, Guildford, UK
| | - Christina Schmidt
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
- Psychology and Cognitive Neuroscience Research Unit, University of Liège, Liège, Belgium
| | - Christophe Phillips
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
- In Silico Medicine Unit, GIGA-Institute, University of Liège, Liège, Belgium
| | - Pierre Maquet
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium
- Department of Neurology, CHU of Liège, Liège, Belgium
| | - Siya Sherif
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium
| | - Gilles Vandewalle
- GIGA-Institute, CRC-In Vivo Imaging Unit, Bâtiment B30, Université de Liège, 4000, Liège, Belgium.
| |
Collapse
|
15
|
Axelrod S, Li X, Sun Y, Lincoln S, Terceros A, O’Neil J, Wang Z, Nguyen A, Vora A, Spicer C, Shapiro B, Young MW. The Drosophila blood-brain barrier regulates sleep via Moody G protein-coupled receptor signaling. Proc Natl Acad Sci U S A 2023; 120:e2309331120. [PMID: 37831742 PMCID: PMC10589661 DOI: 10.1073/pnas.2309331120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/28/2023] [Indexed: 10/15/2023] Open
Abstract
Sleep is vital for most animals, yet its mechanism and function remain unclear. We found that permeability of the BBB (blood-brain barrier)-the organ required for the maintenance of homeostatic levels of nutrients, ions, and other molecules in the brain-is modulated by sleep deprivation (SD) and can cell-autonomously effect sleep changes. We observed increased BBB permeability in known sleep mutants as well as in acutely sleep-deprived animals. In addition to molecular tracers, SD-induced BBB changes also increased the penetration of drugs used in the treatment of brain pathologies. After chronic/genetic or acute SD, rebound sleep or administration of the sleeping aid gaboxadol normalized BBB permeability, showing that SD effects on the BBB are reversible. Along with BBB permeability, RNA levels of the BBB master regulator moody are modulated by sleep. Conversely, altering BBB permeability alone through glia-specific modulation of moody, gαo, loco, lachesin, or neuroglian-each a well-studied regulator of BBB function-was sufficient to induce robust sleep phenotypes. These studies demonstrate a tight link between BBB permeability and sleep and indicate a unique role for the BBB in the regulation of sleep.
Collapse
Affiliation(s)
- Sofia Axelrod
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Xiaoling Li
- International Personalized Cancer Center, Tianjin Cancer Hospital Airport Hospital, Tianjin300308, China
| | - Yingwo Sun
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Samantha Lincoln
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Andrea Terceros
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Jenna O’Neil
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Zikun Wang
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Andrew Nguyen
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Aabha Vora
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Carmen Spicer
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Benjamin Shapiro
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Michael W. Young
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| |
Collapse
|
16
|
Ajayi OM, Wynne NE, Chen SC, Vinauger C, Benoit JB. Sleep: An Essential and Understudied Process in the Biology of Blood-Feeding Arthropods. Integr Comp Biol 2023; 63:530-547. [PMID: 37429615 PMCID: PMC10503478 DOI: 10.1093/icb/icad097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 07/12/2023] Open
Abstract
Understanding the biology of blood-feeding arthropods is critical to managing them as vectors of etiological agents. Circadian rhythms act in the regulation of behavioral and physiological aspects such as blood feeding, immunity, and reproduction. However, the impact of sleep on these processes has been largely ignored in blood-feeding arthropods, but recent studies in mosquitoes show that sleep-like states directly impact host landing and blood feeding. Our focus in this review is on discussing the relationship between sleep and circadian rhythms in blood-feeding arthropods along with how unique aspects such as blood gluttony and dormancy can impact sleep-like states. We highlight that sleep-like states are likely to have profound impacts on vector-host interactions but will vary between lineages even though few direct studies have been conducted. A myriad of factors, such as artificial light, could directly impact the time and levels of sleep in blood-feeding arthropods and their roles as vectors. Lastly, we discuss underlying factors that make sleep studies in blood-feeding arthropods difficult and how these can be bypassed. As sleep is a critical factor in the fitness of animal systems, a lack of focus on sleep in blood-feeding arthropods represents a significant oversight in understanding their behavior and its role in pathogen transmission.
Collapse
Affiliation(s)
- Oluwaseun M Ajayi
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Nicole E Wynne
- Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Shyh-Chi Chen
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Clément Vinauger
- Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Joshua B Benoit
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45221, USA
| |
Collapse
|
17
|
Li Y, Li C, Liu Y, Yu J, Yang J, Cui Y, Wang TV, Li C, Jiang L, Song M, Rao Y. Sleep need, the key regulator of sleep homeostasis, is indicated and controlled by phosphorylation of threonine 221 in salt-inducible kinase 3. Genetics 2023; 225:iyad136. [PMID: 37477881 DOI: 10.1093/genetics/iyad136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 06/11/2023] [Accepted: 07/11/2023] [Indexed: 07/22/2023] Open
Abstract
Sleep need drives sleep and plays a key role in homeostatic regulation of sleep. So far sleep need can only be inferred by animal behaviors and indicated by electroencephalography (EEG). Here we report that phosphorylation of threonine (T) 221 of the salt-inducible kinase 3 (SIK3) increased the catalytic activity and stability of SIK3. T221 phosphorylation in the mouse brain indicates sleep need: more sleep resulting in less phosphorylation and less sleep more phosphorylation during daily sleep/wake cycle and after sleep deprivation (SD). Sleep need was reduced in SIK3 loss of function (LOF) mutants and by T221 mutation to alanine (T221A). Rebound after SD was also decreased in SIK3 LOF and T221A mutant mice. By contrast, SIK1 and SIK2 do not satisfy criteria to be both an indicator and a controller of sleep need. Our results reveal SIK3-T221 phosphorylation as a chemical modification which indicates and controls sleep need.
Collapse
Affiliation(s)
- Yang Li
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Institute of Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518067, China
- Capital Medical University, Beijing 10069, China
- Chinese Institute for Brain Research, Changping Laboratory, Yard 28, Science Park Road, ZGC Life Science Park, Changping District, Beijing 102206, China
| | - Chengang Li
- Institute of Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518067, China
- Capital Medical University, Beijing 10069, China
- Chinese Institute for Brain Research, Changping Laboratory, Yard 28, Science Park Road, ZGC Life Science Park, Changping District, Beijing 102206, China
| | - Yuxiang Liu
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Institute of Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518067, China
- Capital Medical University, Beijing 10069, China
- Chinese Institute for Brain Research, Changping Laboratory, Yard 28, Science Park Road, ZGC Life Science Park, Changping District, Beijing 102206, China
| | - Jianjun Yu
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Jingqun Yang
- Institute of Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518067, China
- Capital Medical University, Beijing 10069, China
- Chinese Institute for Brain Research, Changping Laboratory, Yard 28, Science Park Road, ZGC Life Science Park, Changping District, Beijing 102206, China
| | - Yunfeng Cui
- Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing 102206, China
| | - Tao V Wang
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Chaoyi Li
- Institute of Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518067, China
- Capital Medical University, Beijing 10069, China
- Chinese Institute for Brain Research, Changping Laboratory, Yard 28, Science Park Road, ZGC Life Science Park, Changping District, Beijing 102206, China
| | - Lifen Jiang
- Institute of Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518067, China
- Capital Medical University, Beijing 10069, China
- Chinese Institute for Brain Research, Changping Laboratory, Yard 28, Science Park Road, ZGC Life Science Park, Changping District, Beijing 102206, China
| | - Meilin Song
- Institute of Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518067, China
- Capital Medical University, Beijing 10069, China
- Chinese Institute for Brain Research, Changping Laboratory, Yard 28, Science Park Road, ZGC Life Science Park, Changping District, Beijing 102206, China
| | - Yi Rao
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Institute of Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518067, China
- Capital Medical University, Beijing 10069, China
- Chinese Institute for Brain Research, Changping Laboratory, Yard 28, Science Park Road, ZGC Life Science Park, Changping District, Beijing 102206, China
- Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing 102206, China
| |
Collapse
|
18
|
Barlow IL, Mackay E, Wheater E, Goel A, Lim S, Zimmerman S, Woods I, Prober DA, Rihel J. The zebrafish mutant dreammist implicates sodium homeostasis in sleep regulation. eLife 2023; 12:RP87521. [PMID: 37548652 PMCID: PMC10406431 DOI: 10.7554/elife.87521] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023] Open
Abstract
Sleep is a nearly universal feature of animal behaviour, yet many of the molecular, genetic, and neuronal substrates that orchestrate sleep/wake transitions lie undiscovered. Employing a viral insertion sleep screen in larval zebrafish, we identified a novel gene, dreammist (dmist), whose loss results in behavioural hyperactivity and reduced sleep at night. The neuronally expressed dmist gene is conserved across vertebrates and encodes a small single-pass transmembrane protein that is structurally similar to the Na+,K+-ATPase regulator, FXYD1/Phospholemman. Disruption of either fxyd1 or atp1a3a, a Na+,K+-ATPase alpha-3 subunit associated with several heritable movement disorders in humans, led to decreased night-time sleep. Since atpa1a3a and dmist mutants have elevated intracellular Na+ levels and non-additive effects on sleep amount at night, we propose that Dmist-dependent enhancement of Na+ pump function modulates neuronal excitability to maintain normal sleep behaviour.
Collapse
Affiliation(s)
- Ida L Barlow
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Eirinn Mackay
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Emily Wheater
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Aimee Goel
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Sumi Lim
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Steve Zimmerman
- Department of Molecular and Cellular Biology, Harvard UniversityCambridgeUnited States
| | | | - David A Prober
- Division of Biology and Biological Engineering, California Institute of TechnologyPasadenaUnited States
| | - Jason Rihel
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| |
Collapse
|
19
|
Gerstner JR, Flores CC, Lefton M, Rogers B, Davis CJ. FABP7: a glial integrator of sleep, circadian rhythms, plasticity, and metabolic function. Front Syst Neurosci 2023; 17:1212213. [PMID: 37404868 PMCID: PMC10315501 DOI: 10.3389/fnsys.2023.1212213] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/02/2023] [Indexed: 07/06/2023] Open
Abstract
Sleep and circadian rhythms are observed broadly throughout animal phyla and influence neural plasticity and cognitive function. However, the few phylogenetically conserved cellular and molecular pathways that are implicated in these processes are largely focused on neuronal cells. Research on these topics has traditionally segregated sleep homeostatic behavior from circadian rest-activity rhythms. Here we posit an alternative perspective, whereby mechanisms underlying the integration of sleep and circadian rhythms that affect behavioral state, plasticity, and cognition reside within glial cells. The brain-type fatty acid binding protein, FABP7, is part of a larger family of lipid chaperone proteins that regulate the subcellular trafficking of fatty acids for a wide range of cellular functions, including gene expression, growth, survival, inflammation, and metabolism. FABP7 is enriched in glial cells of the central nervous system and has been shown to be a clock-controlled gene implicated in sleep/wake regulation and cognitive processing. FABP7 is known to affect gene transcription, cellular outgrowth, and its subcellular localization in the fine perisynaptic astrocytic processes (PAPs) varies based on time-of-day. Future studies determining the effects of FABP7 on behavioral state- and circadian-dependent plasticity and cognitive processes, in addition to functional consequences on cellular and molecular mechanisms related to neural-glial interactions, lipid storage, and blood brain barrier integrity will be important for our knowledge of basic sleep function. Given the comorbidity of sleep disturbance with neurological disorders, these studies will also be important for our understanding of the etiology and pathophysiology of how these diseases affect or are affected by sleep.
Collapse
Affiliation(s)
- Jason R. Gerstner
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Steve Gleason Institute for Neuroscience, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Carlos C. Flores
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Micah Lefton
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Brooke Rogers
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Christopher J. Davis
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Steve Gleason Institute for Neuroscience, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| |
Collapse
|
20
|
Shekhar S, Moehlman AT, Park B, Ewnetu M, Tracy C, Titos I, Pawłowski K, Tagliabracci VS, Krämer H. Allnighter pseudokinase-mediated feedback links proteostasis and sleep in Drosophila. Nat Commun 2023; 14:2932. [PMID: 37217484 DOI: 10.1038/s41467-023-38485-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 05/04/2023] [Indexed: 05/24/2023] Open
Abstract
In nervous systems, retrograde signals are key for organizing circuit activity and maintaining neuronal homeostasis. We identify the conserved Allnighter (Aln) pseudokinase as a cell non-autonomous regulator of proteostasis responses necessary for normal sleep and structural plasticity of Drosophila photoreceptors. In aln mutants exposed to extended ambient light, proteostasis is dysregulated and photoreceptors develop striking, but reversible, dysmorphology. The aln gene is widely expressed in different neurons, but not photoreceptors. However, secreted Aln protein is retrogradely endocytosed by photoreceptors. Inhibition of photoreceptor synaptic release reduces Aln levels in lamina neurons, consistent with secreted Aln acting in a feedback loop. In addition, aln mutants exhibit reduced night time sleep, providing a molecular link between dysregulated proteostasis and sleep, two characteristics of ageing and neurodegenerative diseases.
Collapse
Affiliation(s)
- Shashank Shekhar
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX; O'Donnell Brain Institute, Dallas, USA.
| | - Andrew T Moehlman
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX; O'Donnell Brain Institute, Dallas, USA
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Brenden Park
- Department of Molecular Biology UT Southwestern Medical Center, Dallas, TX, USA
| | - Michael Ewnetu
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX; O'Donnell Brain Institute, Dallas, USA
| | - Charles Tracy
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX; O'Donnell Brain Institute, Dallas, USA
| | - Iris Titos
- Molecular Medicine Program, University of Utah, School of Medicine, Salt Lake City, UT, USA
| | - Krzysztof Pawłowski
- Department of Molecular Biology UT Southwestern Medical Center, Dallas, TX, USA
- Department of Biochemistry and Microbiology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, 02-776, Poland
| | - Vincent S Tagliabracci
- Department of Molecular Biology UT Southwestern Medical Center, Dallas, TX, USA
- Howard Hughes Medical Institute, Maryland, USA
| | - Helmut Krämer
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX; O'Donnell Brain Institute, Dallas, USA.
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
21
|
Untiet V, Beinlich FRM, Kusk P, Kang N, Ladrón-de-Guevara A, Song W, Kjaerby C, Andersen M, Hauglund N, Bojarowska Z, Sigurdsson B, Deng S, Hirase H, Petersen NC, Verkhratsky A, Nedergaard M. Astrocytic chloride is brain state dependent and modulates inhibitory neurotransmission in mice. Nat Commun 2023; 14:1871. [PMID: 37015909 PMCID: PMC10073105 DOI: 10.1038/s41467-023-37433-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 03/15/2023] [Indexed: 04/06/2023] Open
Abstract
Information transfer within neuronal circuits depends on the balance and recurrent activity of excitatory and inhibitory neurotransmission. Chloride (Cl-) is the major central nervous system (CNS) anion mediating inhibitory neurotransmission. Astrocytes are key homoeostatic glial cells populating the CNS, although the role of these cells in regulating excitatory-inhibitory balance remains unexplored. Here we show that astrocytes act as a dynamic Cl- reservoir regulating Cl- homoeostasis in the CNS. We found that intracellular chloride concentration ([Cl-]i) in astrocytes is high and stable during sleep. In awake mice astrocytic [Cl-]i is lower and exhibits large fluctuation in response to both sensory input and motor activity. Optogenetic manipulation of astrocytic [Cl-]i directly modulates neuronal activity during locomotion or whisker stimulation. Astrocytes thus serve as a dynamic source of extracellular Cl- available for GABAergic transmission in awake mice, which represents a mechanism for modulation of the inhibitory tone during sustained neuronal activity.
Collapse
Affiliation(s)
- Verena Untiet
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark.
| | - Felix R M Beinlich
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Peter Kusk
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Ning Kang
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Antonio Ladrón-de-Guevara
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Wei Song
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Celia Kjaerby
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Mie Andersen
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Natalie Hauglund
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Zuzanna Bojarowska
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Björn Sigurdsson
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Saiyue Deng
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, P.R. China
| | - Hajime Hirase
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Nicolas C Petersen
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Alexei Verkhratsky
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark.
- Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Rd, Manchester, M13 9PL, UK.
- Achucarro Centre for Neuroscience, IKERBASQUE, Basque Foundation for Science, Plaza Euskadi 5, 48009, Bilbao, Spain.
| | - Maiken Nedergaard
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, 2200, Copenhagen, Denmark.
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| |
Collapse
|
22
|
Chu C, Holst SC, Elmenhorst EM, Foerges AL, Li C, Lange D, Hennecke E, Baur DM, Beer S, Hoffstaedter F, Knudsen GM, Aeschbach D, Bauer A, Landolt HP, Elmenhorst D. Total Sleep Deprivation Increases Brain Age Prediction Reversibly in Multisite Samples of Young Healthy Adults. J Neurosci 2023; 43:2168-2177. [PMID: 36804738 PMCID: PMC10039745 DOI: 10.1523/jneurosci.0790-22.2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 01/13/2023] [Accepted: 01/18/2023] [Indexed: 02/22/2023] Open
Abstract
Sleep loss pervasively affects the human brain at multiple levels. Age-related changes in several sleep characteristics indicate that reduced sleep quality is a frequent characteristic of aging. Conversely, sleep disruption may accelerate the aging process, yet it is not known what will happen to the age status of the brain if we can manipulate sleep conditions. To tackle this question, we used an approach of brain age to investigate whether sleep loss would cause age-related changes in the brain. We included MRI data of 134 healthy volunteers (mean chronological age of 25.3 between the age of 19 and 39 years, 42 females/92 males) from five datasets with different sleep conditions. Across three datasets with the condition of total sleep deprivation (>24 h of prolonged wakefulness), we consistently observed that total sleep deprivation increased brain age by 1-2 years regarding the group mean difference with the baseline. Interestingly, after one night of recovery sleep, brain age was not different from baseline. We also demonstrated the associations between the change in brain age after total sleep deprivation and the sleep variables measured during the recovery night. By contrast, brain age was not significantly changed by either acute (3 h time-in-bed for one night) or chronic partial sleep restriction (5 h time-in-bed for five continuous nights). Together, the convergent findings indicate that acute total sleep loss changes brain morphology in an aging-like direction in young participants and that these changes are reversible by recovery sleep.SIGNIFICANCE STATEMENT Sleep is fundamental for humans to maintain normal physical and psychological functions. Experimental sleep deprivation is a variable-controlling approach to engaging the brain among different sleep conditions for investigating the responses of the brain to sleep loss. Here, we quantified the response of the brain to sleep deprivation by using the change of brain age predictable with brain morphologic features. In three independent datasets, we consistently found increased brain age after total sleep deprivation, which was associated with the change in sleep variables. Moreover, no significant change in brain age was found after partial sleep deprivation in another two datasets. Our study provides new evidence to explain the brainwide effect of sleep loss in an aging-like direction.
Collapse
Affiliation(s)
- Congying Chu
- Institute of Neuroscience and Medicine (INM-2), Forschungszentrum Jülich, 52428 Jülich, Germany
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Sebastian C Holst
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
- Institute of Pharmacology and Toxicology, University of Zurich, CH-8006 Zurich, Switzerland
| | - Eva-Maria Elmenhorst
- Department of Sleep and Human Factors Research, Institute of Aerospace Medicine, German Aerospace Center, 51147 Cologne, Germany
- Institute for Occupational, Social and Environmental Medicine, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
| | - Anna L Foerges
- Institute of Neuroscience and Medicine (INM-2), Forschungszentrum Jülich, 52428 Jülich, Germany
- Department of Neurophysiology, Institute of Zoology (Bio-II), RWTH Aachen University, 52074 Aachen, Germany
| | - Changhong Li
- Institute of Neuroscience and Medicine (INM-2), Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Denise Lange
- Department of Sleep and Human Factors Research, Institute of Aerospace Medicine, German Aerospace Center, 51147 Cologne, Germany
| | - Eva Hennecke
- Department of Sleep and Human Factors Research, Institute of Aerospace Medicine, German Aerospace Center, 51147 Cologne, Germany
| | - Diego M Baur
- Institute of Pharmacology and Toxicology, University of Zurich, CH-8006 Zurich, Switzerland
| | - Simone Beer
- Institute of Neuroscience and Medicine (INM-2), Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Felix Hoffstaedter
- Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
- Institute of Neuroscience and Medicine, Brain and Behaviour (INM-7), Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Gitte M Knudsen
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
- Institute of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Daniel Aeschbach
- Department of Sleep and Human Factors Research, Institute of Aerospace Medicine, German Aerospace Center, 51147 Cologne, Germany
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, Massachusetts 02115
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts 02115
- Institute of Experimental Epileptology and Cognition Research, Faculty of Medicine, University of Bonn, 53127, Bonn, Germany
| | - Andreas Bauer
- Institute of Neuroscience and Medicine (INM-2), Forschungszentrum Jülich, 52428 Jülich, Germany
- Neurological Department, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Hans-Peter Landolt
- Institute of Pharmacology and Toxicology, University of Zurich, CH-8006 Zurich, Switzerland
- Sleep & Health Zurich, University Center of Competence, University of Zurich, Zurich, Switzerland
| | - David Elmenhorst
- Institute of Neuroscience and Medicine (INM-2), Forschungszentrum Jülich, 52428 Jülich, Germany
- Department of Nuclear Medicine, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
- Division of Medical Psychology, Rheinische Friedrich-Wilhelms-University Bonn, Bonn, 53127 Germany
| |
Collapse
|
23
|
Mocci I, Casu MA, Sogos V, Liscia A, Angius R, Cadeddu F, Fanti M, Muroni P, Talani G, Diana A, Collu M, Setzu MD. Effects of memantine on mania-like phenotypes exhibited by Drosophila Shaker mutants. CNS Neurosci Ther 2023. [PMID: 36942502 DOI: 10.1111/cns.14145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/17/2023] [Accepted: 02/19/2023] [Indexed: 03/23/2023] Open
Abstract
INTRODUCTION Increased glutamate levels and electrolytic fluctuations have been observed in acutely manic patients. Despite some efficacy of the non-competitive NMDA receptor antagonist memantine (Mem), such as antidepressant-like and mood-stabilizer drugs in clinical studies, its specific mechanisms of action are still uncertain. The present study aims to better characterize the Drosophila melanogaster fly Shaker mutants (SH), as a translational model of manic episodes within bipolar disorder in humans, and to investigate the potential anti-manic properties of Mem. METHODS AND RESULTS Our findings showed typical behavioral abnormalities in SH, which mirrored with the overexpression of NMDAR-NR1 protein subunit, matched well to glutamate up-regulation. Such molecular features were associated to a significant reduction of SH brain volume in comparison to Wild Type strain flies (WT). Here we report on the ability of Mem treatment to ameliorate behavioral aberrations of SH (similar to that of Lithium), and its ability to reduce NMDAR-NR1 over-expression. CONCLUSIONS Our results show the involvement of the glutamatergic system in the SH, given the interaction between the Shaker channel and the NMDA receptor, suggesting this model as a promising tool for studying the neurobiology of bipolar disorders. Moreover, our results show Mem as a potential disease-modifying therapy, providing insight on new mechanisms of action.
Collapse
Affiliation(s)
- Ignazia Mocci
- Institute of Translational Pharmacology, National Research Council, Science and Technology Park of Sardinia, Cagliari, Italy
| | - Maria Antonietta Casu
- Institute of Translational Pharmacology, National Research Council, Science and Technology Park of Sardinia, Cagliari, Italy
| | - Valeria Sogos
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Anna Liscia
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Rossella Angius
- Unit of Biomedical Research Support, NMR Laboratory and Bioanalytical Technologies, Sardegna Ricerche, Science and Technology Park of Sardinia, Cagliari, Italy
| | - Francesca Cadeddu
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Maura Fanti
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Patrizia Muroni
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Giuseppe Talani
- Institute of Neuroscience, National Research Council, Monserrato, Italy
| | - Andrea Diana
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Maria Collu
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Maria Dolores Setzu
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| |
Collapse
|
24
|
Mariano V, Kanellopoulos AK, Aiello G, Lo AC, Legius E, Achsel T, Bagni C. SREBP modulates the NADP +/NADPH cycle to control night sleep in Drosophila. Nat Commun 2023; 14:763. [PMID: 36808152 PMCID: PMC9941135 DOI: 10.1038/s41467-022-35577-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 12/12/2022] [Indexed: 02/22/2023] Open
Abstract
Sleep behavior is conserved throughout evolution, and sleep disturbances are a frequent comorbidity of neuropsychiatric disorders. However, the molecular basis underlying sleep dysfunctions in neurological diseases remains elusive. Using a model for neurodevelopmental disorders (NDDs), the Drosophila Cytoplasmic FMR1 interacting protein haploinsufficiency (Cyfip85.1/+), we identify a mechanism modulating sleep homeostasis. We show that increased activity of the sterol regulatory element-binding protein (SREBP) in Cyfip85.1/+ flies induces an increase in the transcription of wakefulness-associated genes, such as the malic enzyme (Men), causing a disturbance in the daily NADP+/NADPH ratio oscillations and reducing sleep pressure at the night-time onset. Reduction in SREBP or Men activity in Cyfip85.1/+ flies enhances the NADP+/NADPH ratio and rescues the sleep deficits, indicating that SREBP and Men are causative for the sleep deficits in Cyfip heterozygous flies. This work suggests modulation of the SREBP metabolic axis as a new avenue worth exploring for its therapeutic potential in sleep disorders.
Collapse
Affiliation(s)
- Vittoria Mariano
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, 1005, Switzerland.,Department of Human Genetics, KU Leuven, Leuven, 3000, Belgium
| | | | - Giuseppe Aiello
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, 1005, Switzerland
| | - Adrian C Lo
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, 1005, Switzerland
| | - Eric Legius
- Department of Human Genetics, KU Leuven, Leuven, 3000, Belgium
| | - Tilmann Achsel
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, 1005, Switzerland
| | - Claudia Bagni
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, 1005, Switzerland. .,Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, 00133, Italy.
| |
Collapse
|
25
|
Turner KL, Gheres KW, Drew PJ. Relating Pupil Diameter and Blinking to Cortical Activity and Hemodynamics across Arousal States. J Neurosci 2023; 43:949-964. [PMID: 36517240 PMCID: PMC9908322 DOI: 10.1523/jneurosci.1244-22.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Arousal state affects neural activity and vascular dynamics in the cortex, with sleep associated with large changes in the local field potential and increases in cortical blood flow. We investigated the relationship between pupil diameter and blink rate with neural activity and blood volume in the somatosensory cortex in male and female unanesthetized, head-fixed mice. We monitored these variables while the mice were awake, during periods of rapid eye movement (REM), and non-rapid eye movement (NREM) sleep. Pupil diameter was smaller during sleep than in the awake state. Changes in pupil diameter were coherent with both gamma-band power and blood volume in the somatosensory cortex, but the strength and sign of this relationship varied with arousal state. We observed a strong negative correlation between pupil diameter and both gamma-band power and blood volume during periods of awake rest and NREM sleep, although the correlations between pupil diameter and these signals became positive during periods of alertness, active whisking, and REM. Blinking was associated with increases in arousal and decreases in blood volume when the mouse was asleep. Bilateral coherence in gamma-band power and in blood volume dropped following awake blinking, indicating a reset of neural and vascular activity. Using only eye metrics (pupil diameter and eye motion), we could determine the arousal state of the mouse ('Awake,' 'NREM,' 'REM') with >90% accuracy with a 5 s resolution. There is a strong relationship between pupil diameter and hemodynamics signals in mice, reflecting the pronounced effects of arousal on cerebrovascular dynamics.SIGNIFICANCE STATEMENT Determining arousal state is a critical component of any neuroscience experiment. Pupil diameter and blinking are influenced by arousal state, as are hemodynamics signals in the cortex. We investigated the relationship between cortical hemodynamics and pupil diameter and found that pupil diameter was strongly related to the blood volume in the cortex. Mice were more likely to be awake after blinking than before, and blinking resets neural activity. Pupil diameter and eye motion can be used as a reliable, noninvasive indicator of arousal state. As mice transition from wake to sleep and back again over a timescale of seconds, monitoring pupil diameter and eye motion permits the noninvasive detection of sleep events during behavioral or resting-state experiments.
Collapse
Affiliation(s)
- Kevin L Turner
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802
- Center for Neural Engineering, Pennsylvania State University, University Park, Pennsylvania 16802
| | - Kyle W Gheres
- Center for Neural Engineering, Pennsylvania State University, University Park, Pennsylvania 16802
- Departments of Engineering Science and Mechanics
| | - Patrick J Drew
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802
- Center for Neural Engineering, Pennsylvania State University, University Park, Pennsylvania 16802
- Departments of Engineering Science and Mechanics
- Biology and Neurosurgery, Pennsylvania State University, University Park, Pennsylvania 16802
| |
Collapse
|
26
|
Lee YY, Endale M, Wu G, Ruben MD, Francey LJ, Morris AR, Choo NY, Anafi RC, Smith DF, Liu AC, Hogenesch JB. Integration of genome-scale data identifies candidate sleep regulators. Sleep 2023; 46:zsac279. [PMID: 36462188 PMCID: PMC9905783 DOI: 10.1093/sleep/zsac279] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/02/2022] [Indexed: 12/05/2022] Open
Abstract
STUDY OBJECTIVES Genetics impacts sleep, yet, the molecular mechanisms underlying sleep regulation remain elusive. In this study, we built machine learning models to predict sleep genes based on their similarity to genes that are known to regulate sleep. METHODS We trained a prediction model on thousands of published datasets, representing circadian, immune, sleep deprivation, and many other processes, using a manually curated list of 109 sleep genes. RESULTS Our predictions fit with prior knowledge of sleep regulation and identified key genes and pathways to pursue in follow-up studies. As an example, we focused on the NF-κB pathway and showed that chronic activation of NF-κB in a genetic mouse model impacted the sleep-wake patterns. CONCLUSION Our study highlights the power of machine learning in integrating prior knowledge and genome-wide data to study genetic regulation of complex behaviors such as sleep.
Collapse
Affiliation(s)
- Yin Yeng Lee
- Divisions of Human Genetics and Immunobiology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Mehari Endale
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Gang Wu
- Divisions of Human Genetics and Immunobiology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Marc D Ruben
- Divisions of Human Genetics and Immunobiology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Lauren J Francey
- Divisions of Human Genetics and Immunobiology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Andrew R Morris
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Natalie Y Choo
- Division of Pediatric Otolaryngology-Head and Neck Surgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Ron C Anafi
- Department of Medicine, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David F Smith
- Division of Pediatric Otolaryngology-Head and Neck Surgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Division of Pulmonary Medicine and the Sleep Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Center for Circadian Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Otolaryngology - Head and Neck Surgery, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Andrew C Liu
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - John B Hogenesch
- Divisions of Human Genetics and Immunobiology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
- Center for Circadian Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
27
|
Konakanchi S, Raavi V, Ml HK, Shankar Ms V. Impact of chronic sleep deprivation and sleep recovery on hippocampal oligodendrocytes, anxiety-like behavior, spatial learning and memory of rats. Brain Res Bull 2023; 193:59-71. [PMID: 36494056 DOI: 10.1016/j.brainresbull.2022.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 11/21/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022]
Abstract
Sleep and its quality play an important role in memory, cognition, and quality of life. Sleep deprivation-induced changes in hippocampal neurons and behavior have been studied widely, in contrast, the extent of damage to oligodendrocytes have not been fully understood. The present study aims to investigate chronic sleep deprivation (CSD) and sleep recovery-induced changes in oligodendrocytes of the hippocampus, cognition, and behavior of rats. Male Sprague-Dawley rats (n = 48) were grouped as control, sham control (SC), CSD, and CSD+sleep recovery (CSD+SR) (n = 12/group). CSD and CSD+SR group rats were sleep deprived for 21-days. After CSD, the CSD+SR group rats sleep recovered for 21-days. Oxidative markers, CNPase+ve oligodendrocytes, CNPase intensity, and CNPase gene expression were measured in the hippocampus, and the anxiety-like behavior, spatial learning, and memory were assessed. The 21-days of CSD significantly (p < 0.001) increased oxidative stress and significantly (p < 0.001) reduced the number of CNPase+ve oligodendrocytes, CNPase intensity, and CNPase gene expression when compared to controls. The increased oxidative stress was correlated with reduced CNPase+ve oligodendrocytes, CNPase intensity, and CNPase gene expression (r = -0.9). In-line with cellular changes, an increased (p < 0.01) anxiety-like behavior and impaired spatial memory were observed in the CSD group compared to controls. The 21-days of sleep recovery significantly (p < 0.01) reduced oxidative stress and anxiety-like behavior, improved spatial memory, increased CNPase intensity and CNPase gene expression, and non-significant (p > 0.05) increase in CNPase+ve oligodendrocytes compared to CSD. Overall, the 21-days of CSD reduced the number of CNPase+ve oligodendrocytes in the hippocampus, increased anxiety, and impaired spatial memory in rats. Though the 21-day sleep recovery showed an improvement in all parameters, it was not sufficient to completely reverse the CSD-induced changes to the control level.
Collapse
Affiliation(s)
- Suresh Konakanchi
- Department of Physiology, Sri Devaraj Urs Medical College, Sri Devaraj Urs Academy of Higher Education and Research (Deemed to be University), Kolar 563103, Karnataka, India.
| | - Venkateswarlu Raavi
- Department of Cell Biology and Molecular Genetics, Sri Devaraj Urs Academy of Higher Education and Research (Deemed to be University), Kolar 563103, Karnataka, India.
| | - Harendra Kumar Ml
- Department of Pathology, Sri Devaraj Urs Medical College, Sri Devaraj Urs Academy of Higher Education and Research (Deemed to be University), Kolar 563103, Karnataka, India.
| | - Vinutha Shankar Ms
- Department of Physiology, Sri Devaraj Urs Medical College, Sri Devaraj Urs Academy of Higher Education and Research (Deemed to be University), Kolar 563103, Karnataka, India.
| |
Collapse
|
28
|
Lane JM, Qian J, Mignot E, Redline S, Scheer FAJL, Saxena R. Genetics of circadian rhythms and sleep in human health and disease. Nat Rev Genet 2023; 24:4-20. [PMID: 36028773 PMCID: PMC10947799 DOI: 10.1038/s41576-022-00519-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2022] [Indexed: 12/13/2022]
Abstract
Circadian rhythms and sleep are fundamental biological processes integral to human health. Their disruption is associated with detrimental physiological consequences, including cognitive, metabolic, cardiovascular and immunological dysfunctions. Yet many of the molecular underpinnings of sleep regulation in health and disease have remained elusive. Given the moderate heritability of circadian and sleep traits, genetics offers an opportunity that complements insights from model organism studies to advance our fundamental molecular understanding of human circadian and sleep physiology and linked chronic disease biology. Here, we review recent discoveries of the genetics of circadian and sleep physiology and disorders with a focus on those that reveal causal contributions to complex diseases.
Collapse
Affiliation(s)
- Jacqueline M Lane
- Center for Genomic Medicine and Department of Anaesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital; and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Jingyi Qian
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital; and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Emmanuel Mignot
- Center for Narcolepsy, Stanford University, Palo Alto, California, USA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital; and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - 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, MA, USA.
| | - Richa Saxena
- Center for Genomic Medicine and Department of Anaesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital; and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA.
| |
Collapse
|
29
|
King AL, Shuboni-Mulligan DD, Vera E, Crandon S, Acquaye AA, Boris L, Burton E, Choi A, Christ A, Grajkowska E, Jammula V, Leeper HE, Lollo N, Penas-Prado M, Reyes J, Theeler B, Wall K, Wu J, Gilbert MR, Armstrong TS. Exploring the prevalence and burden of sleep disturbance in primary brain tumor patients. Neurooncol Pract 2022; 9:526-535. [PMID: 36388423 PMCID: PMC9665069 DOI: 10.1093/nop/npac049] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023] Open
Abstract
BACKGROUND Sleep disturbance (SD) is common in patients with cancer and has been associated with worse clinical outcomes. This cross-sectional study explored the prevalence of SD in a primary brain tumor (PBT) population, identified associated demographic and clinical characteristics, and investigated co-occurrence of SD with other symptoms and mood disturbance. METHODS Demographic, clinical characteristics, MD Anderson Symptom Inventory-Brain Tumor, and Patient Reported Outcome Measurement Information System Depression and Anxiety Short-Forms were collected from PBT patients at study entry. Descriptive statistics, Chi-square tests, and independent t-tests were used to report results. RESULTS The sample included 424 patients (58% male, 81% Caucasian) with a mean age of 49 years (range 18-81) and 58% with high-grade gliomas. Moderate-severe SD was reported in 19% of patients and was associated with younger age, poor Karnofsky Performance Status, tumor progression on MRI, and active corticosteroid use. Those with moderate-severe SD had higher overall symptom burden and reported more moderate-severe symptoms. These individuals also reported higher severity in affective and mood disturbance domains, with 3 to 4 times higher prevalence of depressive and anxiety symptoms, respectively. The most frequently co-occurring symptoms with SD were, drowsiness, and distress, though other symptoms typically associated with tumor progression also frequently co-occurred. CONCLUSIONS PBT patients with moderate-severe SD are more symptomatic, have worse mood disturbance, and have several co-occurring symptoms. Targeting interventions for sleep could potentially alleviate other co-occurring symptoms, which may improve life quality for PBT patients. Future longitudinal work examining objective and detailed subjective sleep reports, as well as underlying genetic risk factors, will be important.
Collapse
Affiliation(s)
- Amanda L King
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Dorela D Shuboni-Mulligan
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Elizabeth Vera
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Sonja Crandon
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Alvina A Acquaye
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Lisa Boris
- Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland, USA
| | - Eric Burton
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Anna Choi
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Alexa Christ
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ewa Grajkowska
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Varna Jammula
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Heather E Leeper
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Nicole Lollo
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Marta Penas-Prado
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jennifer Reyes
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Brett Theeler
- Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Kathleen Wall
- Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland, USA
| | - Jing Wu
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Terri S Armstrong
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
30
|
Kinase signalling in excitatory neurons regulates sleep quantity and depth. Nature 2022; 612:512-518. [PMID: 36477539 DOI: 10.1038/s41586-022-05450-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/14/2022] [Indexed: 12/12/2022]
Abstract
Progress has been made in the elucidation of sleep and wakefulness regulation at the neurocircuit level1,2. However, the intracellular signalling pathways that regulate sleep and the neuron groups in which these intracellular mechanisms work remain largely unknown. Here, using a forward genetics approach in mice, we identify histone deacetylase 4 (HDAC4) as a sleep-regulating molecule. Haploinsufficiency of Hdac4, a substrate of salt-inducible kinase 3 (SIK3)3, increased sleep. By contrast, mice that lacked SIK3 or its upstream kinase LKB1 in neurons or with a Hdac4S245A mutation that confers resistance to phosphorylation by SIK3 showed decreased sleep. These findings indicate that LKB1-SIK3-HDAC4 constitute a signalling cascade that regulates sleep and wakefulness. We also performed targeted manipulation of SIK3 and HDAC4 in specific neurons and brain regions. This showed that SIK3 signalling in excitatory neurons located in the cerebral cortex and the hypothalamus positively regulates EEG delta power during non-rapid eye movement sleep (NREMS) and NREMS amount, respectively. A subset of transcripts biased towards synaptic functions was commonly regulated in cortical glutamatergic neurons through the expression of a gain-of-function allele of Sik3 and through sleep deprivation. These findings suggest that NREMS quantity and depth are regulated by distinct groups of excitatory neurons through common intracellular signals. This study provides a basis for linking intracellular events and circuit-level mechanisms that control NREMS.
Collapse
|
31
|
Zheng L, Zhang L. The molecular mechanism of natural short sleep: A path towards understanding why we need to sleep. BRAIN SCIENCE ADVANCES 2022. [DOI: 10.26599/bsa.2022.9050003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Sleep constitutes a third of human life and it is increasingly recognized as important for health. Over the past several decades, numerous genes have been identified to be involved in sleep regulation in animal models, but most of these genes when disturbed impair not only sleep but also health and physiological functions. Human natural short sleepers are individuals with lifelong short sleep and no obvious adverse outcomes associated with the lack of sleep. These traits appear to be heritable, and thus characterization of the genetic basis of natural short sleep provides an opportunity to study not only the genetic mechanism of human sleep but also the relationship between sleep and physiological function. This review focuses on the current understanding of mutations associated with the natural short sleep trait and the mechanisms by which they contribute to this trait.
Collapse
Affiliation(s)
- Liubin Zheng
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - Luoying Zhang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, Hubei, China
| |
Collapse
|
32
|
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
|
33
|
Liu Z, Jiang L, Li C, Li C, Yang J, Yu J, Mao R, Rao Y. LKB1 Is Physiologically Required for Sleep from Drosophila melanogaster to the Mus musculus. Genetics 2022; 221:6586797. [PMID: 35579349 DOI: 10.1093/genetics/iyac082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/10/2022] [Indexed: 11/14/2022] Open
Abstract
Liver Kinase B1 (LKB1) is known as a master kinase for 14 kinases related to the adenosine monophosphate (AMP)-activated protein kinase (AMPK). Two of them salt inducible kinase 3 (SIK3) and AMPKα have previously been implicated in sleep regulation. We generated loss-of-function (LOF) mutants for Lkb1 in both Drosophila and mice. Sleep, but not circadian rhythms, was reduced in Lkb1-mutant flies and in flies with neuronal deletion of Lkb1. Genetic interactions between Lkb1 and Threonine to Alanine mutation at residue 184 of AMPK in Drosophila sleep or those between Lkb1 and Threonine to Glutamic Acid mutation at residue 196 of SIK3 in Drosophila viability have been observed. Sleep was reduced in mice after virally mediated reduction of Lkb1 in the brain. Electroencephalography (EEG) analysis showed that non-rapid eye movement (NREM) sleep and sleep need were both reduced in Lkb1-mutant mice. These results indicate that LKB1 plays a physiological role in sleep regulation conserved from flies to mice.
Collapse
Affiliation(s)
- Ziyi Liu
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, PKU-IDG/McGovern Institute for Brain Research, School of Chemistry and Molecular Engineering, School of Pharmaceutical Sciences, Peking University, Beijing 100871, China
- Chinese Institute for Brain Research, Beijing, China
- Capital Medical University, Beijing, China
- Changping Laboratory, Beijing, China
| | - Lifen Jiang
- Shenzhen Bay Laboratory, Institute of Molecular Physiology, Shenzhen, Guangdong, China
| | - Chaoyi Li
- Shenzhen Bay Laboratory, Institute of Molecular Physiology, Shenzhen, Guangdong, China
| | - Chengang Li
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, PKU-IDG/McGovern Institute for Brain Research, School of Chemistry and Molecular Engineering, School of Pharmaceutical Sciences, Peking University, Beijing 100871, China
- Chinese Institute for Brain Research, Beijing, China
- Capital Medical University, Beijing, China
- Changping Laboratory, Beijing, China
| | - Jingqun Yang
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, PKU-IDG/McGovern Institute for Brain Research, School of Chemistry and Molecular Engineering, School of Pharmaceutical Sciences, Peking University, Beijing 100871, China
- Chinese Institute for Brain Research, Beijing, China
- Capital Medical University, Beijing, China
- Changping Laboratory, Beijing, China
| | - Jianjun Yu
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, PKU-IDG/McGovern Institute for Brain Research, School of Chemistry and Molecular Engineering, School of Pharmaceutical Sciences, Peking University, Beijing 100871, China
- Chinese Institute for Brain Research, Beijing, China
- Capital Medical University, Beijing, China
- Changping Laboratory, Beijing, China
| | - Renbo Mao
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, PKU-IDG/McGovern Institute for Brain Research, School of Chemistry and Molecular Engineering, School of Pharmaceutical Sciences, Peking University, Beijing 100871, China
- Chinese Institute for Brain Research, Beijing, China
- Capital Medical University, Beijing, China
- Changping Laboratory, Beijing, China
| | - Yi Rao
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, PKU-IDG/McGovern Institute for Brain Research, School of Chemistry and Molecular Engineering, School of Pharmaceutical Sciences, Peking University, Beijing 100871, China
- Chinese Institute for Brain Research, Beijing, China
- Capital Medical University, Beijing, China
- Changping Laboratory, Beijing, China
| |
Collapse
|
34
|
Sheardown E, Mech AM, Petrazzini MEM, Leggieri A, Gidziela A, Hosseinian S, Sealy IM, Torres-Perez JV, Busch-Nentwich EM, Malanchini M, Brennan CH. Translational relevance of forward genetic screens in animal models for the study of psychiatric disease. Neurosci Biobehav Rev 2022; 135:104559. [PMID: 35124155 PMCID: PMC9016269 DOI: 10.1016/j.neubiorev.2022.104559] [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: 07/08/2021] [Revised: 12/10/2021] [Accepted: 02/01/2022] [Indexed: 12/16/2022]
Abstract
Psychiatric disorders represent a significant burden in our societies. Despite the convincing evidence pointing at gene and gene-environment interaction contributions, the role of genetics in the etiology of psychiatric disease is still poorly understood. Forward genetic screens in animal models have helped elucidate causal links. Here we discuss the application of mutagenesis-based forward genetic approaches in common animal model species: two invertebrates, nematodes (Caenorhabditis elegans) and fruit flies (Drosophila sp.); and two vertebrates, zebrafish (Danio rerio) and mice (Mus musculus), in relation to psychiatric disease. We also discuss the use of large scale genomic studies in human populations. Despite the advances using data from human populations, animal models coupled with next-generation sequencing strategies are still needed. Although with its own limitations, zebrafish possess characteristics that make them especially well-suited to forward genetic studies exploring the etiology of psychiatric disorders.
Collapse
Affiliation(s)
- Eva Sheardown
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Aleksandra M Mech
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | | | - Adele Leggieri
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Agnieszka Gidziela
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Saeedeh Hosseinian
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Ian M Sealy
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK
| | - Jose V Torres-Perez
- UK Dementia Research Institute at Imperial College London and Department of Brain Sciences, Imperial College London, 86 Wood Lane, London W12 0BZ, UK
| | - Elisabeth M Busch-Nentwich
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Margherita Malanchini
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Caroline H Brennan
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK.
| |
Collapse
|
35
|
Wang J, Fan JY, Zhao Z, Dissel S, Price J. DBT affects sleep in both circadian and non-circadian neurons. PLoS Genet 2022; 18:e1010035. [PMID: 35139068 PMCID: PMC8827452 DOI: 10.1371/journal.pgen.1010035] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 01/12/2022] [Indexed: 11/23/2022] Open
Abstract
Sleep is a very important behavior observed in almost all animals. Importantly, sleep is subject to both circadian and homeostatic regulation. The circadian rhythm determines the daily alternation of the sleep-wake cycle, while homeostasis mediates the rise and dissipation of sleep pressure during the wake and sleep period. As an important kinase, dbt plays a central role in both circadian rhythms and development. We investigated the sleep patterns of several ethyl methanesulfonate-induced dbt mutants and discuss the possible reasons why different sleep phenotypes were shown in these mutants. In order to reduce DBT in all neurons in which it is expressed, CRISPR-Cas9 was used to produce flies that expressed GAL4 in frame with the dbt gene at its endogenous locus, and knock-down of DBT with this construct produced elevated sleep during the day and reduced sleep at night. Loss of sleep at night is mediated by dbt loss during the sleep/wake cycle in the adult, while the increased sleep during the day is produced by reductions in dbt during development and not by reductions in the adult. Additionally, using targeted RNA interference, we uncovered the contribution of dbt on sleep in different subsets of neurons in which dbt is normally expressed. Reduction of dbt in circadian neurons produced less sleep at night, while lower expression of dbt in noncircadian neurons produced increased sleep during the day. Importantly, independently of the types of neurons where dbt affects sleep, we demonstrate that the PER protein is involved in DBT mediated sleep regulation. Doubletime (dbt) is known as a kinase orthologous to mammalian Casein Kinase I ε (CKIε) and Casein Kinase I δ (CKIδ), which are involved in various biological processes and play an important role in regulation of circadian rhythm. In this study, we first analyzed the role of dbt on sleep in Drosophila, and then mapped its expression pattern and further neuronal mechanisms, in which DBT importantly regulates sleep through PER in both non-clock neurons and clock neurons.
Collapse
Affiliation(s)
- Jialin Wang
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Jin-Yuan Fan
- School of Biological and Chemical Sciences, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Zhangwu Zhao
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
- * E-mail: (ZZ); (SD); (JP)
| | - Stephane Dissel
- School of Biological and Chemical Sciences, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
- * E-mail: (ZZ); (SD); (JP)
| | - Jeffrey Price
- School of Biological and Chemical Sciences, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
- * E-mail: (ZZ); (SD); (JP)
| |
Collapse
|
36
|
Crislip GR, Johnston JG, Douma LG, Costello HM, Juffre A, Boyd K, Li W, Maugans CC, Gutierrez-Monreal M, Esser KA, Bryant AJ, Liu AC, Gumz ML. Circadian Rhythm Effects on the Molecular Regulation of Physiological Systems. Compr Physiol 2021; 12:2769-2798. [PMID: 34964116 PMCID: PMC11514412 DOI: 10.1002/cphy.c210011] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Nearly every system within the body contains an intrinsic cellular circadian clock. The circadian clock contributes to the regulation of a variety of homeostatic processes in mammals through the regulation of gene expression. Circadian disruption of physiological systems is associated with pathophysiological disorders. Here, we review the current understanding of the molecular mechanisms contributing to the known circadian rhythms in physiological function. This article focuses on what is known in humans, along with discoveries made with cell and rodent models. In particular, the impact of circadian clock components in metabolic, cardiovascular, endocrine, musculoskeletal, immune, and central nervous systems are discussed. © 2021 American Physiological Society. Compr Physiol 11:1-30, 2021.
Collapse
Affiliation(s)
- G. Ryan Crislip
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation
| | - Jermaine G. Johnston
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation
| | | | - Hannah M. Costello
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation
| | | | - Kyla Boyd
- Department of Biochemistry and Molecular Biology
| | - Wendy Li
- Department of Biochemistry and Molecular Biology
| | | | | | - Karyn A. Esser
- Department of Physiology and Functional Genomics
- Myology Institute
| | | | - Andrew C. Liu
- Department of Physiology and Functional Genomics
- Myology Institute
| | - Michelle L. Gumz
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation
- Department of Biochemistry and Molecular Biology
- Department of Physiology and Functional Genomics
- Center for Integrative Cardiovascular and Metabolic Disease
| |
Collapse
|
37
|
Shola-Dare O, Bailess S, Flores CC, Vanderheyden WM, Gerstner JR. Glitazone Treatment Rescues Phenotypic Deficits in a Fly Model of Gaucher/Parkinson's Disease. Int J Mol Sci 2021; 22:ijms222312740. [PMID: 34884544 PMCID: PMC8657993 DOI: 10.3390/ijms222312740] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/19/2021] [Accepted: 11/23/2021] [Indexed: 12/14/2022] Open
Abstract
Parkinson’s Disease (PD) is the most common movement disorder, and the strongest genetic risk factor for PD is mutations in the glucocerebrosidase gene (GBA). Mutations in GBA also lead to the development of Gaucher Disease (GD), the most common type of lysosomal storage disorder. Current therapeutic approaches fail to address neurological GD symptoms. Therefore, identifying therapeutic strategies that improve the phenotypic traits associated with GD/PD in animal models may provide an opportunity for treating neurological manifestations of GD/PD. Thiazolidinediones (TZDs, also called glitazones) are a class of compounds targeted for the treatment of type 2 diabetes, and have also shown promise for the treatment of neurodegenerative disease, including PD. Here, we tested the efficacy of glitazone administration during development in a fly GD model with deletions in the GBA homolog, dGBA1b (GBA1ΔTT/ΔTT). We observed an optimal dose of pioglitazone (PGZ) at a concentration of 1 μM that reduced sleep deficits, locomotor impairments, climbing defects, and restoration of normal protein levels of Ref(2)P, a marker of autophagic flux, in GBA1ΔTT/ΔTT mutant flies, compared to GBA1+/+ control flies. These data suggest that PGZ may represent a potential compound with which to treat GD/PD by improving function of lysosomal-autophagy pathways, a cellular process that removes misfolded or aggregated proteins.
Collapse
Affiliation(s)
- Oluwanifemi Shola-Dare
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (O.S.-D.); (S.B.); (C.C.F.); (W.M.V.)
| | - Shelby Bailess
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (O.S.-D.); (S.B.); (C.C.F.); (W.M.V.)
| | - Carlos C. Flores
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (O.S.-D.); (S.B.); (C.C.F.); (W.M.V.)
| | - William M. Vanderheyden
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (O.S.-D.); (S.B.); (C.C.F.); (W.M.V.)
| | - Jason R. Gerstner
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (O.S.-D.); (S.B.); (C.C.F.); (W.M.V.)
- Steve Gleason Institute for Neuroscience, Washington State University, Spokane, WA 99202, USA
- Correspondence:
| |
Collapse
|
38
|
Van De Poll MN, van Swinderen B. Balancing Prediction and Surprise: A Role for Active Sleep at the Dawn of Consciousness? Front Syst Neurosci 2021; 15:768762. [PMID: 34803618 PMCID: PMC8602873 DOI: 10.3389/fnsys.2021.768762] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/08/2021] [Indexed: 11/14/2022] Open
Abstract
The brain is a prediction machine. Yet the world is never entirely predictable, for any animal. Unexpected events are surprising, and this typically evokes prediction error signatures in mammalian brains. In humans such mismatched expectations are often associated with an emotional response as well, and emotional dysregulation can lead to cognitive disorders such as depression or schizophrenia. Emotional responses are understood to be important for memory consolidation, suggesting that positive or negative 'valence' cues more generally constitute an ancient mechanism designed to potently refine and generalize internal models of the world and thereby minimize prediction errors. On the other hand, abolishing error detection and surprise entirely (as could happen by generalization or habituation) is probably maladaptive, as this might undermine the very mechanism that brains use to become better prediction machines. This paradoxical view of brain function as an ongoing balance between prediction and surprise suggests a compelling approach to study and understand the evolution of consciousness in animals. In particular, this view may provide insight into the function and evolution of 'active' sleep. Here, we propose that active sleep - when animals are behaviorally asleep but their brain seems awake - is widespread beyond mammals and birds, and may have evolved as a mechanism for optimizing predictive processing in motile creatures confronted with constantly changing environments. To explore our hypothesis, we progress from humans to invertebrates, investigating how a potential role for rapid eye movement (REM) sleep in emotional regulation in humans could be re-examined as a conserved sleep function that co-evolved alongside selective attention to maintain an adaptive balance between prediction and surprise. This view of active sleep has some interesting implications for the evolution of subjective awareness and consciousness in animals.
Collapse
Affiliation(s)
| | - Bruno van Swinderen
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
39
|
Choi J, Kim SJ, Fujiyama T, Miyoshi C, Park M, Suzuki-Abe H, Yanagisawa M, Funato H. The Role of Reproductive Hormones in Sex Differences in Sleep Homeostasis and Arousal Response in Mice. Front Neurosci 2021; 15:739236. [PMID: 34621154 PMCID: PMC8491770 DOI: 10.3389/fnins.2021.739236] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 08/19/2021] [Indexed: 11/13/2022] Open
Abstract
There are various sex differences in sleep/wake behaviors in mice. However, it is unclear whether there are sex differences in sleep homeostasis and arousal responses and whether gonadal hormones are involved in these sex differences. Here, we examined sleep/wake behaviors under baseline condition, after sleep deprivation by gentle handling, and arousal responses to repeated cage changes in male and female C57BL/6 mice that are hormonally intact, gonadectomized, or gonadectomized with hormone supplementation. Compared to males, females had longer wake time, shorter non-rapid eye movement sleep (NREMS) time, and longer rapid eye movement sleep (REMS) episodes. After sleep deprivation, males showed an increase in NREMS delta power, NREMS time, and REMS time, but females showed a smaller increase. Females and males showed similar arousal responses. Gonadectomy had only a modest effect on homeostatic sleep regulation in males but enhanced it in females. Gonadectomy weakened arousal response in males and females. With hormone replacement, baseline sleep in gonadectomized females was similar to that of intact females, and baseline sleep in gonadectomized males was close to that of intact males. Gonadal hormone supplementation restored arousal response in males but not in females. These results indicate that male and female mice differ in their baseline sleep-wake behavior, homeostatic sleep regulation, and arousal responses to external stimuli, which are differentially affected by reproductive hormones.
Collapse
Affiliation(s)
- Jinhwan Choi
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Japan
| | - Staci J Kim
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Japan
| | - Tomoyuki Fujiyama
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Japan
| | - Chika Miyoshi
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Japan
| | - Minjeong Park
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Japan
| | - Haruka Suzuki-Abe
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Japan.,Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Japan
| | - Hiromasa Funato
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Japan.,Department of Anatomy, Faculty of Medicine, Toho University, Tokyo, Japan
| |
Collapse
|
40
|
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
|
41
|
Dai X, Zhou E, Yang W, Mao R, Zhang W, Rao Y. Molecular resolution of a behavioral paradox: sleep and arousal are regulated by distinct acetylcholine receptors in different neuronal types in Drosophila. Sleep 2021; 44:6119684. [PMID: 33493349 DOI: 10.1093/sleep/zsab017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 12/21/2020] [Indexed: 02/02/2023] Open
Abstract
Sleep and arousal are both important for animals. The neurotransmitter acetylcholine (ACh) has long been found to promote both sleep and arousal in mammals, an apparent paradox which has also been found to exist in flies, causing much confusion in understanding sleep and arousal. Here, we have systematically studied all 13 ACh receptors (AChRs) in Drosophila to understand mechanisms underlying ACh function in sleep and arousal. We found that exogenous stimuli-induced arousal was decreased in nAChRα3 mutants, whereas sleep was decreased in nAChRα2 and nAChRβ2 mutants. nAChRα3 functions in dopaminergic neurons to promote exogenous stimuli-induced arousal, whereas nAChRα2 and β2 function in octopaminergic neurons to promote sleep. Our studies have revealed that a single transmitter can promote endogenous sleep and exogenous stimuli-induced arousal through distinct receptors in different types of downstream neurons.
Collapse
Affiliation(s)
- Xihuimin Dai
- PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Beijing Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Beijing, China.,Chinese Institute for Brain Research, Beijing, China.,Howard Hughes Medical Institute, Department of Biology, Brandeis University, Waltham, MA
| | - Enxing Zhou
- PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Beijing Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Beijing, China
| | - Wei Yang
- PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Beijing Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Beijing, China.,Chinese Institute for Brain Research, Beijing, China
| | - Renbo Mao
- Graduate School of Peking Union Medical College, Beijing, China.,National Institute of Biological Sciences, Beijing, China
| | - Wenxia Zhang
- PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Beijing Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Beijing, China
| | - Yi Rao
- PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Beijing Advanced Innovation Center for Genomics, Peking University School of Life Sciences, Beijing, China.,Chinese Institute for Brain Research, Beijing, China
| |
Collapse
|
42
|
Sleep in disorders of consciousness: diagnostic, prognostic, and therapeutic considerations. Curr Opin Neurol 2021; 33:684-690. [PMID: 33177374 DOI: 10.1097/wco.0000000000000870] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
PURPOSE OF REVIEW Sleep is important in the evaluation of patients with disorders of consciousness (DOC). However, it remains unclear whether reconstitution of sleep could enable consciousness or vice versa. Here we synthesize recent evidence on natural recovery of sleep in DOC, and sleep-promoting therapeutic interventions for recovery of consciousness. RECENT FINDINGS In subacute DOC, physiological sleep--wake cycles and complex sleep patterns are related to better outcomes. Moreover, structured rapid-eye-movement (REM), non-REM (NREM) stages, and presence of sleep spindles correlate with full or partial recovery. In chronic DOC, sleep organization may reflect both integrity of consciousness-supporting brain networks and engagement of those networks during wakefulness. Therapeutic strategies have integrated improvement of sleep and sleep--wake cycles in DOC patients; use of bright light stimulation or drugs enhancing sleep and/or vigilance, treatment of sleep apneas, and neuromodulatory stimulations are promising tools to promote healthy sleep architecture and wakeful recovery. SUMMARY Sleep features and sleep--wake cycles are important prognostic markers in subacute DOC and can provide insight into covert recovery in chronic DOC. Although large-scale studies are needed, preliminary studies in limited patients suggest that therapeutic options restoring sleep and/or sleep--wake cycles may improve cognitive function and outcomes in DOC.
Collapse
|
43
|
Huber R, Jacobson DA. The Phylogenetic Roots of Addiction: Compulsive Drug Seeking, Natural and Drug-Sensitive Reward, and the Acquisition of Learned Habits. BRAIN, BEHAVIOR AND EVOLUTION 2021; 95:217-221. [PMID: 34082419 DOI: 10.1159/000517121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 05/06/2021] [Indexed: 11/19/2022]
Affiliation(s)
- Robert Huber
- Department of Biological Sciences, Bowling Green State University, Bowling Green, Ohio, USA
| | - Daniel A Jacobson
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee, USA.,Department of Psychology, University of Tennessee-Knoxville, Knoxville, Tennessee, USA
| |
Collapse
|
44
|
Jin X, Tian Y, Zhang ZC, Gu P, Liu C, Han J. A subset of DN1p neurons integrates thermosensory inputs to promote wakefulness via CNMa signaling. Curr Biol 2021; 31:2075-2087.e6. [PMID: 33740429 DOI: 10.1016/j.cub.2021.02.048] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 12/15/2020] [Accepted: 02/17/2021] [Indexed: 11/29/2022]
Abstract
Sleep is an essential and evolutionarily conserved behavior that is modulated by many environmental factors. Ambient temperature shifting usually occurs during climatic or seasonal change or travel from high-latitude area to low-latitude area that affects animal physiology. Increasing ambient temperature modulates sleep in both humans and Drosophila. Although several thermosensory molecules and neurons have been identified, the neural mechanisms that integrate temperature sensation into the sleep neural circuit remain poorly understood. Here, we reveal that prolonged increasing of ambient temperature induces a reversible sleep reduction and impaired sleep consolidation in Drosophila via activating the internal thermosensory anterior cells (ACs). ACs form synaptic contacts with a subset of posterior dorsal neuron 1 (DN1p) neurons and release acetylcholine to promote wakefulness. Furthermore, we identify that this subset of DN1ps promotes wakefulness by releasing CNMamide (CNMa) neuropeptides to inhibit the Dh44-positive pars intercerebralis (PI) neurons through CNMa receptors. Our study demonstrates that the AC-DN1p-PI neural circuit is responsible for integrating thermosensory inputs into the sleep neural circuit. Moreover, we identify the CNMa signaling pathway as a newly recognized wakefulness-promoting DN1 pathway.
Collapse
Affiliation(s)
- Xi Jin
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, 2 Sipailou Road, Nanjing 210096, China
| | - Yao Tian
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, 2 Sipailou Road, Nanjing 210096, China
| | - Zi Chao Zhang
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, 2 Sipailou Road, Nanjing 210096, China
| | - Pengyu Gu
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, 2 Sipailou Road, Nanjing 210096, China
| | - Chang Liu
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Junhai Han
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, 2 Sipailou Road, Nanjing 210096, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226021, China.
| |
Collapse
|
45
|
Keenan BT, Galante RJ, Lian J, Simecek P, Gatti DM, Zhang L, Lim DC, Svenson KL, Churchill GA, Pack AI. High-throughput sleep phenotyping produces robust and heritable traits in Diversity Outbred mice and their founder strains. Sleep 2021; 43:5740842. [PMID: 32074270 DOI: 10.1093/sleep/zsz278] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/25/2019] [Indexed: 12/14/2022] Open
Abstract
STUDY OBJECTIVES This study describes high-throughput phenotyping strategies for sleep and circadian behavior in mice, including examinations of robustness, reliability, and heritability among Diversity Outbred (DO) mice and their eight founder strains. METHODS We performed high-throughput sleep and circadian phenotyping in male mice from the DO population (n = 338) and their eight founder strains: A/J (n = 6), C57BL/6J (n = 14), 129S1/SvlmJ (n = 6), NOD/LtJ (n = 6), NZO/H1LtJ (n = 6), CAST/EiJ (n = 8), PWK/PhJ (n = 8), and WSB/EiJ (n = 6). Using infrared beam break systems, we defined sleep as at least 40 s of continuous inactivity and quantified sleep-wake amounts and bout characteristics. We developed assays to measure sleep latency in a new environment and during a modified Murine Multiple Sleep Latency Test, and estimated circadian period from wheel-running experiments. For each trait, broad-sense heritability (proportion of variability explained by all genetic factors) was derived in founder strains, while narrow-sense heritability (proportion of variability explained by additive genetic effects) was calculated in DO mice. RESULTS Phenotypes were robust to different inactivity durations to define sleep. Differences across founder strains and moderate/high broad-sense heritability were observed for most traits. There was large phenotypic variability among DO mice, and phenotypes were reliable, although estimates of heritability were lower than in founder mice. This likely reflects important nonadditive genetic effects. CONCLUSIONS A high-throughput phenotyping strategy in mice, based primarily on monitoring of activity patterns, provides reliable and heritable estimates of sleep and circadian traits. This approach is suitable for discovery analyses in DO mice, where genetic factors explain some proportion of phenotypic variation.
Collapse
Affiliation(s)
- Brendan T Keenan
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Raymond J Galante
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jie Lian
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Petr Simecek
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic.,Jackson Laboratory, Bar Harbor, ME
| | | | - Lin Zhang
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Diane C Lim
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | | | | | - Allan I Pack
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
46
|
Panchin Y, Kovalzon VM. Total Wake: Natural, Pathological, and Experimental Limits to Sleep Reduction. Front Neurosci 2021; 15:643496. [PMID: 33897357 PMCID: PMC8058214 DOI: 10.3389/fnins.2021.643496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/26/2021] [Indexed: 11/16/2022] Open
Abstract
Sleep is not considered a pathological state, but it consumes a third of conscious human life. This share is much more than most optimistic life extension forecasts that biotechnologies or experimental and medical interventions can offer. Are there insurmountable physical or biological limitations to reducing the duration of sleep? How far can it be avoided without fatal consequences? What means can reduce the length of sleep? It is widely accepted that sleep is necessary for long-term survival. Here we review the limited yet intriguing evidence that is not consistent with this notion. We concentrate on clinical cases of complete and partial loss of sleep and on human mutations that result in a short sleep phenotype. These observations are supported by new animal studies and are discussed from the perspective of sleep evolution. Two separate hypotheses suggest distinct approaches for remodeling our sleep machinery. If sleep serves an unidentified vital physiological function, this indispensable function has to be identified before "sleep prosthesis" (technical, biological, or chemical) can be developed. If sleep has no vital function, but rather represents a timing mechanism for adaptive inactivity, sleep could be reduced by forging the sleep generation system itself, with no adverse effects.
Collapse
Affiliation(s)
- Yuri Panchin
- Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia
- Department of Mathematical Methods in Biology, Belozersky Institute, Lomonosov Moscow State University, Moscow, Russia
| | - Vladimir M. Kovalzon
- Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia
- Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
47
|
Induction of Mutant Sik3Sleepy Allele in Neurons in Late Infancy Increases Sleep Need. J Neurosci 2021; 41:2733-2746. [PMID: 33558433 DOI: 10.1523/jneurosci.1004-20.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 12/06/2020] [Accepted: 12/10/2020] [Indexed: 01/12/2023] Open
Abstract
Sleep is regulated in a homeostatic manner. Sleep deprivation increases sleep need, which is compensated mainly by increased EEG δ power during non-rapid eye movement sleep (NREMS) and, to a lesser extent, by increased sleep amount. Although genetic factors determine the constitutive level of sleep need and sleep amount in mice and humans, the molecular entity behind sleep need remains unknown. Recently, we found that a gain-of-function Sleepy (Slp) mutation in the salt-inducible kinase 3 (Sik3) gene, which produces the mutant SIK3(SLP) protein, leads to an increase in NREMS EEG δ power and sleep amount. Since Sik3Slp mice express SIK3(SLP) in various types of cells in the brain as well as multiple peripheral tissues from the embryonic stage, the cell type and developmental stage responsible for the sleep phenotype in Sik3Slp mice remain to be elucidated. Here, we generated two mouse lines, synapsin1CreERT2 and Sik3ex13flox mice, which enable inducible Cre-mediated, conditional expression of SIK3(SLP) in neurons on tamoxifen administration. Administration of tamoxifen to synapsin1CreERT2 mice during late infancy resulted in higher recombination efficiency than administration during adolescence. SIK3(SLP) expression after late infancy increased NREMS and NREMS δ power in male synapsin1CreERT2; Sik3 ex13flox/+ mice. The expression of SIK3(SLP) after adolescence led to a higher NREMS δ power without a significant change in NREMS amounts. Thus, neuron-specific expression of SIK3(SLP) after late infancy is sufficient to increase sleep.SIGNIFICANCE STATEMENT The propensity to accumulate sleep need during wakefulness and to dissipate it during sleep underlies the homeostatic regulation of sleep. However, little is known about the developmental stage and cell types involved in determining the homeostatic regulation of sleep. Here, we show that Sik3Slp allele induction in mature neurons in late infancy is sufficient to increase non-rapid eye movement sleep amount and non-rapid eye movement sleep δ power. SIK3 signaling in neurons constitutes an intracellular mechanism to increase sleep.
Collapse
|
48
|
Shaw JC, Crombie GK, Palliser HK, Hirst JJ. Impaired Oligodendrocyte Development Following Preterm Birth: Promoting GABAergic Action to Improve Outcomes. Front Pediatr 2021; 9:618052. [PMID: 33634057 PMCID: PMC7901941 DOI: 10.3389/fped.2021.618052] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/12/2021] [Indexed: 11/21/2022] Open
Abstract
Preterm birth is associated with poor long-term neurodevelopmental and behavioral outcomes, even in the absence of obvious brain injury at the time of birth. In particular, behavioral disorders characterized by inattention, social difficulties and anxiety are common among children and adolescents who were born moderately to late preterm (32-37 weeks' gestation). Diffuse deficits in white matter microstructure are thought to play a role in these poor outcomes with evidence suggesting that a failure of oligodendrocytes to mature and myelinate axons is responsible. However, there remains a major knowledge gap over the mechanisms by which preterm birth interrupts normal oligodendrocyte development. In utero neurodevelopment occurs in an inhibitory-dominant environment due to the action of placentally derived neurosteroids on the GABAA receptor, thus promoting GABAergic inhibitory activity and maintaining the fetal behavioral state. Following preterm birth, and the subsequent premature exposure to the ex utero environment, this action of neurosteroids on GABAA receptors is greatly reduced. Coinciding with a reduction in GABAergic inhibition, the preterm neonatal brain is also exposed to ex utero environmental insults such as periods of hypoxia and excessive glucocorticoid concentrations. Together, these insults may increase levels of the excitatory neurotransmitter glutamate in the developing brain and result in a shift in the balance of inhibitory: excitatory activity toward excitatory. This review will outline the normal development of oligodendrocytes, how it is disrupted under excitation-dominated conditions and highlight how shifting the balance back toward an inhibitory-dominated environment may improve outcomes.
Collapse
Affiliation(s)
- Julia C Shaw
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia.,Mothers and Babies Research Centre, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Gabrielle K Crombie
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia.,Mothers and Babies Research Centre, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Hannah K Palliser
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia.,Mothers and Babies Research Centre, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Jonathan J Hirst
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia.,Mothers and Babies Research Centre, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
49
|
Orzylowski M, Fujiwara E, Mousseau DD, Baker GB. An Overview of the Involvement of D-Serine in Cognitive Impairment in Normal Aging and Dementia. Front Psychiatry 2021; 12:754032. [PMID: 34707525 PMCID: PMC8542907 DOI: 10.3389/fpsyt.2021.754032] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/02/2021] [Indexed: 12/14/2022] Open
Abstract
Dementia, of which Alzheimer's disease (AD) is the most common form, is characterized by progressive cognitive deterioration, including profound memory loss, which affects functioning in many aspects of life. Although cognitive deterioration is relatively common in aging and aging is a risk factor for AD, the condition is not necessarily a part of the aging process. The N-methyl-D-aspartate glutamate receptor (NMDAR) and its co-agonist D-serine are currently of great interest as potential important contributors to cognitive function in normal aging and dementia. D-Serine is necessary for activation of the NMDAR and in maintenance of long-term potentiation (LTP) and is involved in brain development, neuronal connectivity, synaptic plasticity and regulation of learning and memory. In this paper, we review evidence, from both preclinical and human studies, on the involvement of D-serine (and the enzymes involved in its metabolism) in regulation of cognition. Potential mechanisms of action of D-serine are discussed in the context of normal aging and in dementia, as is the potential for using D-serine as a potential biomarker and/or therapeutic agent in dementia. Although there is some controversy in the literature, it has been proposed that in normal aging there is decreased expression of serine racemase and decreased levels of D-serine and down-regulation of NMDARs, resulting in impaired synaptic plasticity and deficits in learning and memory. In contrast, in AD there appears to be activation of serine racemase, increased levels of D-serine and overstimulation of NMDARs, resulting in cytotoxicity, synaptic deficits, and dementia.
Collapse
Affiliation(s)
- Magdalena Orzylowski
- Villa Caritas Geriatric Psychiatry Hospital, Edmonton, AB, Canada.,Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
| | - Esther Fujiwara
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Darrell D Mousseau
- Department of Psychiatry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Glen B Baker
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
50
|
Dopamine Signaling in Wake-Promoting Clock Neurons Is Not Required for the Normal Regulation of Sleep in Drosophila. J Neurosci 2020; 40:9617-9633. [PMID: 33172977 DOI: 10.1523/jneurosci.1488-20.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/15/2020] [Accepted: 10/21/2020] [Indexed: 11/21/2022] Open
Abstract
Dopamine is a wake-promoting neuromodulator in mammals and fruit flies. In Drosophila melanogaster, the network of clock neurons that drives sleep/activity cycles comprises both wake-promoting and sleep-promoting cell types. The large ventrolateral neurons (l-LNvs) and small ventrolateral neurons (s-LNvs) have been identified as wake-promoting neurons within the clock neuron network. The l-LNvs are innervated by dopaminergic neurons, and earlier work proposed that dopamine signaling raises cAMP levels in the l-LNvs and thus induces excitatory electrical activity (action potential firing), which results in wakefulness and inhibits sleep. Here, we test this hypothesis by combining cAMP imaging and patch-clamp recordings in isolated brains. We find that dopamine application indeed increases cAMP levels and depolarizes the l-LNvs, but, surprisingly, it does not result in increased firing rates. Downregulation of the excitatory D1-like dopamine receptor (Dop1R1) in the l-LNvs and s-LNvs, but not of Dop1R2, abolished the depolarization of l-LNvs in response to dopamine. This indicates that dopamine signals via Dop1R1 to the l-LNvs. Downregulation of Dop1R1 or Dop1R2 in the l-LNvs and s-LNvs does not affect sleep in males. Unexpectedly, we find a moderate decrease of daytime sleep with downregulation of Dop1R1 and of nighttime sleep with downregulation of Dop1R2. Since the l-LNvs do not use Dop1R2 receptors and the s-LNvs also respond to dopamine, we conclude that the s-LNvs are responsible for the observed decrease in nighttime sleep. In summary, dopamine signaling in the wake-promoting LNvs is not required for daytime arousal, but likely promotes nighttime sleep via the s-LNvs.SIGNIFICANCE STATEMENT In insect and mammalian brains, sleep-promoting networks are intimately linked to the circadian clock, and the mechanisms underlying sleep and circadian timekeeping are evolutionarily ancient and highly conserved. Here we show that dopamine, one important sleep modulator in flies and mammals, plays surprisingly complex roles in the regulation of sleep by clock-containing neurons. Dopamine inhibits neurons in a central brain sleep center to promote sleep and excites wake-promoting circadian clock neurons. It is therefore predicted to promote wakefulness through both of these networks. Nevertheless, our results reveal that dopamine acting on wake-promoting clock neurons promotes sleep, revealing a previously unappreciated complexity in the dopaminergic control of sleep.
Collapse
|