1
|
Yoon S, Lee S, Joo Y, Ha E, Hong H, Song Y, Lee H, Kim S, Suh C, Lee CJ, Lyoo IK. Variations in brain glutamate and glutamine levels throughout the sleep-wake cycle. Biol Psychiatry 2024:S0006-3223(24)01785-2. [PMID: 39643103 DOI: 10.1016/j.biopsych.2024.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 10/25/2024] [Accepted: 11/21/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Glutamatergic signaling is essential for modulating synaptic plasticity and cognition. However, the dynamics of glutamatergic activity over the 24-hour sleep-wake cycle, particularly in relation to sleep, remain poorly understood. This study aims to investigate diurnal variations in brain Glx levels-representing the combined concentrations of glutamate and glutamine-in humans and to explore their implications for cognitive performance and sleep pressure. METHODS We conducted two independent experiments to measure Glx levels across the sleep-wake cycle using proton magnetic resonance spectroscopy. In Experiment 1, 14 participants underwent 13 hours of Glx measurements during a typical sleep-wake cycle. Experiment 2 extended these measurements to an around-the-clock observation over a 6-day period. This period included two days of normal sleep-wake cycles, 24 hours of enforced wakefulness, and a three-day recovery phase. Seven participants took part in Experiment 2. RESULTS The study observed that brain Glx levels increased during wakefulness and decreased during sleep. Notably, Glx levels were lower during enforced wakefulness compared to those during normal wakefulness. Reduced Glx levels were associated with diminished cognitive performance, while greater Glx exposure over the preceding 24 hours correlated with increased sleep pressure. CONCLUSIONS These findings suggest that Glx accumulation may contribute to increased sleep pressure, while its reduction appears to support wakefulness. These observations, together with the diurnal variations in Glx levels, underscore the dynamic nature of glutamatergic activity across the daily cycle. Further research is warranted to explore the potential role of sleep in regulating glutamatergic homeostasis.
Collapse
Affiliation(s)
- Sujung Yoon
- Ewha Brain Institute, Ewha W. University, Seoul 03760, South Korea; Department of Brain and Cognitive Sciences, Ewha W. University, Seoul 03760, South Korea
| | - Suji Lee
- College of Pharmacy, Dongduk W. University, Seoul 02748, South Korea
| | - Yoonji Joo
- Ewha Brain Institute, Ewha W. University, Seoul 03760, South Korea
| | - Eunji Ha
- Ewha Brain Institute, Ewha W. University, Seoul 03760, South Korea
| | - Haejin Hong
- Ewha Brain Institute, Ewha W. University, Seoul 03760, South Korea
| | - Yumi Song
- Ewha Brain Institute, Ewha W. University, Seoul 03760, South Korea; Department of Brain and Cognitive Sciences, Ewha W. University, Seoul 03760, South Korea
| | - Hyangwon Lee
- Ewha Brain Institute, Ewha W. University, Seoul 03760, South Korea; Department of Brain and Cognitive Sciences, Ewha W. University, Seoul 03760, South Korea
| | - Shinhye Kim
- Ewha Brain Institute, Ewha W. University, Seoul 03760, South Korea; Department of Brain and Cognitive Sciences, Ewha W. University, Seoul 03760, South Korea
| | - Chaewon Suh
- Ewha Brain Institute, Ewha W. University, Seoul 03760, South Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, South Korea
| | - In Kyoon Lyoo
- Ewha Brain Institute, Ewha W. University, Seoul 03760, South Korea; Department of Brain and Cognitive Sciences, Ewha W. University, Seoul 03760, South Korea; Graduate School of Pharmaceutical Sciences, Ewha W. University, Seoul 03760, South Korea.
| |
Collapse
|
2
|
Braga A, Chiacchiaretta M, Pellerin L, Kong D, Haydon PG. Astrocytic metabolic control of orexinergic activity in the lateral hypothalamus regulates sleep and wake architecture. Nat Commun 2024; 15:5979. [PMID: 39013907 PMCID: PMC11252394 DOI: 10.1038/s41467-024-50166-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 06/17/2024] [Indexed: 07/18/2024] Open
Abstract
Neuronal activity undergoes significant changes during vigilance states, accompanied by an accommodation of energy demands. While the astrocyte-neuron lactate shuttle has shown that lactate is the primary energy substrate for sustaining neuronal activity in multiple brain regions, its role in regulating sleep/wake architecture is not fully understood. Here we investigated the involvement of astrocytic lactate supply in maintaining consolidated wakefulness by downregulating, in a cell-specific manner, the expression of monocarboxylate transporters (MCTs) in the lateral hypothalamus of transgenic mice. Our results demonstrate that reduced expression of MCT4 in astrocytes disrupts lactate supply to wake-promoting orexin neurons, impairing wakefulness stability. Additionally, we show that MCT2-mediated lactate uptake is necessary for maintaining tonic firing of orexin neurons and stabilizing wakefulness. Our findings provide both in vivo and in vitro evidence supporting the role of astrocyte-to-orexinergic neuron lactate shuttle in regulating proper sleep/wake stability.
Collapse
Affiliation(s)
- Alice Braga
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Martina Chiacchiaretta
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, 02111, USA.
| | - Luc Pellerin
- Inserm U1313, University and CHU of Poitiers, 86021, Poitiers, France
| | - Dong Kong
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, 02111, USA
- Division of Endocrinology, Department of Pediatrics, F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Philip G Haydon
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, 02111, USA.
| |
Collapse
|
3
|
Hagihara H, Shoji H, Hattori S, Sala G, Takamiya Y, Tanaka M, Ihara M, Shibutani M, Hatada I, Hori K, Hoshino M, Nakao A, Mori Y, Okabe S, Matsushita M, Urbach A, Katayama Y, Matsumoto A, Nakayama KI, Katori S, Sato T, Iwasato T, Nakamura H, Goshima Y, Raveau M, Tatsukawa T, Yamakawa K, Takahashi N, Kasai H, Inazawa J, Nobuhisa I, Kagawa T, Taga T, Darwish M, Nishizono H, Takao K, Sapkota K, Nakazawa K, Takagi T, Fujisawa H, Sugimura Y, Yamanishi K, Rajagopal L, Hannah ND, Meltzer HY, Yamamoto T, Wakatsuki S, Araki T, Tabuchi K, Numakawa T, Kunugi H, Huang FL, Hayata-Takano A, Hashimoto H, Tamada K, Takumi T, Kasahara T, Kato T, Graef IA, Crabtree GR, Asaoka N, Hatakama H, Kaneko S, Kohno T, Hattori M, Hoshiba Y, Miyake R, Obi-Nagata K, Hayashi-Takagi A, Becker LJ, Yalcin I, Hagino Y, Kotajima-Murakami H, Moriya Y, Ikeda K, Kim H, Kaang BK, Otabi H, Yoshida Y, Toyoda A, Komiyama NH, Grant SGN, Ida-Eto M, Narita M, Matsumoto KI, Okuda-Ashitaka E, Ohmori I, Shimada T, Yamagata K, Ageta H, Tsuchida K, Inokuchi K, Sassa T, Kihara A, Fukasawa M, Usuda N, Katano T, Tanaka T, Yoshihara Y, Igarashi M, Hayashi T, Ishikawa K, Yamamoto S, Nishimura N, Nakada K, Hirotsune S, Egawa K, Higashisaka K, Tsutsumi Y, Nishihara S, Sugo N, Yagi T, Ueno N, Yamamoto T, Kubo Y, Ohashi R, Shiina N, Shimizu K, Higo-Yamamoto S, Oishi K, Mori H, Furuse T, Tamura M, Shirakawa H, Sato DX, Inoue YU, Inoue T, Komine Y, Yamamori T, Sakimura K, Miyakawa T. Large-scale animal model study uncovers altered brain pH and lactate levels as a transdiagnostic endophenotype of neuropsychiatric disorders involving cognitive impairment. eLife 2024; 12:RP89376. [PMID: 38529532 PMCID: PMC10965225 DOI: 10.7554/elife.89376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024] Open
Abstract
Increased levels of lactate, an end-product of glycolysis, have been proposed as a potential surrogate marker for metabolic changes during neuronal excitation. These changes in lactate levels can result in decreased brain pH, which has been implicated in patients with various neuropsychiatric disorders. We previously demonstrated that such alterations are commonly observed in five mouse models of schizophrenia, bipolar disorder, and autism, suggesting a shared endophenotype among these disorders rather than mere artifacts due to medications or agonal state. However, there is still limited research on this phenomenon in animal models, leaving its generality across other disease animal models uncertain. Moreover, the association between changes in brain lactate levels and specific behavioral abnormalities remains unclear. To address these gaps, the International Brain pH Project Consortium investigated brain pH and lactate levels in 109 strains/conditions of 2294 animals with genetic and other experimental manipulations relevant to neuropsychiatric disorders. Systematic analysis revealed that decreased brain pH and increased lactate levels were common features observed in multiple models of depression, epilepsy, Alzheimer's disease, and some additional schizophrenia models. While certain autism models also exhibited decreased pH and increased lactate levels, others showed the opposite pattern, potentially reflecting subpopulations within the autism spectrum. Furthermore, utilizing large-scale behavioral test battery, a multivariate cross-validated prediction analysis demonstrated that poor working memory performance was predominantly associated with increased brain lactate levels. Importantly, this association was confirmed in an independent cohort of animal models. Collectively, these findings suggest that altered brain pH and lactate levels, which could be attributed to dysregulated excitation/inhibition balance, may serve as transdiagnostic endophenotypes of debilitating neuropsychiatric disorders characterized by cognitive impairment, irrespective of their beneficial or detrimental nature.
Collapse
Affiliation(s)
- Hideo Hagihara
- Division of Systems Medical Science, Center for Medical Science, Fujita Health UniversityToyoakeJapan
| | - Hirotaka Shoji
- Division of Systems Medical Science, Center for Medical Science, Fujita Health UniversityToyoakeJapan
| | - Satoko Hattori
- Division of Systems Medical Science, Center for Medical Science, Fujita Health UniversityToyoakeJapan
| | - Giovanni Sala
- Division of Systems Medical Science, Center for Medical Science, Fujita Health UniversityToyoakeJapan
| | - Yoshihiro Takamiya
- Division of Systems Medical Science, Center for Medical Science, Fujita Health UniversityToyoakeJapan
| | - Mika Tanaka
- Division of Systems Medical Science, Center for Medical Science, Fujita Health UniversityToyoakeJapan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular CenterSuitaJapan
| | - Mihiro Shibutani
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashiJapan
| | - Izuho Hatada
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashiJapan
| | - Kei Hori
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and PsychiatryKodairaJapan
| | - Mikio Hoshino
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and PsychiatryKodairaJapan
| | - Akito Nakao
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto UniversityKyotoJapan
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto UniversityKyotoJapan
| | - Shigeo Okabe
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of TokyoTokyoJapan
| | - Masayuki Matsushita
- Department of Molecular Cellular Physiology, Graduate School of Medicine, University of the RyukyusNishiharaJapan
| | - Anja Urbach
- Department of Neurology, Jena University HospitalJenaGermany
| | - Yuta Katayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu UniversityFukuokaJapan
| | - Akinobu Matsumoto
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu UniversityFukuokaJapan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu UniversityFukuokaJapan
| | - Shota Katori
- Laboratory of Mammalian Neural Circuits, National Institute of GeneticsMishimaJapan
| | - Takuya Sato
- Laboratory of Mammalian Neural Circuits, National Institute of GeneticsMishimaJapan
| | - Takuji Iwasato
- Laboratory of Mammalian Neural Circuits, National Institute of GeneticsMishimaJapan
| | - Haruko Nakamura
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of MedicineYokohamaJapan
| | - Yoshio Goshima
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of MedicineYokohamaJapan
| | - Matthieu Raveau
- Laboratory for Neurogenetics, RIKEN Center for Brain ScienceWakoJapan
| | - Tetsuya Tatsukawa
- Laboratory for Neurogenetics, RIKEN Center for Brain ScienceWakoJapan
| | - Kazuhiro Yamakawa
- Laboratory for Neurogenetics, RIKEN Center for Brain ScienceWakoJapan
- Department of Neurodevelopmental Disorder Genetics, Institute of Brain Sciences, Nagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Noriko Takahashi
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of TokyoTokyoJapan
- Department of Physiology, Kitasato University School of MedicineSagamiharaJapan
| | - Haruo Kasai
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of TokyoTokyoJapan
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of TokyoTokyoJapan
| | - Johji Inazawa
- Research Core, Tokyo Medical and Dental UniversityTokyoJapan
| | - Ikuo Nobuhisa
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental UniversityTokyoJapan
| | - Tetsushi Kagawa
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental UniversityTokyoJapan
| | - Tetsuya Taga
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental UniversityTokyoJapan
| | - Mohamed Darwish
- Department of Biochemistry, Faculty of Pharmacy, Cairo UniversityCairoEgypt
- Department of Behavioral Physiology, Graduate School of Innovative Life Science, University of ToyamaToyamaJapan
| | | | - Keizo Takao
- Department of Behavioral Physiology, Graduate School of Innovative Life Science, University of ToyamaToyamaJapan
- Department of Behavioral Physiology, Faculty of Medicine, University of ToyamaToyamaJapan
| | - Kiran Sapkota
- Department of Neuroscience, Southern ResearchBirminghamUnited States
| | | | - Tsuyoshi Takagi
- Institute for Developmental Research, Aichi Developmental Disability CenterKasugaiJapan
| | - Haruki Fujisawa
- Department of Endocrinology, Diabetes and Metabolism, School of Medicine, Fujita Health UniversityToyoakeJapan
| | - Yoshihisa Sugimura
- Department of Endocrinology, Diabetes and Metabolism, School of Medicine, Fujita Health UniversityToyoakeJapan
| | - Kyosuke Yamanishi
- Department of Neuropsychiatry, Hyogo Medical University School of MedicineNishinomiyaJapan
| | - Lakshmi Rajagopal
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of MedicineChicagoUnited States
| | - Nanette Deneen Hannah
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of MedicineChicagoUnited States
| | - Herbert Y Meltzer
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of MedicineChicagoUnited States
| | - Tohru Yamamoto
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa UniversityKita-gunJapan
| | - Shuji Wakatsuki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and PsychiatryTokyoJapan
| | - Toshiyuki Araki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and PsychiatryTokyoJapan
| | - Katsuhiko Tabuchi
- Department of Molecular & Cellular Physiology, Shinshu University School of MedicineMatsumotoJapan
| | - Tadahiro Numakawa
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and PsychiatryKodairaJapan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and PsychiatryKodairaJapan
- Department of Psychiatry, Teikyo University School of MedicineTokyoJapan
| | - Freesia L Huang
- Program of Developmental Neurobiology, National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Atsuko Hayata-Takano
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka UniversitySuitaJapan
- Department of Pharmacology, Graduate School of Dentistry, Osaka UniversitySuitaJapan
- United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of FukuiSuitaJapan
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka UniversitySuitaJapan
- United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of FukuiSuitaJapan
- Division of Bioscience, Institute for Datability Science, Osaka UniversitySuitaJapan
- Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka UniversitySuitaJapan
- Department of Molecular Pharmaceutical Science, Graduate School of Medicine, Osaka UniversitySuitaJapan
| | - Kota Tamada
- RIKEN Brain Science InstituteWakoJapan
- Department of Physiology and Cell Biology, Kobe University School of MedicineKobeJapan
| | - Toru Takumi
- RIKEN Brain Science InstituteWakoJapan
- Department of Physiology and Cell Biology, Kobe University School of MedicineKobeJapan
| | - Takaoki Kasahara
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Center for Brain ScienceWakoJapan
- Institute of Biology and Environmental Sciences, Carl von Ossietzky University of OldenburgOldenburgGermany
| | - Tadafumi Kato
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Center for Brain ScienceWakoJapan
- Department of Psychiatry and Behavioral Science, Juntendo University Graduate School of MedicineTokyoJapan
| | - Isabella A Graef
- Department of Pathology, Stanford University School of MedicineStanfordUnited States
| | - Gerald R Crabtree
- Department of Pathology, Stanford University School of MedicineStanfordUnited States
| | - Nozomi Asaoka
- Department of Pharmacology, Kyoto Prefectural University of MedicineKyotoJapan
| | - Hikari Hatakama
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto UniversityKyotoJapan
| | - Shuji Kaneko
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto UniversityKyotoJapan
| | - Takao Kohno
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City UniversityNagoyaJapan
| | - Mitsuharu Hattori
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City UniversityNagoyaJapan
| | - Yoshio Hoshiba
- Laboratory of Medical Neuroscience, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashiJapan
| | - Ryuhei Miyake
- Laboratory for Multi-scale Biological Psychiatry, RIKEN Center for Brain ScienceWakoJapan
| | - Kisho Obi-Nagata
- Laboratory for Multi-scale Biological Psychiatry, RIKEN Center for Brain ScienceWakoJapan
| | - Akiko Hayashi-Takagi
- Laboratory of Medical Neuroscience, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashiJapan
- Laboratory for Multi-scale Biological Psychiatry, RIKEN Center for Brain ScienceWakoJapan
| | - Léa J Becker
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Université de StrasbourgStrasbourgFrance
| | - Ipek Yalcin
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Université de StrasbourgStrasbourgFrance
| | - Yoko Hagino
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | | | - Yuki Moriya
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | - Kazutaka Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | - Hyopil Kim
- Department of Biological Sciences, College of Natural Sciences, Seoul National UniversitySeoulRepublic of Korea
- Department of Biomedical Engineering, Johns Hopkins School of MedicineBaltimoreUnited States
| | - Bong-Kiun Kaang
- Department of Biological Sciences, College of Natural Sciences, Seoul National UniversitySeoulRepublic of Korea
- Center for Cognition and Sociality, Institute for Basic Science (IBS)DaejeonRepublic of Korea
| | - Hikari Otabi
- College of Agriculture, Ibaraki UniversityAmiJapan
- United Graduate School of Agricultural Science, Tokyo University of Agriculture and TechnologyFuchuJapan
| | - Yuta Yoshida
- College of Agriculture, Ibaraki UniversityAmiJapan
| | - Atsushi Toyoda
- College of Agriculture, Ibaraki UniversityAmiJapan
- United Graduate School of Agricultural Science, Tokyo University of Agriculture and TechnologyFuchuJapan
- Ibaraki University Cooperation between Agriculture and Medical Science (IUCAM)IbarakiJapan
| | - Noboru H Komiyama
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of EdinburghEdinburghUnited Kingdom
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of EdinburghEdinburghUnited Kingdom
| | - Seth GN Grant
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of EdinburghEdinburghUnited Kingdom
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of EdinburghEdinburghUnited Kingdom
| | - Michiru Ida-Eto
- Department of Developmental and Regenerative Medicine, Mie University, Graduate School of MedicineTsuJapan
| | - Masaaki Narita
- Department of Developmental and Regenerative Medicine, Mie University, Graduate School of MedicineTsuJapan
| | - Ken-ichi Matsumoto
- Department of Biosignaling and Radioisotope Experiment, Interdisciplinary Center for Science Research, Organization for Research and Academic Information, Shimane UniversityIzumoJapan
| | | | - Iori Ohmori
- Department of Physiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayamaJapan
| | - Tadayuki Shimada
- Child Brain Project, Tokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | - Kanato Yamagata
- Child Brain Project, Tokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | - Hiroshi Ageta
- Division for Therapies Against Intractable Diseases, Center for Medical Science, Fujita Health UniversityToyoakeJapan
| | - Kunihiro Tsuchida
- Division for Therapies Against Intractable Diseases, Center for Medical Science, Fujita Health UniversityToyoakeJapan
| | - Kaoru Inokuchi
- Research Center for Idling Brain Science, University of ToyamaToyamaJapan
- Department of Biochemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of ToyamaToyamaJapan
- Core Research for Evolutionary Science and Technology (CREST), Japan Science and Technology Agency (JST), University of ToyamaToyamaJapan
| | - Takayuki Sassa
- Faculty of Pharmaceutical Sciences, Hokkaido UniversitySapporoJapan
| | - Akio Kihara
- Faculty of Pharmaceutical Sciences, Hokkaido UniversitySapporoJapan
| | - Motoaki Fukasawa
- Department of Anatomy II, Fujita Health University School of MedicineToyoakeJapan
| | - Nobuteru Usuda
- Department of Anatomy II, Fujita Health University School of MedicineToyoakeJapan
| | - Tayo Katano
- Department of Medical Chemistry, Kansai Medical UniversityHirakataJapan
| | - Teruyuki Tanaka
- Department of Developmental Medical Sciences, Graduate School of Medicine, The University of TokyoTokyoJapan
| | - Yoshihiro Yoshihara
- Laboratory for Systems Molecular Ethology, RIKEN Center for Brain ScienceWakoJapan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, School of Medicine, and Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
- Transdiciplinary Research Program, Niigata UniversityNiigataJapan
| | - Takashi Hayashi
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST)TsukubaJapan
| | - Kaori Ishikawa
- Institute of Life and Environmental Sciences, University of TsukubaTsukubaJapan
- Graduate School of Science and Technology, University of TsukubaTsukubaJapan
| | - Satoshi Yamamoto
- Integrated Technology Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company, LtdFujisawaJapan
| | - Naoya Nishimura
- Integrated Technology Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company, LtdFujisawaJapan
| | - Kazuto Nakada
- Institute of Life and Environmental Sciences, University of TsukubaTsukubaJapan
- Graduate School of Science and Technology, University of TsukubaTsukubaJapan
| | - Shinji Hirotsune
- Department of Genetic Disease Research, Osaka City University Graduate School of MedicineOsakaJapan
| | - Kiyoshi Egawa
- Department of Pediatrics, Hokkaido University Graduate School of MedicineSapporoJapan
| | - Kazuma Higashisaka
- Laboratory of Toxicology and Safety Science, Graduate School of Pharmaceutical Sciences, Osaka UniversitySuitaJapan
| | - Yasuo Tsutsumi
- Laboratory of Toxicology and Safety Science, Graduate School of Pharmaceutical Sciences, Osaka UniversitySuitaJapan
| | - Shoko Nishihara
- Glycan & Life Systems Integration Center (GaLSIC), Soka UniversityTokyoJapan
| | - Noriyuki Sugo
- Graduate School of Frontier Biosciences, Osaka UniversitySuitaJapan
| | - Takeshi Yagi
- Graduate School of Frontier Biosciences, Osaka UniversitySuitaJapan
| | - Naoto Ueno
- Laboratory of Morphogenesis, National Institute for Basic BiologyOkazakiJapan
| | - Tomomi Yamamoto
- Division of Biophysics and Neurobiology, National Institute for Physiological SciencesOkazakiJapan
| | - Yoshihiro Kubo
- Division of Biophysics and Neurobiology, National Institute for Physiological SciencesOkazakiJapan
| | - Rie Ohashi
- Laboratory of Neuronal Cell Biology, National Institute for Basic BiologyOkazakiJapan
- Department of Basic Biology, SOKENDAI (Graduate University for Advanced Studies)OkazakiJapan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural SciencesOkazakiJapan
| | - Nobuyuki Shiina
- Laboratory of Neuronal Cell Biology, National Institute for Basic BiologyOkazakiJapan
- Department of Basic Biology, SOKENDAI (Graduate University for Advanced Studies)OkazakiJapan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural SciencesOkazakiJapan
| | - Kimiko Shimizu
- Department of Biological Sciences, School of Science, The University of TokyoTokyoJapan
| | - Sayaka Higo-Yamamoto
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST)TsukubaJapan
| | - Katsutaka Oishi
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST)TsukubaJapan
- Department of Applied Biological Science, Graduate School of Science and Technology, Tokyo University of ScienceNodaJapan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of TokyoKashiwaJapan
- School of Integrative and Global Majors (SIGMA), University of TsukubaTsukubaJapan
| | - Hisashi Mori
- Department of Molecular Neuroscience, Graduate School of Medicine and Pharmaceutical Sciences, University of ToyamaToyamaJapan
| | - Tamio Furuse
- Mouse Phenotype Analysis Division, Japan Mouse Clinic, RIKEN BioResource Research Center (BRC)TsukubaJapan
| | - Masaru Tamura
- Mouse Phenotype Analysis Division, Japan Mouse Clinic, RIKEN BioResource Research Center (BRC)TsukubaJapan
| | - Hisashi Shirakawa
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto UniversityKyotoJapan
| | - Daiki X Sato
- Division of Systems Medical Science, Center for Medical Science, Fujita Health UniversityToyoakeJapan
- Graduate School of Life Sciences, Tohoku UniversitySendaiJapan
| | - Yukiko U Inoue
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and PsychiatryKodairaJapan
| | - Takayoshi Inoue
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and PsychiatryKodairaJapan
| | - Yuriko Komine
- Young Researcher Support Group, Research Enhancement Strategy Office, National Institute for Basic Biology, National Institute of Natural SciencesOkazakiJapan
- Division of Brain Biology, National Institute for Basic BiologyOkazakiJapan
| | - Tetsuo Yamamori
- Division of Brain Biology, National Institute for Basic BiologyOkazakiJapan
- Laboratory for Molecular Analysis of Higher Brain Function, RIKEN Center for Brain ScienceWakoJapan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata UniversityNiigataJapan
- Department of Animal Model Development, Brain Research Institute, Niigata UniversityNiigataJapan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Center for Medical Science, Fujita Health UniversityToyoakeJapan
| |
Collapse
|
4
|
Fernandes M, Spanetta M, Placidi F, Izzi F, Negri F, Nuccetelli M, Bernardini S, Mercuri NB, Liguori C. A preliminary study investigating the clinical potential of measuring cerebrospinal-fluid lactate levels in patients with narcolepsy type 1 and 2. Physiol Behav 2023; 272:114371. [PMID: 37802459 DOI: 10.1016/j.physbeh.2023.114371] [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: 08/18/2023] [Revised: 10/03/2023] [Accepted: 10/04/2023] [Indexed: 10/10/2023]
Abstract
STUDY OBJECTIVES Besides the quantification of orexin-A/hypocretin-1 cerebrospinal fluid (CSF) levels in narcolepsy for diagnostic purposes, several other CSF biomarkers have been evaluated, although with controversial results. Since CSF lactate concentrations fluctuate according to the sleep-wake cycle with higher levels during wakefulness and lower levels during sleep, as documented in animal model studies, the present study aimed at quantifying the CSF lactate levels in patients with narcolepsy type 1 (NT1) and 2 (NT2), which are two sleep disorders featured by excessive daytime sleepiness (EDS). METHODS Patients with NT1 and NT2 were enrolled in this study and compared to a control group of similar age and sex. All the subjects included in the study underwent a polysomnographic study followed by lumbar puncture for the quantification of CSF lactate levels at awakening. RESULTS 23 NT1 (43.5 % male; 36.43 ± 11.89 years) and 15 NT2 patients (46.7 % male; 37.8 ± 14.1 years) were compared to 17 controls (58.8 % male; 32.3 ± 8.4 years). CSF lactate concentrations were reduced in patients with NT1 and NT2 compared to controls but no differences were found between the two groups of patients. ROC curves analysis showed that CSF lactate ≤1.3 mmol/l had a sensitivity of 96.49 and a specificity of 82.35 % for discriminating patients with narcolepsy from controls. CONCLUSIONS The present study showed a decrease in CSF lactate levels in patients with narcolepsy. Notably, the reduction of lactate levels was present in both NT1 and NT2 patients, independently of CSF orexin levels. Narcolepsy patients present EDS with daytime napping and REM-related episodes, possibly substantiating the CSF lactate levels reduction related to the impaired daytime wakefulness which was demonstrated in animal studies. Moreover, CSF lactate levels present a good sensitivity and adequate specificity for differentiating narcolepsy from controls. Further studies are needed to understand the role of CSF lactate and its usefulness for monitoring daytime vigilance in patients with narcolepsy.
Collapse
Affiliation(s)
- Mariana Fernandes
- Department of Systems Medicine, University of Rome Tor Vergata, Italy
| | - Matteo Spanetta
- Department of Systems Medicine, University of Rome Tor Vergata, Italy
| | - Fabio Placidi
- Department of Systems Medicine, University of Rome Tor Vergata, Italy; Sleep Medicine Centre, Neurology Unit, University Hospital of Rome Tor Vergata, Italy
| | - Francesca Izzi
- Sleep Medicine Centre, Neurology Unit, University Hospital of Rome Tor Vergata, Italy
| | - Francesco Negri
- Sleep Medicine Centre, Neurology Unit, University Hospital of Rome Tor Vergata, Italy
| | - Marzia Nuccetelli
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Italy
| | - Sergio Bernardini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Italy
| | - Nicola Biagio Mercuri
- Department of Systems Medicine, University of Rome Tor Vergata, Italy; Sleep Medicine Centre, Neurology Unit, University Hospital of Rome Tor Vergata, Italy
| | - Claudio Liguori
- Department of Systems Medicine, University of Rome Tor Vergata, Italy; Sleep Medicine Centre, Neurology Unit, University Hospital of Rome Tor Vergata, Italy.
| |
Collapse
|
5
|
Ingiosi AM, Frank MG. Noradrenergic Signaling in Astrocytes Influences Mammalian Sleep Homeostasis. Clocks Sleep 2022; 4:332-345. [PMID: 35892990 PMCID: PMC9326550 DOI: 10.3390/clockssleep4030028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/22/2022] [Accepted: 07/01/2022] [Indexed: 02/01/2023] Open
Abstract
Astrocytes influence sleep expression and regulation, but the cellular signaling pathways involved in these processes are poorly defined. We proposed that astrocytes detect and integrate a neuronal signal that accumulates during wakefulness, thereby leading to increased sleep drive. Noradrenaline (NA) satisfies several criteria for a waking signal integrated by astrocytes. We therefore investigated the role of NA signaling in astrocytes in mammalian sleep. We conditionally knocked out (cKO) β2-adrenergic receptors (β2-AR) selectively in astrocytes in mice and recorded electroencephalographic and electromyographic activity under baseline conditions and in response to sleep deprivation (SDep). cKO of astroglial β2-ARs increased active phase siesta duration under baseline conditions and reduced homeostatic compensatory changes in sleep consolidation and non-rapid eye movement slow-wave activity (SWA) after SDep. Overall, astroglial NA β2-ARs influence mammalian sleep homeostasis in a manner consistent with our proposed model of neuronal-astroglial interactions.
Collapse
Affiliation(s)
- Ashley M. Ingiosi
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA;
| | - Marcos G. Frank
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA;
- Gleason Institute for Neuroscience, Washington State University, Spokane, WA 99202, USA
| |
Collapse
|
6
|
Abstract
The detection of neurotransmitter release from reprogrammed human cell is an important demonstration of their functionality. Electrochemistry has the distinct advantages over alternative methods that it allows for the measuring of the analyte of interest at a high temporal resolution. This is necessary for fast events, such as neurotransmitter release and reuptake, which happen in the order of milliseconds to seconds. The precise description of these kinetic events can lead to insights into the function of cells in health and disease and allows for the exploration of events that might be missed using methods that look at absolute concentration values or methods that have a slower sampling rate. In the present chapter, we describe the use of constant potential amperometry and enzyme-coated multielectrode arrays for the detection of glutamate in vitro. These biosensors have the distinct advantage of "self-referencing," a method providing high selectivity while retaining outstanding temporal resolution. Here, we provide a step-by-step user guide for a commercially available system and its application for in vitro systems such as reprogrammed cells.
Collapse
|
7
|
Ruggiero A, Katsenelson M, Slutsky I. Mitochondria: new players in homeostatic regulation of firing rate set points. Trends Neurosci 2021; 44:605-618. [PMID: 33865626 DOI: 10.1016/j.tins.2021.03.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/10/2021] [Accepted: 03/11/2021] [Indexed: 10/21/2022]
Abstract
Neural circuit functions are stabilized by homeostatic processes at long timescales in response to changes in behavioral states, experience, and learning. However, it remains unclear which specific physiological variables are being stabilized and which cellular or neural network components compose the homeostatic machinery. At this point, most evidence suggests that the distribution of firing rates among neurons in a neuronal circuit is the key variable that is maintained around a set-point value in a process called 'firing rate homeostasis.' Here, we review recent findings that implicate mitochondria as central players in mediating firing rate homeostasis. While mitochondria are known to regulate neuronal variables such as synaptic vesicle release or intracellular calcium concentration, the mitochondrial signaling pathways that are essential for firing rate homeostasis remain largely unknown. We used basic concepts of control theory to build a framework for classifying possible components of the homeostatic machinery that stabilizes firing rate, and we particularly emphasize the potential role of sleep and wakefulness in this homeostatic process. This framework may facilitate the identification of new homeostatic pathways whose malfunctions drive instability of neural circuits in distinct brain disorders.
Collapse
Affiliation(s)
- Antonella Ruggiero
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Maxim Katsenelson
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel.
| |
Collapse
|
8
|
The why and how of sleep-dependent synaptic down-selection. Semin Cell Dev Biol 2021; 125:91-100. [PMID: 33712366 PMCID: PMC8426406 DOI: 10.1016/j.semcdb.2021.02.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/28/2021] [Accepted: 02/28/2021] [Indexed: 12/22/2022]
Abstract
Sleep requires that we disconnect from the environment, losing the ability to promptly respond to stimuli. There must be at least one essential function that justifies why we take this risk every day, and that function must depend on the brain being offline. We have proposed that this function is to renormalize synaptic weights after learning has led to a net increase in synaptic strength in many brain circuits. Without this renormalization, synaptic activity would become energetically too expensive and saturation would prevent new learning. There is converging evidence from molecular, electrophysiological, and ultrastructural experiments showing a net increase in synaptic strength after the major wake phase, and a net decline after sleep. The evidence also suggests that sleep-dependent renormalization is a smart process of synaptic down-selection, comprehensive and yet specific, which could explain the many beneficial effects of sleep on cognition. Recently, a key molecular mechanism that allows broad synaptic weakening during sleep was identified. Other mechanisms still being investigated should eventually explain how sleep can weaken most synapses but afford protection to some, including those directly activated by learning. That synaptic down-selection takes place during sleep is by now established; why it should take place during sleep has a plausible explanation; how it happens is still work in progress.
Collapse
|
9
|
Bingul D, Kalra K, Murata EM, Belser A, Dash MB. Persistent changes in extracellular lactate dynamics following synaptic potentiation. Neurobiol Learn Mem 2020; 175:107314. [PMID: 32961277 PMCID: PMC7655607 DOI: 10.1016/j.nlm.2020.107314] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/27/2020] [Accepted: 09/14/2020] [Indexed: 12/19/2022]
Abstract
A diverse array of neurometabolic coupling mechanisms exist within the brain to ensure that sufficient metabolite availability is present to meet both acute and chronic energetic demands. Excitatory synaptic activity, which produces the majority of the brain's energetic demands, triggers a rapid metabolic response including a characteristic shift towards aerobic glycolysis. Herein, astrocytically derived lactate appears to serve as an important metabolite to meet the extensive metabolic needs of activated neurons. Despite a wealth of literature characterizing lactate's role in mediating these acute metabolic needs, the extent to which lactate supports chronic energetic demands of neurons remains unclear. We hypothesized that synaptic potentiation, a ubiquitous brain phenomenon that can produce chronic alterations in synaptic activity, could necessitate persistent alterations in brain energetics. In freely-behaving rats, we induced long-term potentiation (LTP) of synapses within the dentate gyrus through high-frequency electrical stimulation (HFS) of the medial perforant pathway. Before, during, and after LTP induction, we continuously recorded extracellular lactate concentrations within the dentate gyrus to assess how changes in synaptic strength alter local glycolytic activity. Synaptic potentiation 1) altered the acute response of extracellular lactate to transient neuronal activation as evident by a larger initial dip and subsequent overshoot and 2) chronically increased local lactate availability. Although synapses were potentiated immediately following HFS, observed changes in lactate dynamics were only evident beginning ~24 h later. Once observed, however, both synaptic potentiation and altered lactate dynamics persisted for the duration of the experiment (~72 h). Persistent alterations in synaptic strength, therefore, appear to be associated with metabolic plasticity in the form of persistent augmentation of glycolytic activity.
Collapse
Affiliation(s)
- D Bingul
- Program in Neuroscience, Middlebury College, Middlebury, VT 05753, United States
| | - K Kalra
- Program in Neuroscience, Middlebury College, Middlebury, VT 05753, United States
| | - E M Murata
- Program in Neuroscience, Middlebury College, Middlebury, VT 05753, United States
| | - A Belser
- Program in Neuroscience, Middlebury College, Middlebury, VT 05753, United States
| | - M B Dash
- Program in Neuroscience, Middlebury College, Middlebury, VT 05753, United States; Department of Psychology, Middlebury College, Middlebury, VT 05753, United States.
| |
Collapse
|
10
|
Kovalzon VM, Panchin YV. [D-lactate as a novel somnogenic factor?]. Zh Nevrol Psikhiatr Im S S Korsakova 2020; 120:22-25. [PMID: 33076641 DOI: 10.17116/jnevro202012009222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To evaluate an influence of intracerebral L-lactate concentration on sleep-wake cycle. MATERIAL AND METHODS Twenty adult male white rats preliminary implanted (under general anesthesia) with the electrodes for neocortical EEG and a single cannula to a lateral ventricle were used as subjects. A 5 µl bolus of either saline or a solution of sodium L- or D-lactate (0.1 mg, 0.2 M, Sigma-Aldrich) was injected through the cannula and followed by a 6-hr recording. RESULTS AND CONCLUSION Administration of L-lactate does not influence sleep-wake cycle of experimental animals. At the same time, its artificial optical analog D-lactate induces the significant (as compared to the control) decrease in wake (34.8% to 26.5%) and increase in slow wave sleep (57.4% to 69.2%). It has been suggested that D-lactate may be the antagonist of one or several L-lactate receptors.
Collapse
Affiliation(s)
- V M Kovalzon
- Severtsov Institute of Ecology and Evolution of the Russian Academy of Sciences, Moscow, Russia.,Kharkhevich Institute of Information Transmission of the Russian Academy of Sciences, Moscow, Russia
| | - Yu V Panchin
- Kharkhevich Institute of Information Transmission of the Russian Academy of Sciences, Moscow, Russia.,Belozerskiy Institute of Physical-Chemical Biology of Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
11
|
Carroll CM, Hsiang H, Snyder S, Forsberg J, Dash MB. Cortical zeta-inhibitory peptide injection reduces local sleep need. Sleep 2020; 42:5306948. [PMID: 30722054 DOI: 10.1093/sleep/zsz028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 01/28/2019] [Indexed: 11/14/2022] Open
Abstract
Local sleep need within cortical circuits exhibits extensive interregional variability and appears to increase following learning during preceding waking. Although the biological mechanisms responsible for generating sleep need are unclear, this local variability could arise as a consequence of wake-dependent synaptic plasticity. To test whether cortical synaptic strength is a proximate driver of sleep homeostasis, we developed a novel experimental approach to alter local sleep need. One hour prior to light onset, we injected zeta-inhibitory peptide (ZIP), a pharmacological antagonist of protein kinase Mζ, which can produce pronounced synaptic depotentiation, into the right motor cortex of freely behaving rats. When compared with saline control, ZIP selectively reduced slow-wave activity (SWA; the best electrophysiological marker of sleep need) within the injected motor cortex without affecting SWA in a distal cortical site. This local reduction in SWA was associated with a significant reduction in the slope and amplitude of individual slow waves. Local ZIP injection did not significantly alter the amount of time spent in each behavioral state, locomotor activity, or EEG/LFP power during waking or REM sleep. Thus, local ZIP injection selectively produced a local reduction in sleep need; synaptic strength, therefore, may play a causal role in generating local homeostatic sleep need within the cortex.
Collapse
Affiliation(s)
| | | | - Sam Snyder
- Program in Neuroscience, Middlebury College, Middlebury, VT
| | - Jade Forsberg
- Program in Neuroscience, Middlebury College, Middlebury, VT
| | - Michael B Dash
- Program in Neuroscience, Middlebury College, Middlebury, VT.,Department of Psychology, Middlebury College, Middlebury, VT
| |
Collapse
|
12
|
Garofalo S, Picard K, Limatola C, Nadjar A, Pascual O, Tremblay MÈ. Role of Glia in the Regulation of Sleep in Health and Disease. Compr Physiol 2020; 10:687-712. [PMID: 32163207 DOI: 10.1002/cphy.c190022] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Sleep is a naturally occurring physiological state that is required to sustain physical and mental health. Traditionally viewed as strictly regulated by top-down control mechanisms, sleep is now known to also originate locally. Glial cells are emerging as important contributors to the regulation of sleep-wake cycles, locally and among dedicated neural circuits. A few pioneering studies revealed that astrocytes and microglia may influence sleep pressure, duration as well as intensity, but the precise involvement of these two glial cells in the regulation of sleep remains to be fully addressed, across contexts of health and disease. In this overview article, we will first summarize the literature pertaining to the role of astrocytes and microglia in the regulation of sleep under normal physiological conditions. Afterward, we will discuss the beneficial and deleterious consequences of glia-mediated neuroinflammation, whether it is acute, or chronic and associated with brain diseases, on the regulation of sleep. Sleep disturbances are a main comorbidity in neurodegenerative diseases, and in several brain diseases that include pain, epilepsy, and cancer. Identifying the relationships between glia-mediated neuroinflammation, sleep-wake rhythm disruption and brain diseases may have important implications for the treatment of several disorders. © 2020 American Physiological Society. Compr Physiol 10:687-712, 2020.
Collapse
Affiliation(s)
- Stefano Garofalo
- Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Katherine Picard
- Nutrition et Neurobiologie Intégrée, UMR 1286, Institut National de la Recherche Agronomique, Bordeaux University, Bordeaux, France.,Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| | - Agnès Nadjar
- Nutrition et Neurobiologie Intégrée, UMR 1286, Institut National de la Recherche Agronomique, Bordeaux University, Bordeaux, France
| | - Olivier Pascual
- INSERM U1028, CNRS UMR5292, Lyon Neuroscience Research Center, Université Claude Bernard Lyon, Lyon, France
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada.,Départment de médecine moleculaire, Faculté de médecine, Université Laval, Québec, Quebec, Canada
| |
Collapse
|
13
|
Scavuzzo CJ, Rakotovao I, Dickson CT. Differential effects of L- and D-lactate on memory encoding and consolidation: Potential role of HCAR1 signaling. Neurobiol Learn Mem 2019; 168:107151. [PMID: 31881352 DOI: 10.1016/j.nlm.2019.107151] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 11/14/2019] [Accepted: 12/23/2019] [Indexed: 01/11/2023]
Abstract
The process of memory consolidation is energy-demanding and brain energy deficits result in memory impairments. Indeed, L-lactate, a preferred neuronal energy substrate, enhances the formation of memory, while blockade of the neuronal uptake of L-lactate by either pharmacological means or using its enantiomer D-lactate, impairs memory. Beyond metabolism, both enantiomers of lactate also have signaling properties through the hydroxycarboxylic acid receptor 1 (HCAR1). Thus far, paradigms testing for an effect of lactate on memory modulation have ignored HCAR1 signaling while also mainly performing manipulations before learning and using intracranial administration techniques. Using an inhibitory avoidance (IA) memory protocol, the present study examined the effects of systemic administration of both L- and D-lactate as well as the specific HCAR1 agonist 3,5-dihydroxybenzoic acid (3,5-DHBA) across pre- and post-training periods. We found that post-training subcutaneous injections of either 3,5-DHBA or D-lactate significantly enhanced memory compared to saline controls, whereas L-lactate had no effect, suggesting that HCAR1 signaling in the absence of lactate metabolism supports memory consolidation processes. When administered 15 minutes prior to training, D-lactate and 3,5-DHBA impaired memory compared to saline controls. In contrast, L-lactate treated rats showed memory enhancements as compared to D-lactate-treated rats. Taken together, these results suggest different roles for lactate at different memory stages. It is likely that a metabolic role is at play during learning while HCAR1 signaling may play a greater role during consolidation.
Collapse
Affiliation(s)
- Claire J Scavuzzo
- Department of Psychology, University of Alberta, Edmonton, Alberta T6G 2E9, Canada.
| | - Irina Rakotovao
- Department of Psychology, University of Alberta, Edmonton, Alberta T6G 2E9, Canada
| | - Clayton T Dickson
- Department of Psychology, University of Alberta, Edmonton, Alberta T6G 2E9, Canada; Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta T6G 2E1, Canada; Department of Physiology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| |
Collapse
|
14
|
Carroll CM, Macauley SL. The Interaction Between Sleep and Metabolism in Alzheimer's Disease: Cause or Consequence of Disease? Front Aging Neurosci 2019; 11:258. [PMID: 31616284 PMCID: PMC6764218 DOI: 10.3389/fnagi.2019.00258] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 08/30/2019] [Indexed: 01/21/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and affects over 45 million people worldwide. Both type-2-diabetes (T2D), a metabolic condition associated with aging, and disrupted sleep are implicated in the pathogenesis of AD, but how sleep and metabolism interact to affect AD progression remains unclear. In the healthy brain, sleep/wake cycles are a well-coordinated interaction between metabolic and neuronal activity, but when disrupted, are associated with a myriad of health-related issues, including metabolic syndrome, cardiovascular disease, T2D, and AD. Therefore, this review will explore our current understanding of the relationship between metabolism, sleep, and AD-related pathology to identify the causes and consequences of disease progression in AD. Moreover, sleep disturbances and metabolic dysfunction could serve as potential therapeutic targets to mitigate the increased risk of AD in individuals with T2D or offer a novel approach for treating AD.
Collapse
Affiliation(s)
| | - Shannon L. Macauley
- Section of Gerontology and Geriatric Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
15
|
Bellesi M, Haswell JD, de Vivo L, Marshall W, Roseboom PH, Tononi G, Cirelli C. Myelin modifications after chronic sleep loss in adolescent mice. Sleep 2019; 41:4850494. [PMID: 29741724 DOI: 10.1093/sleep/zsy034] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Indexed: 01/28/2023] Open
Abstract
Study Objectives Previous studies found that sleep loss can suppress the expression of genes implicated in myelination and can have adverse effects on oligodendrocyte precursor cells. On the other hand, sleep may favor myelination by promoting the expression of genes involved in its formation and maintenance. Albeit limited, these results suggest that sleep loss can have detrimental effects on the formation and maintenance of myelin. Methods Here, we tested this hypothesis by evaluating ultrastructural modifications of myelin in two brain regions (corpus callosum and lateral olfactory tract) of mice exposed to different periods of sleep loss, from a few hours of sleep deprivation to ~5 days of chronic sleep restriction. In addition, we measured the internodal length-the distance between consecutive nodes of Ranvier along the axon-and plasma corticosterone levels. Results We find that g-ratio-the ratio of the diameter of the axon itself to the outer diameter of the myelinated fiber-increases after chronic sleep loss. This effect is mediated by a reduction in myelin thickness and is not associated with changes in the internodal length. Relative to sleep, plasma corticosterone levels increase after acute sleep deprivation, but show only a trend to increase after chronic sleep loss. Conclusions Chronic sleep loss may negatively affect myelin.
Collapse
Affiliation(s)
- Michele Bellesi
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI.,Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy
| | | | - Luisa de Vivo
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI
| | - William Marshall
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI
| | | | - Giulio Tononi
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI
| | - Chiara Cirelli
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
16
|
Panagiotou M, Deboer T. Chronic high-caloric diet accentuates age-induced sleep alterations in mice. Behav Brain Res 2019; 362:131-139. [PMID: 30639608 DOI: 10.1016/j.bbr.2019.01.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 01/09/2019] [Accepted: 01/10/2019] [Indexed: 11/15/2022]
Abstract
Obesity and sleep disturbances comprise major health problems which are likely interrelated. Diet-induced obesity in young mice has been demonstrated to lead towards an altered sleep homeostasis. In the current study, we investigated the effect of chronic (12 weeks) high-caloric diet (HCD, 45% fat) consumption on sleep and the sleep electroencephalogram (EEG) in young and older mice (6-month-old, n = 9; 18-month-old, n = 8 and 24-month-old, n = 4) and compared with age-matched controls on normal chow (n = 11, n = 9 and n = 9 respectively). Half of the 24-month-old mice did not cope well with HCD, therefore this group has a lower n and limited statistical power. We recorded EEG and the electromyogram for continuous 48-h and performed a 6-h sleep deprivation during the second day. In aged HCD fed mice (18 months old) compared to young, an aging effect was still evident, characterized by decreased waking and increased NREM sleep in the dark period, decreased REM sleep during the light period, as well as increased slow-wave-activity (SWA, EEG power in NREM sleep in 0.5-4.0 Hz). Additionally, aged HCD treated mice showed increased NREM sleep and decreased waking, compared to age-matched controls, denoting an enhanced aging phenotype in the sleep architecture. Notably, an overall increase was found in the slow component of SWA (0.5-2.5 Hz) in aged HCD fed mice compared to age-matched controls. Our data suggest that the effect of aging is the dominant variable irrespective of diet. However, a synergistic effect of aging and diet is noted indicating that chronic HCD consumption exacerbates age-associated sleep alterations.
Collapse
Affiliation(s)
- M Panagiotou
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Centre, Leiden, the Netherlands
| | - T Deboer
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Centre, Leiden, the Netherlands.
| |
Collapse
|
17
|
Abstract
Mechanisms for elimination of metabolites from ISF include metabolism, blood-brain barrier transport and non-selective, perivascular efflux, this last being assessed by measuring the clearance of markers like inulin. Clearance describes elimination. Clearance of a metabolite generated within the brain is determined as its elimination rate divided by its concentration in interstitial fluid (ISF). However, the more frequently measured parameter is the rate constant for elimination determined as elimination rate divided by amount present, which thus depends on both the elimination processes and the distribution of the metabolite in the brain. The relative importance of the various elimination mechanisms depends on the particular metabolite. Little is known about the effects of sleep on clearance via metabolism or blood-brain barrier transport, but studies with inulin in mice comparing perivascular effluxes during sleep and wakefulness reveal a 4.2-fold increase in clearance. Amongst the important brain metabolites considered, CO2 is eliminated so rapidly across the blood-brain barrier that clearance is blood flow limited and elimination quickly balances production. Glutamate is removed from ISF primarily by uptake into astrocytes and conversion to glutamine, but also by transport across the blood-brain barrier. Both lactate and amyloid-β are eliminated by metabolism, blood-brain barrier transport and perivascular efflux and both show decreased production, decreased ISF concentration and increased perivascular clearance during sleep. Taken altogether available data indicate that sleep increases perivascular and non-perivascular clearances for amyloid-β which reduces its concentration and may have long-term consequences for the formation of plaques and cerebral arterial deposits.
Collapse
Affiliation(s)
- Stephen B Hladky
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK.
| | - Margery A Barrand
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK
| |
Collapse
|
18
|
Abstract
The cellular mechanisms governing the expression, regulation, and function of sleep are not entirely understood. The traditional view is that these mechanisms are neuronal. An alternative view is that glial brain cells may play important roles in these processes. Their ubiquity in the central nervous system makes them well positioned to modulate neuronal circuits that gate sleep and wake. Their ability to respond to chemical neuronal signals suggests that they form feedback loops with neurons that may globally regulate neuronal activity. Their potential role in detoxifying the brain, regulating neuronal metabolism, and promoting synaptic plasticity raises the intriguing possibility that glia mediate important functions ascribed to sleep.
Collapse
Affiliation(s)
- Marcos G Frank
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University Spokane, Spokane, WA, USA.
| |
Collapse
|
19
|
Bourdon AK, Spano GM, Marshall W, Bellesi M, Tononi G, Serra PA, Baghdoyan HA, Lydic R, Campagna SR, Cirelli C. Metabolomic analysis of mouse prefrontal cortex reveals upregulated analytes during wakefulness compared to sleep. Sci Rep 2018; 8:11225. [PMID: 30046159 PMCID: PMC6060152 DOI: 10.1038/s41598-018-29511-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/03/2018] [Indexed: 12/18/2022] Open
Abstract
By identifying endogenous molecules in brain extracellular fluid metabolomics can provide insight into the regulatory mechanisms and functions of sleep. Here we studied how the cortical metabolome changes during sleep, sleep deprivation and spontaneous wakefulness. Mice were implanted with electrodes for chronic sleep/wake recording and with microdialysis probes targeting prefrontal and primary motor cortex. Metabolites were measured using ultra performance liquid chromatography-high resolution mass spectrometry. Sleep/wake changes in metabolites were evaluated using partial least squares discriminant analysis, linear mixed effects model analysis of variance, and machine-learning algorithms. More than 30 known metabolites were reliably detected in most samples. When used by a logistic regression classifier, the profile of these metabolites across sleep, spontaneous wake, and enforced wake was sufficient to assign mice to their correct experimental group (pair-wise) in 80-100% of cases. Eleven of these metabolites showed significantly higher levels in awake than in sleeping mice. Some changes extend previous findings (glutamate, homovanillic acid, lactate, pyruvate, tryptophan, uridine), while others are novel (D-gluconate, N-acetyl-beta-alanine, N-acetylglutamine, orotate, succinate/methylmalonate). The upregulation of the de novo pyrimidine pathway, gluconate shunt and aerobic glycolysis may reflect a wake-dependent need to promote the synthesis of many essential components, from nucleic acids to synaptic membranes.
Collapse
Affiliation(s)
- Allen K Bourdon
- Department of Chemistry, University of Tennessee, Knoxville, TN, United States
| | - Giovanna Maria Spano
- Department of Psychiatry, University of Wisconsin, Madison, Madison, WI, United States
| | - William Marshall
- Department of Psychiatry, University of Wisconsin, Madison, Madison, WI, United States
| | - Michele Bellesi
- Department of Psychiatry, University of Wisconsin, Madison, Madison, WI, United States.,Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy
| | - Giulio Tononi
- Department of Psychiatry, University of Wisconsin, Madison, Madison, WI, United States
| | - Pier Andrea Serra
- Department of Clinical and Experimental Medicine, University of Sassari, Sassari, Italy
| | - Helen A Baghdoyan
- Department of Anesthesiology and Psychology, University of Tennessee, Knoxville, TN, United States.,Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Ralph Lydic
- Department of Anesthesiology and Psychology, University of Tennessee, Knoxville, TN, United States.,Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Shawn R Campagna
- Department of Chemistry, University of Tennessee, Knoxville, TN, United States. .,Biological and Small Molecule Mass Spectrometry Core, University of Tennessee, Knoxville, TN, United States.
| | - Chiara Cirelli
- Department of Psychiatry, University of Wisconsin, Madison, Madison, WI, United States.
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW The goal of the present paper is to review current literature supporting the occurrence of fundamental changes in brain energy metabolism during the transition from wakefulness to sleep. RECENT FINDINGS Latest research in the field indicates that glucose utilization and the concentrations of several brain metabolites consistently change across the sleep-wake cycle. Lactate, a product of glycolysis that is involved in synaptic plasticity, has emerged as a good biomarker of brain state. Sleep-induced changes in cerebral metabolite levels result from a shift in oxidative metabolism, which alters the reliance of brain metabolism upon carbohydrates. We found wide support for the notion that brain energetics is state dependent. In particular, fatty acids and ketone bodies partly replace glucose as cerebral energy source during sleep. This mechanism plausibly accounts for increases in biosynthetic pathways and functional alterations in neuronal activity associated with sleep. A better account of brain energy metabolism during sleep might help elucidate the long mysterious restorative effects of sleep for the whole organism.
Collapse
Affiliation(s)
- Nadia Nielsen Aalling
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, Faculty of Health and Medical Sciences, University of Copenhagen, Nørre Allé 14, 2200, Copenhagen N, Denmark
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, Faculty of Health and Medical Sciences, University of Copenhagen, Nørre Allé 14, 2200, Copenhagen N, Denmark.,Center for Translational Neuromedicine, University of Rochester Medical School, Rochester, NY, 14640, USA
| | - Mauro DiNuzzo
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, Faculty of Health and Medical Sciences, University of Copenhagen, Nørre Allé 14, 2200, Copenhagen N, Denmark.
| |
Collapse
|
21
|
Wigren HK, Porkka-Heiskanen T. Novel concepts in sleep regulation. Acta Physiol (Oxf) 2018; 222:e13017. [PMID: 29253320 DOI: 10.1111/apha.13017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 12/07/2017] [Accepted: 12/08/2017] [Indexed: 12/12/2022]
Abstract
Knowledge regarding the cellular mechanisms of sleep regulation is accumulating rapidly. In addition to neurones, also non-neuronal brain cells (astrocytes and microglia) are emerging as potential players. New techniques, particularly optogenetics and designed receptors activated by artificial ligands (DREADD), have provided also sleep research with important additional tools to study the effect of either silencing or activating specific neuronal groups/neuronal networks by opening or shutting ion channels on cells. The advantages of these strategies are the possibility to genetically target specific cell populations and the possibility to either activate or inhibit them with inducing light signal into the brain. Studies probing circuits of NREM and REM sleep regulation, as well as their role in memory consolidation, have been conducted recently. In addition, fundamentally new thoughts and potential mechanisms have been introduced to the field. The role of non-neuronal tissues in the regulation of many brain functions has become evident. These non-neuronal cells, particularly astrocytes, integrate large number of neurones, and it has been suggested that one of their functions is to integrate the (neural) activity in larger brain areas-a feature that is one of the prominent features of also the state of sleep.
Collapse
Affiliation(s)
- H.-K. Wigren
- Department of Physiology; University of Helsinki; Helsinki Finland
| | | |
Collapse
|
22
|
Wei Q, Ta G, He W, Wang W, Wu Q. Stilbene Glucoside, a Putative Sleep Promoting Constituent from Polygonum multiflorum Affects Sleep Homeostasis by Affecting the Activities of Lactate Dehydrogenase and Salivary Alpha Amylase. Chem Pharm Bull (Tokyo) 2017; 65:1011-1019. [PMID: 29093287 DOI: 10.1248/cpb.c17-00275] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Chinese herbal medicine (CHM) has been used for treating insomnia for centuries. The most used CHM for insomnia was Polygonum multiflorum. However, the molecular mechanism for CHM preventing insomnia is unknown. Stilbene glucoside (THSG), an important active component of P. multiflorum, may play an important role for treating insomnia. To test the hypothesis, Kunming mice were treated with different dosages of THSG. To examine the sleep duration, a computer-controlled sleep-wake detection system was implemented. Electroencephalogram (EEG) and electromyogram (EMG) electrodes were implanted to determine sleep-wake state. RT-PCR and Western blot was used to measure the levels of lactate dehydrogenase (LDH) and saliva alpha amylase. Spearman's rank correlation coefficient was used to identify the strength of correlation between the variables. The results showed that THSG significantly prolonged the sleep time of the mice (p<0.01). THSG changed sleep profile by reducing wake and rapid eye movement (REM) period, and increasing non-REM period. RT-PCR and Western blot analysis showed that THSG could down-regulate the levels of LDH and saliva alpha amylase (p<0.05). The level of lactate and glucose was positively related with the activity of LDH and saliva alpha amylase (p<0.05), respectively. On the other hand, the activities of LDH and amylase were negatively associated with sleep duration (p<0.05). The levels of lactate and glucose affect sleep homeostasis. Thus, THSG may prevent insomnia by regulating sleep duration via LDH and salivary alpha amylase.
Collapse
Affiliation(s)
- Qian Wei
- Heart Disease Diagnosis and Treatment Center, The Affiliated Hospital to Changchun University of Chinese Medicine
| | - Guang Ta
- Department of Emergency and ICU, The Affiliated Hospital to Changchun University of Chinese Medicine
| | - Wenjing He
- Department of Emergency and ICU, The Affiliated Hospital to Changchun University of Chinese Medicine
| | - Wei Wang
- Department of Emergency and ICU, The Affiliated Hospital to Changchun University of Chinese Medicine
| | - Qiucheng Wu
- Department of Emergency and ICU, The Affiliated Hospital to Changchun University of Chinese Medicine
| |
Collapse
|
23
|
DiNuzzo M, Nedergaard M. Brain energetics during the sleep-wake cycle. Curr Opin Neurobiol 2017; 47:65-72. [PMID: 29024871 PMCID: PMC5732842 DOI: 10.1016/j.conb.2017.09.010] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/06/2017] [Accepted: 09/16/2017] [Indexed: 12/11/2022]
Abstract
Brain activity during wakefulness is associated with high metabolic rates that are believed to support information processing and memory encoding. In spite of loss of consciousness, sleep still carries a substantial energy cost. Experimental evidence supports a cerebral metabolic shift taking place during sleep that suppresses aerobic glycolysis, a hallmark of environment-oriented waking behavior and synaptic plasticity. Recent studies reveal that glial astrocytes respond to the reduction of wake-promoting neuromodulators by regulating volume, composition and glymphatic drainage of interstitial fluid. These events are accompanied by changes in neuronal discharge patterns, astrocyte-neuron interactions, synaptic transactions and underlying metabolic features. Internally-generated neuronal activity and network homeostasis are proposed to account for the high sleep-related energy demand.
Collapse
Affiliation(s)
- Mauro DiNuzzo
- Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Maiken Nedergaard
- Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Center for Translational Neuromedicine, University of Rochester Medical School, Rochester, NY 14640, USA
| |
Collapse
|
24
|
Lundgaard I, Lu ML, Yang E, Peng W, Mestre H, Hitomi E, Deane R, Nedergaard M. Glymphatic clearance controls state-dependent changes in brain lactate concentration. J Cereb Blood Flow Metab 2017; 37:2112-2124. [PMID: 27481936 PMCID: PMC5464705 DOI: 10.1177/0271678x16661202] [Citation(s) in RCA: 203] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Brain lactate concentration is higher during wakefulness than in sleep. However, it is unknown why arousal is linked to an increase in brain lactate and why lactate declines within minutes of sleep. Here, we show that the glymphatic system is responsible for state-dependent changes in brain lactate concentration. Suppression of glymphatic function via acetazolamide treatment, cisterna magna puncture, aquaporin 4 deletion, or changes in body position reduced the decline in brain lactate normally observed when awake mice transition into sleep or anesthesia. Concurrently, the same manipulations diminished accumulation of lactate in cervical, but not in inguinal lymph nodes when mice were anesthetized. Thus, our study suggests that brain lactate is an excellent biomarker of the sleep-wake cycle and increases further during sleep deprivation, because brain lactate is inversely correlated with glymphatic-lymphatic clearance. This analysis provides fundamental new insight into brain energy metabolism by demonstrating that glucose that is not fully oxidized can be exported as lactate via glymphatic-lymphatic fluid transport.
Collapse
Affiliation(s)
- Iben Lundgaard
- 1 Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester, Rochester, NY, USA
| | - Minh Lon Lu
- 1 Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester, Rochester, NY, USA.,2 Medical Research Council Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Ezra Yang
- 1 Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester, Rochester, NY, USA
| | - Weiguo Peng
- 1 Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester, Rochester, NY, USA
| | - Humberto Mestre
- 1 Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester, Rochester, NY, USA
| | - Emi Hitomi
- 1 Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester, Rochester, NY, USA
| | - Rashid Deane
- 1 Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester, Rochester, NY, USA
| | - Maiken Nedergaard
- 1 Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester, Rochester, NY, USA.,3 Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
25
|
Korf EM, Mölle M, Born J, Ngo HVV. Blindfolding during wakefulness causes decrease in sleep slow wave activity. Physiol Rep 2017; 5:5/7/e13239. [PMID: 28408638 PMCID: PMC5392525 DOI: 10.14814/phy2.13239] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 03/13/2017] [Indexed: 12/18/2022] Open
Abstract
Slow wave activity (SWA, 0.5–4 Hz) represents the predominant EEG oscillatory activity during slow wave sleep (SWS). Its amplitude is considered in part a reflection of synaptic potentiation in cortical networks due to encoding of information during prior waking, with higher amplitude indicating stronger potentiation. Previous studies showed that increasing and diminishing specific motor behaviors produced corresponding changes in SWA in the respective motor cortical areas during subsequent SWS. Here, we tested whether this relationship can be generalized to the visual system, that is, whether diminishing encoding of visual information likewise leads to a localized decrease in SWA over the visual cortex. Experiments were performed in healthy men whose eyes on two different days were or were not covered for 10.5 h before bedtime. The subject's EEG was recorded during sleep and, after sleep, visual evoked potentials (VEPs) were recorded. SWA during nonrapid eye movement sleep (NonREM sleep) was lower after blindfolding than after eyes open (P < 0.01). The decrease in SWA that was most consistent during the first 20 min of NonREM sleep, did not remain restricted to visual cortex regions, with changes over frontal and parietal cortical regions being even more pronounced. In the morning after sleep, the N75‐P100 peak‐to‐peak‐amplitude of the VEP was significantly diminished in the blindfolded condition. Our findings confirm a link between reduced wake encoding and diminished SWA during ensuing NonREM sleep, although this link appears not to be restricted to sensory cortical areas.
Collapse
Affiliation(s)
| | - Matthias Mölle
- Center of Brain, Behavior and Metabolism University of Lübeck, Lübeck, Germany
| | - Jan Born
- Department of Neuroendocrinology, University of Lübeck, Lübeck, Germany .,Institute for Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany
| | - Hong-Viet V Ngo
- Institute for Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany .,School of Psychology University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
26
|
Abstract
Sleep is a complex physiological process that is regulated globally, regionally, and locally by both cellular and molecular mechanisms. It occurs to some extent in all animals, although sleep expression in lower animals may be co-extensive with rest. Sleep regulation plays an intrinsic part in many behavioral and physiological functions. Currently, all researchers agree there is no single physiological role sleep serves. Nevertheless, it is quite evident that sleep is essential for many vital functions including development, energy conservation, brain waste clearance, modulation of immune responses, cognition, performance, vigilance, disease, and psychological state. This review details the physiological processes involved in sleep regulation and the possible functions that sleep may serve. This description of the brain circuitry, cell types, and molecules involved in sleep regulation is intended to further the reader's understanding of the functions of sleep.
Collapse
Affiliation(s)
- Mark R. Zielinski
- Veterans Affairs Boston Healthcare System, West Roxbury, MA 02132, USA and Harvard Medical School, Department of Psychiatry
| | - James T. McKenna
- Veterans Affairs Boston Healthcare System, West Roxbury, MA 02132, USA and Harvard Medical School, Department of Psychiatry
| | - Robert W. McCarley
- Veterans Affairs Boston Healthcare System, Brockton, MA 02301, USA and Harvard Medical School, Department of Psychiatry
| |
Collapse
|
27
|
de Vivo L, Nelson AB, Bellesi M, Noguti J, Tononi G, Cirelli C. Loss of Sleep Affects the Ultrastructure of Pyramidal Neurons in the Adolescent Mouse Frontal Cortex. Sleep 2016; 39:861-74. [PMID: 26715225 DOI: 10.5665/sleep.5644] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 11/21/2015] [Indexed: 11/03/2022] Open
Abstract
STUDY OBJECTIVE The adolescent brain may be uniquely affected by acute sleep deprivation (ASD) and chronic sleep restriction (CSR), but direct evidence is lacking. We used electron microscopy to examine how ASD and CSR affect pyramidal neurons in the frontal cortex of adolescent mice, focusing on mitochondria, endosomes, and lysosomes that together perform most basic cellular functions, from nutrient intake to prevention of cellular stress. METHODS Adolescent (1-mo-old) mice slept (S) or were sleep deprived (ASD, with novel objects and running wheels) during the first 6-8 h of the light period, chronically sleep restricted (CSR) for > 4 days (using novel objects, running wheels, social interaction, forced locomotion, caffeinated water), or allowed to recover sleep (RS) for ∼32 h after CSR. Ultrastructural analysis of 350 pyramidal neurons was performed (S = 82; ASD = 86; CSR = 103; RS = 79; 4 to 5 mice/group). RESULTS Several ultrastructural parameters differed in S versus ASD, S versus CSR, CSR versus RS, and S versus RS, although the different methods used to enforce wake may have contributed to some of the differences between short and long sleep loss. Differences included larger cytoplasmic area occupied by mitochondria in CSR versus S, and higher number of secondary lysosomes in CSR versus S and RS. We also found that sleep loss may unmask interindividual differences not obvious during baseline sleep. Moreover, using a combination of 11 ultrastructural parameters, we could predict in up to 80% of cases whether sleep or wake occurred at the single cell level. CONCLUSIONS Ultrastructural analysis may be a powerful tool to identify which cellular organelles, and thus which cellular functions, are most affected by sleep and sleep loss.
Collapse
Affiliation(s)
- Luisa de Vivo
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI
| | - Aaron B Nelson
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI
| | - Michele Bellesi
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI
| | - Juliana Noguti
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI
| | - Giulio Tononi
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI
| | - Chiara Cirelli
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
28
|
Bellesi M, Tononi G, Cirelli C, Serra PA. Region-Specific Dissociation between Cortical Noradrenaline Levels and the Sleep/Wake Cycle. Sleep 2016; 39:143-54. [PMID: 26237776 DOI: 10.5665/sleep.5336] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 06/28/2015] [Indexed: 12/20/2022] Open
Abstract
STUDY OBJECTIVES The activity of the noradrenergic system of the locus coeruleus (LC) is high in wake and low in sleep. LC promotes arousal and EEG activation, as well as attention, working memory, and cognitive flexibility. These functions rely on prefrontal cortex and are impaired by sleep deprivation, but the extent to which LC activity changes during wake remains unclear. Moreover, it is unknown whether noradrenergic neurons can sustain elevated firing during extended wake. Recent studies show that relative to LC neurons targeting primary motor cortex (M1), those projecting to medial prefrontal cortex (mPFC) have higher spontaneous firing rates and are more excitable. These results suggest that noradrenaline (NA) levels should be higher in mPFC than M1, and that during prolonged wake LC cells targeting mPFC may fatigue more, but direct evidence is lacking. METHODS We performed in vivo microdialysis experiments in adult (9-10 weeks old) C57BL/6 mice implanted for chronic electroencephalographic recordings. Cortical NA levels were measured during spontaneous sleep and wake (n = 8 mice), and in the course of sleep deprivation (n = 6). RESULTS We found that absolute NA levels are higher in mPFC than in M1. Moreover, in both areas they decline during sleep and increase during wake, but these changes are faster in M1 than mPFC. Finally, by the end of sleep deprivation NA levels decline only in mPFC. CONCLUSIONS Locus coeruleus (LC) neurons targeting prefrontal cortex may fatigue more markedly, or earlier, than other LC cells, suggesting one of the mechanisms underlying the cognitive impairment and the increased sleep presure associated with sleep deprivation. COMMENTARY A commentary on this article appears in this issue on page 11.
Collapse
Affiliation(s)
- Michele Bellesi
- Dept. of Psychiatry, University of Wisconsin-Madison, Madison, WI
| | - Giulio Tononi
- Dept. of Psychiatry, University of Wisconsin-Madison, Madison, WI
| | - Chiara Cirelli
- Dept. of Psychiatry, University of Wisconsin-Madison, Madison, WI
| | - Pier Andrea Serra
- Dept. of Clinical and Experimental Medicine, University of Sassari, Italy
| |
Collapse
|
29
|
Pace M, Baracchi F, Gao B, Bassetti C. Identification of Sleep-Modulated Pathways Involved in Neuroprotection from Stroke. Sleep 2015; 38:1707-18. [PMID: 26085290 DOI: 10.5665/sleep.5148] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 04/13/2015] [Indexed: 12/21/2022] Open
Abstract
STUDY OBJECTIVES Sleep deprivation (SDp) performed before stroke induces an ischemic tolerance state as observed in other forms of preconditioning. As the mechanisms underlying this effect are not well understood, we used DNA oligonucleotide microarray analysis to identify the genes and the gene-pathways underlying SDp preconditioning effects. DESIGN Gene expression was analyzed 3 days after stroke in 4 experimental groups: (i) SDp performed before focal cerebral ischemia (IS) induction; (ii) SDp performed before sham surgery; (iii) IS without SDp; and (iv) sham surgery without SDp. SDp was performed by gentle handling during the last 6 h of the light period, and ischemia was induced immediately after. SETTINGS Basic sleep research laboratory. MEASUREMENTS AND RESULTS Stroke induced a massive alteration in gene expression both in sleep deprived and non-sleep deprived animals. However, compared to animals that underwent ischemia alone, SDp induced a general reduction in transcriptional changes with a reduction in the upregulation of genes involved in cell cycle regulation and immune response. Moreover, an upregulation of a new neuroendocrine pathway which included melanin concentrating hormone, glycoprotein hormones-α-polypeptide and hypocretin was observed exclusively in rats sleep deprived before stroke. CONCLUSION Our data indicate that sleep deprivation before stroke reprogrammed the signaling response to injury. The inhibition of cell cycle regulation and inflammation are neuroprotective mechanisms reported also for other forms of preconditioning treatment, whereas the implication of the neuroendocrine function is novel and has never been described before. These results therefore provide new insights into neuroprotective mechanisms involved in ischemic tolerance mechanisms.
Collapse
Affiliation(s)
- Marta Pace
- ZEN - Zentrum für Experimentelle Neurologie, Inselspital, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Francesca Baracchi
- ZEN - Zentrum für Experimentelle Neurologie, Inselspital, Bern, Switzerland
| | - Bo Gao
- ZEN - Zentrum für Experimentelle Neurologie, Inselspital, Bern, Switzerland
| | - Claudio Bassetti
- ZEN - Zentrum für Experimentelle Neurologie, Inselspital, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| |
Collapse
|
30
|
Bellesi M, de Vivo L, Tononi G, Cirelli C. Effects of sleep and wake on astrocytes: clues from molecular and ultrastructural studies. BMC Biol 2015; 13:66. [PMID: 26303010 PMCID: PMC4548305 DOI: 10.1186/s12915-015-0176-7] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 08/03/2015] [Indexed: 12/22/2022] Open
Abstract
Background Astrocytes can mediate neurovascular coupling, modulate neuronal excitability, and promote synaptic maturation and remodeling. All these functions are likely to be modulated by the sleep/wake cycle, because brain metabolism, neuronal activity and synaptic turnover change as a function of behavioral state. Yet, little is known about the effects of sleep and wake on astrocytes. Results Here we show that sleep and wake strongly affect both astrocytic gene expression and ultrastructure in the mouse brain. Using translating ribosome affinity purification technology and microarrays, we find that 1.4 % of all astrocytic transcripts in the forebrain are dependent on state (three groups, sleep, wake, short sleep deprivation; six mice per group). Sleep upregulates a few select genes, like Cirp and Uba1, whereas wake upregulates many genes related to metabolism, the extracellular matrix and cytoskeleton, including Trio, Synj2 and Gem, which are involved in the elongation of peripheral astrocytic processes. Using serial block face scanning electron microscopy (three groups, sleep, short sleep deprivation, chronic sleep restriction; three mice per group, >100 spines per mouse, 3D), we find that a few hours of wake are sufficient to bring astrocytic processes closer to the synaptic cleft, while chronic sleep restriction also extends the overall astrocytic coverage of the synapse, including at the axon–spine interface, and increases the available astrocytic surface in the neuropil. Conclusions Wake-related changes likely reflect an increased need for glutamate clearance, and are consistent with an overall increase in synaptic strength when sleep is prevented. The reduced astrocytic coverage during sleep, instead, may favor glutamate spillover, thus promoting neuronal synchronization during non-rapid eye movement sleep. Electronic supplementary material The online version of this article (doi:10.1186/s12915-015-0176-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Michele Bellesi
- Department of Psychiatry, University of Wisconsin-Madison, 6001 Research Park Blvd, Madison, WI, 53719, USA.
| | - Luisa de Vivo
- Department of Psychiatry, University of Wisconsin-Madison, 6001 Research Park Blvd, Madison, WI, 53719, USA.
| | - Giulio Tononi
- Department of Psychiatry, University of Wisconsin-Madison, 6001 Research Park Blvd, Madison, WI, 53719, USA.
| | - Chiara Cirelli
- Department of Psychiatry, University of Wisconsin-Madison, 6001 Research Park Blvd, Madison, WI, 53719, USA.
| |
Collapse
|
31
|
Effects of growth hormone-releasing hormone on sleep and brain interstitial fluid amyloid-β in an APP transgenic mouse model. Brain Behav Immun 2015; 47:163-71. [PMID: 25218899 PMCID: PMC4362875 DOI: 10.1016/j.bbi.2014.09.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Revised: 08/13/2014] [Accepted: 09/03/2014] [Indexed: 11/21/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by impairment of cognitive function, extracellular amyloid plaques, intracellular neurofibrillary tangles, and synaptic and neuronal loss. There is substantial evidence that the aggregation of amyloid β (Aβ) in the brain plays a key role in the pathogenesis of AD and that Aβ aggregation is a concentration dependent process. Recently, it was found that Aβ levels in the brain interstitial fluid (ISF) are regulated by the sleep-wake cycle in both humans and mice; ISF Aβ is higher during wakefulness and lower during sleep. Intracerebroventricular infusion of orexin increased wakefulness and ISF Aβ levels, and chronic sleep deprivation significantly increased Aβ plaque formation in amyloid precursor protein transgenic (APP) mice. Growth hormone-releasing hormone (GHRH) is a well-documented sleep regulatory substance which promotes non-rapid eye movement sleep. GHRHR(lit/lit) mice that lack functional GHRH receptor have shorter sleep duration and longer wakefulness during light periods. The current study was undertaken to determine whether manipulating sleep by interfering with GHRH signaling affects brain ISF Aβ levels in APPswe/PS1ΔE9 (PS1APP) transgenic mice that overexpress mutant forms of APP and PSEN1 that cause autosomal dominant AD. We found that intraperitoneal injection of GHRH at dark onset increased sleep and decreased ISF Aβ and that delivery of a GHRH antagonist via reverse-microdialysis suppressed sleep and increased ISF Aβ. The diurnal fluctuation of ISF Aβ in PS1APP/GHRHR(lit/lit) mice was significantly smaller than that in PS1APP/GHRHR(lit/+) mice. However despite decreased sleep in GHRHR deficient mice, this was not associated with an increase in Aβ accumulation later in life. One of several possibilities for the finding is the fact that GHRHR deficient mice have GHRH-dependent but sleep-independent factors which protect against Aβ deposition.
Collapse
|
32
|
Morland C, Lauritzen KH, Puchades M, Holm-Hansen S, Andersson K, Gjedde A, Attramadal H, Storm-Mathisen J, Bergersen LH. The lactate receptor, G-protein-coupled receptor 81/hydroxycarboxylic acid receptor 1: Expression and action in brain. J Neurosci Res 2015; 93:1045-55. [DOI: 10.1002/jnr.23593] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 03/21/2015] [Accepted: 03/23/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Cecilie Morland
- The Brain and Muscle Energy Group; Department of Anatomy; Institute of Basic Medical Sciences, University of Oslo; Oslo Norway
- The Brain and Muscle Energy Group; Department of Oral Biology; University of Oslo; Oslo Norway
| | - Knut Husø Lauritzen
- The Brain and Muscle Energy Group; Department of Anatomy; Institute of Basic Medical Sciences, University of Oslo; Oslo Norway
| | - Maja Puchades
- The Brain and Muscle Energy Group; Department of Anatomy; Institute of Basic Medical Sciences, University of Oslo; Oslo Norway
| | - Signe Holm-Hansen
- Department of Neuroscience and Pharmacology; University of Copenhagen; Copenhagen Denmark
| | - Krister Andersson
- The Brain and Muscle Energy Group; Department of Anatomy; Institute of Basic Medical Sciences, University of Oslo; Oslo Norway
| | - Albert Gjedde
- Department of Neuroscience and Pharmacology; University of Copenhagen; Copenhagen Denmark
| | - Håvard Attramadal
- Institute for Surgical Research, Oslo University Hospital; Oslo Norway
- Center for Heart Failure Research, University of Oslo; Oslo Norway
| | - Jon Storm-Mathisen
- The Brain and Muscle Energy Group; Department of Anatomy; Institute of Basic Medical Sciences, University of Oslo; Oslo Norway
| | - Linda Hildegard Bergersen
- The Brain and Muscle Energy Group; Department of Anatomy; Institute of Basic Medical Sciences, University of Oslo; Oslo Norway
- Department of Neuroscience and Pharmacology; University of Copenhagen; Copenhagen Denmark
- Center for Healthy Aging; Faculty of Health Sciences; University of Copenhagen; Copenhagen Denmark
| |
Collapse
|
33
|
Rempe MJ, Wisor JP. Cerebral lactate dynamics across sleep/wake cycles. Front Comput Neurosci 2015; 8:174. [PMID: 25642184 PMCID: PMC4294128 DOI: 10.3389/fncom.2014.00174] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 12/23/2014] [Indexed: 12/17/2022] Open
Abstract
Cerebral metabolism varies dramatically as a function of sleep state. Brain concentration of lactate, the end product of glucose utilization via glycolysis, varies as a function of sleep state, and like slow wave activity (SWA) in the electroencephalogram (EEG), increases as a function of time spent awake or in rapid eye movement sleep and declines as a function of time spent in slow wave sleep (SWS). We sought to determine whether lactate concentration exhibits homeostatic dynamics akin to those of SWA in SWS. Lactate concentration in the cerebral cortex was measured by indwelling enzymatic biosensors. A set of equations based conceptually on Process S (previously used to quantify the homeostatic dynamics of SWA) was used to predict the sleep/wake state-dependent dynamics of lactate concentration in the cerebral cortex. Additionally, we applied an iterative parameter space-restricting algorithm (the Nelder-Mead method) to reduce computational time to find the optimal values of the free parameters. Compared to an exhaustive search, this algorithm reduced the computation time required by orders of magnitude. We show that state-dependent lactate concentration dynamics can be described by a homeostatic model, but that the optimal time constants for describing lactate dynamics are much smaller than those of SWA. This disconnect between lactate dynamics and SWA dynamics does not support the concept that lactate concentration is a biochemical mediator of sleep homeostasis. However, lactate synthesis in the cerebral cortex may nonetheless be informative with regard to sleep function, since the impact of glycolysis on sleep slow wave regulation is only just now being investigated.
Collapse
Affiliation(s)
- Michael J Rempe
- Mathematics and Computer Science, Whitworth University Spokane, WA, USA ; Department of Integrative Physiology and Neuroscience, College of Medical Sciences, Washington State University Spokane Spokane, WA, USA
| | - Jonathan P Wisor
- Department of Integrative Physiology and Neuroscience, College of Medical Sciences, Washington State University Spokane Spokane, WA, USA
| |
Collapse
|
34
|
Rempe MJ, Clegern WC, Wisor JP. An automated sleep-state classification algorithm for quantifying sleep timing and sleep-dependent dynamics of electroencephalographic and cerebral metabolic parameters. Nat Sci Sleep 2015; 7:85-99. [PMID: 26366107 PMCID: PMC4562753 DOI: 10.2147/nss.s84548] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION Rodent sleep research uses electroencephalography (EEG) and electromyography (EMG) to determine the sleep state of an animal at any given time. EEG and EMG signals, typically sampled at >100 Hz, are segmented arbitrarily into epochs of equal duration (usually 2-10 seconds), and each epoch is scored as wake, slow-wave sleep (SWS), or rapid-eye-movement sleep (REMS), on the basis of visual inspection. Automated state scoring can minimize the burden associated with state and thereby facilitate the use of shorter epoch durations. METHODS We developed a semiautomated state-scoring procedure that uses a combination of principal component analysis and naïve Bayes classification, with the EEG and EMG as inputs. We validated this algorithm against human-scored sleep-state scoring of data from C57BL/6J and BALB/CJ mice. We then applied a general homeostatic model to characterize the state-dependent dynamics of sleep slow-wave activity and cerebral glycolytic flux, measured as lactate concentration. RESULTS More than 89% of epochs scored as wake or SWS by the human were scored as the same state by the machine, whether scoring in 2-second or 10-second epochs. The majority of epochs scored as REMS by the human were also scored as REMS by the machine. However, of epochs scored as REMS by the human, more than 10% were scored as SWS by the machine and 18 (10-second epochs) to 28% (2-second epochs) were scored as wake. These biases were not strain-specific, as strain differences in sleep-state timing relative to the light/dark cycle, EEG power spectral profiles, and the homeostatic dynamics of both slow waves and lactate were detected equally effectively with the automated method or the manual scoring method. Error associated with mathematical modeling of temporal dynamics of both EEG slow-wave activity and cerebral lactate either did not differ significantly when state scoring was done with automated versus visual scoring, or was reduced with automated state scoring relative to manual classification. CONCLUSIONS Machine scoring is as effective as human scoring in detecting experimental effects in rodent sleep studies. Automated scoring is an efficient alternative to visual inspection in studies of strain differences in sleep and the temporal dynamics of sleep-related physiological parameters.
Collapse
Affiliation(s)
- Michael J Rempe
- Mathematics and Computer Science, Whitworth University, Spokane, WA, USA ; College of Medical Sciences and Sleep and Performance Research Center, Washington State University, Spokane, WA, USA
| | - William C Clegern
- College of Medical Sciences and Sleep and Performance Research Center, Washington State University, Spokane, WA, USA
| | - Jonathan P Wisor
- College of Medical Sciences and Sleep and Performance Research Center, Washington State University, Spokane, WA, USA
| |
Collapse
|
35
|
Jakubcakova V, Curzi ML, Flachskamm C, Hambsch B, Landgraf R, Kimura M. The glycolytic metabolite methylglyoxal induces changes in vigilance by generating low-amplitude non-REM sleep. J Psychopharmacol 2013; 27:1070-5. [PMID: 23828824 DOI: 10.1177/0269881113495596] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Methylglyoxal (MG), an essential by-product of glycolysis, is a highly reactive endogenous α-oxoaldehyde. Although high levels of MG are cytotoxic, physiological doses of MG were shown to reduce anxiety-related behavior through selective activation of γ-aminobutyric acid type A (GABAA) receptors. Because the latter play a major role in sleep induction, this study examined the potential of MG to regulate sleep. Specifically, we assessed how MG influences sleep-wake behavior in CD1 mice that received intracerebroventricular injections of either vehicle or 0.7 µmol MG at onset of darkness. We used electroencephalogram (EEG) and electromyogram (EMG) recordings to monitor changes in vigilance states, sleep architecture and the EEG spectrum, for 24 h after receipt of injections. Administration of MG rapidly induced non-rapid eye movement sleep (NREMS) and, concomitantly, decreased wakefulness and suppressed EEG delta power during NREMS. In addition, MG robustly enhanced the amount and number of episodes of an unclassified state of vigilance in which EMG, as well as EEG delta and theta power, were very low. MG did not affect overall rapid eye movement sleep (REMS) in a given 24-h period, but significantly reduced the power of theta activity during REMS. Our results provide the first evidence that MG can exert sleep-promoting properties by triggering low-amplitude NREMS.
Collapse
|
36
|
Petit JM, Gyger J, Burlet-Godinot S, Fiumelli H, Martin JL, Magistretti PJ. Genes involved in the astrocyte-neuron lactate shuttle (ANLS) are specifically regulated in cortical astrocytes following sleep deprivation in mice. Sleep 2013; 36:1445-58. [PMID: 24082304 DOI: 10.5665/sleep.3034] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
STUDY OBJECTIVES There is growing evidence indicating that in order to meet the neuronal energy demands, astrocytes provide lactate as an energy substrate for neurons through a mechanism called "astrocyte-neuron lactate shuttle" (ANLS). Since neuronal activity changes dramatically during vigilance states, we hypothesized that the ANLS may be regulated during the sleep-wake cycle. To test this hypothesis we investigated the expression of genes associated with the ANLS specifically in astrocytes following sleep deprivation. Astrocytes were purified by fluorescence-activated cell sorting from transgenic mice expressing the green fluorescent protein (GFP) under the control of the human astrocytic GFAP-promoter. DESIGN 6-hour instrumental sleep deprivation (TSD). SETTING Animal sleep research laboratory. PARTICIPANTS Young (P23-P27) FVB/N-Tg (GFAP-GFP) 14Mes/J (Tg) mice of both sexes and 7-8 week male Tg and FVB/Nj mice. INTERVENTIONS Basal sleep recordings and sleep deprivation achieved using a modified cage where animals were gently forced to move. MEASUREMENTS AND RESULTS Since Tg and FVB/Nj mice displayed a similar sleep-wake pattern, we performed a TSD in young Tg mice. Total RNA was extracted from the GFP-positive and GFP-negative cells sorted from cerebral cortex. Quantitative RT-PCR analysis showed that levels of Glut1, α-2-Na/K pump, Glt1, and Ldha mRNAs were significantly increased following TSD in GFP-positive cells. In GFP-negative cells, a tendency to increase, although not significant, was observed for Ldha, Mct2, and α-3-Na/K pump mRNAs. CONCLUSIONS This study shows that TSD induces the expression of genes associated with ANLS specifically in astrocytes, underlying the important role of astrocytes in the maintenance of the neuro-metabolic coupling across the sleep-wake cycle.
Collapse
Affiliation(s)
- Jean-Marie Petit
- LNDC, Brain Mind Institute, Life Sciences Faculty, Swiss Federal Institute of Technology, Lausanne, Switzerland ; Center for Psychiatric Neuroscience, Department of Psychiatry CHUV, Prilly, Switzerland
| | | | | | | | | | | |
Collapse
|
37
|
Kostin A, McGinty D, Szymusiak R, Alam MN. Sleep-wake and diurnal modulation of nitric oxide in the perifornical-lateral hypothalamic area: real-time detection in freely behaving rats. Neuroscience 2013; 254:275-84. [PMID: 24056193 DOI: 10.1016/j.neuroscience.2013.09.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 09/09/2013] [Accepted: 09/10/2013] [Indexed: 01/12/2023]
Abstract
Nitric oxide (NO) has been implicated in the regulation of sleep. The perifornical-lateral hypothalamic area (PF-LHA) is a key wake-promoting region and contains neurons that are active during behavioral or cortical activation. Recently, we found higher levels of NO metabolites (NOx), an indirect measure of NO levels, in the PF-LHA during prolonged waking (SD). However, NO is highly reactive and diffuses rapidly and the NOx assay is not sensitive enough to detect rapid-changes in NO levels across spontaneous sleep-waking states. We used a novel Nafion®-modified Platinum (NF-PT) electrode for real-time detection of NO levels in the PF-LHA across sleep-wake cycles, dark-light phases, and during SD. Sprague-Dawley male rats were surgically prepared for chronic sleep-wake recording and implantation of NF-PT electrode into the PF-LHA. Electroencephalogram (EEG), electromyogram (EMG), and electrochemical current generated by NF-PT electrode were continuously acquired for 5-7days including one day with 3h of SD. In the PF-LHA, NO levels exhibited a waking>rapid eye movement (REM)>non-rapid eye movement (nonREM) sleep pattern (0.56±0.03μM>0.47±0.02μM>0.42±0.02μM; p<0.01). NO levels were also higher during the dark- as compared to the light-phase (0.53±0.03μM vs. 0.44±0.02μM; p<0.01). NO levels increased during 3h of SD as compared to undisturbed control (0.58±0.04μM vs. 0.47±0.01μM; p<0.05). The findings indicate that in the PF-LHA, NO is produced during behavioral or cortical activation. Since elevated levels of NO inhibits most of the PF-LHA neurons that are active during cortical activation, these findings support a hypothesis that NO produced in conjunction with the activation of PF-LHA neurons during waking/SD, inhibits the same neuronal population to promote sleep.
Collapse
Affiliation(s)
- A Kostin
- Research Service, Veterans Affairs Greater Los Angeles Healthcare System, Sepulveda, CA 91343, USA
| | | | | | | |
Collapse
|
38
|
The changing brain--insights into the mechanisms of neural and behavioral adaptation to the environment. Neuroscience 2013; 247:412-22. [PMID: 23602885 DOI: 10.1016/j.neuroscience.2013.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The Kavli Prize in Neuroscience was awarded for the third time in September 2012, by the Norwegian Academy of Science and Letters in Oslo. The accompanying Kavli Prize Symposium on Neuroscience, held in Bergen and Trondheim, was a showcase of excellence in neuroscience research. The common theme of the Symposium presentations was the mechanisms by which animals adapt to their environment. The symposium speakers--Michael Greenberg, Erin Schuman, Chiara Cirelli, Michael Meaney, Catherine Dulac, Hopi Hoekstra, and Stanislas Dehaene--covered topics ranging from the molecular and cellular levels to the systems level and behavior. Thus a single amino acid change in a transcriptional repressor can disrupt gene regulation through neural activity (Greenberg). Deep sequencing analysis of the neuropil transcriptome indicates that a large fraction of the synaptic proteome is synthesized in situ in axons and dendrites, permitting local regulation (Schuman). The nature of the 'reset' function that makes animals dependent of sleep is being revealed (Cirelli). Maternal behavior can cause changes in gene expression that stably modify behavior in the offspring (Meaney). Removal of a single sensory channel protein in the vomero-nasal organ can switch off male-specific and switch on female-specific innate behavior of mice in response to environmental stimulation (Dulac). Innate behaviors can be stably transmitted from parent to offspring through generations even when those behaviors cannot be expressed, as illustrated by the elaborate burrowing behavior in a rodent species, in which independent genetic regions regulate distinct modules of the burrowing pattern (Hoekstra). Finally, at the other extreme of the nature-nurture scale, functional magnetic resonance imaging (fMRI) analysis in children and adults identified a brain area specifically involved in reading (Dehaene). As the area must originally have developed for a purpose other than reading, such as shape recognition, this illustrates the use of a previously formed neural structure to tackle a new challenge.
Collapse
|
39
|
Porkka-Heiskanen T, Zitting KM, Wigren HK. Sleep, its regulation and possible mechanisms of sleep disturbances. Acta Physiol (Oxf) 2013; 208:311-28. [PMID: 23746394 DOI: 10.1111/apha.12134] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 05/16/2013] [Accepted: 06/04/2013] [Indexed: 12/22/2022]
Abstract
The state of sleep consists of different phases that proceed in successive, tightly regulated order through the night forming a physiological program, which for each individual is different but stabile from one night to another. Failure to accomplish this program results in feeling of unrefreshing sleep and tiredness in the morning. The program core is constructed by genetic factors but regulated by circadian rhythm and duration and intensity of day time brain activity. Many environmental factors modulate sleep, including stress, health status and ingestion of vigilance-affecting nutrients or medicines (e.g. caffeine). Acute sleep loss results in compromised cognitive performance, memory deficits, depressive mood and involuntary sleep episodes during the day. Moreover, prolonged sleep curtailment has many adverse health effects, as evidenced by both epidemiological and experimental studies. These effects include increased risk for depression, type II diabetes, obesity and cardiovascular diseases. In addition to voluntary restriction of sleep, shift work, irregular working hours, jet lag and stress are important factors that induce curtailed or bad quality sleep and/or insomnia. This review covers the current theories on the function of normal sleep and describes current knowledge on the physiologic effects of sleep loss. It provides insights into the basic mechanisms of the regulation of wakefulness and sleep creating a theoretical background for understanding different disturbances of sleep.
Collapse
Affiliation(s)
| | - K.-M. Zitting
- Institute of Biomedicine; University of Helsinki; Helsinki; Finland
| | - H.-K. Wigren
- Institute of Biomedicine; University of Helsinki; Helsinki; Finland
| |
Collapse
|
40
|
Frank MG. Astroglial regulation of sleep homeostasis. Curr Opin Neurobiol 2013; 23:812-8. [PMID: 23518138 DOI: 10.1016/j.conb.2013.02.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 02/20/2013] [Accepted: 02/21/2013] [Indexed: 12/12/2022]
Abstract
Mammalian sleep is regulated by two distinct mechanisms. A circadian oscillator provides timing signals that organize sleep and wake across the 24 hour day. A homeostatic mechanism increases sleep drive and sleep amounts (or intensity) as a function of prior time awake. The cellular mechanisms of sleep homeostasis are poorly defined, but are thought to be primarily neuronal. According to one view, sleep homeostasis arises from interactions between subcortical neurons that register sleep pressure and other neurons that promote either sleep or wakefulness. Alternatively, sleep drive may arise independently among neurons throughout the brain in a use-dependent fashion. Implicit in both views is the idea that sleep homeostasis is solely the product of neurons. In this article, I discuss an emerging view that glial astrocytes may play an essential role in sleep homeostasis.
Collapse
Affiliation(s)
- Marcos G Frank
- University of Pennsylvania, Perelman School of Medicine, Department of Neuroscience, 215 Stemmler Hall, 35th & Hamilton Walk, Philadelphia, PA 19104-6074, United States.
| |
Collapse
|
41
|
Dash MB, Bellesi M, Tononi G, Cirelli C. Sleep/wake dependent changes in cortical glucose concentrations. J Neurochem 2012; 124:79-89. [PMID: 23106535 DOI: 10.1111/jnc.12063] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 09/17/2012] [Accepted: 10/12/2012] [Indexed: 11/30/2022]
Abstract
Most of the energy in the brain comes from glucose and supports glutamatergic activity. The firing rate of cortical glutamatergic neurons, as well as cortical extracellular glutamate levels, increase with time spent awake and decline throughout non rapid eye movement sleep, raising the question whether glucose levels reflect behavioral state and sleep/wake history. Here chronic (2-3 days) electroencephalographic recordings in the rat cerebral cortex were coupled with fixed-potential amperometry to monitor the extracellular concentration of glucose ([gluc]) on a second-by-second basis across the spontaneous sleep-wake cycle and in response to 3 h of sleep deprivation. [Gluc] progressively increased during non rapid eye movement sleep and declined during rapid eye movement sleep, while during wake an early decline in [gluc] was followed by an increase 8-15 min after awakening. There was a significant time of day effect during the dark phase, when rats are mostly awake, with [gluc] being significantly lower during the last 3-4 h of the night relative to the first 3-4 h. Moreover, the duration of the early phase of [gluc] decline during wake was longer after prolonged wake than after consolidated sleep. Thus, the sleep/wake history may affect the levels of glucose available to the brain upon awakening.
Collapse
Affiliation(s)
- Michael B Dash
- Department of Psychiatry, University of Wisconsin, Madison, WI 53719, USA
| | | | | | | |
Collapse
|
42
|
Bellesi M, Vyazovskiy VV, Tononi G, Cirelli C, Conti F. Reduction of EEG theta power and changes in motor activity in rats treated with ceftriaxone. PLoS One 2012; 7:e34139. [PMID: 22479544 PMCID: PMC3316604 DOI: 10.1371/journal.pone.0034139] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 02/22/2012] [Indexed: 12/31/2022] Open
Abstract
The glutamate transporter GLT-1 is responsible for the largest proportion of total glutamate transport. Recently, it has been demonstrated that ceftriaxone (CEF) robustly increases GLT-1 expression. In addition, physiological studies have shown that GLT-1 up-regulation strongly affects synaptic plasticity, and leads to an impairment of the prepulse inhibition, a simple form of information processing, thus suggesting that GLT-1 over-expression may lead to dysfunctions of large populations of neurons. To test this possibility, we assessed whether CEF affects cortical electrical activity by using chronic electroencephalographic (EEG) recordings in male WKY rats. Spectral analysis showed that 8 days of CEF treatment resulted in a delayed reduction in EEG theta power (7–9 Hz) in both frontal and parietal derivations. This decrease peaked at day 10, i.e., 2 days after the end of treatment, and disappeared by day 16. In addition, we found that the same CEF treatment increased motor activity, especially when EEG changes are more prominent. Taken together, these data indicate that GLT-1 up-regulation, by modulating glutamatergic transmission, impairs the activity of widespread neural circuits. In addition, the increased motor activity and prepulse inhibition alterations previously described suggest that neural circuits involved in sensorimotor control are particularly sensitive to GLT-1 up-regulation.
Collapse
Affiliation(s)
- Michele Bellesi
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy
| | | | | | | | | |
Collapse
|