1
|
Coluk Y, Yildirim G, Yildirmak S, Peker EGG. Altered brain-derived neurotrophic factor levels and oxidative stress in REM sleep deprivation: a rat model study. BMC Neurol 2025; 25:122. [PMID: 40119302 PMCID: PMC11927282 DOI: 10.1186/s12883-025-04127-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 03/07/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) is among the modulators associated with cognition and sleep that play a role in sleep disorders. This study aimed at investigating the effects of chronic sleep deprivation and REM sleep deprivation on BDNF levels and oxidative stress markers. METHODS A total of 24 healthy male Wistar albino rats were separated into 3 groups as REM sleep deprivation group, control sleep deprivation group and control group. To create models of 21-day REM sleep deprivation and control sleep deprivation, we used the platform technique. After 21 days blood BDNF, brain tissue BDNF, brain tissue malondialdehyde, glutathione, ascorbic acid, nitrite and nitrate were evaluated. RESULTS Compared with the control group, control sleep deprivation group showed a significant increase in brain tissue levels of BDNF (p = 0.038), whereas a significant decrease was observed in the levels of glutathione (GSH) and nitric oxide (NO) (p:0.036). No statistical difference was observed between the blood levels of BDNF in either group (p: 0.795). CONCLUSION Our results showed decreases in GSH and NO levels and increases in malondialdehyde levels in the sleep deprivation models, reflecting oxidative stress in the brain. Additionally, we observed increases in brain BDNF levels in the control sleep deprivation model.
Collapse
Affiliation(s)
- Yonca Coluk
- Department of Otorhinolaryngology, Faculty of Medicine, Giresun University, Giresun, 28200, Turkey.
| | | | - Sembol Yildirmak
- Department of Biochemistry, Faculty of Medicine, Mersin University, Mersin, 33000, Turkey
| | | |
Collapse
|
2
|
Muzumdar N, Jackson KM, Buckman JF, Spaeth AM, Sokolovsky AW, Pawlak AP, White HR. Elucidating the alcohol-sleep-hangover relationship in college students using a daily diary approach. Pharmacol Biochem Behav 2025; 247:173910. [PMID: 39579876 DOI: 10.1016/j.pbb.2024.173910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/30/2024] [Accepted: 11/18/2024] [Indexed: 11/25/2024]
Abstract
This daily diary study expands knowledge of the pharmacological alcohol-sleep relationship using a multilevel modeling approach. The interplay between alcohol and sleep on hangover susceptibility is also explored. College students (n = 337; 52 % female) provided 2976 days of self-reported alcohol use. We regressed sleep duration onto accumulated sleep debt, prior night sleep duration, and estimated blood alcohol concentration (eBAC) at bedtime; linear mixed models disaggregated day and person-level effects. Binomial models, assessing days after drinking when eBAC = 0 % versus when eBAC>0 % at waketime, regressed hangover susceptibility onto the same predictors plus sleep duration. More accumulated sleep debt predicted slightly longer same-night sleep. Greater than average bedtime intoxication predicted longer than average same-night sleep when drinking ceased early, but later drinking attenuated the relationship. People who typically stopped drinking later in the night reported typically shorter sleep durations on drinking nights. When waketime eBAC = 0 %, higher eBAC at bedtime and drinking later on a given night predicted greater next-day hangover susceptibility. Typical bedtime eBAC and typically later drinking predicted typically greater hangover susceptibility. When waketime eBAC>0 %, longer sleep duration predicted more likely hangovers. Bedtime eBAC and sleep debt interacted, such that more sleep debt attenuated the positive association between intoxication and next-day hangover susceptibility. Late-night drinking appeared to reduce sleep duration and increase hangover susceptibility. Accumulated sleep debt complicated the alcohol-sleep-hangover relationship. External factors influencing sleep behaviors were not assessed, but the results highlight the need to deconstruct sleep into acute and chronic processes. Future studies should better subdivide physiological processes related to hangovers.
Collapse
Affiliation(s)
- Neel Muzumdar
- Department of Kinesiology and Health, Rutgers University - New Brunswick, New Brunswick, NJ 08901, United States of America
| | - Kristina M Jackson
- Rutgers Addiction Research Center, Rutgers University - New Brunswick, Piscataway, NJ 08854, United States of America.
| | - Jennifer F Buckman
- Department of Kinesiology and Health, Rutgers University - New Brunswick, New Brunswick, NJ 08901, United States of America
| | - Andrea M Spaeth
- Department of Kinesiology and Health, Rutgers University - New Brunswick, New Brunswick, NJ 08901, United States of America
| | - Alexander W Sokolovsky
- Center for Alcohol and Addiction Studies, Brown University School of Public Health, Providence, RI 02903, United States of America
| | - Anthony P Pawlak
- Department of Kinesiology and Health, Rutgers University - New Brunswick, New Brunswick, NJ 08901, United States of America; Rutgers Addiction Research Center, Rutgers University - New Brunswick, Piscataway, NJ 08854, United States of America
| | - Helene R White
- Center of Alcohol and Substance Use Studies, Rutgers University - New Brunswick, Piscataway, NJ 08854, United States of America
| |
Collapse
|
3
|
Zheng Y, Bao J, Tang L, Chen C, Guo W, Li K, Zhang R, Wu J. Association between weekend catch-up sleep and depression of the United States population from 2017 to 2018: A cross-sectional study. Sleep Med 2024; 119:9-16. [PMID: 38631161 DOI: 10.1016/j.sleep.2024.04.012] [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: 12/11/2023] [Revised: 04/02/2024] [Accepted: 04/08/2024] [Indexed: 04/19/2024]
Abstract
Insufficient sleep on weekdays has become a societal norm, and studies have shown that sleep deprivation increases the risk of depression. Although individuals often resort to weekend catch-up sleep (CUS) as a compensatory measure, the present evidence supporting its efficacy in mitigating the risk of depression is limited. This article attempts to explore the relationship between CUS and depression. In this study, a total of 5510 participants were included, characterized into two groups: nondepressed (n = 5051) and depressed (n = 459), with data extracted from the National Health and Nutrition Examination Survey (NHANES). Compared with people without CUS, those practicing CUS exhibited a significantly lower risk of depression (OR = 0.81, P = 0.048). In subgroup analysis, this reduction effect was only observed in males (OR = 0.70, 95 % CI 0.05 to 0.99, P = 0.04), middle-aged (>40, ≤60) (OR: 0.57, 95 % CI: 0.40 to 0.81, P = 0.002), married or living with parents (OR: 0.61, 95 % CI: 0.44 to 0.86, P = 0.004), groups with three or more family members (OR: 0.69, 95 % CI: 0.52 to 0.93, P = 0.01), and individuals without alcohol intake (OR: 0.24,95 % CI: 0.09 to 0.67, P = 0.006). Therefore, in the realm of depression treatment, doctors may consider advising patients to get adequate sleep on weekends as part of their overall treatment plan. At the same time, individuals can also choose weekend sleep as a proactive strategy for regulating their psychological status.
Collapse
Affiliation(s)
- Yanxu Zheng
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China; Xiangya School of Medicine, Central South University, Hunan, 410013, China
| | - Jing Bao
- Xiangya School of Medicine, Central South University, Hunan, 410013, China
| | - Long Tang
- Xiangya School of Medicine, Central South University, Hunan, 410013, China
| | - Chuhua Chen
- Xiangya School of Medicine, Central South University, Hunan, 410013, China
| | - Weiqin Guo
- Xiangya School of Medicine, Central South University, Hunan, 410013, China
| | - Kecheng Li
- Xiangya School of Medicine, Central South University, Hunan, 410013, China
| | - Ruxu Zhang
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China.
| | - Jinze Wu
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
4
|
Xiao X, Rui Y, Jin Y, Chen M. Relationship of Sleep Disorder with Neurodegenerative and Psychiatric Diseases: An Updated Review. Neurochem Res 2024; 49:568-582. [PMID: 38108952 DOI: 10.1007/s11064-023-04086-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 12/19/2023]
Abstract
Sleep disorders affect many people worldwide and can accompany neurodegenerative and psychiatric diseases. Sleep may be altered before the clinical manifestations of some of these diseases appear. Moreover, some sleep disorders affect the physiological organization and function of the brain by influencing gene expression, accelerating the accumulation of abnormal proteins, interfering with the clearance of abnormal proteins, or altering the levels of related hormones and neurotransmitters, which can cause or may be associated with the development of neurodegenerative and psychiatric diseases. However, the detailed mechanisms of these effects are unclear. This review mainly focuses on the relationship between and mechanisms of action of sleep in Alzheimer's disease, depression, and anxiety, as well as the relationships between sleep and Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. This summary of current research hotspots may provide researchers with better clues and ideas to develop treatment solutions for neurodegenerative and psychiatric diseases associated with sleep disorders.
Collapse
Affiliation(s)
- Xiao Xiao
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, Anhui, China
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Yimin Rui
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, Anhui, China
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Yu Jin
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Ming Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
5
|
Zhu J, Chen C, Liu X, He M, Fang Y, Wang L, Jia J, Guo J, Zhao Z, Gao C, He J, Xu C, Xu F, Ma D, Wang J, Zhang Z. Cerebellar Purkinje cell firing promotes conscious recovery from anesthesia state through coordinating neuronal communications with motor cortex. Theranostics 2024; 14:480-495. [PMID: 38169536 PMCID: PMC10758059 DOI: 10.7150/thno.89592] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/11/2023] [Indexed: 01/05/2024] Open
Abstract
Background: The neurobiological basis of gaining consciousness from unconscious state induced by anesthetics remains unknown. This study was designed to investigate the involvement of the cerebello-thalamus-motor cortical loop mediating consciousness transitions from the loss of consciousness (LOC) induced by an inhalational anesthetic sevoflurane in mice. Methods: The neural tracing and fMRI together with opto-chemogenetic manipulation were used to investigate the potential link among cerebello-thalamus-motor cortical brain regions. The fiber photometry of calcium and neurotransmitters, including glutamate (Glu), γ-aminobutyric acid (GABA) and norepinephrine (NE), were monitored from the motor cortex (M1) and the 5th lobule of the cerebellar vermis (5Cb) during unconsciousness induced by sevoflurane and gaining consciousness after sevoflurane exposure. Cerebellar Purkinje cells were optogenetically manipulated to investigate their influence on consciousness transitions during and after sevoflurane exposure. Results: Activation of 5Cb Purkinje cells increased the Ca2+ flux in the M1 CaMKIIα+ neurons, but this increment was significantly reduced by inactivation of posterior and parafascicular thalamic nucleus. The 5Cb and M1 exhibited concerted calcium flux, and glutamate and GABA release during transitions from wakefulness, loss of consciousness, burst suppression to conscious recovery. Ca2+ flux and Glu release in the M1, but not in the 5Cb, showed a strong synchronization with the EEG burst suppression, particularly, in the gamma-band range. In contrast, the Glu, GABA and NE release and Ca2+ oscillations were coherent with the EEG gamma band activity only in the 5Cb during the pre-recovery of consciousness period. The optogenetic activation of Purkinje cells during burst suppression significantly facilitated emergence from anesthesia while the optogenetic inhibition prolonged the time to gaining consciousness. Conclusions: Our data indicate that cerebellar neuronal communication integrated with motor cortex through thalamus promotes consciousness recovery from anesthesia which may likely serve as arousal regulation.
Collapse
Affiliation(s)
- Jinpiao Zhu
- Department of Anesthesiology, Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
- Department of Anesthesiology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Chang Chen
- Department of Anesthesiology, Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Xiaodong Liu
- Department of Anesthesia and Intensive Care, Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, P.R. China
| | - Mengying He
- Department of Anesthesiology, Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Yuanyuan Fang
- Department of Anesthesiology, Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Li Wang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan 430071, China
| | - Junke Jia
- Department of Anesthesiology, Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Juan Guo
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ziyue Zhao
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan 430071, China
| | - Chenyi Gao
- Department of Anesthesiology, Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Jingang He
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan 430071, China
| | - Chengshi Xu
- Department of Anesthesiology, Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Fuqiang Xu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Daqing Ma
- Department of Anesthesiology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Division of Anesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Jie Wang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zongze Zhang
- Department of Anesthesiology, Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| |
Collapse
|
6
|
Ding W, Yang L, Shi E, Kim B, Low S, Hu K, Gao L, Chen P, Ding W, Borsook D, Luo A, Choi JH, Wang C, Akeju O, Yang J, Ran C, Schreiber KL, Mao J, Chen Q, Feng G, Shen S. The endocannabinoid N-arachidonoyl dopamine is critical for hyperalgesia induced by chronic sleep disruption. Nat Commun 2023; 14:6696. [PMID: 37880241 PMCID: PMC10600211 DOI: 10.1038/s41467-023-42283-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 10/05/2023] [Indexed: 10/27/2023] Open
Abstract
Chronic pain is highly prevalent and is linked to a broad range of comorbidities, including sleep disorders. Epidemiological and clinical evidence suggests that chronic sleep disruption (CSD) leads to heightened pain sensitivity, referred to as CSD-induced hyperalgesia. However, the underlying mechanisms are unclear. The thalamic reticular nucleus (TRN) has unique integrative functions in sensory processing, attention/arousal and sleep spindle generation. We report that the TRN played an important role in CSD-induced hyperalgesia in mice, through its projections to the ventroposterior region of the thalamus. Metabolomics revealed that the level of N-arachidonoyl dopamine (NADA), an endocannabinoid, was decreased in the TRN after CSD. Using a recently developed CB1 receptor (cannabinoid receptor 1) activity sensor with spatiotemporal resolution, CB1 receptor activity in the TRN was found to be decreased after CSD. Moreover, CSD-induced hyperalgesia was attenuated by local NADA administration to the TRN. Taken together, these results suggest that TRN NADA signaling is critical for CSD-induced hyperalgesia.
Collapse
Affiliation(s)
- Weihua Ding
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Liuyue Yang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eleanor Shi
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Bowon Kim
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sarah Low
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kun Hu
- Department of Pathology, Tuft University School of Medicine, Boston, MA, USA
| | - Lei Gao
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ping Chen
- College of Science and Mathematics, University of Massachusetts Boston, Boston, MA, USA
| | - Wei Ding
- College of Science and Mathematics, University of Massachusetts Boston, Boston, MA, USA
| | - David Borsook
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew Luo
- Summer Intern Program of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, currently at Brandeis University, Boston, MA, USA
| | - Jee Hyun Choi
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul, South Korea
| | - Changning Wang
- Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Oluwaseun Akeju
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jun Yang
- Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Chongzhao Ran
- Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kristin L Schreiber
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jianren Mao
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Qian Chen
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Guoping Feng
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Shiqian Shen
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Ranjan A, Biswas S, Mallick BN. Rapid eye movement sleep loss associated cytomorphometric changes and neurodegeneration. Sleep Med 2023; 110:25-34. [PMID: 37524037 DOI: 10.1016/j.sleep.2023.07.022] [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: 02/20/2023] [Revised: 07/10/2023] [Accepted: 07/22/2023] [Indexed: 08/02/2023]
Abstract
Rapid eye movement sleep (REMS) is essential for leading normal healthy living at least in higher-order mammals, including humans. In this review, we briefly survey the available literature for evidence linking cytomorphometric changes in the brain due to loss of REMS. As a mechanism of action, we add evidence that REMS loss elevates noradrenaline (NA) levels in the brain, which affects neuronal cytomorphology. These changes may be a compensatory mechanism as the changes return to normal after the subjects recover from the loss of REMS or if during REMS deprivation, the subjects are treated with NA-adrenoceptor antagonist prazosin (PRZ). We had proposed earlier that one of the fundamental functions of REMS is to maintain the level of NA in the brain. We elaborate on this idea to propose that if REMS loss continues without recovery, the sustained level of NA breaks down neurophysiologically active compensatory mechanism/s starting with changes in the neuronal cytomorphology, followed by their degeneration, leading to acute and chronic pathological conditions. Identification of neuronal cytomorphological changes could prove to be of significance for predicting future neuronal (brain) damage as well as an indicator for REMS health. Although current brain imaging techniques may not enable us to visualize changes in neuronal cytomorphology, given the rapid technological progress including use of artificial intelligence, we are optimistic that it may be a reality soon. Finally, we propose that maintenance of optimum REMS must be considered a criterion for leading a healthy life.
Collapse
Affiliation(s)
- Amit Ranjan
- Department of Zoology, Mahatma Gandhi Central University, Motihari, East Champaran, Bihar, 845401, India.
| | - Sudipta Biswas
- Math, Science, Engineering Department, South Mountain Community College, 7050 S 24th St, Phoenix, AZ, 85042, USA
| | - Birendra Nath Mallick
- Amity Institute of Neuropsychology & Neurosciences, Amity University Campus, Sector 125, Gautam Budh Nagar, Noida, 201313, Uttar Pradesh, India
| |
Collapse
|
8
|
Ma WX, Yuan PC, Zhang H, Kong LX, Lazarus M, Qu WM, Wang YQ, Huang ZL. Adenosine and P1 receptors: Key targets in the regulation of sleep, torpor, and hibernation. Front Pharmacol 2023; 14:1098976. [PMID: 36969831 PMCID: PMC10036772 DOI: 10.3389/fphar.2023.1098976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 02/27/2023] [Indexed: 03/12/2023] Open
Abstract
Graphical AbstractAdenosine mediates sleep, torpor and hibernation through P1 receptors. Recent reasearch has shown that P1 receptors play a vital role in the regulation of sleep-wake, torpor and hibernation-like states. In this review, we focus on the roles and neurobiological mechanisms of the CNS adenosine and P1 receptors in these three states. Among them, A1 and A2A receptors are key targets for sleep-wake regulation, A1Rs and A3Rs are very important for torpor induction, and activation of A1Rs is sufficient for hibernation-like state.
Collapse
Affiliation(s)
- Wei-Xiang Ma
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Department of Pharmacology, School of Basic Medical Sciences, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ping-Chuan Yuan
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, School of Pharmacy, Wannan Medical College, Wuhu, China
| | - Hui Zhang
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, School of Pharmacy, Wannan Medical College, Wuhu, China
| | - Ling-Xi Kong
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Department of Pharmacology, School of Basic Medical Sciences, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Michael Lazarus
- International Institute for Integrative Sleep Medicine (WPI-IIIS) and Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Wei-Min Qu
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Department of Pharmacology, School of Basic Medical Sciences, Institutes of Brain Science, Fudan University, Shanghai, China
- *Correspondence: Wei-Min Qu, ; Yi-Qun Wang, ; Zhi-Li Huang,
| | - Yi-Qun Wang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Department of Pharmacology, School of Basic Medical Sciences, Institutes of Brain Science, Fudan University, Shanghai, China
- *Correspondence: Wei-Min Qu, ; Yi-Qun Wang, ; Zhi-Li Huang,
| | - Zhi-Li Huang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Department of Pharmacology, School of Basic Medical Sciences, Institutes of Brain Science, Fudan University, Shanghai, China
- *Correspondence: Wei-Min Qu, ; Yi-Qun Wang, ; Zhi-Li Huang,
| |
Collapse
|
9
|
Li T, Gao Y, He M, Gui Z, Zhao B, Cao Y, Chen T, Zhu J, Wang J, Zhong Q, Zhang Z. P2X7 receptor-activated microglia in cortex is critical for sleep disorder under neuropathic pain. Front Neurosci 2023; 17:1095718. [PMID: 36816134 PMCID: PMC9936193 DOI: 10.3389/fnins.2023.1095718] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Neuropathic pain (NP) is associated with sleep disturbances, which may substantially influence the quality of life. Clinical and animal studies demonstrated that neurotransmitter is one of the main contributors to cause sleep disturbances induced by NP. Recently, it was reported that P2X7 receptors (P2X7R) are widely expressed in microglia, which serves crucial role in neuronal activity in the pain and sleep-awake cycle. In this study, we adopted the chronic constriction injury (CCI) model to establish the progress of chronic pain and investigated whether P2X7R of microglia in cortex played a critical role in sleep disturbance induced by NP. At electroencephalogram (EEG) level, sleep disturbance was observed in mice treated with CCI as they exhibited mechanical and thermal hypersensitivity, and inhibition of P2X7R ameliorated these changes. We showed a dramatic high level of P2X7R and Iba-1 co-expression in the cortical region, and the inhibition of P2X7R also adversely affected it. Furthermore, the power of LFPs in ventral posterior nucleus (VP) and primary somatosensory cortex (S1) which changed in the CCI group was adverse after the inhibition of P2X7R. Furthermore, inhibition of P2X7R also decreased the VP-S1 coherence which increased in CCI group. Nuclear magnetic resonance demonstrated inhibition of P2X7R decreased glutamate (Glu) levels in thalamic and cortical regions which were significantly increased in the CCI mice. Our findings provide evidence that NP has a critical effect on neuronal activity linked to sleep and may built up a new target for the development of sleep disturbances under chronic pain conditions.
Collapse
Affiliation(s)
- Tingting Li
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Yunling Gao
- Xiangyang Central Hospital, Institute of Neuroscience and Brain Diseases, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Mengying He
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China,Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, China
| | - Zhu Gui
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, China,University of Chinese Academy of Sciences, Beijing, China
| | - Bingchu Zhao
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, China,School of Computer Science, Wuhan University, Wuhan, Hubei, China
| | - Yue Cao
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Ting Chen
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Jinpiao Zhu
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China,Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, China
| | - Jie Wang
- Xiangyang Central Hospital, Institute of Neuroscience and Brain Diseases, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China,Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, China,University of Chinese Academy of Sciences, Beijing, China
| | - Qi Zhong
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China,*Correspondence: Qi Zhong,
| | - Zongze Zhang
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China,Zongze Zhang,
| |
Collapse
|
10
|
Yu Z, Li B, Tang W, Dong Z, Liu R, Yu S. Acute sleep deprivation aggravates nitroglycerin-evoked hyperalgesia in mice. Mol Pain 2023; 19:17448069221149645. [PMID: 36550614 PMCID: PMC9830572 DOI: 10.1177/17448069221149645] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Sleep deprivation can trigger migraine, and migraineurs often choose to sleep to relieve headaches during acute migraine. This study aimed to explore the effect of acute sleep deprivation on hyperalgesia induced by nitroglycerin in mice. In part one, after either 6-h sleep deprivation or 6-h normal sleep, mice were intraperitoneally injected with nitroglycerin or saline. The mechanical pain threshold and withdrawal latency of the hindpaw were measured every 30 min for 6 h. Next, the same sleep deprivation and injection procedure was performed with new mice, and mice were sacrificed 4.5 h after injection. The trigeminal nucleus caudalis and upper cervical spinal segments were taken for immunofluorescence Fos staining. In part two, after injection of saline or nitroglycerin, the mice were either deprived of sleep for 6 h or allowed to sleep without interference. The mechanical and thermal pain threshold were measured after 6 h. In part three, we compared the sleep time of mice after intraperitoneal injection of saline or nitroglycerin without interference. Sleep deprivation for 6 h did not cause any changes in the baseline pain thresholds in mice. However, pretreatment with 6-h sleep deprivation significantly prolonged the duration of hyperalgesia induced by nitroglycerin. Additionally, the expression of Fos at 4.5 h was significantly higher in the 6-h sleep deprivation and nitroglycerin group than in the other three groups. When intraperitoneal injection was given first, the mechanical pain threshold of the hind paw was significantly lower in the group that received nitroglycerin with 6-h sleep deprivation than in the other groups. Compared to the saline injection, one-time nitroglycerin injection would result in a significant increase in sleep latency and decrease in sleep duration for the normal mice. Acute sleep deprivation significantly aggravated the hyperalgesia induced by nitroglycerin in mice, which highlights the importance of sleep disorders for migraine.
Collapse
Affiliation(s)
- Zhe Yu
- International Headache Center, Department of Neurology, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Bozhi Li
- International Headache Center, Department of Neurology, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Wenjing Tang
- International Headache Center, Department of Neurology, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Zhao Dong
- International Headache Center, Department of Neurology, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Ruozhuo Liu
- International Headache Center, Department of Neurology, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Shengyuan Yu
- International Headache Center, Department of Neurology, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
11
|
Guzeev MA, Kurmazov NS, Ekimova IV. [Chronic sleep restriction in rats leads to a weakening of compensatory reactions in response to acute sleep deprivation]. Zh Nevrol Psikhiatr Im S S Korsakova 2023; 123:35-42. [PMID: 37275996 DOI: 10.17116/jnevro202312305235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To identify features in the compensatory mechanisms of sleep regulation in response to acute sleep deprivation after chronic sleep restriction in rats. MATERIAL AND METHODS Male Wistar rats 7-8 months old underwent 5-day sleep restriction: 3 h of sleep deprivation and 1 h of sleep opportunity repeating throughout each day. Six-hour acute total sleep deprivation was performed at the beginning of daylight hours on the 3rd day after sleep restriction. Polysomnogramms were recorded throughout the day before chronic sleep restriction, on the 2nd recovery day after chronic sleep restriction and after acute sleep deprivation. The control group was not subjected to chronic sleep restriction. RESULTS The animals after chronic sleep restriction had the compensatory increase in total sleep time in response to acute sleep deprivation weaker than in control animals. Animals after sleep restriction had the compensatory increase in the time of slow-wave sleep (SWS) only in the first 6 hours after acute sleep deprivation, whereas in control animals the period of compensation of SWS lasted 12 hours. A compensatory increase in slow-wave activity (SWA) was observed in both groups of animals, but in animals experiencing chronic sleep restriction the amplitude of SWA after acute sleep deprivation was less than in control animals. A compensatory increase in REM sleep in sleep restricted animals occurred immediately after acute sleep deprivation and coincides with a compensatory increase in SWS and SWA, whereas in control conditions these processes are spaced in time. CONCLUSION Compensatory reactions in response to acute sleep deprivation (sleep homeostasis) are weakened in animals subjected to chronic sleep restriction, as the reaction time and amplitude are reduced.
Collapse
Affiliation(s)
- M A Guzeev
- Sechenov Institute of Evolutionary Physiology and Biochemistry, St Petersburg, Russia
| | - N S Kurmazov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, St Petersburg, Russia
| | - I V Ekimova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, St Petersburg, Russia
| |
Collapse
|
12
|
Zamore Z, Veasey SC. Neural consequences of chronic sleep disruption. Trends Neurosci 2022; 45:678-691. [PMID: 35691776 PMCID: PMC9388586 DOI: 10.1016/j.tins.2022.05.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/09/2022] [Accepted: 05/20/2022] [Indexed: 12/25/2022]
Abstract
Recent studies in both humans and animal models call into question the completeness of recovery after chronic sleep disruption. Studies in humans have identified cognitive domains particularly vulnerable to delayed or incomplete recovery after chronic sleep disruption, including sustained vigilance and episodic memory. These findings, in turn, provide a focus for animal model studies to critically test the lasting impact of sleep loss on the brain. Here, we summarize the human response to sleep disruption and then discuss recent findings in animal models examining recovery responses in circuits pertinent to vigilance and memory. We then propose pathways of injury common to various forms of sleep disruption and consider the implications of this injury in aging and in neurodegenerative disorders.
Collapse
Affiliation(s)
- Zachary Zamore
- Chronobiology and Sleep Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sigrid C Veasey
- Chronobiology and Sleep Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
13
|
López-Muciño LA, García-García F, Cueto-Escobedo J, Acosta-Hernández M, Venebra-Muñoz A, Rodríguez-Alba JC. Sleep loss and addiction. Neurosci Biobehav Rev 2022; 141:104832. [PMID: 35988803 DOI: 10.1016/j.neubiorev.2022.104832] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 11/25/2022]
Abstract
Reducing sleep hours is a risk factor for developing cardiovascular, metabolic, and psychiatric disorders. Furthermore, previous studies have shown that reduction in sleep time is a factor that favors relapse in addicted patients. Additionally, animal models have demonstrated that both sleep restriction and sleep deprivation increase the preference for alcohol, methylphenidate, and the self-administration of cocaine. Therefore, the present review discusses current knowledge about the influence of sleep hours reduction on addictivebehaviors; likewise, we discuss the neuronal basis underlying the sleep reduction-addiction relationship, like the role of the orexin and dopaminergic system and neuronal plasticity (i.e., delta FosB expression). Potentially, chronic sleep restriction could increase brain vulnerability and promote addictive behavior.
Collapse
Affiliation(s)
- Luis Angel López-Muciño
- Health Sciences Ph.D. Program, Health Sciences Institute, Veracruzana University, Xalapa, VER 91190, Mexico.
| | - Fabio García-García
- Department of Biomedicine, Health Sciences Institute, Veracruzana University, Xalapa, VER 91190, Mexico.
| | - Jonathan Cueto-Escobedo
- Department of Clinical and Translational Research, Health Sciences Institute, Veracruzana University, Xalapa, VER 91190, Mexico.
| | - Mario Acosta-Hernández
- Department of Biomedicine, Health Sciences Institute, Veracruzana University, Xalapa, VER 91190, Mexico.
| | - Arturo Venebra-Muñoz
- Laboratory of Neurobiology of Addiction and Brain Plasticity, Faculty of Science, Autonomous University of Mexico State, Edomex 50295, Mexico.
| | - Juan Carlos Rodríguez-Alba
- Department of Biomedicine, Health Sciences Institute, Veracruzana University, Xalapa, VER 91190, Mexico.
| |
Collapse
|
14
|
Bjorness TE, Greene RW. Arousal-Mediated Sleep Disturbance Persists During Cocaine Abstinence in Male Mice. Front Neurosci 2022; 16:868049. [PMID: 35812231 PMCID: PMC9260276 DOI: 10.3389/fnins.2022.868049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Acute cocaine disturbs sleep on a dose-dependent basis; however, the consequences of chronic cocaine remain unclear. While the arousal promotion following cocaine has been well-established, effects of cocaine on sleep after termination of chronic cocaine exposure appear variable in human subjects with few studies in non-human subjects. Here, a within-subjects design (outcomes normalized to baseline, undisturbed behavior) and between-subjects design (repeated experimenter-administered cocaine vs. experimenter-administered saline) was used to investigate sleep homeostasis and sleep/waking under repeated cocaine/saline exposure and prolonged forced abstinence conditions in mice. Overall, during the forced abstinence period increases in arousal, as determined by sleep latency and gamma energy, persisted for 2 weeks. However, the sleep response to externally enforced sleep deprivation was unchanged suggesting that sleep disruptions during the forced abstinence period were driven by enhancement of arousal in the absence of changes in sleep homeostatic responses.
Collapse
Affiliation(s)
- Theresa E. Bjorness
- Research Service, Veterans Affairs (VA) North Texas Health Care System, Dallas, TX, United States
- Department of Psychiatry, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern, Dallas, TX, United States
- *Correspondence: Theresa E. Bjorness,
| | - Robert W. Greene
- Department of Psychiatry, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern, Dallas, TX, United States
- Department of Neuroscience, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
15
|
Fernandes GL, Araujo P, Tufik S, Andersen ML. SLEEPINESS PROFILES IN MICE SUBMITTED TO ACUTE AND CHRONIC SLEEP DEPRIVATION. Behav Processes 2022; 200:104661. [PMID: 35618241 DOI: 10.1016/j.beproc.2022.104661] [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: 02/25/2021] [Revised: 05/09/2022] [Accepted: 05/16/2022] [Indexed: 11/29/2022]
Abstract
Sleepiness is a behavioural consequence of sleep pressure, which shows interindividual variation, a characteristic possibly related to central sleep mechanisms. However, there is a lack of evidence linking progressive sleep need and sleepiness with factors of individual variability, which could be tested by total acute and chronic sleep deprivation. Thus, the objective of the study was to investigate the development of sleepiness in sleep deprived mice. Male C57BL/6J mice were distributed in sleep deprivation, sleep rebound and control groups. Animals underwent acute sleep deprivation for 3, 6, 9 or 12hours or chronic sleep deprivation for 6hours for 5 consecutive days. Sleep rebound groups had a sleep opportunity for 1, 2, 3, or 4hours after acute sleep deprivation or 24hours after chronic sleep deprivation. During the protocols, sleep attempts were counted to calculate a sleepiness index. After euthanasia, blood was collected for corticosterone assessment. Using the average of group sleep attempts, it was possible to differentiate between sleepy (mean>group average) and resistant animals (mean<group average). Resistant mice were more frequent in all protocols. Individual variation accounted for 52% of sleepiness variance during chronic sleep deprivation and extended wakefulness explained 68% of sleepiness variance during acute sleep deprivation. A normal corticosterone peak was observed at the start of the dark phase, independent of sleep deprivation. Different profiles of sleepiness emerged in sleep deprived mice. Sleep deprivation was the main factor for sleepiness during acute sleep deprivation whereas in chronic deprivation individual variation was more relevant.
Collapse
Affiliation(s)
| | - Paula Araujo
- Departamento de Psicobiologia - Universidade Federal de São Paulo - São Paulo, Brazil; Departamento de Ciências Fisiológicas - Escola de Ciências Médicas, Santa Casa de São Paulo - São Paulo, Brazil
| | - Sergio Tufik
- Departamento de Psicobiologia - Universidade Federal de São Paulo - São Paulo, Brazil
| | - Monica Levy Andersen
- Departamento de Psicobiologia - Universidade Federal de São Paulo - São Paulo, Brazil.
| |
Collapse
|
16
|
Atrooz F, Alrousan G, Hassan A, Salim S. Early-Life Sleep Deprivation Enhanced Alcohol Consumption in Adolescent Rats. Front Neurosci 2022; 16:856120. [PMID: 35546871 PMCID: PMC9081815 DOI: 10.3389/fnins.2022.856120] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 02/24/2022] [Indexed: 12/05/2022] Open
Abstract
Evidence in the literature suggests that sleep deprivation during early-life developmental stages, by impacting important processes such as the reward circuit maturation, may increase the vulnerability for alcohol and substance use. The mechanisms involved are not fully understood. In this study, utilizing our previously established model, we examined the impact of early-life sleep deprivation on alcohol consumption in adolescent rats. Male Sprague Dawley rats served as either the control (CON) or sleep-deprived (SD) group. Sleep deprivation was induced using a Pinnacle automated sleep deprivation apparatus. The SD group of rats was sleep deprived for 6–8 h/day for 14 days from postnatal day (PND)19 to PND32. At PND33, anxiety- and depression-like behaviors were assessed in rats using elevated plus maze and sucrose splash test, respectively. At PND39, alcohol consumption was assessed in rats for five consecutive days using the two-bottle choice paradigm, water versus 5% ethanol. SD rats exhibited significant anxiety- and depression-like behaviors as compared to CON rats. Interestingly, SD rats consumed a larger volume of alcohol when compared to CON rats, which was significantly higher at day 5 (mean of alcohol consumption (ml) ± SD; CON = 6.67 ± 3.42; SD = 19.00 ± 6.05, p = 0.0126). SD rats also showed high preference for alcohol over water, which was significantly higher at day 5 (mean of alcohol preference (%) ± SD; CON = 26.85 ± 14.97; SD = 57.69 ± 5.61, p = 0.014). Our data suggest that early-life sleep deprivation enhanced alcohol consumption in adolescent rats.
Collapse
Affiliation(s)
- Fatin Atrooz
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, United States
| | - Ghalya Alrousan
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, United States
| | - Arham Hassan
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, United States
| | - Samina Salim
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, United States
| |
Collapse
|
17
|
Seungmagalgeun-Tang, a Traditional Herbal Formula, Alleviates Skin Inflammation and Depression-Like Behavior in Atopic Dermatitis Mice under Sleep Deprivation Conditions. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1307173. [PMID: 35368752 PMCID: PMC8967505 DOI: 10.1155/2022/1307173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/05/2022] [Indexed: 11/29/2022]
Abstract
Atopic dermatitis (AD) is a common inflammatory skin disease, which can be worsened under sleep deprivation (SD) conditions. This study investigated the efficacy and the mechanism of action of the traditional herbal formula Seungmagalgeun-tang (SMGGT) on the inflammation and behavioral changes in a mouse model of AD exposed to SD. SMGGT decreased levels of IgE, TNF-α, IL-4, IL-13, and mast cell infiltration and reduced the expression of CD3 in the mouse skin. SMGGT also reversed the SD-induced increase in corticosterone and decrease in melatonin level. Furthermore, SMGGT reduced the immobility time in the tail suspension test significantly. HaCaT cells and HMC-1 cells were used to investigate the effects of SMGGT on cell signaling pathways. In TNF-α/IFN-γ (TI) treated HaCaT cells, SMGGT reduced production of TARC/CCL17 and MDC/CCL22 and suppressed the p38 MAPK, STAT1, and NF-κB pathways. In substance P (SP)/CRH-stimulated HMC-1 cells, SMGGT decreased VEGF production and inhibited ERK phosphorylation. Network pharmacology and molecular docking analysis revealed that puerarin and paeoniflorin might contribute to the effects of SMGGT by targeting several AD-related molecules and pathways. Puerarin and paeoniflorin exerted anti-inflammatory effects by decreasing production of MDC/CCL22 and IL-6 in TI-treated HaCaT cells and VEGF production in SP/CRH-stimulated HMC-1 cells. This study suggests that SMGGT with puerarin and paeoniflorin as main bioactive components alleviates skin inflammation and depression-like behavior in a sleep-deprived mouse model of AD.
Collapse
|
18
|
Deurveilher S, Antonchuk M, Saumure BSC, Baldin A, Semba K. No loss of orexin/hypocretin, melanin-concentrating hormone or locus coeruleus noradrenergic neurons in a rat model of chronic sleep restriction. Eur J Neurosci 2021; 54:6027-6043. [PMID: 34355453 DOI: 10.1111/ejn.15412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/13/2021] [Accepted: 07/29/2021] [Indexed: 12/31/2022]
Abstract
Chronic sleep restriction (CSR) is common in modern society, adversely affecting cognitive performance and health. Yet how it impacts neurons regulating sleep remains unclear. Several studies using mice reported substantial losses of wake-active orexin/hypocretin and locus coeruleus (LC) noradrenergic neurons, but not rapid eye movement sleep-active melanin-concentrating hormone (MCH) neurons, following CSR. Here, we used immunohistochemistry and stereology to examine orexin, MCH and LC noradrenergic neurons in a rat model of CSR that uses programmed wheel rotation (3 h on/1 h off; '3/1' protocol). Adult male Wistar rats underwent one or four cycles of the 4-day 3/1 CSR protocol, with 2-day recovery between cycles in home cages. Time-matched control rats were housed in locked wheels/home cages. We found no significant differences in the numbers of orexin, MCH and LC noradrenergic neurons following either one- or four-cycle CSR protocol compared to respective controls. Similarly, the four-cycle CSR protocol had no effect on the densities of orexin axon terminals in the LC, noradrenergic dendrites in the LC and noradrenergic axon terminals in the frontal cortex. Body weights, however, decreased after one cycle of CSR and then increased with diminishing slope over the next three cycles. Thus, we found no evidence for loss of orexin or LC noradrenergic neurons following one and four cycles of the 4-day 3/1 CSR protocol in rats. Differences in CSR protocols and/or possible species differences in neuronal vulnerability to sleep loss may account for the discrepancy between the current results in rats and previous findings in mice.
Collapse
Affiliation(s)
- Samuel Deurveilher
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Michael Antonchuk
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Brock St C Saumure
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Andrew Baldin
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kazue Semba
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
19
|
Guzeev MA, Kurmazov NS, Simonova VV, Pastukhov YF, Ekimova IV. [Modeling of chronic sleep restriction for translational studies]. Zh Nevrol Psikhiatr Im S S Korsakova 2021; 121:6-13. [PMID: 34078853 DOI: 10.17116/jnevro20211214026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To develop of a chronic sleep restriction model in rats by repeated sleep deprivation using an orbital shaker and to determine whether this model leads to disturbances in sleep homeostatic mechanisms. MATERIAL AND METHODS Male Wistar rats (7-8 months old) underwent sleep restriction for five consecutive days: 3 h of sleep deprivation and 1 h of sleep opportunity repeating throughout each day. Polysomnograms were recorded telemetrically throughout the day before sleep restriction (baseline), on the 1st, 3rd, 5th day of sleep restriction and 2 days after the end of sleep restriction (recovery period). RESULTS During the period of sleep restriction, the total amount of slow-wave sleep (SWS) and rapid eye movement (REM) sleep decreased by 61% and 55%, respectively, compared to baseline. On the first day of recovery, amount of SWS increased mainly in the dark (active) phase of the day, while REM sleep increased in both light and dark phases; there was no marked rebound of daily SWS amount, while REM sleep increased by 30% from baseline. On the first day of recovery, an elevation of EEG beta and sigma power in sleep states was observed mainly in the light phase of the day. The loss of deep SWS throughout the sleep restriction period increased from 50% on 1st day to 75% on 5th day. The level of deep SWS remained below the baseline by 15-20% on the two subsequent days of recovery. The findings suggest that homeostatic mechanisms of SWS are persistently impaired after 5-day chronic sleep restriction. Besides, a decline of wakefulness accompanied by an increase of SWS in the active phase of the recovery period indicates a disruption in circadian rhythm. CONCLUSION The proposed model leads to the disruption of sleep homeostatic mechanisms, which, in turn, impede compensation of SWS loss caused by chronic insufficient sleep.
Collapse
Affiliation(s)
- M A Guzeev
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg, Russia
| | - N S Kurmazov
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg, Russia
| | - V V Simonova
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg, Russia
| | - Yu F Pastukhov
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg, Russia
| | - I V Ekimova
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg, Russia
| |
Collapse
|
20
|
Skorucak J, Weber N, Carskadon MA, Reynolds C, Coussens S, Achermann P, Short MA. Homeostatic Response to Sleep Restriction in Adolescents. Sleep 2021; 44:6249597. [PMID: 33893807 DOI: 10.1093/sleep/zsab106] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/21/2021] [Indexed: 11/12/2022] Open
Abstract
The high prevalence of chronic sleep restriction in adolescents underscores the importance of understanding how adolescent sleep is regulated under such conditions. One component of sleep regulation is a homeostatic process: if sleep is restricted, then sleep intensity increases. Our knowledge of this process is primarily informed by total sleep deprivation studies and has been incorporated in mathematical models of human sleep regulation. Several animal studies, however, suggest that adaptation occurs in chronic sleep restriction conditions, showing an attenuated or even decreased homeostatic response. We investigated the homeostatic response of adolescents to different sleep opportunities. Thirty-four participants were allocated to one of three groups with 5, 7.5 or 10 h of sleep opportunity per night for 5 nights. Each group underwent a protocol of 9 nights designed to mimic a school week between 2 weekends: 2 baseline nights (10 h sleep opportunity), 5 condition nights (5, 7.5 or 10 h), and two recovery nights (10 h). Measures of sleep homeostasis (slow-wave activity and slow-wave energy) were calculated from frontal and central EEG derivations and compared to predictions derived from simulations of the homeostatic process of the two-process model of sleep regulation. Only minor differences were found between empirical data and model predictions, indicating that sleep homeostasis is preserved under chronic sleep restriction in adolescents. These findings improve our understanding of effects of repetitive short sleep in adolescents.
Collapse
Affiliation(s)
- Jelena Skorucak
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.,Sleep and Health Zurich, University of Zurich, Zurich, Switzerland
| | - Nathan Weber
- School of Psychology, Flinders University, Adelaide, South Australia
| | - Mary A Carskadon
- E.P. Bradley Hospital, Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Chelsea Reynolds
- School of Psychology, Flinders University, Adelaide, South Australia
| | - Scott Coussens
- Cognitive Neuroscience Laboratory, University of South Australia, Adelaide, South Australia
| | - Peter Achermann
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.,Sleep and Health Zurich, University of Zurich, Zurich, Switzerland.,The KEY Institute for Brain Mind Research, Department of Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry, Zurich, Switzerland
| | - Michelle A Short
- School of Psychology, Flinders University, Adelaide, South Australia
| |
Collapse
|
21
|
Deurveilher S, Shewchuk SM, Semba K. Homeostatic sleep and body temperature responses to acute sleep deprivation are preserved following chronic sleep restriction in rats. J Sleep Res 2021; 30:e13348. [PMID: 33783043 DOI: 10.1111/jsr.13348] [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: 12/28/2020] [Revised: 03/10/2021] [Accepted: 03/17/2021] [Indexed: 11/30/2022]
Abstract
Chronic sleep insufficiency is common in our society and has negative cognitive and health impacts. It can also alter sleep regulation, yet whether it affects subsequent homeostatic responses to acute sleep loss is unclear. We assessed sleep and thermoregulatory responses to acute sleep deprivation before and after a '3/1' chronic sleep restriction protocol in adult male Wistar rats. The 3/1 protocol consisted of continuous cycles of wheel rotations (3 h on/1 h off) for 4 days. Sleep latency in a 2-h multiple sleep latency test starting 26 h post-3/1 was unchanged, whereas non-rapid eye movement sleep (NREMS) and associated electroencephalogram delta power (a measure of sleep need) over a 24-h period beginning 54 h post-3/1 were reduced, compared to respective pre-3/1 baseline levels. However, in response to acute sleep deprivation (6 h by 'gentle handling') starting 78 h post-3/1, the compensatory rebounds in NREMS and rapid eye movement sleep (REMS) amounts and NREMS delta power were unaltered. Body temperature increased progressively across the 3/1 protocol and returned to baseline levels on the second day post-3/1. The acute sleep deprivation also increased body temperature, followed by a decline below baseline levels, with no difference between before and after 3/1 sleep restriction. Non-sleep-restricted control rats showed responses to acute sleep deprivation similar to those observed in the sleep-restricted animals. These results suggest that the process of sleep homeostasis is altered on the third recovery day after a 4-day 3/1 sleep restriction protocol, whereas subsequent homeostatic sleep and temperature responses to brief sleep deprivation are not affected.
Collapse
Affiliation(s)
- Samuel Deurveilher
- Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada
| | | | - Kazue Semba
- Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada.,Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, Canada.,Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
22
|
Changed signals of blood adenosine and cytokines are associated with parameters of sleep and/or cognition in the patients with chronic insomnia disorder. Sleep Med 2021; 81:42-51. [PMID: 33636543 DOI: 10.1016/j.sleep.2021.02.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 01/13/2021] [Accepted: 02/01/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVES This study aimed to investigate whether plasma levels of adenosine, adenosine deaminase (ADA), and certain cytokines change in patients with chronic insomnia disorder (CID), and if so, whether these alterations are associated with poor sleep quality and cognitive dysfunction. METHODS Fifty-five CID patients were selected for the study, along with fifty-five healthy controls (HC) matched to the patients according to their basic data. All subjects completed sleep, emotion, and cognition assessments, with some CID patients also completing an overnight polysomnography. The plasma level of adenosine was measured using liquid chromatography-tandem mass spectrometry, while ADA level was quantified using a quantitative sandwich enzyme-linked immunosorbent assay. Levels of cytokines, including IL-1β, IL-2, IL-4, IL-6, IL-10, IL-12, TNF-α, and IFN-γ, were measured using Luminex liquid chip technology. RESULTS CID patients had a lower adenosine level, and higher levels of ADA and some of the cytokines (IL-1β, IL-2, IL-6, IL-10 and TNF-α) compared with controls. In the CID group, plasma concentrations of adenosine were negatively correlated with Pittsburgh Sleep Quality Index scores, while concentrations of IL-1β, IL-6 and TNF-α were positively correlated with these scores. Concentrations of IL-1β and TNF-α were negatively correlated with scores on the Chinese-Beijing Version of the Montreal Cognitive Assessment. Moreover, levels of IL-1β, TNF-α, IL-6, and IL-2 were positively correlated with memory test errors by CID patients after controlling for confounding factors. CONCLUSIONS The reduced adenosine and elevated cytokine levels of CID patients were associated with the severity of insomnia and/or cognitive dysfunction.
Collapse
|
23
|
Cortical astrocytes regulate ethanol consumption and intoxication in mice. Neuropsychopharmacology 2021; 46:500-508. [PMID: 32464636 PMCID: PMC8027025 DOI: 10.1038/s41386-020-0721-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/18/2022]
Abstract
Astrocytes are fundamental building blocks of the central nervous system. Their dysfunction has been implicated in many psychiatric disorders, including alcohol use disorder, yet our understanding of their functional role in ethanol intoxication and consumption is very limited. Astrocytes regulate behavior through multiple intracellular signaling pathways, including G-protein coupled-receptor (GPCR)-mediated calcium signals. To test the hypothesis that GPCR-induced calcium signaling is also involved in the behavioral effects of ethanol, we expressed astrocyte-specific excitatory DREADDs in the prefrontal cortex (PFC) of mice. Activating Gq-GPCR signaling in PFC astrocytes increased drinking in ethanol-naïve mice, but not in mice with a history of ethanol drinking. In contrast, reducing calcium signaling with an astrocyte-specific calcium extruder reduced ethanol intake. Cortical astrocyte calcium signaling also altered the acute stimulatory and sedative-hypnotic effects of ethanol. Astrocyte-specific Gq-DREADD activation increased both the locomotor-activating effects of low dose ethanol and the sedative-hypnotic effects of a high dose, while reduced astrocyte calcium signaling diminished sensitivity to the hypnotic effects. In addition, we found that adenosine A1 receptors were required for astrocyte calcium activation to increase ethanol sedation. These results support integral roles for PFC astrocytes in the behavioral actions of ethanol that are due, at least in part, to adenosine receptor activation.
Collapse
|
24
|
Nagai J, Yu X, Papouin T, Cheong E, Freeman MR, Monk KR, Hastings MH, Haydon PG, Rowitch D, Shaham S, Khakh BS. Behaviorally consequential astrocytic regulation of neural circuits. Neuron 2020; 109:576-596. [PMID: 33385325 DOI: 10.1016/j.neuron.2020.12.008] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/23/2020] [Accepted: 12/07/2020] [Indexed: 12/16/2022]
Abstract
Astrocytes are a large and diverse population of morphologically complex cells that exist throughout nervous systems of multiple species. Progress over the last two decades has shown that astrocytes mediate developmental, physiological, and pathological processes. However, a long-standing open question is how astrocytes regulate neural circuits in ways that are behaviorally consequential. In this regard, we summarize recent studies using Caenorhabditis elegans, Drosophila melanogaster, Danio rerio, and Mus musculus. The data reveal diverse astrocyte mechanisms operating in seconds or much longer timescales within neural circuits and shaping multiple behavioral outputs. We also refer to human diseases that have a known primary astrocytic basis. We suggest that including astrocytes in mechanistic, theoretical, and computational studies of neural circuits provides new perspectives to understand behavior, its regulation, and its disease-related manifestations.
Collapse
Affiliation(s)
- Jun Nagai
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; RIKEN Center for Brain Science, 2-1 Hirosawa Wako City, Saitama 351-0198, Japan
| | - Xinzhu Yu
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, 514 Burrill Hall, 407 S. Goodwin Ave, Urbana, IL 61801, USA
| | - Thomas Papouin
- Department of Neuroscience, Washington University in St. Louis, School of Medicine, Campus Box 8108, 660 South Euclid Ave., St. Louis, MO 63110, USA
| | - Eunji Cheong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Marc R Freeman
- The Vollum Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Kelly R Monk
- The Vollum Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Michael H Hastings
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Philip G Haydon
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - David Rowitch
- Department of Paediatrics, Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK; Departments of Pediatrics and Neurosurgery, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
| | - Shai Shaham
- Laboratory of Developmental Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Baljit S Khakh
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA.
| |
Collapse
|
25
|
Roehrs TA, Auciello J, Tseng J, Whiteside G. Current and potential pharmacological treatment options for insomnia in patients with alcohol use disorder in recovery. Neuropsychopharmacol Rep 2020; 40:211-223. [PMID: 32543111 PMCID: PMC7722668 DOI: 10.1002/npr2.12117] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/28/2020] [Accepted: 05/07/2020] [Indexed: 01/12/2023] Open
Abstract
Alcohol use disorder (AUD) is characterized by dysfunction in motivational, mood-stress regulation, and sleep systems that interact in complex ways to heighten the risk of relapse during abstinence. Emerging data suggest that excessive and chronic alcohol use disrupts sleep homeostasis and, in abstinence, subjects with AUD are known to experience insomnia that may persist for weeks to years, which we propose to refer to as insomnia associated with alcohol cessation (IAAC). The purpose of this review is to provide an update of pharmacological approaches to therapy including compounds in development, to raise awareness of the prevalence of and unmet need in IAAC and highlight differences in treatment consideration for IAAC as compared to insomnia disorder. We performed a search of select electronic databases to identify studies of pharmacological agents used to treat sleep disturbances in abstinent or treatment-seeking patients with alcohol use disorder. The search, conducted in June 2019 and updated in December 2019, yielded 1,188 abstracts after duplicates were removed, of which 36 full-text articles were assessed for eligibility. Eighteen studies were included, 15 randomized controlled trials and three open-label studies. Several classes of medications including antidepressants, anticonvulsants, and antipsychotics have been evaluated for their effectiveness in treating sleep disturbances in abstinent or treatment-seeking patients with AUD. None of these medications are approved by the FDA for the treatment of IAAC, and the currently available evidence for these agents is limited. Randomized, controlled clinical trials are warranted to evaluate the efficacy and safety of medications in the treatment of IAAC.
Collapse
Affiliation(s)
- Timothy A. Roehrs
- Henry Ford Health SystemSleep Disorders and Research CenterDetroitMIUSA
- Department of Psychiatry and Behavioral NeuroscienceSchool of MedicineWayne State UniversityDetroitMIUSA
| | | | | | | |
Collapse
|
26
|
Differential modulation of NREM sleep regulation and EEG topography by chronic sleep restriction in mice. Sci Rep 2020; 10:18. [PMID: 31924847 PMCID: PMC6954245 DOI: 10.1038/s41598-019-54790-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/18/2019] [Indexed: 11/26/2022] Open
Abstract
Compensatory elevation in NREM sleep EEG delta power has been typically observed following prolonged wakefulness and widely used as a sleep homeostasis indicator. However, recent evidence in human and rodent chronic sleep restriction (CSR) studies suggests that NREM delta power is not progressively increased despite of accumulated sleep loss over days. In addition, there has been little progress in understanding how sleep EEG in different brain regions responds to CSR. Using novel high-density EEG electrode arrays in the mouse model of CSR where mice underwent 18-h sleep deprivation per day for 5 consecutive days, we performed an extensive analysis of topographical NREM sleep EEG responses to the CSR condition, including period-amplitude analysis of individual slow waves. As previously reported in our analysis of REM sleep responses, we found different patterns of changes: (i) progressive decrease in NREM sleep duration and consolidation, (ii) persistent enhancement in NREM delta power especially in the frontal and parietal regions, and (iii) progressive increases in individual slow wave slope and frontal fast oscillation power. These results suggest that multiple sleep-wake regulatory systems exist in a brain region-specific manner, which can be modulated independently, especially in the CSR condition.
Collapse
|
27
|
Serchov T, Schwarz I, Theiss A, Sun L, Holz A, Döbrössy MD, Schwarz MK, Normann C, Biber K, van Calker D. Enhanced adenosine A 1 receptor and Homer1a expression in hippocampus modulates the resilience to stress-induced depression-like behavior. Neuropharmacology 2019; 162:107834. [PMID: 31682853 DOI: 10.1016/j.neuropharm.2019.107834] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 10/19/2019] [Accepted: 10/29/2019] [Indexed: 11/30/2022]
Abstract
Resilience to stress is critical for the development of depression. Enhanced adenosine A1 receptor (A1R) signaling mediates the antidepressant effects of acute sleep deprivation (SD). However, chronic SD causes long-lasting upregulation of brain A1R and increases the risk of depression. To investigate the effects of A1R on mood, we utilized two transgenic mouse lines with inducible A1R overexpression in forebrain neurons. These two lines have identical levels of A1R increase in the cortex, but differ in the transgenic A1R expression in the hippocampus. Switching on the transgene promotes robust antidepressant and anxiolytic effects in both lines. The mice of the line without transgenic A1R overexpression in the hippocampus (A1Hipp-) show very strong resistance towards development of stress-induced chronic depression-like behavior. In contrast, the mice of the line in which A1R upregulation extends to the hippocampus (A1Hipp+), exhibit decreased resilience to depression as compared to A1Hipp-. Similarly, automatic analysis of reward behavior of the two lines reveals that depression resistant A1Hipp-transgenic mice exhibit high sucrose preference, while mice of the vulnerable A1Hipp + line developed stress-induced anhedonic phenotype. The A1Hipp + mice have increased Homer1a expression in hippocampus, correlating with impaired long-term potentiation in the CA1 region, mimicking the stressed mice. Furthermore, virus-mediated overexpression of Homer1a in the hippocampus decreases stress resilience. Taken together our data indicate for first time that increased expression of A1R and Homer1a in the hippocampus modulates the resilience to stress-induced depression and thus might potentially mediate the detrimental effects of chronic sleep restriction on mood.
Collapse
Affiliation(s)
- Tsvetan Serchov
- Department of Stereotactic and Functional Neurosurgery, Medical Center - University Freiburg, Faculty of Medicine, University of Freiburg, Breisacher Str. 64, 79106, Freiburg, Germany.
| | - Inna Schwarz
- Functional Neuroconnectomics Group, Department of Experimental Epileptology and Cognition Research, Life and Brain Centre, University of Bonn, Medical School, 53105, Bonn, Germany
| | - Alice Theiss
- Department for Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hauptstrasse 5, 79104, Freiburg, Germany
| | - Lu Sun
- Department for Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hauptstrasse 5, 79104, Freiburg, Germany; Department Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, 9713, AV Groningen, the Netherlands
| | - Amrei Holz
- Department for Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hauptstrasse 5, 79104, Freiburg, Germany; Faculty of Biology, University of Freiburg, Schaenzlestr. 1, 79104, Freiburg, Germany
| | - Mate D Döbrössy
- Department of Stereotactic and Functional Neurosurgery, Medical Center - University Freiburg, Faculty of Medicine, University of Freiburg, Breisacher Str. 64, 79106, Freiburg, Germany
| | - Martin K Schwarz
- Functional Neuroconnectomics Group, Department of Experimental Epileptology and Cognition Research, Life and Brain Centre, University of Bonn, Medical School, 53105, Bonn, Germany
| | - Claus Normann
- Department for Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hauptstrasse 5, 79104, Freiburg, Germany
| | - Knut Biber
- Department for Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hauptstrasse 5, 79104, Freiburg, Germany; Department Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, 9713, AV Groningen, the Netherlands
| | - Dietrich van Calker
- Department for Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hauptstrasse 5, 79104, Freiburg, Germany
| |
Collapse
|
28
|
Calker D, Biber K, Domschke K, Serchov T. The role of adenosine receptors in mood and anxiety disorders. J Neurochem 2019; 151:11-27. [DOI: 10.1111/jnc.14841] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/15/2019] [Accepted: 07/18/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Dietrich Calker
- Department for Psychiatry and Psychotherapy, Medical Center ‐ University of Freiburg, Faculty of Medicine University of Freiburg Freiburg Germany
| | - Knut Biber
- Section Medical Physiology, Department of Neuroscience University Medical Center Groningen, University of Groningen Groningen The Netherlands
| | - Katharina Domschke
- Department for Psychiatry and Psychotherapy, Medical Center ‐ University of Freiburg, Faculty of Medicine University of Freiburg Freiburg Germany
- Centre for Basics in Neuromodulation, Faculty of Medicine University of Freiburg Freiburg Germany
| | - Tsvetan Serchov
- Department of Stereotactic and Functional Neurosurgery, Faculty of Medicine, Medical Center ‐ University Freiburg University of Freiburg Freiburg Germany
| |
Collapse
|
29
|
Lazarus M, Oishi Y, Bjorness TE, Greene RW. Gating and the Need for Sleep: Dissociable Effects of Adenosine A 1 and A 2A Receptors. Front Neurosci 2019; 13:740. [PMID: 31379490 PMCID: PMC6650574 DOI: 10.3389/fnins.2019.00740] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 07/02/2019] [Indexed: 12/20/2022] Open
Abstract
Roughly one-third of the human lifetime is spent in sleep, yet the reason for sleep remains unclear. Understanding the physiologic function of sleep is crucial toward establishing optimal health. Several proposed concepts address different aspects of sleep physiology, including humoral and circuit-based theories of sleep-wake regulation, the homeostatic two-process model of sleep regulation, the theory of sleep as a state of adaptive inactivity, and observations that arousal state and sleep homeostasis can be dissociated in pathologic disorders. Currently, there is no model that places the regulation of arousal and sleep homeostasis in a unified conceptual framework. Adenosine is well known as a somnogenic substance that affects normal sleep-wake patterns through several mechanisms in various brain locations via A1 or A2A receptors (A1Rs or A2ARs). Many cells and processes appear to play a role in modulating the extracellular concentration of adenosine at neuronal A1R or A2AR sites. Emerging evidence suggests that A1Rs and A2ARs have different roles in the regulation of sleep. In this review, we propose a model in which A2ARs allow the brain to sleep, i.e., these receptors provide sleep gating, whereas A1Rs modulate the function of sleep, i.e., these receptors are essential for the expression and resolution of sleep need. In this model, sleep is considered a brain state established in the absence of arousing inputs.
Collapse
Affiliation(s)
- Michael Lazarus
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Yo Oishi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Theresa E Bjorness
- Research and Development, VA North Texas Health Care System, Dallas, TX, United States.,Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Robert W Greene
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan.,Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
30
|
Mathews HL, Stitzel JA. The effects of oral nicotine administration and abstinence on sleep in male C57BL/6J mice. Psychopharmacology (Berl) 2019; 236:1335-1347. [PMID: 30564868 PMCID: PMC7372999 DOI: 10.1007/s00213-018-5139-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 11/29/2018] [Indexed: 11/24/2022]
Abstract
BACKGROUND Sleep disturbances are common in smoking cessation attempts and are predictive of relapse. Despite this knowledge, there is no established animal model to study the effect of nicotine abstinence on sleep and EEG parameters. OBJECTIVES The present study was conducted to characterize sleep and wakefulness in male C57BL/6J mice during periods of oral nicotine administration and abstinence. METHODS Male C57BL/6J mice were implanted with EEG/EMG recording devices. EEG/EMG data were recorded continuously for a period of 4 weeks. At the beginning of week 2, 200 μg/ml of nicotine was added to the 0.2% saccharin vehicle drinking solution. Following a 2-week period of oral nicotine administration, abstinence was initiated by excluding the nicotine from the 0.2% saccharin vehicle drinking solution. EEG/EMG were analyzed at pre-nicotine baseline, during nicotine administration, and on days 1, 2, and 5 of abstinence from nicotine. RESULTS Oral nicotine administration decreased total sleep time during the active phase, consistent with the stimulant actions of nicotine. In contrast, NREM sleep quantity was increased during the active phase on nicotine abstinence day 1 and REM sleep was decreased during days 2 and 5 of abstinence. Further, sleep fragmentation was increased during the inactive phase on all days of abstinence. Oral nicotine administration and abstinence from nicotine also altered EEG relative power frequencies during the inactive and active phase. CONCLUSIONS Both oral nicotine administration and abstinence lead to sleep disturbances in mice. Similarities between this model and human reports on the effect of nicotine/nicotine withdrawal on sleep support its utility in examining the molecular mechanisms that modulate the relationship between sleep, nicotine, and nicotine abstinence/withdrawal.
Collapse
Affiliation(s)
- Hunter L Mathews
- Department of Psychology and Neuroscience, The University of Colorado Boulder, Institute for Behavioral Genetics, 1480 30th Street, Boulder, CO, 80309, USA.
| | - Jerry A Stitzel
- Department of Integrative Physiology, The University of Colorado Boulder, Institute for Behavioral Genetics, 1480 30th Street, Boulder, CO, 80309, USA
| |
Collapse
|
31
|
Bjorness TE, Greene RW. Sleep deprivation alters the time course but not magnitude of locomotor sensitization to cocaine. Sci Rep 2018; 8:17672. [PMID: 30518935 PMCID: PMC6281608 DOI: 10.1038/s41598-018-36002-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 11/13/2018] [Indexed: 12/11/2022] Open
Abstract
Repeated exposure to drugs of abuse progressively increases the response to the same stimuli, a process known as sensitization. Behavioral sensitization to cocaine administration is often measured in non-human subjects via locomotor activity which is easily quantifiable. The effects of four hours of sleep deprivation on repeated cocaine (five daily and one challenge) showed attenuated hyperactivity on the first day only, compared to the non-deprived group. Both groups reached the same final level of sensitization, indicating that sleep deprivation altered the time course, but not magnitude of locomotor sensitization.
Collapse
Affiliation(s)
- Theresa E Bjorness
- Research Service, North Texas VA Health Care System, Dallas, TX, 75216, USA.
- Department of Psychiatry, University of Texas Southwestern, Dallas, TX, 75390, USA.
| | - Robert W Greene
- Department of Psychiatry, University of Texas Southwestern, Dallas, TX, 75390, USA
- Department of Neuroscience, University of Texas Southwestern, Dallas, TX, 75390, USA
- Research Service, North Texas VA Health Care System, Dallas, TX, 75216, USA
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, 305-8577, Japan
| |
Collapse
|
32
|
Cognitive impairments by alcohol and sleep deprivation indicate trait characteristics and a potential role for adenosine A 1 receptors. Proc Natl Acad Sci U S A 2018; 115:8009-8014. [PMID: 30012607 DOI: 10.1073/pnas.1803770115] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Trait-like differences in cognitive performance after sleep loss put some individuals more at risk than others, the basis of such disparities remaining largely unknown. Similarly, interindividual differences in impairment in response to alcohol intake have been observed. We tested whether performance impairments due to either acute or chronic sleep loss can be predicted by an individual's vulnerability to acute alcohol intake. Also, we used positron emission tomography (PET) to test whether acute alcohol infusion results in an up-regulation of cerebral A1 adenosine receptors (A1ARs), similar to the changes previously observed following sleep deprivation. Sustained attention in the psychomotor vigilance task (PVT) was tested in 49 healthy volunteers (26 ± 5 SD years; 15 females) (i) under baseline conditions: (ii) after ethanol intake, and after either (iii) total sleep deprivation (TSD; 35 hours awake; n = 35) or (iv) partial sleep deprivation (PSD; four nights with 5 hours scheduled sleep; n = 14). Ethanol- versus placebo-induced changes in cerebral A1AR availability were measured in 10 healthy male volunteers (31 ± 9 years) with [18F]8-cyclopentyl-3-(3-fluoropropyl)-1-propylxanthine (CPFPX) PET. Highly significant correlations between the performance impairments induced by ethanol and sleep deprivation were found for various PVT parameters, including mean speed (TSD, r = 0.62; PSD, r = 0.84). A1AR availability increased up to 26% in several brain regions with ethanol infusion. Our studies revealed individual trait characteristics for being either vulnerable or resilient to both alcohol and to sleep deprivation. Both interventions induce gradual increases in cerebral A1AR availability, pointing to a potential common molecular response mechanism.
Collapse
|
33
|
Onaolapo OJ, Onaolapo AY. Melatonin in drug addiction and addiction management: Exploring an evolving multidimensional relationship. World J Psychiatry 2018; 8:64-74. [PMID: 29988891 PMCID: PMC6033744 DOI: 10.5498/wjp.v8.i2.64] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/06/2018] [Accepted: 05/10/2018] [Indexed: 02/05/2023] Open
Abstract
Melatonin is a pleiotropic signalling molecule that regulates several physiological functions, and synchronises biological rhythms. Recent evidences are beginning to reveal that a dysregulation of endogenous melatonin rhythm or action may play a larger role in the aetiology and behavioural expression of drug addiction, than was previously considered. Also, the findings from a number of animal studies suggest that exogenous melatonin supplementation and therapeutic manipulation of melatonin/melatonin receptor interactions may be beneficial in the management of behavioural manifestations of drug addiction. However, repeated exogenous melatonin administration may cause a disruption of its endogenous rhythm and be associated with potential drawbacks that might limit its usefulness. In this review, we examine the roles of melatonin and its receptors in addictive behaviours; discussing how our understanding of melatonin’s modulatory effects on the brain rewards system and crucial neurotransmitters such as dopamine has evolved over the years. Possible indications(s) for melatonergic agents in addiction management, and how manipulations of the endogenous melatonin system may be of benefit are also discussed. Finally, the potential impediments to application of melatonin in the management of addictive behaviours are considered.
Collapse
Affiliation(s)
- Olakunle J Onaolapo
- Behavioural Neuroscience/Neuropharmacology Unit, Department of Pharmacology and Therapeutics, Ladoke Akintola University of Technology, Osogbo 230263, Osun State, Nigeria
| | - Adejoke Y Onaolapo
- Behavioural Neuroscience/Neurobiology Unit, Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho 210211, Oyo State, Nigeria
| |
Collapse
|
34
|
Skorucak J, Arbon EL, Dijk DJ, Achermann P. Response to chronic sleep restriction, extension, and subsequent total sleep deprivation in humans: adaptation or preserved sleep homeostasis? Sleep 2018; 41:4990768. [DOI: 10.1093/sleep/zsy078] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 04/30/2018] [Indexed: 12/22/2022] Open
Affiliation(s)
- Jelena Skorucak
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
- Zurich Center for Interdisciplinary Sleep Research, University of Zurich, Zurich, Switzerland
| | - Emma L Arbon
- Surrey Sleep Research Centre, University of Surrey, Guildford, United Kingdom
| | - Derk-Jan Dijk
- Surrey Sleep Research Centre, University of Surrey, Guildford, United Kingdom
| | - Peter Achermann
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
- Zurich Center for Interdisciplinary Sleep Research, University of Zurich, Zurich, Switzerland
| |
Collapse
|
35
|
Schwartz MD, Palmerston JB, Lee DL, Hoener MC, Kilduff TS. Deletion of Trace Amine-Associated Receptor 1 Attenuates Behavioral Responses to Caffeine. Front Pharmacol 2018; 9:35. [PMID: 29456505 PMCID: PMC5801540 DOI: 10.3389/fphar.2018.00035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/12/2018] [Indexed: 12/18/2022] Open
Abstract
Trace amines (TAs), endogenous amino acid metabolites that are structurally similar to the biogenic amines, are endogenous ligands for trace amine-associated receptor 1 (TAAR1), a GPCR that modulates dopaminergic, serotonergic, and glutamatergic activity. Selective TAAR1 full and partial agonists exhibit similar pro-cognitive, antidepressant- and antipsychotic-like properties in rodents and non-human primates, suggesting TAAR1 as a novel target for the treatment of neurological and psychiatric disorders. We previously reported that TAAR1 partial agonists are wake-promoting in rats and mice, and that TAAR1 knockout (KO) and overexpressing mice exhibit altered sleep-wake and EEG spectral composition. Here, we report that locomotor and EEG spectral responses to the psychostimulants modafinil and caffeine are attenuated in TAAR1 KO mice. TAAR1 KO mice and WT littermates were instrumented for EEG and EMG recording and implanted with telemetry transmitters for monitoring locomotor activity (LMA) and core body temperature (Tb). Following recovery, mice were administered modafinil (25, 50, 100 mg/kg), caffeine (2.5, 10, 20 mg/kg) or vehicle p.o. at ZT6 in balanced order. In WT mice, both modafinil and caffeine dose-dependently increased LMA for up to 6 h following dosing, whereas only the highest dose of each drug increased LMA in KO mice, and did so for less time after dosing. This effect was particularly pronounced following caffeine, such that total LMA response was significantly attenuated in KO mice compared to WT at all doses of caffeine and did not differ from Vehicle treatment. Tb increased comparably in both genotypes in a dose-dependent manner. TAAR1 deletion was associated with reduced wake consolidation following both drugs, but total time in wakefulness did not differ between KO and WT mice. Furthermore, gamma band EEG activity following both modafinil and caffeine treatment was attenuated in TAAR1 KO compared to WT mice. Our results show that TAAR1 is a critical component of the behavioral and cortical arousal associated with two widely used psychostimulants with very different mechanisms of action. Together with our previous findings, these data suggest that TAAR1 is a previously unrecognized component of an endogenous wake-modulating system.
Collapse
Affiliation(s)
- Michael D Schwartz
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA, United States
| | - Jeremiah B Palmerston
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA, United States
| | - Diana L Lee
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA, United States
| | - Marius C Hoener
- Neuroscience, Ophthalmology and Rare Diseases Discovery and Translational Area, Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Ltd., Basel, Switzerland
| | - Thomas S Kilduff
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA, United States
| |
Collapse
|
36
|
Modeling the adenosine system as a modulator of cognitive performance and sleep patterns during sleep restriction and recovery. PLoS Comput Biol 2017; 13:e1005759. [PMID: 29073206 PMCID: PMC5675465 DOI: 10.1371/journal.pcbi.1005759] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 11/07/2017] [Accepted: 09/01/2017] [Indexed: 11/18/2022] Open
Abstract
Sleep loss causes profound cognitive impairments and increases the concentrations of adenosine and adenosine A1 receptors in specific regions of the brain. Time courses for performance impairment and recovery differ between acute and chronic sleep loss, but the physiological basis for these time courses is unknown. Adenosine has been implicated in pathways that generate sleepiness and cognitive impairments, but existing mathematical models of sleep and cognitive performance do not explicitly include adenosine. Here, we developed a novel receptor-ligand model of the adenosine system to test the hypothesis that changes in both adenosine and A1 receptor concentrations can capture changes in cognitive performance during acute sleep deprivation (one prolonged wake episode), chronic sleep restriction (multiple nights with insufficient sleep), and subsequent recovery. Parameter values were estimated using biochemical data and reaction time performance on the psychomotor vigilance test (PVT). The model closely fit group-average PVT data during acute sleep deprivation, chronic sleep restriction, and recovery. We tested the model's ability to reproduce timing and duration of sleep in a separate experiment where individuals were permitted to sleep for up to 14 hours per day for 28 days. The model accurately reproduced these data, and also correctly predicted the possible emergence of a split sleep pattern (two distinct sleep episodes) under these experimental conditions. Our findings provide a physiologically plausible explanation for observed changes in cognitive performance and sleep during sleep loss and recovery, as well as a new approach for predicting sleep and cognitive performance under planned schedules.
Collapse
|
37
|
Doty TJ, So CJ, Bergman EM, Trach SK, Ratcliffe RH, Yarnell AM, Capaldi VF, Moon JE, Balkin TJ, Quartana PJ. Limited Efficacy of Caffeine and Recovery Costs During and Following 5 Days of Chronic Sleep Restriction. Sleep 2017; 40:4344846. [DOI: 10.1093/sleep/zsx171] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
38
|
Adenosine A 2A receptor mediates hypnotic effects of ethanol in mice. Sci Rep 2017; 7:12678. [PMID: 28978989 PMCID: PMC5627250 DOI: 10.1038/s41598-017-12689-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 09/13/2017] [Indexed: 12/25/2022] Open
Abstract
Ethanol has extensive effects on sleep and daytime alertness, causing premature disability and death. Adenosine, as a potent sleep-promoting substance, is involved in many cellular and behavioral responses to ethanol. However, the mechanisms of hypnotic effects of ethanol remain unclear. In this study, we investigated the role of adenosine in ethanol-induced sleep using C57BL/6Slac mice, adenosine A2A receptor (A2AR) knockout mice, and their wild-type littermates. The results showed that intraperitoneal injection of ethanol (3.0 g/kg) at 21:00 decreased the latency to non-rapid eye movement (NREM) sleep and increased the duration of NREM sleep for 5 h. Ethanol dose-dependently increased NREM sleep, which was consistent with decreases in wakefulness in C57BL/6Slac mice compared with their own control. Caffeine (5, 10, or 15 mg/kg), a nonspecific adenosine receptor antagonist, dose-dependently and at high doses completely blocked ethanol-induced NREM sleep when administered 30 min prior to (but not after) ethanol injection. Moreover, ethanol-induced NREM sleep was completely abolished in A2AR knockout mice compared with wild-type mice. These findings strongly indicate that A2AR is a key receptor for the hypnotic effects of ethanol, and pretreatment of caffeine might be a strategy to counter the hypnotic effects of ethanol.
Collapse
|
39
|
Carrier J, Semba K, Deurveilher S, Drogos L, Cyr-Cronier J, Lord C, Sekerovick Z. Sex differences in age-related changes in the sleep-wake cycle. Front Neuroendocrinol 2017; 47:66-85. [PMID: 28757114 DOI: 10.1016/j.yfrne.2017.07.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 06/09/2017] [Accepted: 07/19/2017] [Indexed: 02/06/2023]
Abstract
Age-related changes in sleep and circadian regulation occur as early as the middle years of life. Research also suggests that sleep and circadian rhythms are regulated differently between women and men. However, does sleep and circadian rhythms regulation age similarly in men and women? In this review, we present the mechanisms underlying age-related differences in sleep and the current state of knowledge on how they interact with sex. We also address how testosterone, estrogens, and progesterone fluctuations across adulthood interact with sleep and circadian regulation. Finally, we will propose research avenues to unravel the mechanisms underlying sex differences in age-related effects on sleep.
Collapse
Affiliation(s)
- Julie Carrier
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Montréal, Québec, Canada; Institut Universitaire de Gériatrie de Montréal, Université de Montréal, Montréal, Québec, Canada; Département de psychologie, Université de Montréal, Montréal, Québec, Canada.
| | - Kazue Semba
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Psychology & Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Samuel Deurveilher
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Lauren Drogos
- Departments of Physiology & Pharmacology and Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Jessica Cyr-Cronier
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Montréal, Québec, Canada
| | - Catherine Lord
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Montréal, Québec, Canada
| | - Zoran Sekerovick
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Montréal, Québec, Canada
| |
Collapse
|
40
|
Connexin 43-Mediated Astroglial Metabolic Networks Contribute to the Regulation of the Sleep-Wake Cycle. Neuron 2017; 95:1365-1380.e5. [PMID: 28867552 DOI: 10.1016/j.neuron.2017.08.022] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 06/29/2017] [Accepted: 08/14/2017] [Indexed: 01/13/2023]
Abstract
Astrocytes produce and supply metabolic substrates to neurons through gap junction-mediated astroglial networks. However, the role of astroglial metabolic networks in behavior is unclear. Here, we demonstrate that perturbation of astroglial networks impairs the sleep-wake cycle. Using a conditional Cre-Lox system in mice, we show that knockout of the gap junction subunit connexin 43 in astrocytes throughout the brain causes excessive sleepiness and fragmented wakefulness during the nocturnal active phase. This astrocyte-specific genetic manipulation silenced the wake-promoting orexin neurons located in the lateral hypothalamic area (LHA) by impairing glucose and lactate trafficking through astrocytic networks. This global wakefulness instability was mimicked with viral delivery of Cre recombinase to astrocytes in the LHA and rescued by in vivo injections of lactate. Our findings propose a novel regulatory mechanism critical for maintaining normal daily cycle of wakefulness and involving astrocyte-neuron metabolic interactions.
Collapse
|
41
|
Abstract
Sleep homeostasis is a fundamental property of vigilance state regulation that is highly conserved across species. Neuronal systems and circuits that underlie sleep homeostasis are not well understood. In Drosophila, a neuronal circuit involving neurons in the ellipsoid body and in the dorsal Fan-shaped body is a candidate for both tracing sleep need during waking and translating it to increased sleep drive and expression. Sleep homeostasis in rats and mice involves multiple neuromodulators acting on multiple wake- and sleep-promoting neuronal systems. A functional central homeostat emerges from A1 receptor mediated actions of adenosine on wake-promoting neurons in the basal forebrain and hypothalamus, and A2A adenosine receptor-mediated actions on sleep-promoting neurons in the preoptic hypothalamus and nucleus accumbens.
Collapse
|
42
|
Decreased alertness due to sleep loss increases pain sensitivity in mice. Nat Med 2017; 23:768-774. [PMID: 28481358 DOI: 10.1038/nm.4329] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 03/17/2017] [Indexed: 12/11/2022]
Abstract
Extended daytime and nighttime activities are major contributors to the growing sleep deficiency epidemic, as is the high prevalence of sleep disorders like insomnia. The consequences of chronic insufficient sleep for health remain uncertain. Sleep quality and duration predict presence of pain the next day in healthy subjects, suggesting that sleep disturbances alone may worsen pain, and experimental sleep deprivation in humans supports this claim. We demonstrate that sleep loss, but not sleep fragmentation, in healthy mice increases sensitivity to noxious stimuli (referred to as 'pain') without general sensory hyper-responsiveness. Moderate daily repeated sleep loss leads to a progressive accumulation of sleep debt and also to exaggerated pain responses, both of which are rescued after restoration of normal sleep. Caffeine and modafinil, two wake-promoting agents that have no analgesic activity in rested mice, immediately normalize pain sensitivity in sleep-deprived animals, without affecting sleep debt. The reversibility of mild sleep-loss-induced pain by wake-promoting agents reveals an unsuspected role for alertness in setting pain sensitivity. Clinically, insufficient or poor-quality sleep may worsen pain and this enhanced pain may be reduced not by analgesics, whose effectiveness is reduced, but by increasing alertness or providing better sleep.
Collapse
|
43
|
Luong L, Bannon NM, Redenti A, Chistiakova M, Volgushev M. Very low concentrations of ethanol suppress excitatory synaptic transmission in rat visual cortex. Eur J Neurosci 2017; 45:1333-1342. [PMID: 28263415 DOI: 10.1111/ejn.13557] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 02/25/2017] [Accepted: 02/27/2017] [Indexed: 01/12/2023]
Abstract
Ethanol is one of the most commonly used substances in the world. Behavioral effects of alcohol are well described, however, cellular mechanisms of its action are poorly understood. There is an apparent contradiction between measurable behavioral changes produced by low concentrations of ethanol, and lack of evidence of synaptic changes at these concentrations. Furthermore, effects of ethanol on synaptic transmission in the neocortex are poorly understood. Here, we set to determine effects of ethanol on excitatory synaptic transmission in the neocortex. We show that 1-50 mm ethanol suppresses excitatory synaptic transmission to layer 2/3 pyramidal neurons in rat visual cortex in a concentration-dependent manner. To the best of our knowledge, this is the first demonstration of the effects of very low concentrations of ethanol (from 1 mm) on synaptic transmission in the neocortex. We further show that a selective antagonist of A1 adenosine receptors, 8-cyclopentyl-1,3-dipropylxanthine (DPCPX), blocks effects of 1-10 mm ethanol on synaptic transmission. However, the reduction in excitatory postsynaptic potential amplitude by 50 mm ethanol was not affected by DPCPX. We propose that ethanol depresses excitatory synaptic transmission in the neocortex by at least two mechanisms, engaged at different concentrations: low concentrations of ethanol reduce synaptic transmission via A1 R-dependent mechanism and involve presynaptic changes, while higher concentrations activate additional, adenosine-independent mechanisms with predominantly postsynaptic action. Involvement of adenosine signaling in mediating effects of low concentrations of ethanol may have important implications for understanding alcohol's effects on brain function, and provide a mechanistic explanation to the interaction between alcohol and caffeine.
Collapse
Affiliation(s)
- Lucas Luong
- Department of Psychological Sciences, University of Connecticut, 406 Babbidge Road Unit 1020, Storrs, CT, 06268, USA
| | - Nicholas M Bannon
- Department of Psychological Sciences, University of Connecticut, 406 Babbidge Road Unit 1020, Storrs, CT, 06268, USA
| | - Andrew Redenti
- Department of Psychological Sciences, University of Connecticut, 406 Babbidge Road Unit 1020, Storrs, CT, 06268, USA
| | - Marina Chistiakova
- Department of Psychological Sciences, University of Connecticut, 406 Babbidge Road Unit 1020, Storrs, CT, 06268, USA
| | - Maxim Volgushev
- Department of Psychological Sciences, University of Connecticut, 406 Babbidge Road Unit 1020, Storrs, CT, 06268, USA
| |
Collapse
|
44
|
Foley J, Blutstein T, Lee S, Erneux C, Halassa MM, Haydon P. Astrocytic IP 3/Ca 2+ Signaling Modulates Theta Rhythm and REM Sleep. Front Neural Circuits 2017; 11:3. [PMID: 28167901 PMCID: PMC5253379 DOI: 10.3389/fncir.2017.00003] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 01/09/2017] [Indexed: 12/27/2022] Open
Abstract
Rapid eye movement (REM) sleep onset is triggered by disinhibition of cholinergic neurons in the pons. During REM sleep, the brain exhibits prominent activity in the 5–8 Hz (theta) frequency range. How REM sleep onset and theta waves are regulated is poorly understood. Astrocytes, a non-neuronal cell type in the brain, respond to cholinergic signals by elevating their intracellular Ca2+ concentration. The goal of this study was to assess the sleep architecture of mice with attenuated IP3 mediated Ca2+ signaling in astrocytes. Vigilance states and cortical electroencephalograph power were measured in wild type mice and mice with attenuated IP3/Ca2+ signaling. Attenuating IP3/Ca2+ signaling specifically in astrocytes caused mice to spend more time in REM sleep and enter this state more frequently during their inactive phase. These mice also exhibited greater power in the theta frequency range. These data suggest a role for astrocytic IP3/Ca2+ signaling in modulating REM sleep and the associated physiological state of the cortex.
Collapse
Affiliation(s)
- Jeannine Foley
- Department of Neuroscience, Tufts University, Boston MA, USA
| | | | - SoYoung Lee
- Department of Neuroscience, Tufts University, Boston MA, USA
| | - Christophe Erneux
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Université Libre de Bruxelles Brussels, Belgium
| | - Michael M Halassa
- Departments of Psychiatry, Neuroscience and Physiology, Neuroscience Institute, New York University, New York NY, USA
| | - Philip Haydon
- Department of Neuroscience, Tufts University, Boston MA, USA
| |
Collapse
|
45
|
Hall S, Deurveilher S, Ko KR, Burns J, Semba K. Region-specific increases in FosB/ΔFosB immunoreactivity in the rat brain in response to chronic sleep restriction. Behav Brain Res 2017; 322:9-17. [PMID: 28089853 DOI: 10.1016/j.bbr.2017.01.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 01/05/2017] [Accepted: 01/11/2017] [Indexed: 01/23/2023]
Abstract
Using a rat model of chronic sleep restriction (CSR) featuring periodic sleep deprivation with slowly rotating wheels (3h on/1h off), we previously observed that 99h of this protocol induced both homeostatic and allostatic (adaptive) changes in physiological and behavioural measures. Notably, the initial changes in sleep intensity and attention performance gradually adapted during CSR despite accumulating sleep loss. To identify brain regions involved in these responses, we used FosB/ΔFosB immunohistochemistry as a marker of chronic neuronal activation. Adult male rats were housed in motorized activity wheels and underwent the 3/1 CSR protocol for 99h, or 99h followed by 6 or 12days of recovery. Control rats were housed in home cages, locked activity wheels, or unlocked activity wheels that the animals could turn freely. Immunohistochemistry was conducted using an antibody that recognized both FosB and ΔFosB, and 24 brain regions involved in sleep/wake, autonomic, and limbic functions were examined. The number of darkly-stained FosB/ΔFosB-immunoreactive cells was increased immediately following 99h of CSR in 8/24 brain regions, including the medial preoptic and perifornical lateral hypothalamic areas, dorsomedial and paraventricular hypothalamic nuclei, and paraventricular thalamic nucleus. FosB/ΔFosB labeling was at control levels in all 8 brain areas following 6 or 12 recovery days, suggesting that most of the immunoreactivity immediately after CSR reflected FosB, the more transient marker of chronic neuronal activation. This region-specific induction of FosB/ΔFosB following CSR may be involved in the mechanisms underlying the allostatic changes in behavioural and physiological responses to CSR.
Collapse
Affiliation(s)
- Shannon Hall
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Samüel Deurveilher
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kristin Robin Ko
- School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Joan Burns
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kazue Semba
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Psychology & Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
46
|
Skatchkov SN, Antonov SM, Eaton MJ. Glia and glial polyamines. Role in brain function in health and disease. BIOCHEMISTRY MOSCOW SUPPLEMENT SERIES A-MEMBRANE AND CELL BIOLOGY 2016. [DOI: 10.1134/s1990747816010116] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
47
|
Dubowy C, Moravcevic K, Yue Z, Wan JY, Van Dongen HP, Sehgal A. Genetic Dissociation of Daily Sleep and Sleep Following Thermogenetic Sleep Deprivation in Drosophila. Sleep 2016; 39:1083-95. [PMID: 26951392 PMCID: PMC4835307 DOI: 10.5665/sleep.5760] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 01/11/2016] [Indexed: 12/23/2022] Open
Abstract
STUDY OBJECTIVES Sleep rebound-the increase in sleep that follows sleep deprivation-is a hallmark of homeostatic sleep regulation that is conserved across the animal kingdom. However, both the mechanisms that underlie sleep rebound and its relationship to habitual daily sleep remain unclear. To address this, we developed an efficient thermogenetic method of inducing sleep deprivation in Drosophila that produces a substantial rebound, and applied the newly developed method to assess sleep rebound in a screen of 1,741 mutated lines. We used data generated by this screen to identify lines with reduced sleep rebound following thermogenetic sleep deprivation, and to probe the relationship between habitual sleep amount and sleep following thermogenetic sleep deprivation in Drosophila. METHODS To develop a thermogenetic method of sleep deprivation suitable for screening, we thermogenetically stimulated different populations of wake-promoting neurons labeled by Gal4 drivers. Sleep rebound following thermogenetically-induced wakefulness varies across the different sets of wake-promoting neurons that were stimulated, from very little to quite substantial. Thermogenetic activation of neurons marked by the c584-Gal4 driver produces both strong sleep loss and a substantial rebound that is more consistent within genotypes than rebound following mechanical or caffeine-induced sleep deprivation. We therefore used this driver to induce sleep deprivation in a screen of 1,741 mutagenized lines generated by the Drosophila Gene Disruption Project. Flies were subjected to 9 h of sleep deprivation during the dark period and released from sleep deprivation 3 h before lights-on. Recovery was measured over the 15 h following sleep deprivation. Following identification of lines with reduced sleep rebound, we characterized baseline sleep and sleep depth before and after sleep deprivation for these hits. RESULTS We identified two lines that consistently exhibit a blunted increase in the duration and depth of sleep after thermogenetic sleep deprivation. Neither of the two genotypes has reduced total baseline sleep. Statistical analysis across all screened lines shows that genotype is a strong predictor of recovery sleep, independent from effects of genotype on baseline sleep. CONCLUSIONS Our data show that rebound sleep following thermogenetic sleep deprivation can be genetically separated from sleep at baseline. This suggests that genetically controlled mechanisms of sleep regulation not manifest under undisturbed conditions contribute to sleep rebound following thermogenetic sleep deprivation.
Collapse
Affiliation(s)
- Christine Dubowy
- Cell and Molecular Biology Graduate Group, Biomedical Graduate Studies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Katarina Moravcevic
- Department of Neuroscience, HHMI, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Zhifeng Yue
- Department of Neuroscience, HHMI, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Joy Y. Wan
- Department of Neuroscience, HHMI, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Hans P.A. Van Dongen
- Sleep and Performance Research Center and Elson S. Floyd College of Medicine, Washington State University, Spokane, WA
| | - Amita Sehgal
- Department of Neuroscience, HHMI, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
48
|
Transportation in the Interstitial Space of the Brain Can Be Regulated by Neuronal Excitation. Sci Rep 2015; 5:17673. [PMID: 26631412 PMCID: PMC4668547 DOI: 10.1038/srep17673] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 10/30/2015] [Indexed: 01/03/2023] Open
Abstract
The transportation of substances in the interstitial space (ISS) is crucial for the maintenance of brain homeostasis, however its link to neuronal activity remains unclear. Here, we report a marked reduction in substance transportation in the ISS after neuronal excitation. Using a tracer-based method, water molecules in the interstitial fluid (ISF) could be specifically visualized in magnetic resonance (MR) imaging. We first observed the flow of ISF in the thalamus and caudate nucleus of a rat. The ISF flow was then modulated using a painful stimulation model. We demonstrated that the flow of ISF slowed significantly following neuronal activity in the thalamus. This reduction in ISF flow continued for hours and was not accompanied by slow diffusion into the ISS. This observation suggests that the transportation of substances into the ISS can be regulated with a selective external stimulation.
Collapse
|
49
|
Kim Y, Elmenhorst D, Weisshaupt A, Wedekind F, Kroll T, McCarley RW, Strecker RE, Bauer A. Chronic sleep restriction induces long-lasting changes in adenosine and noradrenaline receptor density in the rat brain. J Sleep Res 2015; 24:549-558. [PMID: 25900125 PMCID: PMC4583343 DOI: 10.1111/jsr.12300] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 03/15/2015] [Indexed: 01/10/2023]
Abstract
Although chronic sleep restriction frequently produces long-lasting behavioural and physiological impairments in humans, the underlying neural mechanisms are unknown. Here we used a rat model of chronic sleep restriction to investigate the role of brain adenosine and noradrenaline systems, known to regulate sleep and wakefulness, respectively. The density of adenosine A1 and A2a receptors and β-adrenergic receptors before, during and following 5 days of sleep restriction was assessed with autoradiography. Rats (n = 48) were sleep-deprived for 18 h day(-1) for 5 consecutive days (SR1-SR5), followed by 3 unrestricted recovery sleep days (R1-R3). Brains were collected at the beginning of the light period, which was immediately after the end of sleep deprivation on sleep restriction days. Chronic sleep restriction increased adenosine A1 receptor density significantly in nine of the 13 brain areas analysed with elevations also observed on R3 (+18 to +32%). In contrast, chronic sleep restriction reduced adenosine A2a receptor density significantly in one of the three brain areas analysed (olfactory tubercle which declined 26-31% from SR1 to R1). A decrease in β-adrenergic receptors density was seen in substantia innominata and ventral pallidum which remained reduced on R3, but no changes were found in the anterior cingulate cortex. These data suggest that chronic sleep restriction can induce long-term changes in the brain adenosine and noradrenaline receptors, which may underlie the long-lasting neurocognitive impairments observed in chronic sleep restriction.
Collapse
Affiliation(s)
- Youngsoo Kim
- Department of Psychiatry, VA Boston Healthcare System, Research Service and Harvard Medical School, Brockton, MA, USA
| | - David Elmenhorst
- Department of Psychiatry, VA Boston Healthcare System, Research Service and Harvard Medical School, Brockton, MA, USA
- Institute for Neuroscience and Medicine (INM-2), Forschungszentrum Jülich, Jülich, Germany
| | - Angela Weisshaupt
- Institute for Neuroscience and Medicine (INM-2), Forschungszentrum Jülich, Jülich, Germany
| | - Franziska Wedekind
- Institute for Neuroscience and Medicine (INM-2), Forschungszentrum Jülich, Jülich, Germany
| | - Tina Kroll
- Institute for Neuroscience and Medicine (INM-2), Forschungszentrum Jülich, Jülich, Germany
| | - Robert W McCarley
- Department of Psychiatry, VA Boston Healthcare System, Research Service and Harvard Medical School, Brockton, MA, USA
| | - Robert E Strecker
- Department of Psychiatry, VA Boston Healthcare System, Research Service and Harvard Medical School, Brockton, MA, USA
| | - Andreas Bauer
- Institute for Neuroscience and Medicine (INM-2), Forschungszentrum Jülich, Jülich, Germany
- Neurological Department, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
50
|
Willoughby AR, de Zambotti M, Baker FC, Colrain IM. Partial K-Complex Recovery Following Short-Term Abstinence in Individuals with Alcohol Use Disorder. Alcohol Clin Exp Res 2015; 39:1417-24. [PMID: 26175209 DOI: 10.1111/acer.12769] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 05/05/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND The K-complex (KC) is a brain potential characteristic of nonrapid eye movement (NREM) sleep resulting from the synchronous activity of a large population of neurons and hypothesized to reflect brain integrity. KC amplitude is lower in individuals with alcohol use disorder (AUD) compared with age-matched controls, but its recovery with short-term abstinence has not been studied. Therefore, we investigated whether the KC shows significant recovery over the first 4 months of abstinence in individuals with AUD. METHODS A total of 16 recently abstinent AUD individuals (46.6 ± 9.3 years) and 13 gender and age-matched healthy controls (41.6 ± 8.3 years) were studied on 3 occasions: the Initial session was within 1 month of the AUD individuals' last drink, then 1 and 3 months later. Overnight electroencephalogram was recorded while participants were presented with tones during stage 2 NREM sleep to elicit KCs. RESULTS At the Initial session, AUD participants showed significantly lower KC amplitude and incidence compared with controls. In the AUD individuals, KC amplitude increased significantly from the Initial to the 1-month session. KC incidence showed a marginally significant increase. Neither KC amplitude nor incidence changed from the 1-month to the 3-month session. No changes in KC amplitude or incidence across sessions were observed in the control group. CONCLUSIONS Our results demonstrate partial KC recovery during the first 2 months of abstinence. This recovery is consistent with the time course of structural brain recovery in abstinent AUD individuals demonstrated by recent neuroimaging results.
Collapse
Affiliation(s)
| | | | - Fiona C Baker
- Center for Health Sciences , SRI International, Menlo Park, California.,Brain Function Research Group, University of the Witwatersrand, Johannesburg, South Africa
| | - Ian M Colrain
- Melbourne School of Psychological Sciences University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|