1
|
Schaefke B, Li J, Zhao B, Wang L, Tseng YT. Slumber under pressure: REM sleep and stress response. Prog Neurobiol 2025; 249:102771. [PMID: 40273975 DOI: 10.1016/j.pneurobio.2025.102771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 04/17/2025] [Accepted: 04/17/2025] [Indexed: 04/26/2025]
Abstract
Sleep, a state of reduced responsiveness and distinct brain activity, is crucial across the animal kingdom. This review explores the potential adaptive functions of REM sleep in adapting to stress, emphasizing its role in memory consolidation, emotional regulation, and threat processing. We further explore the underlying neural mechanisms linking stress responses to REM sleep. By synthesizing current findings, we propose that REM sleep allows animals to "rehearse" or simulate responses to danger in a secure, offline state, while also maintaining emotional balance. Environmental factors, such as predation risk and social dynamics, further influence REM sleep. This modulation may enhance survival by optimizing stress responses while fulfilling physiological needs in animals. Insights into REM sleep's role in animals may shed light on human sleep in the context of modern stressors and sleep disruptions. This review also explores the complex interplay between stress, immunity, sleep disruptions-particularly involving REM sleep-and their evolutionary underpinnings.
Collapse
Affiliation(s)
- Bernhard Schaefke
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Jingfei Li
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Science, Beijing 10049, China
| | - Binghao Zhao
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Liping Wang
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, China.
| | - Yu-Ting Tseng
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, China.
| |
Collapse
|
2
|
Ammothumkandy A, Cayce A, Shariq M, Bonaguidi MA. Astroglia's role in synchronized spontaneous neuronal activity: from physiology to pathology. Front Cell Neurosci 2025; 19:1544460. [PMID: 40177583 PMCID: PMC11961896 DOI: 10.3389/fncel.2025.1544460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
The nervous system relies on a balance of excitatory and inhibitory signals. Aberrant neuronal hyperactivity is a pathological phenotype associated with several neurological disorders, with its most severe effects observed in epilepsy patients. This review explores the literature on spontaneous synchronized neuronal activity, its physiological role, and its aberrant forms in disease. Emphasizing the importance of targeting underlying disease mechanisms beyond traditional neuron-focused therapies, the review delves into the role of astroglia in epilepsy progression. We detail how astroglia transitions from a normal to a pathological state, leading to epileptogenic seizures and cognitive decline. Astroglia activity is correlated with epileptiform activity in both animal models and human tissue, indicating their potential role in seizure induction and modulation. Understanding astroglia's dual beneficial and detrimental roles could lead to novel treatments for epilepsy and other neurological disorders with aberrant neuronal activity as the underlying disease substrate.
Collapse
Affiliation(s)
- Aswathy Ammothumkandy
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, United States
| | - Alisha Cayce
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, United States
| | - Mohammad Shariq
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, United States
| | - Michael A. Bonaguidi
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, United States
- Keck School of Medicine, Neurorestoration Center, University of Southern California, Los Angeles, CA, United States
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
- Department of Gerontology, University of Southern California, Los Angeles, CA, United States
- Keck School of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
3
|
Tiwari J, Sur S, Naseem A, Gupta P, Fatima S, Rani S, Malik S. Effect of daytime light intensity on daily behaviours and concurrent hypothalamic gene expressions in migratory redheaded bunting. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 262:113081. [PMID: 39689406 DOI: 10.1016/j.jphotobiol.2024.113081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/25/2024] [Accepted: 12/07/2024] [Indexed: 12/19/2024]
Abstract
Animals use photic cues to time their daily and seasonal activity. The role of photoperiod has been much investigated in seasonal responses, but the role of light intensity is less understood in passerine finches. We investigated if and how daytime light intensity influences photoinduced migratory phenologies and hypothalamic mRNA expressions in a Palearctic-Indian migratory finch, redheaded bunting (Emberiza bruniceps). Photoperiodic manipulations were employed to induce winter-nonmigratory (NM), premigratory (PM), and migratory (MIG) states in photosensitive buntings. In each life history state, the birds were further subjected to 0.055 (low), 0.277 (medium), or 1.11 W/m2 (high) (N = 5 each) light intensity treatment. The low daytime light intensity dampened the locomotor activity rhythm and delayed the onset of Zugunruhe. We found life history-dependant but not light intensity-dependant changes in body mass, fat score, and testis volume. Plasma corticosterone levels were increased under the low-light intensity group in the migratory state. The buntings were foraging throughout the night in the migratory state, aiding body fattening. Front and back sleep were drastically reduced during the migratory phase under all three light intensities. In the migratory state, we found elevated hypothalamic IL1B and IL6 expression in medium and high-light intensity groups, which had significantly reduced sleep duration. In the winter nonmigratory state, the expression of CAMK2 correlated with daytime activity and active wakefulness of buntings. The decreased GHRH expression correlates with the reduction in total sleep in migrating buntings. Overall, daytime light intensity emerges as a key factor that fine-tunes the photoperiodic response and regulates active and sleep behaviour in migratory buntings.
Collapse
Affiliation(s)
- Jyoti Tiwari
- Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh 226007, India
| | - Sayantan Sur
- Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh 226007, India; School of Biodiversity, One Health & Veterinary Medicine, University of Glasgow, Glasgow, United Kingdom
| | - Asma Naseem
- Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh 226007, India
| | - Preeti Gupta
- Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh 226007, India; Department of Physiology, King George's Medical University, Lucknow 226003, India
| | - Shirin Fatima
- Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh 226007, India
| | - Sangeeta Rani
- Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh 226007, India
| | - Shalie Malik
- Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh 226007, India.
| |
Collapse
|
4
|
Zhu Y, Ma J, Li Y, Gu M, Feng X, Shao Y, Tan L, Lou HF, Sun L, Liu Y, Zeng LH, Qiu Z, Li XM, Duan S, Yu YQ. Adenosine-Dependent Arousal Induced by Astrocytes in a Brainstem Circuit. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407706. [PMID: 39494592 DOI: 10.1002/advs.202407706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/20/2024] [Indexed: 11/05/2024]
Abstract
Astrocytes play a crucial role in regulating sleep-wake behavior. However, how astrocytes govern a specific sleep-arousal circuit remains unknown. Here, the authors show that parafacial zone (PZ) astrocytes responded to sleep-wake cycles with state-differential Ca2+ activity, peaking during transitions from sleep to wakefulness. Using chemogenetic and optogenetic approaches, they find that activating PZ astrocytes elicited and sustained wakefulness by prolonging arousal episodes while impeding transitions from wakefulness to non-rapid eye movement (NREM) sleep. Activation of PZ astrocytes specially induced the elevation of extracellular adenosine through the ATP hydrolysis pathway but not equilibrative nucleoside transporter (ENT) mediated transportation. Strikingly, the rise in adenosine levels induced arousal by activating A1 receptors, suggesting a distinct role for adenosine in the PZ beyond its conventional sleep homeostasis modulation observed in the basal forebrain (BF) and cortex. Moreover, at the circuit level, PZ astrocyte activation induced arousal by suppressing the GABA release from the PZGABA neurons, which promote NREM sleep and project to the parabrachial nucleus (PB). Thus, their study unveils a distinctive arousal-promoting effect of astrocytes within the PZ through extracellular adenosine and elucidates the underlying mechanism at the neural circuit level.
Collapse
Affiliation(s)
- Yuwei Zhu
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, 311121, China
| | - Jiale Ma
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Nanhu Brain-computer Interface Institute, Hangzhou, 311100, China
| | - Yulan Li
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Mengyang Gu
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xiang Feng
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, 311121, China
| | - Yujin Shao
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Lei Tan
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Hui-Fang Lou
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Li Sun
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, 311121, China
| | - Yijun Liu
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, 311121, China
| | - Ling-Hui Zeng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Zilong Qiu
- Department of Neurology, Songjiang Hospital, Songjiang Research Institute, MOE-Shanghai Key Laboratory for Children's Environmental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiao-Ming Li
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, 311121, China
- Nanhu Brain-computer Interface Institute, Hangzhou, 311100, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, 310058, China
| | - Shumin Duan
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, 311121, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
- Department of Neurology, Songjiang Hospital, Songjiang Research Institute, MOE-Shanghai Key Laboratory for Children's Environmental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, 310058, China
| | - Yan-Qin Yu
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, 311121, China
- Nanhu Brain-computer Interface Institute, Hangzhou, 311100, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
5
|
Chen J, Peng G, Sun B. Alzheimer's disease and sleep disorders: A bidirectional relationship. Neuroscience 2024; 557:12-23. [PMID: 39137870 DOI: 10.1016/j.neuroscience.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 08/15/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent dementia, pathologically featuring abnormal accumulation of amyloid-β (Aβ) and hyperphosphorylated tau, while sleep, divided into rapid eye movement sleep (REM) and nonrapid eye movement sleep (NREM), plays a key role in consolidating social and spatial memory. Emerging evidence has revealed that sleep disorders such as circadian disturbances and disruption of neuronal rhythm activity are considered as both candidate risks and consequence of AD, suggesting a bidirectional relationship between sleep and AD. This review will firstly grasp basic knowledge of AD pathogenesis, then highlight macrostructural and microstructural alteration of sleep along with AD progression, explain the interaction between accumulation of Aβ and hyperphosphorylated tau, which are two critical neuropathological processes of AD, as well as neuroinflammation and sleep, and finally introduce several methods of sleep enhancement as strategies to reduce AD-associated neuropathology. Although theories about the bidirectional relationship and relevant therapeutic methods in mice have been well developed in recent years, the knowledge in human is still limited. More studies on how to effectively ameliorate AD pathology in patients by sleep enhancement and what specific roles of sleep play in AD are needed.
Collapse
Affiliation(s)
- Junhua Chen
- Chu Kochen Honors College of Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | - Guoping Peng
- Department of Neurology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China.
| | - Binggui Sun
- Department of Anesthesiology of the Children's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Zhejiang University, Hangzhou, Zhejiang Province 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University Hangzhou, Zhejiang Province 310058, China.
| |
Collapse
|
6
|
Huang L, Zhu W, Li N, Zhang B, Dai W, Li S, Xu H. Functions and mechanisms of adenosine and its receptors in sleep regulation. Sleep Med 2024; 115:210-217. [PMID: 38373361 DOI: 10.1016/j.sleep.2024.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/01/2024] [Accepted: 02/03/2024] [Indexed: 02/21/2024]
Abstract
Sleep is a natural and recurring state of life. Long-term insomnia can lead to physical and mental fatigue, inattention, memory loss, anxiety, depression and other symptoms, imposing immense public health and economic burden worldwide. The sleep and awakening regulation system is composed of many nerve nuclei and neurotransmitters in the brain, and it forms a neural network that interacts and restricts each other to regulate the occurrence and maintenance of sleep-wake. Adenosine (AD) is a neurotransmitter in the central nervous system and a driver of sleep. Meanwhile, the functions and mechanisms underlying sleep-promoting effects of adenosine and its receptors are still not entirely clear. However, in recent years, the increasing evidence indicated that adenosine can promote sleep through inhibiting arousal system and activating sleep-promoting system. At the same time, astrocyte-derived adenosine in modulating sleep homeostasis and sleep loss-induced related cognitive and memory deficits plays an important role. This review, therefore, summarizes the current research on the functions and possible mechanisms of adenosine and its receptors in the regulation of sleep and homeostatic control of sleep. Understanding these aspects will provide us better ideas on clinical problems such as insomnia, hypersomnia and other sleep disorders.
Collapse
Affiliation(s)
- Lishan Huang
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Wenwen Zhu
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Nanxi Li
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Bin Zhang
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Wenbin Dai
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Sen Li
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, China.
| | - Houping Xu
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
7
|
Postnov D, Semyachkina-Glushkovskaya O, Litvinenko E, Kurths J, Penzel T. Mechanisms of Activation of Brain's Drainage during Sleep: The Nightlife of Astrocytes. Cells 2023; 12:2667. [PMID: 37998402 PMCID: PMC10670149 DOI: 10.3390/cells12222667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023] Open
Abstract
The study of functions, mechanisms of generation, and pathways of movement of cerebral fluids has a long history, but the last decade has been especially productive. The proposed glymphatic hypothesis, which suggests a mechanism of the brain waste removal system (BWRS), caused an active discussion on both the criticism of some of the perspectives and our intensive study of new experimental facts. It was especially found that the intensity of the metabolite clearance changes significantly during the transition between sleep and wakefulness. Interestingly, at the cellular level, a number of aspects of this problem have been focused on, such as astrocytes-glial cells, which, over the past two decades, have been recognized as equal partners of neurons and perform many important functions. In particular, an important role was assigned to astrocytes within the framework of the glymphatic hypothesis. In this review, we return to the "astrocytocentric" view of the BWRS function and the explanation of its activation during sleep from the viewpoint of new findings over the last decade. Our main conclusion is that the BWRS's action may be analyzed both at the systemic (whole-brain) and at the local (cellular) level. The local level means here that the neuro-glial-vascular unit can also be regarded as the smallest functional unit of sleep, and therefore, the smallest functional unit of the BWRS.
Collapse
Affiliation(s)
- Dmitry Postnov
- Department of Optics and Biophotonics, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia;
| | - Oxana Semyachkina-Glushkovskaya
- Department of Biology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia; (O.S.-G.); (J.K.)
- Physics Department, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany
| | - Elena Litvinenko
- Department of Optics and Biophotonics, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia;
| | - Jürgen Kurths
- Department of Biology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia; (O.S.-G.); (J.K.)
- Physics Department, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany
- Potsdam Institute for Climate Impact Research, Telegrafenberg A31, 14473 Potsdam, Germany
| | - Thomas Penzel
- Department of Biology, Saratov State University, Astrakhanskaya Str. 83, 410012 Saratov, Russia; (O.S.-G.); (J.K.)
- Charité — Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
8
|
Brazhe A, Verisokin A, Verveyko D, Postnov D. Astrocytes: new evidence, new models, new roles. Biophys Rev 2023; 15:1303-1333. [PMID: 37975000 PMCID: PMC10643736 DOI: 10.1007/s12551-023-01145-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 09/08/2023] [Indexed: 11/19/2023] Open
Abstract
Astrocytes have been in the limelight of active research for about 3 decades now. Over this period, ideas about their function and role in the nervous system have evolved from simple assistance in energy supply and homeostasis maintenance to a complex informational and metabolic hub that integrates data on local neuronal activity, sensory and arousal context, and orchestrates many crucial processes in the brain. Rapid progress in experimental techniques and data analysis produces a growing body of data, which can be used as a foundation for formulation of new hypotheses, building new refined mathematical models, and ultimately should lead to a new level of understanding of the contribution of astrocytes to the cognitive tasks performed by the brain. Here, we highlight recent progress in astrocyte research, which we believe expands our understanding of how low-level signaling at a cellular level builds up to processes at the level of the whole brain and animal behavior. We start our review with revisiting data on the role of noradrenaline-mediated astrocytic signaling in locomotion, arousal, sensory integration, memory, and sleep. We then briefly review astrocyte contribution to the regulation of cerebral blood flow regulation, which is followed by a discussion of biophysical mechanisms underlying astrocyte effects on different brain processes. The experimental section is closed by an overview of recent experimental techniques available for modulation and visualization of astrocyte dynamics. We then evaluate how the new data can be potentially incorporated into the new mathematical models or where and how it already has been done. Finally, we discuss an interesting prospect that astrocytes may be key players in important processes such as the switching between sleep and wakefulness and the removal of toxic metabolites from the brain milieu.
Collapse
Affiliation(s)
- Alexey Brazhe
- Department of Biophysics, Biological Faculty, Lomonosov Moscow State University, Leninskie Gory, 1/24, Moscow, 119234 Russia
- Department of Molecular Neurobiology, Institute of Bioorganic Chemistry RAS, GSP-7, Miklukho-Maklay Str., 16/10, Moscow, 117997 Russia
| | - Andrey Verisokin
- Department of Theoretical Physics, Kursk State University, Radishcheva st., 33, Kursk, 305000 Russia
| | - Darya Verveyko
- Department of Theoretical Physics, Kursk State University, Radishcheva st., 33, Kursk, 305000 Russia
| | - Dmitry Postnov
- Department of Optics and Biophotonics, Saratov State University, Astrakhanskaya st., 83, Saratov, 410012 Russia
| |
Collapse
|
9
|
Aframian K, Yousef Yengej D, Nwaobi S, Raman S, Faas GC, Charles A. Effects of chronic caffeine on patterns of brain blood flow and behavior throughout the sleep-wake cycle in freely behaving mice. PNAS NEXUS 2023; 2:pgad303. [PMID: 37780231 PMCID: PMC10538474 DOI: 10.1093/pnasnexus/pgad303] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 09/05/2023] [Indexed: 10/03/2023]
Abstract
Caffeine has significant effects on neurovascular activity and behavior throughout the sleep-wake cycle. We used a minimally invasive microchip/video system to continuously record effects of caffeine in the drinking water of freely behaving mice. Chronic caffeine shifted both rest and active phases by up to 2 h relative to the light-dark cycle in a dose-dependent fashion. There was a particular delay in the onset of rapid eye movement (REM) sleep as compared with non-REM sleep during the rest phase. Chronic caffeine increased wakefulness during the active phase and consolidated sleep during the rest phase; overall, there was no net change in the amount of time spent in the wake, sleep, or REM sleep states during caffeine administration. Despite these effects on wakefulness and sleep, chronic caffeine decreased mean cerebral blood volume (CBV) during the active phase and increased mean CBV during the rest phase. Chronic caffeine also increased heart rate variability in both the sleep and wake states. These results provide new insight into the effects of caffeine on the biology of the sleep-wake cycle. Increased blood flow during sleep caused by chronic caffeine may have implications for its potential neuroprotective effects through vascular mechanisms of brain waste clearance.
Collapse
Affiliation(s)
- Kimiya Aframian
- Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles Young Drive, Los Angeles, CA 90095, USA
| | - Dmitri Yousef Yengej
- Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles Young Drive, Los Angeles, CA 90095, USA
| | - Sinifunanya Nwaobi
- Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles Young Drive, Los Angeles, CA 90095, USA
| | - Shrayes Raman
- Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles Young Drive, Los Angeles, CA 90095, USA
| | - Guido C Faas
- Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles Young Drive, Los Angeles, CA 90095, USA
| | - Andrew Charles
- Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles Young Drive, Los Angeles, CA 90095, USA
| |
Collapse
|
10
|
Liu D, Wang Q, Li Y, Yuan Z, Liu Z, Guo J, Li X, Zhang W, Tao Y, Mei J. Fructus gardeniae ameliorates anxiety-like behaviors induced by sleep deprivation via regulating hippocampal metabolomics and gut microbiota. Front Cell Infect Microbiol 2023; 13:1167312. [PMID: 37377643 PMCID: PMC10291143 DOI: 10.3389/fcimb.2023.1167312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
Fructus gardeniae (FG) is a traditional Chinese medicine and health food for thousands of years of application throughout Chinese history and is still widely used in clinical Chinese medicine. FG has a beneficial impact on anxiety, depression, insomnia, and psychiatric disorders; however, its mechanism of action requires further investigation. This study aimed to investigate the effects and mechanisms of FG on sleep deprivation (SD)-induced anxiety-like behavior in rats. A model of SD-induced anxiety-like behavior in rats was established by intraperitoneal injection of p-chlorophenylalanine (PCPA). This was accompanied by neuroinflammation and metabolic abnormalities in the hippocampus and disturbance of intestinal microbiota. However reduced SD-induced anxiety-like behavior and decreased levels of pro-inflammatory cytokines including TNF-α and IL-1β were observed in the hippocampus of rats after 7 days of FG intervention. In addition, metabolomic analysis demonstrated that FG was able to modulate levels of phosphatidylserine 18, Phosphatidylinositol 18, sn-glycero-3-phosphocholine, deoxyguanylic acid, xylose, betaine and other metabolites in the hippocampus. The main metabolic pathways of hippocampal metabolites after FG intervention involve carbon metabolism, glycolysis/gluconeogenesis, pentose phosphate, and glycerophospholipid metabolism. 16S rRNA sequencing illustrated that FG ameliorated the dysbiosis of gut microbiota in anxious rats, mainly increased the abundance of Muribaculaceae and Lactobacillus, and decreased the abundance of Lachnospiraceae_NK4A136_group. In addition, the correlation analysis demonstrated that there was a close relationship between hippocampal metabolites and intestinal microbiota. In conclusion, FG improved the anxiety behavior and inhibited of neuroinflammation in sleep-deprived rats, and the mechanism may be related to the FG regulation of hippocampal metabolites and intestinal microflora composition.
Collapse
Affiliation(s)
- Dong Liu
- Department of Emergency, The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
- Department of Traditional Chinese Medicine, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Qianfei Wang
- Department of Emergency, The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Ying Li
- Department of Pharmacy, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Zhenshuang Yuan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhiliang Liu
- Department of Emergency, Hebei Yiling Hospital, Shijiazhang, Hebei, China
| | - Junli Guo
- Department of Emergency, The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Xin Li
- Department of Emergency, The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Weichao Zhang
- Department of Emergency, The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Yulei Tao
- Department of Emergency, The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Jianqiang Mei
- Department of Emergency, The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| |
Collapse
|
11
|
Ingiosi AM, Frank MG. Goodnight, astrocyte: waking up to astroglial mechanisms in sleep. FEBS J 2023; 290:2553-2564. [PMID: 35271767 PMCID: PMC9463397 DOI: 10.1111/febs.16424] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/16/2022] [Accepted: 03/07/2022] [Indexed: 01/03/2023]
Abstract
Astrocytes mediate many important aspects of neural homeostasis, but until recently, their role in sleep was largely unknown. The situation has dramatically changed in the last decade. The use of transgenic animals, optogenetics, chemogenetics, brain imaging and sophisticated molecular assays has led to exciting discoveries. Astrocytes dynamically change their activity across the sleep-wake cycle and may encode sleep need via changes in intracellular signalling pathways. Astrocytes also exocytose/secrete sleep-inducing molecules which modulate brain activity, sleep architecture and sleep regulation. Many of these observations have been made in mice and Drosophila melanogaster, indicating that astroglial sleep mechanisms are evolutionarily conserved. We review recent findings and discuss future directions.
Collapse
Affiliation(s)
- Ashley M Ingiosi
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Marcos G Frank
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| |
Collapse
|
12
|
Peng W, Liu X, Ma G, Wu Z, Wang Z, Fei X, Qin M, Wang L, Li Y, Zhang S, Xu M. Adenosine-independent regulation of the sleep-wake cycle by astrocyte activity. Cell Discov 2023; 9:16. [PMID: 36746933 PMCID: PMC9902472 DOI: 10.1038/s41421-022-00498-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 11/20/2022] [Indexed: 02/08/2023] Open
Abstract
Astrocytes play a crucial role in regulating sleep-wake behavior, and adenosine signaling is generally thought to be involved. Here we show multiple lines of evidence supporting that modulation of the sleep-wake behavior by astrocyte Ca2+ activity could occur without adenosine signaling. In the basal forebrain and the brainstem, two brain regions that are known to be essential for sleep-wake regulation, chemogenetically-induced astrocyte Ca2+ elevation significantly modulated the sleep-wake cycle. Although astrocyte Ca2+ level positively correlated with the amount of extracellular adenosine, as revealed by a genetically encoded adenosine sensor, we found no detectable change in adenosine level after suppressing astrocyte Ca2+ elevation, and transgenic mice lacking one of the major extracellular ATP-adenosine conversion enzymes showed similar extracellular adenosine level and astrocyte Ca2+-induced sleep modulation. Furthermore, astrocyte Ca2+ is dependent primarily on local neuronal activity, causing brain region-specific regulation of the sleep-wake cycle. Thus, neural activity-dependent astrocyte activity could regulate the sleep-wake behavior independent of adenosine signaling.
Collapse
Affiliation(s)
- Wanling Peng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, China.
| | - Xiaotong Liu
- grid.9227.e0000000119573309Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Guofen Ma
- grid.16821.3c0000 0004 0368 8293Center for Brain Science of Shanghai Children’s Medical Center, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,grid.16821.3c0000 0004 0368 8293Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhaofa Wu
- grid.11135.370000 0001 2256 9319State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China ,grid.11135.370000 0001 2256 9319Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Ziyue Wang
- grid.16821.3c0000 0004 0368 8293Center for Brain Science of Shanghai Children’s Medical Center, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,grid.16821.3c0000 0004 0368 8293Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiang Fei
- grid.9227.e0000000119573309Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Meiling Qin
- grid.9227.e0000000119573309Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Lizhao Wang
- grid.16821.3c0000 0004 0368 8293Center for Brain Science of Shanghai Children’s Medical Center, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,grid.16821.3c0000 0004 0368 8293Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yulong Li
- grid.11135.370000 0001 2256 9319State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China ,grid.11135.370000 0001 2256 9319Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China ,grid.11135.370000 0001 2256 9319PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Siyu Zhang
- Center for Brain Science of Shanghai Children's Medical Center, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Min Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, China. .,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shangha, China.
| |
Collapse
|
13
|
Picard K, Corsi G, Decoeur F, Di Castro MA, Bordeleau M, Persillet M, Layé S, Limatola C, Tremblay MÈ, Nadjar A. Microglial homeostasis disruption modulates non-rapid eye movement sleep duration and neuronal activity in adult female mice. Brain Behav Immun 2023; 107:153-164. [PMID: 36202169 DOI: 10.1016/j.bbi.2022.09.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 09/12/2022] [Accepted: 09/30/2022] [Indexed: 11/07/2022] Open
Abstract
Sleep is a natural physiological state, tightly regulated through several neuroanatomical and neurochemical systems, which is essential to maintain physical and mental health. Recent studies revealed that the functions of microglia, the resident immune cells of the brain, differ along the sleep-wake cycle. Inflammatory cytokines, such as interleukin-1β and tumor necrosis factor-α, mainly produced by microglia in the brain, are also well-known to promote sleep. However, the contributing role of microglia on sleep regulation remains largely elusive, even more so in females. Given the higher prevalence of various sleep disorders in women, we aimed to determine the role of microglia in regulating the sleep-wake cycle specifically in female mice. Microglia were depleted in adult female mice with inhibitors of the colony-stimulating factor 1 receptor (CSF1R) (PLX3397 or PLX5622), which is required for microglial population maintenance. This led to a 65-73% reduction of the microglial population, as confirmed by immunofluorescence staining against IBA1 (marker of microglia/macrophages) and TMEM119 (microglia-specific marker) in the reticular nucleus of the thalamus and primary motor cortex. The spontaneous sleep-wake cycle was evaluated at steady-state, during microglial homeostasis disruption and after complete microglial repopulation, upon cessation of treatment with the inhibitors of CSF1R, using electroencephalography (EEG) and electromyography (EMG). We found that microglia-depleted female mice spent more time in non-rapid eye movement (NREM) sleep and had an increased number of NREM sleep episodes, which was partially restored after microglial total repopulation. To determine whether microglia could regulate sleep locally by modulating synaptic transmission, we used patch clamp to record spontaneous activity of pyramidal neurons in the primary motor cortex, which showed an increase of excitatory synaptic transmission during the dark phase. These changes in neuronal activity were modulated by microglial depletion in a phase-dependent manner. Altogether, our results indicate that microglia are involved in the sleep regulation of female mice, further strengthening their potential implication in the development and/or progression of sleep disorders. Furthermore, our findings indicate that microglial repopulation can contribute to normalizing sleep alterations caused by their partial depletion.
Collapse
Affiliation(s)
- Katherine Picard
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Département de médecine moléculaire, Université Laval, Québec, QC, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Giorgio Corsi
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Fanny Decoeur
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | | | - Maude Bordeleau
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Marine Persillet
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - Sophie Layé
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Department of Neurophysiology, Neuropharmacology, Inflammaging, IRCCS Neuromed, Pozzilli, Italy
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Département de médecine moléculaire, Université Laval, Québec, QC, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada; Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada; Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
| | - Agnès Nadjar
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France; INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France; Institut Universitaire de France (IUF), France.
| |
Collapse
|
14
|
Sunkaria A, Bhardwaj S. Sleep Disturbance and Alzheimer's Disease: The Glial Connection. Neurochem Res 2022; 47:1799-1815. [PMID: 35303225 DOI: 10.1007/s11064-022-03578-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/27/2022] [Accepted: 03/09/2022] [Indexed: 12/28/2022]
Abstract
Poor quality and quantity of sleep are very common in elderly people throughout the world. Growing evidence has suggested that sleep disturbances could accelerate the process of neurodegeneration. Recent reports have shown a positive correlation between sleep deprivation and amyloid-β (Aβ)/tau aggregation in the brain of Alzheimer's patients. Glial cells have long been implicated in the progression of Alzheimer's disease (AD) and recent findings have also suggested their role in regulating sleep homeostasis. However, how glial cells control the sleep-wake balance and exactly how disturbed sleep may act as a trigger for Alzheimer's or other neurological disorders have recently gotten attention. In an attempt to connect the dots, the present review has highlighted the role of glia-derived sleep regulatory molecules in AD pathogenesis. Role of glia in sleep disturbance and Alzheimer's progression.
Collapse
Affiliation(s)
- Aditya Sunkaria
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab, 143005, India.
| | - Supriya Bhardwaj
- Department of Dermatology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| |
Collapse
|
15
|
Lee R, McGee A, Fernandez FX. Systematic review of drugs that modify the circadian system's phase-shifting responses to light exposure. Neuropsychopharmacology 2022; 47:866-879. [PMID: 34961774 PMCID: PMC8882192 DOI: 10.1038/s41386-021-01251-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/08/2021] [Accepted: 11/30/2021] [Indexed: 11/09/2022]
Abstract
We searched PubMed for primary research quantifying drug modification of light-induced circadian phase-shifting in rodents. This search, conducted for work published between 1960 and 2018, yielded a total of 146 papers reporting results from 901 studies. Relevant articles were those with any extractable data on phase resetting in wildtype (non-trait selected) rodents administered a drug, alongside a vehicle/control group, near or at the time of exposure. Most circadian pharmacology experiments were done using drugs thought to act directly on either the brain's central pacemaker, the suprachiasmatic nucleus (SCN), the SCN's primary relay, the retinohypothalamic tract, secondary pathways originating from the medial/dorsal raphe nuclei and intergeniculate leaflet, or the brain's sleep-arousal centers. While the neurotransmitter systems underlying these circuits were of particular interest, including those involving glutamate, gamma-aminobutyric acid, serotonin, and acetylcholine, other signaling modalities have also been assessed, including agonists and antagonists of receptors linked to dopamine, histamine, endocannabinoids, adenosine, opioids, and second-messenger pathways downstream of glutamate receptor activation. In an effort to identify drugs that unduly influence circadian responses to light, we quantified the net effects of each drug class by ratioing the size of the phase-shift observed after administration to that observed with vehicle in a given experiment. This allowed us to organize data across the literature, compare the relative efficacy of one mechanism versus another, and clarify which drugs might best suppress or potentiate phase resetting. Aggregation of the available data in this manner suggested that several candidates might be clinically relevant as auxiliary treatments to suppress ectopic light responses during shiftwork or amplify the circadian effects of timed bright light therapy. Future empirical research will be necessary to validate these possibilities.
Collapse
Affiliation(s)
- Robert Lee
- Department of Psychology, University of Arizona, Tucson, AZ, USA
| | - Austin McGee
- Department of Psychology, University of Arizona, Tucson, AZ, USA
| | - Fabian-Xosé Fernandez
- Department of Psychology, University of Arizona, Tucson, AZ, USA.
- Department of Neurology, University of Arizona, Tucson, AZ, USA.
- BIO5 and McKnight Brain Research Institutes, Tucson, AZ, USA.
| |
Collapse
|
16
|
Debom GN, Rubenich DS, Braganhol E. Adenosinergic Signaling as a Key Modulator of the Glioma Microenvironment and Reactive Astrocytes. Front Neurosci 2022; 15:648476. [PMID: 35069091 PMCID: PMC8766410 DOI: 10.3389/fnins.2021.648476] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 12/03/2021] [Indexed: 12/13/2022] Open
Abstract
Astrocytes are numerous glial cells of the central nervous system (CNS) and play important roles in brain homeostasis. These cells can directly communicate with neurons by releasing gliotransmitters, such as adenosine triphosphate (ATP) and glutamate, into the multipartite synapse. Moreover, astrocytes respond to tissue injury in the CNS environment. Recently, astrocytic heterogeneity and plasticity have been discussed by several authors, with studies proposing a spectrum of astrocytic activation characterized by A1/neurotoxic and A2/neuroprotective polarization extremes. The fundamental roles of astrocytes in communicating with other cells and sustaining homeostasis are regulated by purinergic signaling. In the CNS environment, the gliotransmitter ATP acts cooperatively with other glial signaling molecules, such as cytokines, which may impact CNS functions by facilitating/inhibiting neurotransmitter release. Adenosine (ADO), the main product of extracellular ATP metabolism, is an important homeostatic modulator and acts as a neuromodulator in synaptic transmission via P1 receptor sensitization. Furthermore, purinergic signaling is a key factor in the tumor microenvironment (TME), as damaged cells release ATP, leading to ADO accumulation in the TME through the ectonucleotidase cascade. Indeed, the enzyme CD73, which converts AMP to ADO, is overexpressed in glioblastoma cells; this upregulation is associated with tumor aggressiveness. Because of the crucial activity of CD73 in these cells, extracellular ADO accumulation in the TME contributes to sustaining glioblastoma immune escape while promoting A2-like activation. The present review describes the importance of ADO in modulating astrocyte polarization and simultaneously promoting tumor growth. We also discuss whether targeting of CD73 to block ADO production can be used as an alternative cancer therapy.
Collapse
Affiliation(s)
- Gabriela N Debom
- Programa de Pós-graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Dominique S Rubenich
- Programa de Pós-graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Elizandra Braganhol
- Programa de Pós-graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil.,Instituto de Cardiologia do Rio Grande do Sul, Instituto de Cardiologia - Fundação Universitária de Cardiologia, Porto Alegre, Brazil
| |
Collapse
|
17
|
Bonilla-Jaime H, Zeleke H, Rojas A, Espinosa-Garcia C. Sleep Disruption Worsens Seizures: Neuroinflammation as a Potential Mechanistic Link. Int J Mol Sci 2021; 22:12531. [PMID: 34830412 PMCID: PMC8617844 DOI: 10.3390/ijms222212531] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 11/16/2021] [Accepted: 11/18/2021] [Indexed: 12/13/2022] Open
Abstract
Sleep disturbances, such as insomnia, obstructive sleep apnea, and daytime sleepiness, are common in people diagnosed with epilepsy. These disturbances can be attributed to nocturnal seizures, psychosocial factors, and/or the use of anti-epileptic drugs with sleep-modifying side effects. Epilepsy patients with poor sleep quality have intensified seizure frequency and disease progression compared to their well-rested counterparts. A better understanding of the complex relationship between sleep and epilepsy is needed, since approximately 20% of seizures and more than 90% of sudden unexpected deaths in epilepsy occur during sleep. Emerging studies suggest that neuroinflammation, (e.g., the CNS immune response characterized by the change in expression of inflammatory mediators and glial activation) may be a potential link between sleep deprivation and seizures. Here, we review the mechanisms by which sleep deprivation induces neuroinflammation and propose that neuroinflammation synergizes with seizure activity to worsen neurodegeneration in the epileptic brain. Additionally, we highlight the relevance of sleep interventions, often overlooked by physicians, to manage seizures, prevent epilepsy-related mortality, and improve quality of life.
Collapse
Affiliation(s)
- Herlinda Bonilla-Jaime
- Departamento de Biología de la Reproducción, Área de Biología Conductual y Reproductiva, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de Mexico CP 09340, Mexico;
| | - Helena Zeleke
- Neuroscience and Behavioral Biology Program, College of Arts and Sciences, Emory University, Atlanta, GA 30322, USA;
| | - Asheebo Rojas
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Claudia Espinosa-Garcia
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
18
|
Mielnicka A, Michaluk P. Exocytosis in Astrocytes. Biomolecules 2021; 11:1367. [PMID: 34572580 PMCID: PMC8471187 DOI: 10.3390/biom11091367] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/10/2021] [Accepted: 09/14/2021] [Indexed: 12/17/2022] Open
Abstract
Until recently, astrocytes were thought to be a part of a simple "brain glue" providing only a supporting role for neurons. However, the discoveries of the last two decades have proven astrocytes to be dynamic partners participating in brain metabolism and actively influencing communication between neurons. The means of astrocyte-neuron communication are diverse, although regulated exocytosis has received the most attention but also caused the most debate. Similar to most of eukaryotic cells, astrocytes have a complex range of vesicular organelles which can undergo exocytosis as well as intricate molecular mechanisms that regulate this process. In this review, we focus on the components needed for regulated exocytosis to occur and summarise the knowledge about experimental evidence showing its presence in astrocytes.
Collapse
Affiliation(s)
| | - Piotr Michaluk
- BRAINCITY, Laboratory of Neurobiology, The Nencki Institute of Experimental Biology, PAS, 02-093 Warsaw, Poland;
| |
Collapse
|
19
|
Corsi G, Picard K, di Castro MA, Garofalo S, Tucci F, Chece G, Del Percio C, Golia MT, Raspa M, Scavizzi F, Decoeur F, Lauro C, Rigamonti M, Iannello F, Ragozzino DA, Russo E, Bernardini G, Nadjar A, Tremblay ME, Babiloni C, Maggi L, Limatola C. Microglia modulate hippocampal synaptic transmission and sleep duration along the light/dark cycle. Glia 2021; 70:89-105. [PMID: 34487590 PMCID: PMC9291950 DOI: 10.1002/glia.24090] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 08/27/2021] [Accepted: 08/27/2021] [Indexed: 01/09/2023]
Abstract
Microglia, the brain's resident macrophages, actively contribute to the homeostasis of cerebral parenchyma by sensing neuronal activity and supporting synaptic remodeling and plasticity. While several studies demonstrated different roles for astrocytes in sleep, the contribution of microglia in the regulation of sleep/wake cycle and in the modulation of synaptic activity in the different day phases has not been deeply investigated. Using light as a zeitgeber cue, we studied the effects of microglial depletion with the colony stimulating factor‐1 receptor antagonist PLX5622 on the sleep/wake cycle and on hippocampal synaptic transmission in male mice. Our data demonstrate that almost complete microglial depletion increases the duration of NREM sleep and reduces the hippocampal excitatory neurotransmission. The fractalkine receptor CX3CR1 plays a relevant role in these effects, because cx3cr1GFP/GFP mice recapitulate what found in PLX5622‐treated mice. Furthermore, during the light phase, microglia express lower levels of cx3cr1 and a reduction of cx3cr1 expression is also observed when cultured microglial cells are stimulated by ATP, a purinergic molecule released during sleep. Our findings suggest that microglia participate in the regulation of sleep, adapting their cx3cr1 expression in response to the light/dark phase, and modulating synaptic activity in a phase‐dependent manner.
Collapse
Affiliation(s)
- Giorgio Corsi
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Katherine Picard
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Quebec City, Quebec, Canada
| | | | - Stefano Garofalo
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Federico Tucci
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,Department of Neurology, San Raffaele of Cassino, Cassino (FR), Italy
| | - Giuseppina Chece
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Claudio Del Percio
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Maria Teresa Golia
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Marcello Raspa
- National Research Council, Institute of Biochemistry and Cell Biology (EMMA/Infrafrontier/IMPC, International Campus "A. Buzzati-Traverso", Rome, Italy
| | - Ferdinando Scavizzi
- National Research Council, Institute of Biochemistry and Cell Biology (EMMA/Infrafrontier/IMPC, International Campus "A. Buzzati-Traverso", Rome, Italy
| | - Fanny Decoeur
- INRAE, Bordeaux INP, NutriNeuro UMR 1286, Bordeaux University, Bordeaux, France
| | - Clotilde Lauro
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | | | | | | | - Eleonora Russo
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | | | - Agnès Nadjar
- INRAE, Bordeaux INP, NutriNeuro UMR 1286, Bordeaux University, Bordeaux, France.,INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, Bordeaux, France
| | - Marie Eve Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Quebec City, Quebec, Canada.,Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada.,The Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Claudio Babiloni
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,Department of Neurology, San Raffaele of Cassino, Cassino (FR), Italy
| | - Laura Maggi
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Istituto Pasteur Italia, Rome, Italy.,Department of Neurophysiology, Neuropharmacology, Inflammaging, IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
20
|
Gobbo D, Scheller A, Kirchhoff F. From Physiology to Pathology of Cortico-Thalamo-Cortical Oscillations: Astroglia as a Target for Further Research. Front Neurol 2021; 12:661408. [PMID: 34177766 PMCID: PMC8219957 DOI: 10.3389/fneur.2021.661408] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/11/2021] [Indexed: 12/21/2022] Open
Abstract
The electrographic hallmark of childhood absence epilepsy (CAE) and other idiopathic forms of epilepsy are 2.5-4 Hz spike and wave discharges (SWDs) originating from abnormal electrical oscillations of the cortico-thalamo-cortical network. SWDs are generally associated with sudden and brief non-convulsive epileptic events mostly generating impairment of consciousness and correlating with attention and learning as well as cognitive deficits. To date, SWDs are known to arise from locally restricted imbalances of excitation and inhibition in the deep layers of the primary somatosensory cortex. SWDs propagate to the mostly GABAergic nucleus reticularis thalami (NRT) and the somatosensory thalamic nuclei that project back to the cortex, leading to the typical generalized spike and wave oscillations. Given their shared anatomical basis, SWDs have been originally considered the pathological transition of 11-16 Hz bursts of neural oscillatory activity (the so-called sleep spindles) occurring during Non-Rapid Eye Movement (NREM) sleep, but more recent research revealed fundamental functional differences between sleep spindles and SWDs, suggesting the latter could be more closely related to the slow (<1 Hz) oscillations alternating active (Up) and silent (Down) cortical activity and concomitantly occurring during NREM. Indeed, several lines of evidence support the fact that SWDs impair sleep architecture as well as sleep/wake cycles and sleep pressure, which, in turn, affect seizure circadian frequency and distribution. Given the accumulating evidence on the role of astroglia in the field of epilepsy in the modulation of excitation and inhibition in the brain as well as on the development of aberrant synchronous network activity, we aim at pointing at putative contributions of astrocytes to the physiology of slow-wave sleep and to the pathology of SWDs. Particularly, we will address the astroglial functions known to be involved in the control of network excitability and synchronicity and so far mainly addressed in the context of convulsive seizures, namely (i) interstitial fluid homeostasis, (ii) K+ clearance and neurotransmitter uptake from the extracellular space and the synaptic cleft, (iii) gap junction mechanical and functional coupling as well as hemichannel function, (iv) gliotransmission, (v) astroglial Ca2+ signaling and downstream effectors, (vi) reactive astrogliosis and cytokine release.
Collapse
Affiliation(s)
- Davide Gobbo
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Anja Scheller
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| |
Collapse
|
21
|
Rijo-Ferreira F, Bjorness TE, Cox KH, Sonneborn A, Greene RW, Takahashi JS. Sleeping Sickness Disrupts the Sleep-Regulating Adenosine System. J Neurosci 2020; 40:9306-9316. [PMID: 33097636 PMCID: PMC7687053 DOI: 10.1523/jneurosci.1046-20.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 09/28/2020] [Accepted: 10/11/2020] [Indexed: 12/16/2022] Open
Abstract
Patients with sleeping sickness, caused by the parasite Trypanosoma brucei, have disruptions in both sleep timing and sleep architecture. However, the underlying cause of these sleep disturbances is not well understood. Here, we assessed the sleep architecture of male mice infected with T. brucei and found that infected mice had drastically altered sleep patterns. Interestingly, T. brucei-infected mice also had a reduced homeostatic sleep response to sleep deprivation, a response modulated by the adenosine system. We found that infected mice had a reduced electrophysiological response to an adenosine receptor antagonist and increased adenosine receptor gene expression. Although the mechanism by which T. brucei infection causes these changes remains to be determined, our findings suggest that the symptoms of sleeping sickness may be because of alterations in homeostatic adenosine signaling.SIGNIFICANCE STATEMENT Sleeping sickness is a fatal disease that disrupts the circadian clock, causes disordered temperature regulation, and induces sleep disturbance. To examine the neurologic effects of infection in the absence of other symptoms, in this study, we used a mouse model of sleeping sickness in which the acute infection was treated but brain infection remained. Using this model, we evaluated the effects of the sleeping sickness parasite, Trypanosoma brucei, on sleep patterns in mice, under both normal and sleep-deprived conditions. Our findings suggest that signaling of adenosine, a neuromodulator involved in mediating homeostatic sleep drive, may be reduced in infected mice.
Collapse
Affiliation(s)
- Filipa Rijo-Ferreira
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
| | - Theresa E Bjorness
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
- Research Service, VA North Texas Health Care System, Dallas, Texas 75216-7167
| | - Kimberly H Cox
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
| | - Alex Sonneborn
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
| | - Robert W Greene
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
| | - Joseph S Takahashi
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9111
| |
Collapse
|
22
|
Abstract
Sleep is a fundamental property conserved across species. The homeostatic induction of sleep indicates the presence of a mechanism that is progressively activated by the awake state and that induces sleep. Several lines of evidence support that such function, namely, sleep need, lies in the neuronal assemblies rather than specific brain regions and circuits. However, the molecular mechanism underlying the dynamics of sleep need is still unclear. This review aims to summarize recent studies mainly in rodents indicating that protein phosphorylation, especially at the synapses, could be the molecular entity associated with sleep need. Genetic studies in rodents have identified a set of kinases that promote sleep. The activity of sleep-promoting kinases appears to be elevated during the awake phase and in sleep deprivation. Furthermore, the proteomic analysis demonstrated that the phosphorylation status of synaptic protein is controlled by the sleep-wake cycle. Therefore, a plausible scenario may be that the awake-dependent activation of kinases modifies the phosphorylation status of synaptic proteins to promote sleep. We also discuss the possible importance of multisite phosphorylation on macromolecular protein complexes to achieve the slow dynamics and physiological functions of sleep in mammals.
Collapse
Affiliation(s)
- Koji L Ode
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroki R Ueda
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics, Osaka, Japan
| |
Collapse
|
23
|
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
|
24
|
Decoeur F, Benmamar-Badel A, Leyrolle Q, Persillet M, Layé S, Nadjar A. Dietary N-3 PUFA deficiency affects sleep-wake activity in basal condition and in response to an inflammatory challenge in mice. Brain Behav Immun 2020; 85:162-169. [PMID: 31100369 DOI: 10.1016/j.bbi.2019.05.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/12/2019] [Accepted: 05/11/2019] [Indexed: 12/22/2022] Open
Abstract
Essential polyunsaturated fatty acids (PUFA) from the n-3 and n-6 series constitute the building blocks of brain cell membranes where they regulate most aspects of cell physiology. They are either biosynthesized from their dietary precursors or can be directly sourced from the diet. An overall increase in the dietary n-6/n-3 PUFA ratio, as observed in the Western diet, leads to reduced n-3 PUFAs in tissues that include the brain. Some clinical studies have shown a positive correlation between dietary n-3 PUFA intake and sleep quantity, yet evidence is still sparse. We here used a preclinical model of dietary n-3 PUFA deficiency to assess the precise relationship between dietary PUFA intake and sleep/wake activity. Using electroencephalography (EEG)/electromyography (EMG) recordings on n-3 PUFA deficient or sufficient mice, we showed that dietary PUFA deficiency affects the architecture of sleep-wake activity and the oscillatory activity of cortical neurons during sleep. In a second part of the study, and since PUFAs are a potent modulator of inflammation, we assessed the effect of dietary n-3 PUFA deficiency on the sleep response to an inflammatory stimulus known to modulate sleep/wake activity. We injected mice with the endotoxin lipopolysaccharide (LPS) and quantified the sleep response across the following 12 h. Our results revealed that n-3 PUFA deficiency affects the sleep response in basal condition and after a peripheral immune challenge. More studies are now required aimed at deciphering the molecular mechanisms underlying the intimate relationship between n-3 PUFAs and sleep/wake activity.
Collapse
Affiliation(s)
- F Decoeur
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A Benmamar-Badel
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - Q Leyrolle
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - M Persillet
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - S Layé
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A Nadjar
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France.
| |
Collapse
|
25
|
Dash MB. Infraslow coordination of slow wave activity through altered neuronal synchrony. Sleep 2019; 42:5540154. [PMID: 31353415 DOI: 10.1093/sleep/zsz170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/29/2019] [Indexed: 11/14/2022] Open
Abstract
Slow wave activity (SWA; the EEG power between 0.5 and 4 Hz during non-rapid eye movement sleep [NREM]) is the best electrophysiological marker of sleep need; SWA dissipates across the night and increases following sleep deprivation. In addition to these well-documented homeostatic SWA trends, SWA exhibits extensive variability across shorter timescales (seconds to minutes) and between local cortical regions. The physiological underpinnings of SWA variability, however, remain poorly characterized. In male Sprague-Dawley rats, we observed that SWA exhibits pronounced infraslow fluctuations (~40- to 120-s periods) that are coordinated across disparate cortical locations. Peaks in SWA across infraslow cycles were associated with increased slope, amplitude, and duration of individual slow waves and a reduction in the total number of waves and proportion of multipeak waves. Using a freely available data set comprised of extracellular unit recordings during consolidated NREM episodes in male Long-Evans rats, we further show that infraslow SWA does not appear to arise as a consequence of firing rate modulation of putative excitatory or inhibitory neurons. Instead, infraslow SWA was associated with alterations in neuronal synchrony surrounding "On"/"Off" periods and changes in the number and duration of "Off" periods. Collectively, these data provide a mechanism by which SWA can be coordinated across disparate cortical locations and thereby connect local and global expression of this patterned neuronal activity. In doing so, infraslow SWA may contribute to the regulation of cortical circuits during sleep and thereby play a critical role in sleep function.
Collapse
Affiliation(s)
- Michael B Dash
- Department of Psychology, Middlebury College, Middlebury, VT
- Program in Neuroscience, Middlebury College, Middlebury, VT
| |
Collapse
|
26
|
Vardjan N, Parpura V, Verkhratsky A, Zorec R. Gliocrine System: Astroglia as Secretory Cells of the CNS. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1175:93-115. [PMID: 31583585 DOI: 10.1007/978-981-13-9913-8_4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Astrocytes are secretory cells, actively participating in cell-to-cell communication in the central nervous system (CNS). They sense signaling molecules in the extracellular space, around the nearby synapses and also those released at much farther locations in the CNS, by their cell surface receptors, get excited to then release their own signaling molecules. This contributes to the brain information processing, based on diffusion within the extracellular space around the synapses and on convection when locales relatively far away from the release sites are involved. These functions resemble secretion from endocrine cells, therefore astrocytes were termed to be a part of the gliocrine system in 2015. An important mechanism, by which astrocytes release signaling molecules is the merger of the vesicle membrane with the plasmalemma, i.e., exocytosis. Signaling molecules stored in astroglial secretory vesicles can be discharged into the extracellular space after the vesicle membrane fuses with the plasma membrane. This leads to a fusion pore formation, a channel that must widen to allow the exit of the Vesiclal cargo. Upon complete vesicle membrane fusion, this process also integrates other proteins, such as receptors, transporters and channels into the plasma membrane, determining astroglial surface signaling landscape. Vesiclal cargo, together with the whole vesicle can also exit astrocytes by the fusion of multivesicular bodies with the plasma membrane (exosomes) or by budding of vesicles (ectosomes) from the plasma membrane into the extracellular space. These astroglia-derived extracellular vesicles can later interact with various target cells. Here, the characteristics of four types of astroglial secretory vesicles: synaptic-like microvesicles, dense-core vesicles, secretory lysosomes, and extracellular vesicles, are discussed. Then machinery for vesicle-based exocytosis, second messenger regulation and the kinetics of exocytotic vesicle content discharge or release of extracellular vesicles are considered. In comparison to rapidly responsive, electrically excitable neurons, the receptor-mediated cytosolic excitability-mediated astroglial exocytotic vesicle-based transmitter release is a relatively slow process.
Collapse
Affiliation(s)
- Nina Vardjan
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, 1000, Ljubljana, Slovenia. .,Celica Biomedical, 1000, Ljubljana, Slovenia.
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK.,Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.,Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Spain
| | - Robert Zorec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, 1000, Ljubljana, Slovenia. .,Celica Biomedical, 1000, Ljubljana, Slovenia.
| |
Collapse
|
27
|
Zhou X, Xiao Q, Xie L, Yang F, Wang L, Tu J. Astrocyte, a Promising Target for Mood Disorder Interventions. Front Mol Neurosci 2019; 12:136. [PMID: 31231189 PMCID: PMC6560156 DOI: 10.3389/fnmol.2019.00136] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/09/2019] [Indexed: 01/03/2023] Open
Abstract
Mood disorders have multiple phenotypes and complex underlying biological mechanisms and, as such, there are no effective therapeutic strategies. A review of recent work on the role of astrocytes in mood disorders is thus warranted, which we embark on here. We argue that there is tremendous potential for novel strategies for therapeutic interventions based on the role of astrocytes. Astrocytes are traditionally considered to have supporting roles within the brain, yet emerging evidence has shown that astrocytes have more direct roles in influencing brain function. Notably, evidence from postmortem human brain tissues has highlighted changes in glial cell morphology, density and astrocyte-related biomarkers and genes following mood disorders, indicating astrocyte involvement in mood disorders. Findings from animal models strongly imply that astrocytes not only change astrocyte morphology and physiological characteristics but also influence neural circuits via synapse structure and formation. This review pays particular attention to interactions between astrocytes and neurons and argues that astrocyte dysfunction affects the monoaminergic system, excitatory–inhibitory balance and neurotrophic states of local networks. Together, these studies provide a foundation of knowledge about the exact role of astrocytes in mood disorders. Importantly, we then change the focus from neurons to glial cells and the interactions between the two, so that we can understand newly proposed mechanisms underlying mood disorders, and to identify more diagnostic indicators or effective targets for treatment of these diseases.
Collapse
Affiliation(s)
- Xinyi Zhou
- Shenzhen Key Lab of Neuropsychiatric Modulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.,Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Qian Xiao
- Shenzhen Key Lab of Neuropsychiatric Modulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.,Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Li Xie
- Shenzhen Key Lab of Neuropsychiatric Modulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Fan Yang
- Shenzhen Key Lab of Neuropsychiatric Modulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Liping Wang
- Shenzhen Key Lab of Neuropsychiatric Modulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Jie Tu
- Shenzhen Key Lab of Neuropsychiatric Modulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| |
Collapse
|
28
|
Sundin EM, Ciubuc JD, Bennet KE, Ochoa K, Manciu FS. Comparative Computational and Experimental Detection of Adenosine Using Ultrasensitive Surface-Enhanced Raman Spectroscopy. SENSORS (BASEL, SWITZERLAND) 2018; 18:E2696. [PMID: 30115871 PMCID: PMC6111885 DOI: 10.3390/s18082696] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 08/11/2018] [Accepted: 08/14/2018] [Indexed: 01/30/2023]
Abstract
To better understand detection and monitoring of the important neurotransmitter adenosine at physiological levels, this study combines quantum chemical density functional modeling and ultrasensitive surface-enhanced Raman spectroscopic (SERS) measurements. Combined simulation results and experimental data for an analyte concentration of about 10-11 molar indicate the presence of all known molecular forms resulting from adenosine's complex redox-reaction. Detailed analysis presented here, besides assessing potential Raman signatures of these adenosinic forms, also sheds light on the analytic redox process and voltammetric detection. Examples of adenosine Raman fingerprints for different molecular orientations with respect to the SERS substrate are the vibrational line around 920 ± 10 cm-1 for analyte physisorption through the carbinol moiety and around 1600 ± 20 cm-1 for its fully oxidized form. However, both hydroxyl/oxygen sites and NH₂/nitrogen sites contribute to molecule's interaction with the SERS environment. Our results also reveal that contributions of partially oxidized adenosine forms and of the standard form are more likely to be detected with the first recorded voltammetric oxidation peak. The fully oxidized adenosine form contributes mostly to the second peak. Thus, this comparative theoretical⁻experimental investigation of adenosine's vibrational signatures provides significant insights for advancing its detection, and for future development of opto-voltammetric biosensors.
Collapse
Affiliation(s)
- Emma M Sundin
- Department of Physics, University of Texas at El Paso, El Paso, TX 79968, USA.
- Department of Biomedical Engineering, University of Texas at El Paso, El Paso, TX 79968, USA.
| | - John D Ciubuc
- Department of Physics, University of Texas at El Paso, El Paso, TX 79968, USA.
- Department of Biomedical Engineering, University of Texas at El Paso, El Paso, TX 79968, USA.
| | - Kevin E Bennet
- Division of Engineering, Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA.
| | - Katia Ochoa
- Department of Physics, University of Texas at El Paso, El Paso, TX 79968, USA.
| | - Felicia S Manciu
- Department of Physics, University of Texas at El Paso, El Paso, TX 79968, USA.
- Department of Biomedical Engineering, University of Texas at El Paso, El Paso, TX 79968, USA.
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA.
| |
Collapse
|
29
|
Szentirmai É, Kapás L. Brown adipose tissue plays a central role in systemic inflammation-induced sleep responses. PLoS One 2018; 13:e0197409. [PMID: 29746591 PMCID: PMC5945014 DOI: 10.1371/journal.pone.0197409] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/01/2018] [Indexed: 01/31/2023] Open
Abstract
We previously identified brown adipose tissue (BAT) as a source of sleep-inducing signals. Pharmacological activation of BAT enhances sleep while sleep loss leads to increased BAT thermogenesis. Recovery sleep after sleep loss is diminished in mice that lack uncoupling protein 1 (UCP-1), and also in wild-type (WT) mice after sensory denervation of the BAT. Systemic inflammation greatly affects metabolism and the function of adipose tissue, and also induces characteristic sleep responses. We hypothesized that sleep responses to acute inflammation are mediated by BAT-derived signals. To test this, we determined the effects of systemic inflammation on sleep and body temperature in UCP-1 knockout (KO) and WT mice. Intraperitoneal injections of lipopolysaccharide, tumor necrosis factor-α, interleukin-1 beta and clodronate containing liposomes were used to induce systemic inflammation. In WT animals, non-rapid-eye movement sleep (NREMS) was elevated in all four inflammatory models. All NREMS responses were completely abolished in UCP-1 KO animals. Systemic inflammation elicited an initial hypothermia followed by fever in WT mice. The hypothermic phase, but not the fever, was abolished in UCP-1 KO mice. The only recognized function of UCP-1 is to promote thermogenesis in brown adipocytes. Present results indicate that the presence of UCP-1 is necessary for increased NREMS but does not contribute to the development of fever in systemic inflammation.
Collapse
Affiliation(s)
- Éva Szentirmai
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, Washington, United States of America
- Sleep and Performance Research Center, Washington State University, Spokane, Washington, United States of America
| | - Levente Kapás
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, Washington, United States of America
- Sleep and Performance Research Center, Washington State University, Spokane, Washington, United States of America
| |
Collapse
|
30
|
The putative role of oxidative stress and inflammation in the pathophysiology of sleep dysfunction across neuropsychiatric disorders: Focus on chronic fatigue syndrome, bipolar disorder and multiple sclerosis. Sleep Med Rev 2018; 41:255-265. [PMID: 29759891 DOI: 10.1016/j.smrv.2018.03.007] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 02/20/2018] [Accepted: 03/27/2018] [Indexed: 12/29/2022]
Abstract
Sleep and circadian abnormalities are prevalent and burdensome manifestations of diverse neuro-immune diseases, and may aggravate the course of several neuropsychiatric disorders. The underlying pathophysiology of sleep abnormalities across neuropsychiatric disorders remains unclear, and may involve the inter-play of several clinical variables and mechanistic pathways. In this review, we propose a heuristic framework in which reciprocal interactions of immune, oxidative and nitrosative stress, and mitochondrial pathways may drive sleep abnormalities across potentially neuroprogressive disorders. Specifically, it is proposed that systemic inflammation may activate microglial cells and astrocytes in brain regions involved in sleep and circadian regulation. Activated glial cells may secrete pro-inflammatory cytokines (for example, interleukin-1 beta and tumour necrosis factor alpha), nitric oxide and gliotransmitters, which may influence the expression of key circadian regulators (e.g., the Circadian Locomotor Output Cycles Kaput (CLOCK) gene). Furthermore, sleep disruption may further aggravate oxidative and nitrosative, peripheral immune activation, and (neuro) inflammation across these disorders in a vicious pathophysiological loop. This review will focus on chronic fatigue syndrome, bipolar disorder, and multiple sclerosis as exemplars of neuro-immune disorders. We conclude that novel therapeutic targets exploring immune and oxidative & nitrosative pathways (p.e. melatonin and molecular hydrogen) hold promise in alleviating sleep and circadian dysfunction in these disorders.
Collapse
|
31
|
Abstract
Astrocytes are neural cells of ectodermal, neuroepithelial origin that provide for homeostasis and defense of the central nervous system (CNS). Astrocytes are highly heterogeneous in morphological appearance; they express a multitude of receptors, channels, and membrane transporters. This complement underlies their remarkable adaptive plasticity that defines the functional maintenance of the CNS in development and aging. Astrocytes are tightly integrated into neural networks and act within the context of neural tissue; astrocytes control homeostasis of the CNS at all levels of organization from molecular to the whole organ.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| | - Maiken Nedergaard
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| |
Collapse
|
32
|
Verkhratsky A, Nedergaard M. Physiology of Astroglia. Physiol Rev 2018; 98:239-389. [PMID: 29351512 PMCID: PMC6050349 DOI: 10.1152/physrev.00042.2016] [Citation(s) in RCA: 1068] [Impact Index Per Article: 152.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/22/2017] [Accepted: 04/27/2017] [Indexed: 02/07/2023] Open
Abstract
Astrocytes are neural cells of ectodermal, neuroepithelial origin that provide for homeostasis and defense of the central nervous system (CNS). Astrocytes are highly heterogeneous in morphological appearance; they express a multitude of receptors, channels, and membrane transporters. This complement underlies their remarkable adaptive plasticity that defines the functional maintenance of the CNS in development and aging. Astrocytes are tightly integrated into neural networks and act within the context of neural tissue; astrocytes control homeostasis of the CNS at all levels of organization from molecular to the whole organ.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| | - Maiken Nedergaard
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| |
Collapse
|
33
|
Kao YH, Lin MS, Chen CM, Wu YR, Chen HM, Lai HL, Chern Y, Lin CJ. Targeting ENT1 and adenosine tone for the treatment of Huntington's disease. Hum Mol Genet 2017; 26:467-478. [PMID: 28069792 DOI: 10.1093/hmg/ddw402] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/21/2016] [Indexed: 11/14/2022] Open
Abstract
Huntington's disease (HD) is caused by an abnormal CAG expansion in the exon 1 of huntingtin gene. The treatment of HD is an unmet medical need. Given the important role of adenosine in modulating brain activity, in this study, levels of adenosine and adenine nucleotides in the cerebral spinal fluid of patients with HD and in the brain of two mouse models of HD (R6/2 and Hdh150Q) were analysed. The expression and activity of ENT1 in the striatum of mice with HD were measured. Targeting adenosine tone for treating HD was examined in R6/2 mice by genetic removal of ENT1 and by giving an ENT1 inhibitor, respectively. The results showed that the adenosine homeostasis is dysregulated in the brain of patients and mice with HD. In patients, the ratio of adenosine/ATP in the cerebral spinal fluid was negatively correlated with the disease duration, and tended to have a positive correlation with independence scale and functional capacity. In comparison to controls, mRNA level of ENT1 was higher in the striatum of R6/2 and Hdh150Q mice. Intrastriatal administration of ENT1 inhibitors increased extracellular level of adenosine in the striatum of R6/2 mice to a much higher level than controls. Chronic inhibition of ENT1 or by genetic removal of ENT1 enhanced the survival of R6/2 mice. Collectively, adenosine homeostasis and ENT1 expression are altered in HD. The inhibition of ENT1 can enhance extracellular adenosine level and be a potential therapeutic approach for treating HD.
Collapse
Affiliation(s)
- Yu-Han Kao
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Meng-Syuan Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center and College of Medicine, Chang-Gung University, Tao-Yuan, Taiwan
| | - Yih-Ru Wu
- Department of Neurology, Chang Gung Memorial Hospital Linkou Medical Center and College of Medicine, Chang-Gung University, Tao-Yuan, Taiwan
| | - Hui-Mei Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hsing-Lin Lai
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yijuang Chern
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chun-Jung Lin
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
34
|
Vesicular nucleotide transporter (VNUT): appearance of an actress on the stage of purinergic signaling. Purinergic Signal 2017; 13:387-404. [PMID: 28616712 DOI: 10.1007/s11302-017-9568-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 05/05/2017] [Indexed: 12/17/2022] Open
Abstract
Vesicular storage of ATP is one of the processes initiating purinergic chemical transmission. Although an active transport mechanism was postulated to be involved in the processes, a transporter(s) responsible for the vesicular storage of ATP remained unidentified for some time. In 2008, SLC17A9, the last identified member of the solute carrier 17 type I inorganic phosphate transporter family, was found to encode the vesicular nucleotide transporter (VNUT) that is responsible for the vesicular storage of ATP. VNUT transports various nucleotides in a membrane potential-dependent fashion and is expressed in the various ATP-secreting cells. Mice with knockout of the VNUT gene lose vesicular storage and release of ATP from neurons and neuroendocrine cells, resulting in blockage of the initiation of purinergic chemical transmission. Thus, VNUT plays an essential role in the vesicular storage and release of ATP. The VNUT knockout mice exhibit resistance for neuropathic pain and a therapeutic effect against diabetes by way of increased insulin sensitivity. Thus, VNUT inhibitors and suppression of VNUT gene expression may be used for therapeutic purposes through suppression of purinergic chemical transmission. This review summarizes the studies to date on VNUT and discusses what we have learned about the relevance of vesicular ATP release as a potential drug target.
Collapse
|
35
|
Tatsuki F, Ode KL, Ueda HR. Ca 2+-dependent hyperpolarization hypothesis for mammalian sleep. Neurosci Res 2017; 118:48-55. [PMID: 28433628 DOI: 10.1016/j.neures.2017.03.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 03/24/2017] [Accepted: 03/27/2017] [Indexed: 11/15/2022]
Abstract
The detailed molecular mechanisms underlying the regulation of sleep/wake cycles in mammals are elusive. In this regulation, at least two mechanisms with fast and slow time scales are involved. In the faster time scale, a state of non-rapid-eye-movement (NREM) sleep can be microscopically characterized by the millisecond-to-second-order electrical behavior of neurons, namely slow-wave oscillations described by electrophysiology. In the slower time scale, the total duration of NREM sleep is homeostatically regulated by sleep pressure (the need for sleep), which is usually sustained for hours or even days and can be macroscopically described by electroencephalogram (EEG). The longer dynamics of sleep regulation are often explained by the accumulation of sleep-inducing substances (SISs). However, we still do not have a concrete model to connect fast, microscopic dynamics and slow, macroscopic dynamics. In this review, we introduce a recent Ca2+-dependent hyperpolarization hypothesis, in which the Ca2+-dependent hyperpolarization of cortical-membrane potential induces slow-wave oscillation. Slow dynamics of the Ca2+-dependent hyperpolarization pathway might be regulated by recently identified sleep-promoting kinases as well as classical SISs. Therefore, cortical Ca2+-dependent hyperpolarization may be a fundamental mechanism connecting fast neural activity to the slow dynamics of sleep pressure.
Collapse
Affiliation(s)
- Fumiya Tatsuki
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan; The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8865, Japan
| | - Koji L Ode
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan; Laboratory for Synthetic Biology, RIKEN Quantitative Biology Center, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hiroki R Ueda
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan; Laboratory for Synthetic Biology, RIKEN Quantitative Biology Center, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
36
|
The Role of Adenosine Signaling in Headache: A Review. Brain Sci 2017; 7:brainsci7030030. [PMID: 28335379 PMCID: PMC5366829 DOI: 10.3390/brainsci7030030] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 03/05/2017] [Accepted: 03/07/2017] [Indexed: 12/18/2022] Open
Abstract
Migraine is the third most prevalent disease on the planet, yet our understanding of its mechanisms and pathophysiology is surprisingly incomplete. Recent studies have built upon decades of evidence that adenosine, a purine nucleoside that can act as a neuromodulator, is involved in pain transmission and sensitization. Clinical evidence and rodent studies have suggested that adenosine signaling also plays a critical role in migraine headache. This is further supported by the widespread use of caffeine, an adenosine receptor antagonist, in several headache treatments. In this review, we highlight evidence that supports the involvement of adenosine signaling in different forms of headache, headache triggers, and basic headache physiology. This evidence supports adenosine A2A receptors as a critical adenosine receptor subtype involved in headache pain. Adenosine A2A receptor signaling may contribute to headache via the modulation of intracellular Cyclic adenosine monophosphate (cAMP) production or 5' AMP-activated protein kinase (AMPK) activity in neurons and glia to affect glutamatergic synaptic transmission within the brainstem. This evidence supports the further study of adenosine signaling in headache and potentially illuminates it as a novel therapeutic target for migraine.
Collapse
|
37
|
Revealing the role of the endocannabinoid system modulators, SR141716A, URB597 and VDM-11, in sleep homeostasis. Neuroscience 2016; 339:433-449. [DOI: 10.1016/j.neuroscience.2016.10.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 10/04/2016] [Accepted: 10/04/2016] [Indexed: 01/16/2023]
|
38
|
Zorec R, Parpura V, Verkhratsky A. Astroglial Vesicular Trafficking in Neurodegenerative Diseases. Neurochem Res 2016; 42:905-917. [DOI: 10.1007/s11064-016-2055-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/30/2016] [Accepted: 08/31/2016] [Indexed: 12/20/2022]
|
39
|
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
|
40
|
Abstract
UNLABELLED An emergent concept in neurosciences consists in considering brain functions as the product of dynamic interactions between neurons and glial cells, particularly astrocytes. Although the role played by astrocytes in synaptic transmission and plasticity is now largely documented, their contribution to neuronal network activity is only beginning to be appreciated. In mouse olfactory bulb slices, we observed that the membrane potential of mitral cells oscillates between UP and DOWN states at a low frequency (<1 Hz). Such slow oscillations are correlated with glomerular local field potentials, indicating spontaneous local network activity. Using a combination of genetic and pharmacological tools, we showed that the activity of astroglial connexin 43 hemichannels, opened in an activity-dependent manner, increases UP state amplitude and impacts mitral cell firing rate. This effect requires functional adenosine A1 receptors, in line with the observation that ATP is released via connexin 43 hemichannels. These results highlight a new mechanism of neuroglial interaction in the olfactory bulb, where astrocyte connexin hemichannels are both targets and modulators of neuronal circuit function. SIGNIFICANCE STATEMENT An emergent concept in neuroscience consists in considering brain function as the product of dynamic interactions between neurons and glial cells, particularly astrocytes. A typical feature of astrocytes is their high expression level of connexins, the molecular constituents of gap junction channels and hemichannels. Although hemichannels represent a powerful medium for intercellular communication between astrocytes and neurons, their function in physiological conditions remains largely unexplored. Our results show that in the olfactory bulb, connexin 43 hemichannel function is promoted by neuronal activity and, in turn, modulates neuronal network slow oscillations. This novel mechanism of neuroglial interaction could influence olfactory information processing by directly impacting the output of the olfactory bulb.
Collapse
|
41
|
Ingiosi AM, Opp MR. Sleep and immunomodulatory responses to systemic lipopolysaccharide in mice selectively expressing interleukin-1 receptor 1 on neurons or astrocytes. Glia 2016; 64:780-91. [PMID: 26775112 DOI: 10.1002/glia.22961] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 11/23/2015] [Accepted: 12/16/2015] [Indexed: 12/22/2022]
Abstract
Sleep-wake behavior is altered in response to immune challenge. Although the precise mechanisms that govern sickness-induced changes in sleep are not fully understood, interleukin-1β (IL-1) is one mediator of these responses. To better understand mechanisms underlying sleep and inflammatory responses to immune challenge, we used two transgenic mouse strains that express IL-1 receptor 1 (IL1R1) only in the central nervous system and selectively on neurons or astrocytes. Electroencephalographic recordings from transgenic and wild-type mice reveal that systemic challenge with lipopolysaccharide (LPS) fragments sleep, suppresses rapid eye movement sleep (REMS), increases non-REMS (NREMS), diminishes NREM delta power, and induces fever in all genotypes. However, the magnitude of REMS suppression is greater in mice expressing IL1R1 on astrocytes compared with mice in which IL1R1 is selectively expressed on neurons. Furthermore, there is a delayed increase in NREM delta power when IL1R1 is expressed on astrocytes. LPS-induced sleep fragmentation is reduced in mice expressing IL1R1 on neurons. Although LPS increases IL-1 and IL-6 in brain of all genotypes, this response is attenuated when IL1R1 is expressed selectively on neurons or on astrocytes. Collectively, these data suggest that in these transgenic mice under the conditions of this study it is neuronal IL1R1 that plays a greater role in LPS-induced suppression of REMS and NREM delta power, whereas astroglial IL1R1 is more important for sleep fragmentation after this immune challenge. Thus, aspects of central responses to LPS are modulated by IL1R1 in a cell type-specific manner.
Collapse
Affiliation(s)
- Ashley M Ingiosi
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan.,Program in Biomedical Sciences, University of Michigan, Ann Arbor, Michigan.,Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington
| | - Mark R Opp
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington.,Graduate Program in Neuroscience, University of Washington, Seattle, Washington
| |
Collapse
|
42
|
Verkhratsky A, Matteoli M, Parpura V, Mothet JP, Zorec R. Astrocytes as secretory cells of the central nervous system: idiosyncrasies of vesicular secretion. EMBO J 2016; 35:239-57. [PMID: 26758544 DOI: 10.15252/embj.201592705] [Citation(s) in RCA: 296] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 12/01/2015] [Indexed: 11/09/2022] Open
Abstract
Astrocytes are housekeepers of the central nervous system (CNS) and are important for CNS development, homeostasis and defence. They communicate with neurones and other glial cells through the release of signalling molecules. Astrocytes secrete a wide array of classic neurotransmitters, neuromodulators and hormones, as well as metabolic, trophic and plastic factors, all of which contribute to the gliocrine system. The release of neuroactive substances from astrocytes occurs through several distinct pathways that include diffusion through plasmalemmal channels, translocation by multiple transporters and regulated exocytosis. As in other eukaryotic cells, exocytotic secretion from astrocytes involves divergent secretory organelles (synaptic-like microvesicles, dense-core vesicles, lysosomes, exosomes and ectosomes), which differ in size, origin, cargo, membrane composition, dynamics and functions. In this review, we summarize the features and functions of secretory organelles in astrocytes. We focus on the biogenesis and trafficking of secretory organelles and on the regulation of the exocytotic secretory system in the context of healthy and diseased astrocytes.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Life Sciences, The University of Manchester, Manchester, UK Achucarro Center for Neuroscience, IKERBASQUE Basque Foundation for Science, Bilbao, Spain Department of Neurosciences, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain University of Nizhny Novgorod, Nizhny Novgorod, Russia Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology University of Ljubljana, Ljubljana, Slovenia Celica BIOMEDICAL, Ljubljana, Slovenia
| | - Michela Matteoli
- CNR Institute of Neuroscience, Milano, Italy Humanitas Research Hospital, Rozzano, Italy
| | - Vladimir Parpura
- Department of Neurobiology, Civitan International Research Center and Center for Glial Biology in Medicine, Evelyn F. McKnight Brain Institute, Atomic Force Microscopy & Nanotechnology Laboratories University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jean-Pierre Mothet
- Team Gliotransmission & Synaptopathies, Aix-Marseille University CNRS, CRN2M UMR7286, Marseille, France
| | - Robert Zorec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology University of Ljubljana, Ljubljana, Slovenia Celica BIOMEDICAL, Ljubljana, Slovenia
| |
Collapse
|
43
|
Sahlender DA, Savtchouk I, Volterra A. What do we know about gliotransmitter release from astrocytes? Philos Trans R Soc Lond B Biol Sci 2015; 369:20130592. [PMID: 25225086 PMCID: PMC4173278 DOI: 10.1098/rstb.2013.0592] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Astrocytes participate in information processing by actively modulating synaptic properties via gliotransmitter release. Various mechanisms of astrocytic release have been reported, including release from storage organelles via exocytosis and release from the cytosol via plasma membrane ion channels and pumps. It is still not fully clear which mechanisms operate under which conditions, but some of them, being Ca2+-regulated, may be physiologically relevant. The properties of Ca2+-dependent transmitter release via exocytosis or via ion channels are different and expected to produce different extracellular transmitter concentrations over time and to have distinct functional consequences. The molecular aspects of these two release pathways are still under active investigation. Here, we discuss the existing morphological and functional evidence in support of either of them. Transgenic mouse models, specific antagonists and localization studies have provided insight into regulated exocytosis, albeit not in a systematic fashion. Even more remains to be uncovered about the details of channel-mediated release. Better functional tools and improved ultrastructural approaches are needed in order fully to define specific modalities and effects of astrocytic gliotransmitter release pathways.
Collapse
Affiliation(s)
- Daniela A Sahlender
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, Lausanne 1005, Switzerland
| | - Iaroslav Savtchouk
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, Lausanne 1005, Switzerland
| | - Andrea Volterra
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, Lausanne 1005, Switzerland
| |
Collapse
|
44
|
Ropert N, Jalil A, Li D. Expression and cellular function of vSNARE proteins in brain astrocytes. Neuroscience 2015; 323:76-83. [PMID: 26518463 DOI: 10.1016/j.neuroscience.2015.10.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 10/07/2015] [Accepted: 10/21/2015] [Indexed: 12/27/2022]
Abstract
Gray matter protoplasmic astrocytes, a major type of glial cell in the mammalian brain, extend thin processes ensheathing neuronal synaptic terminals. Albeit electrically silent, astrocytes respond to neuronal activity with Ca(2+) signals that trigger the release of gliotransmitters, such as glutamate, d-serine, and ATP, which modulate synaptic transmission. It has been suggested that the astrocytic processes, together with neuronal pre- and post-synaptic elements, constitute a tripartite synapse, and that astrocytes actively regulate information processing. Astrocytic vesicles expressing VAMP2 and VAMP3 vesicular SNARE (vSNARE) proteins have been suggested to be a key feature of the tripartite synapse and mediate gliotransmitter release through Ca(2+)-regulated exocytosis. However, the concept of exocytotic release of gliotransmitters by astrocytes has been challenged. Here we review studies investigating the expression profile of VAMP2 and VAMP3 vSNARE proteins in rodent astrocytes, and the functional implication of VAMP2/VAMP3 vesicles in astrocyte signaling. We also discuss our recent data suggesting that astrocytic VAMP3 vesicles regulate the trafficking of glutamate transporters at the plasma membrane and glutamate uptake. A better understanding of the functional consequences of the astrocytic vSNARE vesicles on glutamate signaling, neuronal excitability and plasticity, will require the development of new strategies to selectively interrogate the astrocytic vesicles trafficking in vivo.
Collapse
Affiliation(s)
- N Ropert
- Brain Physiology Laboratory, CNRS UMR8118, Paris F-75006, France; Fédération de Recherche en Neurosciences, FR 3636, Université Paris Descartes, 45 rue des Saints Pères, Paris F-75006, France; Sorbonne Paris Cité, 190, avenue de France, Paris F-75013, France
| | - A Jalil
- Brain Physiology Laboratory, CNRS UMR8118, Paris F-75006, France; Fédération de Recherche en Neurosciences, FR 3636, Université Paris Descartes, 45 rue des Saints Pères, Paris F-75006, France; Sorbonne Paris Cité, 190, avenue de France, Paris F-75013, France
| | - D Li
- Brain Physiology Laboratory, CNRS UMR8118, Paris F-75006, France; Fédération de Recherche en Neurosciences, FR 3636, Université Paris Descartes, 45 rue des Saints Pères, Paris F-75006, France; Sorbonne Paris Cité, 190, avenue de France, Paris F-75013, France.
| |
Collapse
|
45
|
Vardjan N, Parpura V, Zorec R. Loose excitation-secretion coupling in astrocytes. Glia 2015; 64:655-67. [PMID: 26358496 DOI: 10.1002/glia.22920] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 08/26/2015] [Indexed: 12/19/2022]
Abstract
Astrocytes play an important housekeeping role in the central nervous system. Additionally, as secretory cells, they actively participate in cell-to-cell communication, which can be mediated by membrane-bound vesicles. The gliosignaling molecules stored in these vesicles are discharged into the extracellular space after the vesicle membrane fuses with the plasma membrane. This process is termed exocytosis, regulated by SNARE proteins, and triggered by elevations in cytosolic calcium levels, which are necessary and sufficient for exocytosis in astrocytes. For astrocytic exocytosis, calcium is sourced from the intracellular endoplasmic reticulum store, although its entry from the extracellular space contributes to cytosolic calcium dynamics in astrocytes. Here, we discuss calcium management in astrocytic exocytosis and the properties of the membrane-bound vesicles that store gliosignaling molecules, including the vesicle fusion machinery and kinetics of vesicle content discharge. In astrocytes, the delay between the increase in cytosolic calcium activity and the discharge of secretions from the vesicular lumen is orders of magnitude longer than that in neurons. This relatively loose excitation-secretion coupling is likely tailored to the participation of astrocytes in modulating neural network processing.
Collapse
Affiliation(s)
- Nina Vardjan
- Celica Biomedical, Ljubljana, Slovenia.,Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Vladimir Parpura
- Department of Neurobiology, Center for Glial Biology in Medicine, Civitan International Research Center, Atomic Force Microscopy & Nanotechnology Laboratories, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, Alabama
| | - Robert Zorec
- Celica Biomedical, Ljubljana, Slovenia.,Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
46
|
Do stars govern our actions? Astrocyte involvement in rodent behavior. Trends Neurosci 2015; 38:535-49. [DOI: 10.1016/j.tins.2015.07.006] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Revised: 07/24/2015] [Accepted: 07/28/2015] [Indexed: 12/20/2022]
|
47
|
Ode KL, Ueda HR. Seeing the forest and trees: whole-body and whole-brain imaging for circadian biology. Diabetes Obes Metab 2015; 17 Suppl 1:47-54. [PMID: 26332968 DOI: 10.1111/dom.12511] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 05/26/2015] [Indexed: 01/13/2023]
Abstract
Recent advances in methods for making mammalian organs translucent have made possible whole-body fluorescent imaging with single-cell resolution. Because organ-clearing methods can be used to image the heterogeneous nature of cell populations, they are powerful tools to investigate the hierarchical organization of the cellular circadian clock, and how the clock synchronizes a variety of physiological activities. In particular, methods compatible with genetically encoded fluorescent reporters have the potential to detect circadian activity in different brain regions and the circadian-phase distribution across the whole body. In this review, we summarize the current methods and strategy for making organs translucent (removal of lipids, decolourization of haemoglobin and adjusting the refractive index of the specimen). We then discuss possible applications to circadian biology. For example, the coupling of circadian rhythms among different brain regions, brain activity in sleep-wake cycles and the role of migrating cells such as immune cells and cancer cells in chronopharmacology.
Collapse
Affiliation(s)
- K L Ode
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Quantitative Biology Center, RIKEN, Osaka, Japan
| | - H R Ueda
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Quantitative Biology Center, RIKEN, Osaka, Japan
| |
Collapse
|
48
|
Witts EC, Nascimento F, Miles GB. Adenosine-mediated modulation of ventral horn interneurons and spinal motoneurons in neonatal mice. J Neurophysiol 2015; 114:2305-15. [PMID: 26311185 PMCID: PMC4609759 DOI: 10.1152/jn.00574.2014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 08/21/2015] [Indexed: 01/20/2023] Open
Abstract
Neuromodulation allows neural networks to adapt to varying environmental and biomechanical demands. Purinergic signaling is known to be an important modulatory system in many parts of the CNS, including motor control circuitry. We have recently shown that adenosine modulates the output of mammalian spinal locomotor control circuitry (Witts EC, Panetta KM, Miles GB. J Neurophysiol 107: 1925–1934, 2012). Here we investigated the cellular mechanisms underlying this adenosine-mediated modulation. Whole cell patch-clamp recordings were performed on ventral horn interneurons and motoneurons within in vitro mouse spinal cord slice preparations. We found that adenosine hyperpolarized interneurons and reduced the frequency and amplitude of synaptic inputs to interneurons. Both effects were blocked by the A1-type adenosine receptor antagonist DPCPX. Analysis of miniature postsynaptic currents recorded from interneurons revealed that adenosine reduced their frequency but not amplitude, suggesting that adenosine acts on presynaptic receptors to modulate synaptic transmission. In contrast to interneurons, recordings from motoneurons revealed an adenosine-mediated depolarization. The frequency and amplitude of synaptic inputs to motoneurons were again reduced by adenosine, but we saw no effect on miniature postsynaptic currents. Again these effects on motoneurons were blocked by DPCPX. Taken together, these results demonstrate differential effects of adenosine, acting via A1 receptors, in the mouse spinal cord. Adenosine has a general inhibitory action on ventral horn interneurons while potentially maintaining motoneuron excitability. This may allow for adaptation of the locomotor pattern generated by interneuronal networks while helping to ensure the maintenance of overall motor output.
Collapse
Affiliation(s)
- Emily C Witts
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, Fife, United Kingdom
| | - Filipe Nascimento
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, Fife, United Kingdom
| | - Gareth B Miles
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, Fife, United Kingdom
| |
Collapse
|
49
|
Dissel S, Seugnet L, Thimgan MS, Silverman N, Angadi V, Thacher PV, Burnham MM, Shaw PJ. Differential activation of immune factors in neurons and glia contribute to individual differences in resilience/vulnerability to sleep disruption. Brain Behav Immun 2015; 47:75-85. [PMID: 25451614 PMCID: PMC4416079 DOI: 10.1016/j.bbi.2014.09.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 09/26/2014] [Accepted: 09/29/2014] [Indexed: 11/26/2022] Open
Abstract
Individuals frequently find themselves confronted with a variety of challenges that threaten their wellbeing. While some individuals face these challenges efficiently and thrive (resilient) others are unable to cope and may suffer persistent consequences (vulnerable). Resilience/vulnerability to sleep disruption may contribute to the vulnerability of individuals exposed to challenging conditions. With that in mind we exploited individual differences in a fly's ability to form short-term memory (STM) following 3 different types of sleep disruption to identify the underlying genes. Our analysis showed that in each category of flies examined, there are individuals that form STM in the face of sleep loss (resilient) while other individuals show dramatic declines in cognitive behavior (vulnerable). Molecular genetic studies revealed that Antimicrobial Peptides, factors important for innate immunity, were candidates for conferring resilience/vulnerability to sleep deprivation. Specifically, Metchnikowin (Mtk), drosocin (dro) and Attacin (Att) transcript levels seemed to be differentially increased by sleep deprivation in glia (Mtk), neurons (dro) or primarily in the head fat body (Att). Follow-up genetic studies confirmed that expressing Mtk in glia but not neurons, and expressing dro in neurons but not glia, disrupted memory while modulating sleep in opposite directions. These data indicate that various factors within glia or neurons can contribute to individual differences in resilience/vulnerability to sleep deprivation.
Collapse
Affiliation(s)
- Stephane Dissel
- Department of Anatomy & Neurobiology, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, USA
| | - Laurent Seugnet
- Department of Anatomy & Neurobiology, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, USA
| | - Matthew S. Thimgan
- Department of Anatomy & Neurobiology, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, USA
| | - Neal Silverman
- Division of Infectious Diseases, Department of Medicine, University of Massachusetts Medical School, 364 Plantation St., Worcester, MA 01605
| | - Veena Angadi
- Department of Anatomy & Neurobiology, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, USA
| | - Pamela V. Thacher
- Department of Psychology, St Lawrence University, 23 Romoda Drive, Canton, NY 13617
| | | | - Paul J. Shaw
- Department of Anatomy & Neurobiology, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, USA
| |
Collapse
|
50
|
Abstract
Gliotransmission, a process involving active vesicular release of glutamate and other neurotransmitters by astrocytes, is thought to play a critical role in many brain functions. A new paper by Nedergaard et al. (2014) identifies an experimental flaw in these previous studies suggesting that astrocytes may not perform active vesicular release after all.
Collapse
Affiliation(s)
- Steven A Sloan
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305-5125, USA.
| | - Ben A Barres
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305-5125, USA
| |
Collapse
|