1
|
Vastegani SM, Margha ZB, Farbood Y, Sarkaki A, Khoshnam SE. Caffeine and Exercise: A Dual Approach to Combat Cognitive Decline Induced by REM Sleep Deprivation. Mol Neurobiol 2025:10.1007/s12035-025-04845-1. [PMID: 40131698 DOI: 10.1007/s12035-025-04845-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/12/2025] [Indexed: 03/27/2025]
Abstract
Sleep deprivation (SD) is known to impair memory and cognitive functions often linked to heightened oxidative stress and neuroinflammation. While caffeine and exercise individually offer neuroprotective effects, the combined influence of these interventions on preserving cognitive function and reducing hippocampal damage during REM-SD has yet to be fully explored. This study investigates the synergistic potential of caffeine and exercise in mitigating these neurological damages induced by REM-SD. Male Wistar rats were divided into five groups: control, SD, caffeine + SD, exercise + SD, and caffeine + exercise + SD. The rats received caffeine (30 mg/kg, p.o) and underwent a treadmill exercise regimen, running 5 days a week for 4 weeks. Following this, they were placed in a REM-SD apparatus for 72 h. We evaluated the cognitive functions of the animals using standard behavioral tests. In addition, we assessed BBB permeability, brain water content (BWC), oxidative-antioxidative status, inflammatory/anti-inflammatory conditions, and apoptotic and neurotrophic indices in the hippocampus of REM-SD rats. Our findings indicate that the combination of caffeine and exercise significantly enhanced cognitive functions in REM-SD rats. This combination also reduced BBB permeability and BWC, alleviated oxidative stress, lowered inflammatory and apoptotic indices, and increased neurotrophic gene expression in the hippocampus of REM-SD rats. This study highlights the protective effects of caffeine and exercise on cognitive function and BBB integrity, likely through the enhancement of antioxidant and anti-inflammatory indices in the hippocampus, as well as their anti-apoptotic and neurotrophic effects in REM-SD rats.
Collapse
Affiliation(s)
- Sadegh Moradi Vastegani
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zeynab Behdarvand Margha
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Yaghoob Farbood
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Alireza Sarkaki
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
2
|
Wankhede NL, Kale MB, Kyada A, M RM, Chaudhary K, Naidu KS, Rahangdale S, Shende PV, Taksande BG, Khalid M, Gulati M, Umekar MJ, Fareed M, Kopalli SR, Koppula S. Sleep deprivation-induced shifts in gut microbiota: Implications for neurological disorders. Neuroscience 2025; 565:99-116. [PMID: 39622383 DOI: 10.1016/j.neuroscience.2024.11.070] [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: 07/31/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Sleep deprivation is a prevalent issue in contemporary society, with significant ramifications for both physical and mental well-being. Emerging scientific evidence illuminates its intricate interplay with the gut-brain axis, a vital determinant of neurological function. Disruptions in sleep patterns disturb the delicate equilibrium of the gut microbiota, resulting in dysbiosis characterized by alterations in microbial composition and function. This dysbiosis contributes to the exacerbation of neurological disorders such as depression, anxiety, and cognitive decline through multifaceted mechanisms, including heightened neuroinflammation, disturbances in neurotransmitter signalling, and compromised integrity of the gut barrier. In response to these challenges, there is a burgeoning interest in therapeutic interventions aimed at restoring gut microbial balance and alleviating neurological symptoms precipitated by sleep deprivation. Probiotics, dietary modifications, and behavioural strategies represent promising avenues for modulating the gut microbiota and mitigating the adverse effects of sleep disturbances on neurological health. Moreover, the advent of personalized interventions guided by advanced omics technologies holds considerable potential for tailoring treatments to individualized needs and optimizing therapeutic outcomes. Interdisciplinary collaboration and concerted research efforts are imperative for elucidating the underlying mechanisms linking sleep, gut microbiota, and neurological function. Longitudinal studies, translational research endeavours, and advancements in technology are pivotal for unravelling the complex interplay between these intricate systems.
Collapse
Affiliation(s)
- Nitu L Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Mayur B Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Ashishkumar Kyada
- Marwadi University Research Center, Department of Pharmacy, Faculty of Health Sciences Marwadi University, Rajkot 360003, Gujarat, India
| | - Rekha M M
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Kamlesh Chaudhary
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, India
| | - Sandip Rahangdale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Prajwali V Shende
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Brijesh G Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Mohammad Khalid
- Department of Pharmacognosy, College of Pharmacy Prince Sattam Bin Abdulaziz University Alkharj, Saudi Arabia
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 1444411, India; ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 20227, Australia
| | - Milind J Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| |
Collapse
|
3
|
Chai R, Bian WJ. Adolescent sleep and its disruption in depression and anxiety. Front Neurosci 2024; 18:1479420. [PMID: 39575099 PMCID: PMC11578994 DOI: 10.3389/fnins.2024.1479420] [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: 08/12/2024] [Accepted: 10/03/2024] [Indexed: 11/24/2024] Open
Abstract
Adolescence is a pivotal stage during development when one's personality, emotion, and behavioral traits are shaped to a great extent, and the underlying neural circuits undergo substantial developmental organizations. Dramatic and dynamic changes occur in sleep architecture throughout the postnatal developmental course. Insufficient sleep and disruption of sleep/wake coherence are prevalent among the adolescents worldwide, and even so in young patients with neuropsychiatric conditions. Although accumulating evidence has suggested a tight association between sleep disruption and depression/anxiety, the causal relationship remains largely unclear. More importantly, most of these studies focused on adult subjects, and little is known about the role of sleep during the development of mood and behavior. Here we review recent studies investigating the acute and chronic effects of adolescent sleep disruption on depression and anxiety both in humans and rodent models with focuses on the assessment methodology and age. By discussing the findings and unsolved problems, we hope to achieve a better understanding of the relationship between sleep and mental health in adolescents and provide insights for future research.
Collapse
Affiliation(s)
- Ruiming Chai
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| | - Wen-Jie Bian
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| |
Collapse
|
4
|
Kiss MG, Cohen O, McAlpine CS, Swirski FK. Influence of sleep on physiological systems in atherosclerosis. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1284-1300. [PMID: 39528718 PMCID: PMC11567060 DOI: 10.1038/s44161-024-00560-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024]
Abstract
Sleep is a fundamental requirement of life and is integral to health. Deviation from optimal sleep associates with numerous diseases including those of the cardiovascular system. Studies, spanning animal models to humans, show that insufficient, disrupted or inconsistent sleep contribute to poor cardiovascular health by disrupting body systems. Fundamental experiments have begun to uncover the molecular and cellular links between sleep and heart health while large-scale human studies have associated sleep with cardiovascular outcomes in diverse populations. Here, we review preclinical and clinical findings that demonstrate how sleep influences the autonomic nervous, metabolic and immune systems to affect atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Máté G Kiss
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Oren Cohen
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cameron S McAlpine
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Filip K Swirski
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
5
|
Makino Y, Hodgson NW, Doenier E, Serbin AV, Osada K, Artoni P, Dickey M, Sullivan B, Potter-Dickey A, Komanchuk J, Sekhon B, Letourneau N, Ryan ND, Trauth J, Cameron JL, Hensch TK. Sleep-sensitive dopamine receptor expression in male mice underlies attention deficits after a critical period of early adversity. Sci Transl Med 2024; 16:eadh9763. [PMID: 39383245 DOI: 10.1126/scitranslmed.adh9763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/03/2024] [Accepted: 09/13/2024] [Indexed: 10/11/2024]
Abstract
Early life stress (ELS) yields cognitive impairments of unknown molecular and physiological origin. We found that fragmented maternal care of mice during a neonatal critical period from postnatal days P2-9 elevated dopamine receptor D2R and suppressed D4R expression, specifically within the anterior cingulate cortex (ACC) in only the male offspring. This was associated with poor performance on a two-choice visual attention task, which was acutely rescued in adulthood by local or systemic pharmacological rebalancing of D2R/D4R activity. Furthermore, ELS male mice demonstrated heightened hypothalamic orexin and persistently disrupted sleep. Given that acute sleep deprivation in normally reared male mice mimicked the ACC dopamine receptor subtype modulation and disrupted attention of ELS mice, sleep loss likely underlies cognitive deficits in ELS mice. Likewise, sleep impairment mediated the attention deficits associated with early adversity in human children, as demonstrated by path analysis on data collected with multiple questionnaires for a large child cohort. A deeper understanding of the sex-specific cognitive consequences of ELS thus has the potential to reveal therapeutic strategies for overcoming them.
Collapse
Affiliation(s)
- Yuichi Makino
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
- International Research Center for Neurointelligence, UTIAS, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Nathaniel W Hodgson
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Emma Doenier
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Anna Victoria Serbin
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Koya Osada
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Pietro Artoni
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Matthew Dickey
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Breanna Sullivan
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | - Jelena Komanchuk
- School of Nursing, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Bikram Sekhon
- School of Nursing, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Nicole Letourneau
- School of Nursing, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Neal D Ryan
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jeanette Trauth
- Department of Behavioral and Community Health Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Judy L Cameron
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Takao K Hensch
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
- International Research Center for Neurointelligence, UTIAS, University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| |
Collapse
|
6
|
Aghajani M, Aghajani M, Moghaddam EK, Faghihi M, Imani A. Acute sleep deprivation (ASD) and cardioprotection: Impact of ASD on oxytocin-mediated sympathetic nervous activation preceding myocardial infarction. Neuropeptides 2024; 107:102453. [PMID: 38959559 DOI: 10.1016/j.npep.2024.102453] [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: 02/18/2024] [Revised: 05/10/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
INTRODUCTION This study explored how acute sleep deprivation (ASD) before myocardial ischemia influences oxytocin release from paraventricular (PVN) neurons and its correlation with sympathetic nervous system (SNS) activity post-acute sleep loss, impacting subsequent left ventricular (LV) remodeling following myocardial infarction (MI). METHODS The study was conducted in two phases: induction of ASD, inducing MI, blood sampling, euthanizing animals and collecting their heart and brain for histological and gene expression evaluations. The animals in first and second phase were euthanized 24 h and 14 days after MI, respectively. RESULTS Pre-MI ASD, accompanied by increased serum epinephrine levels within 24 h of MI, upregulated oxytocin and cFos expression in the PVN. Also, pre-MI ASD resulted in decreased serum PAB levels 14 days post-MI (P < 0.001). While notable echocardiographic changes were seen in MI versus sham groups, ASD demonstrated protective effects. This was evidenced by reduced infarct size, elevated TIMP1, MMP2, and MMP9 in the LV of SD + MI animals versus MI alone (P < 0.05). Additionally, histological analysis showed reduced LV fibrosis in pre-MI ASD subjects (P < 0.05). CONCLUSION Our study supports the notion that activation of oxytocin neurons within the PVN subsequent to ASD interacts with autonomic centers in the central nervous system. This enhanced sympathetic outflow to the heart prior to MI triggers a preconditioning response, thereby mediating cardioprotection through decreased oxidative stress biomarkers and regulated extracellular matrix (ECM) turnover.
Collapse
Affiliation(s)
- Marjan Aghajani
- Physiology Department, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mozhgan Aghajani
- Rasoole-Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | | | - Mahdieh Faghihi
- Physiology Department, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Imani
- Physiology Department, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Sleep Breathing Disorders Research Center (SBDRC), Tehran University of Medical Sciences, Tehran, Iran; Occupational Sleep Research Center, Baharloo Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Nakata S, Iwasaki K, Funato H, Yanagisawa M, Ozaki H. Neuronal subtype-specific transcriptomic changes in the cerebral neocortex associated with sleep pressure. Neurosci Res 2024; 207:13-25. [PMID: 38537682 DOI: 10.1016/j.neures.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/19/2024] [Accepted: 03/22/2024] [Indexed: 04/07/2024]
Abstract
Sleep is homeostatically regulated by sleep pressure, which increases during wakefulness and dissipates during sleep. Recent studies have suggested that the cerebral neocortex, a six-layered structure composed of various layer- and projection-specific neuronal subtypes, is involved in the representation of sleep pressure governed by transcriptional regulation. Here, we examined the transcriptomic changes in neuronal subtypes in the neocortex upon increased sleep pressure using single-nucleus RNA sequencing datasets and predicted the putative intracellular and intercellular molecules involved in transcriptome alterations. We revealed that sleep deprivation (SD) had the greatest effect on the transcriptome of layer 2 and 3 intratelencephalic (L2/3 IT) neurons among the neocortical glutamatergic neuronal subtypes. The expression of mutant SIK3 (SLP), which is known to increase sleep pressure, also induced profound changes in the transcriptome of L2/3 IT neurons. We identified Junb as a candidate transcription factor involved in the alteration of the L2/3 IT neuronal transcriptome by SD and SIK3 (SLP) expression. Finally, we inferred putative intercellular ligands, including BDNF, LSAMP, and PRNP, which may be involved in SD-induced alteration of the transcriptome of L2/3 IT neurons. We suggest that the transcriptome of L2/3 IT neurons is most impacted by increased sleep pressure among neocortical glutamatergic neuronal subtypes and identify putative molecules involved in such transcriptional alterations.
Collapse
Affiliation(s)
- Shinya Nakata
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kanako Iwasaki
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hiromasa Funato
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan; Department of Anatomy, Graduate School of Medicine, Toho University, Tokyo, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan; Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| | - Haruka Ozaki
- Bioinformatics Laboratory, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan; Center for Artificial Intelligence Research, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| |
Collapse
|
8
|
Jung JH, Kim J, Akber U, Lee NY, Baek JW, Jung J, Park M, Kang J, Jeon S, Park CS, Kim T. Enhanced homeostatic sleep response and decreased neurodegenerative proteins in cereblon knock-out mice. Commun Biol 2024; 7:1218. [PMID: 39349747 PMCID: PMC11442454 DOI: 10.1038/s42003-024-06879-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 09/10/2024] [Indexed: 10/04/2024] Open
Abstract
Energy homeostasis and sleep have a bidirectional relationship. Cereblon (CRBN) regulates energy levels by ubiquitinating the AMP-activated protein kinase(AMPK), an energy sensor. However, whether CRBN participates in sleep is unclear. Here, we examine sleep-wake patterns in Crbn+/+ and Crbn-/- mice during 24-h baseline, 6-h sleep deprivation (SD), and following 6-h recovery sleep (RS). At baseline, overall sleep patterns are similar between genotypes. However, SD decreases CRBN expression in Crbn+/+ mice and increases phospho-Tau, phospho-α-synuclein, DNAJA1 (DJ2), and DNAJB1 (DJ1) in both genotypes, with Crbn-/- mice showing a lesser extent of increase in p-Tau and p-α-synuclein and a higher level of heat shock protein 70 (HSP70), DJ2, and DJ1. During RS, Crbn-/- mice show increased slow-wave activity in the low-delta range (0.5-2.5 Hz), suggesting higher homeostatic sleep propensity associated with AMPK hyperactivation. By illuminating the role of CRBN in regulating sleep-wake behaviors through AMPK, we suggest CRBN as a potential therapeutic target for managing sleep disorders and preventing neurodegeneration.
Collapse
Affiliation(s)
- Jun-Hyung Jung
- School of Life Sciences, Gwangju Institute Science and Technology (GIST), Gwangju, Republic of Korea
| | - Jinhong Kim
- Department of Biomedical Science and Engineering, GIST, Gwangju, Republic of Korea
| | - Uroos Akber
- School of Life Sciences, Gwangju Institute Science and Technology (GIST), Gwangju, Republic of Korea
| | - Na Young Lee
- School of Life Sciences, Gwangju Institute Science and Technology (GIST), Gwangju, Republic of Korea
| | - Jeong-Won Baek
- School of Life Sciences, Gwangju Institute Science and Technology (GIST), Gwangju, Republic of Korea
| | - Jieun Jung
- Department of Biomedical Science and Engineering, GIST, Gwangju, Republic of Korea
| | - Mincheol Park
- Department of Biomedical Science and Engineering, GIST, Gwangju, Republic of Korea
| | - Jiseung Kang
- Department of Biomedical Science and Engineering, GIST, Gwangju, Republic of Korea
| | - Seungje Jeon
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chul-Seung Park
- School of Life Sciences, Gwangju Institute Science and Technology (GIST), Gwangju, Republic of Korea.
| | - Tae Kim
- Department of Biomedical Science and Engineering, GIST, Gwangju, Republic of Korea.
| |
Collapse
|
9
|
Chen ZK, Liu YY, Zhou JC, Chen GH, Liu CF, Qu WM, Huang ZL. Insomnia-related rodent models in drug discovery. Acta Pharmacol Sin 2024; 45:1777-1792. [PMID: 38671193 PMCID: PMC11335876 DOI: 10.1038/s41401-024-01269-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 03/24/2024] [Indexed: 04/28/2024]
Abstract
Despite the widespread prevalence and important medical impact of insomnia, effective agents with few side effects are lacking in clinics. This is most likely due to relatively poor understanding of the etiology and pathophysiology of insomnia, and the lack of appropriate animal models for screening new compounds. As the main homeostatic, circadian, and neurochemical modulations of sleep remain essentially similar between humans and rodents, rodent models are often used to elucidate the mechanisms of insomnia and to develop novel therapeutic targets. In this article, we focus on several rodent models of insomnia induced by stress, diseases, drugs, disruption of the circadian clock, and other means such as genetic manipulation of specific neuronal activity, respectively, which could be used to screen for novel hypnotics. Moreover, important advantages and constraints of some animal models are discussed. Finally, this review highlights that the rodent models of insomnia may play a crucial role in novel drug development to optimize the management of insomnia.
Collapse
Affiliation(s)
- Ze-Ka Chen
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science; Joint International Research Laboratory of Sleep; and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yuan-Yuan Liu
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science; Joint International Research Laboratory of Sleep; and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ji-Chuan Zhou
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science; Joint International Research Laboratory of Sleep; and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Gui-Hai Chen
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, China
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
| | - Wei-Min Qu
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science; Joint International Research Laboratory of Sleep; and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science; Joint International Research Laboratory of Sleep; and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
10
|
Pinto MJ, Bizien L, Fabre JM, Ðukanović N, Lepetz V, Henderson F, Pujol M, Sala RW, Tarpin T, Popa D, Triller A, Léna C, Fabre V, Bessis A. Microglial TNFα controls daily changes in synaptic GABAARs and sleep slow waves. J Cell Biol 2024; 223:e202401041. [PMID: 38695719 PMCID: PMC11070559 DOI: 10.1083/jcb.202401041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/18/2024] [Accepted: 03/28/2024] [Indexed: 05/08/2024] Open
Abstract
Microglia sense the changes in their environment. How microglia actively translate these changes into suitable cues to adapt brain physiology is unknown. We reveal an activity-dependent regulation of cortical inhibitory synapses by microglia, driven by purinergic signaling acting on P2RX7 and mediated by microglia-derived TNFα. We demonstrate that sleep induces microglia-dependent synaptic enrichment of GABAARs in a manner dependent on microglial TNFα and P2RX7. We further show that microglia-specific depletion of TNFα alters slow waves during NREM sleep and blunt memory consolidation in sleep-dependent learning tasks. Together, our results reveal that microglia orchestrate sleep-intrinsic plasticity of synaptic GABAARs, sculpt sleep slow waves, and support memory consolidation.
Collapse
Affiliation(s)
- Maria Joana Pinto
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Lucy Bizien
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Julie M.J. Fabre
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Nina Ðukanović
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Valentin Lepetz
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Fiona Henderson
- Neurosciences Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), CNRS, INSERM, Sorbonne Universités, Paris, France
| | - Marine Pujol
- Neurosciences Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), CNRS, INSERM, Sorbonne Universités, Paris, France
| | - Romain W. Sala
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Thibault Tarpin
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Daniela Popa
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Antoine Triller
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Clément Léna
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Véronique Fabre
- Neurosciences Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), CNRS, INSERM, Sorbonne Universités, Paris, France
| | - Alain Bessis
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| |
Collapse
|
11
|
Giri B, Kinsky N, Kaya U, Maboudi K, Abel T, Diba K. Sleep loss diminishes hippocampal reactivation and replay. Nature 2024; 630:935-942. [PMID: 38867049 PMCID: PMC11472378 DOI: 10.1038/s41586-024-07538-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/07/2024] [Indexed: 06/14/2024]
Abstract
Memories benefit from sleep1, and the reactivation and replay of waking experiences during hippocampal sharp-wave ripples (SWRs) are considered to be crucial for this process2. However, little is known about how these patterns are impacted by sleep loss. Here we recorded CA1 neuronal activity over 12 h in rats across maze exploration, sleep and sleep deprivation, followed by recovery sleep. We found that SWRs showed sustained or higher rates during sleep deprivation but with lower power and higher frequency ripples. Pyramidal cells exhibited sustained firing during sleep deprivation and reduced firing during sleep, yet their firing rates were comparable during SWRs regardless of sleep state. Despite the robust firing and abundance of SWRs during sleep deprivation, we found that the reactivation and replay of neuronal firing patterns was diminished during these periods and, in some cases, completely abolished compared to ad libitum sleep. Reactivation partially rebounded after recovery sleep but failed to reach the levels found in natural sleep. These results delineate the adverse consequences of sleep loss on hippocampal function at the network level and reveal a dissociation between the many SWRs elicited during sleep deprivation and the few reactivations and replays that occur during these events.
Collapse
Affiliation(s)
- Bapun Giri
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Nathaniel Kinsky
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Utku Kaya
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Kourosh Maboudi
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Ted Abel
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - Kamran Diba
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
12
|
Su AX, Ma ZJ, Li ZY, Li XY, Xia L, Ge YJ, Chen GH. Serum levels of neurotensin, pannexin-1, and sestrin-2 and the correlations with sleep quality or/and cognitive function in the patients with chronic insomnia disorder. Front Psychiatry 2024; 15:1360305. [PMID: 38803679 PMCID: PMC11128551 DOI: 10.3389/fpsyt.2024.1360305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/29/2024] [Indexed: 05/29/2024] Open
Abstract
Objectives To examine serum concentrations of neurotensin, pannexin-1 and sestrin-2, and their correlations with subjective and objective sleep quality and cognitive function in the patients with chronic insomnia disorder (CID). Methods Sixty-five CID patients were enrolled continuously and fifty-six good sleepers in the same period were served as healthy controls (HCs). Serum levels of neurotensin, pannexin-1 and sestrin-2 were measured by enzyme-linked immunosorbent assays. Sleep quality was assessed with the Pittsburgh Sleep Quality Index (PSQI) and polysomnography, and mood was evaluated by 17-item Hamilton Depression Rating Scale. General cognitive function was assessed with the Chinese-Beijing Version of Montreal Cognitive Assessment and spatial memory was evaluated by Blue Velvet Arena Test (BVAT). Results Relative to the HCs, the CID sufferers had higher levels of neurotensin (t=5.210, p<0.001) and pannexin-1 (Z=-4.169, p<0.001), and lower level of sestrin-2 (Z=-2.438, p=0.015). In terms of objective sleep measures, pannexin-1 was positively associated with total sleep time (r=0.562, p=0.002) and sleep efficiency (r=0.588, p=0.001), and negatively with wake time after sleep onset (r=-0.590, p=0.001) and wake time (r=-0.590, p=0.001); sestrin-2 was positively associated with percentage of rapid eye movement sleep (r=0.442, p=0.016) and negatively with non-rapid eye movement sleep stage 2 in the percentage (r=-0.394, p=0.034). Adjusted for sex, age and HAMD, pannexin-1 was still associated with the above objective sleep measures, but sestrin-2 was only negatively with wake time (r=-0.446, p=0.022). However, these biomarkers showed no significant correlations with subjective sleep quality (PSQI score). Serum concentrations of neurotensin and pannexin-1 were positively associated with the mean erroneous distance in the BVAT. Adjusted for sex, age and depression, neurotensin was negatively associated with MoCA score (r=-0.257, p=0.044), pannexin-1 was positively associated with the mean erroneous distance in the BVAT (r=0.270, p=0.033). Conclusions The CID patients had increased neurotensin and pannexin-1 and decreased sestrin-2 in the serum levels, indicating neuron dysfunction, which could be related to poor sleep quality and cognitive dysfunction measured objectively.
Collapse
Affiliation(s)
- Ai-Xi Su
- Department of Neurology (Sleep Disorder), The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, China
- Department of General Medicine, the First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Zi-Jie Ma
- Department of Neurology (Sleep Disorder), The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, China
| | - Zong-Yin Li
- Department of Neurology (Sleep Disorder), The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, China
| | - Xue-Yan Li
- Department of Neurology (Sleep Disorder), The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, China
| | - Lan Xia
- Department of Neurology (Sleep Disorder), The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, China
| | - Yi-Jun Ge
- Department of Neurology (Sleep Disorder), The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, China
| | - Gui-Hai Chen
- Department of Neurology (Sleep Disorder), The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, China
| |
Collapse
|
13
|
Sang D, Lin K, Yang Y, Ran G, Li B, Chen C, Li Q, Ma Y, Lu L, Cui XY, Liu Z, Lv SQ, Luo M, Liu Q, Li Y, Zhang EE. Prolonged sleep deprivation induces a cytokine-storm-like syndrome in mammals. Cell 2023; 186:5500-5516.e21. [PMID: 38016470 DOI: 10.1016/j.cell.2023.10.025] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 08/17/2023] [Accepted: 10/25/2023] [Indexed: 11/30/2023]
Abstract
Most animals require sleep, and sleep loss induces serious pathophysiological consequences, including death. Previous experimental approaches for investigating sleep impacts in mice have been unable to persistently deprive animals of both rapid eye movement sleep (REMS) and non-rapid eye movement sleep (NREMS). Here, we report a "curling prevention by water" paradigm wherein mice remain awake 96% of the time. After 4 days of exposure, mice exhibit severe inflammation, and approximately 80% die. Sleep deprivation increases levels of prostaglandin D2 (PGD2) in the brain, and we found that elevated PGD2 efflux across the blood-brain-barrier-mediated by ATP-binding cassette subfamily C4 transporter-induces both accumulation of circulating neutrophils and a cytokine-storm-like syndrome. Experimental disruption of the PGD2/DP1 axis dramatically reduced sleep-deprivation-induced inflammation. Thus, our study reveals that sleep-related changes in PGD2 in the central nervous system drive profound pathological consequences in the peripheral immune system.
Collapse
Affiliation(s)
- Di Sang
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China; National Institute of Biological Sciences, Beijing, China
| | - Keteng Lin
- National Institute of Biological Sciences, Beijing, China; College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yini Yang
- Peking University School of Life Sciences, Beijing, China
| | - Guangdi Ran
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Bohan Li
- Peking-Tsinghua Center for Life Sciences, Beijing, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Chen Chen
- National Institute of Biological Sciences, Beijing, China
| | - Qi Li
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Yan Ma
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Lihui Lu
- National Institute of Biological Sciences, Beijing, China
| | - Xi-Yang Cui
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Zhibo Liu
- Peking-Tsinghua Center for Life Sciences, Beijing, China; Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Sheng-Qing Lv
- Department of Neurosurgery, Xinqiao Hospital, Chongqing, China
| | - Minmin Luo
- National Institute of Biological Sciences, Beijing, China; School of Life Sciences, Tsinghua University, Beijing, China; Chinese Institute for Brain Research, Beijing, China
| | - Qinghua Liu
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Yulong Li
- Peking University School of Life Sciences, Beijing, China; Peking-Tsinghua Center for Life Sciences, Beijing, China; State Key Laboratory of Membrane Biology, Beijing, China; PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Eric Erquan Zhang
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China.
| |
Collapse
|
14
|
Arizanovska D, Emodogo JA, Lally AP, Palavicino-Maggio CB, Liebl DJ, Folorunso OO. Cross species review of the physiological role of D-serine in translationally relevant behaviors. Amino Acids 2023; 55:1501-1517. [PMID: 37833512 PMCID: PMC10689556 DOI: 10.1007/s00726-023-03338-6] [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: 06/01/2023] [Accepted: 09/19/2023] [Indexed: 10/15/2023]
Abstract
Bridging the gap between preclinical models of neurological and psychiatric disorders with their human manifestations is necessary to understand their underlying mechanisms, identify biomarkers, and develop novel therapeutics. Cognitive and social impairments underlie multiple neuropsychiatric and neurological disorders and are often comorbid with sleep disturbances, which can exacerbate poor outcomes. Importantly, many symptoms are conserved between vertebrates and invertebrates, although they may have subtle differences. Therefore, it is essential to determine the molecular mechanisms underlying these behaviors across different species and their translatability to humans. Genome-wide association studies have indicated an association between glutamatergic gene variants and both the risk and frequency of psychiatric disorders such as schizophrenia, bipolar disorder, and autism spectrum disorder. For example, changes in glutamatergic neurotransmission, such as glutamate receptor subtype N-methyl-D-aspartate receptor (NMDAR) hypofunction, have been shown to contribute to the pathophysiology of schizophrenia. Furthermore, in neurological disorders, such as traumatic brain injury and Alzheimer's disease, hyperactivation of NMDARs leads to synaptic damage. In addition to glutamate binding, NMDARs require the binding of a co-agonist D-serine or glycine to the GluN1 subunit to open. D-serine, which is racemized from L-serine by the neuronal enzyme serine racemase (SRR), and both SRR and D-serine are enriched in cortico-limbic brain regions. D-serine is critical for complex behaviors, such as cognition and social behavior, where dysregulation of its synthesis and release has been implicated in many pathological conditions. In this review, we explore the role of D-serine in behaviors that are translationally relevant to multiple psychiatric and neurological disorders in different models across species.
Collapse
Affiliation(s)
- Dena Arizanovska
- The Miami Project to Cure Paralysis, Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jada A Emodogo
- Translational Psychiatry Laboratory, McLean Hospital, Belmont, MA, USA
| | - Anna P Lally
- Translational Neuroscience Laboratory, McLean Hospital, Belmont, MA, USA
| | - Caroline B Palavicino-Maggio
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Neurobiological Mechanisms of Aggression Laboratory, McLean Hospital, Belmont, MA, USA
| | - Daniel J Liebl
- The Miami Project to Cure Paralysis, Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Oluwarotimi O Folorunso
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
- Translational Psychiatry Laboratory, McLean Hospital, Belmont, MA, USA.
| |
Collapse
|
15
|
Neculicioiu VS, Colosi IA, Costache C, Toc DA, Sevastre-Berghian A, Colosi HA, Clichici S. Sleep Deprivation-Induced Oxidative Stress in Rat Models: A Scoping Systematic Review. Antioxidants (Basel) 2023; 12:1600. [PMID: 37627596 PMCID: PMC10451248 DOI: 10.3390/antiox12081600] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/01/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Sleep deprivation is highly prevalent in the modern world, possibly reaching epidemic proportions. While multiple theories regarding the roles of sleep exist (inactivity, energy conservation, restoration, brain plasticity and antioxidant), multiple unknowns still remain regarding the proposed antioxidant roles of sleep. The existing experimental evidence is often contradicting, with studies pointing both toward and against the presence of oxidative stress after sleep deprivation. The main goals of this review were to analyze the existing experimental data regarding the relationship between sleep deprivation and oxidative stress, to attempt to further clarify multiple aspects surrounding this relationship and to identify current knowledge gaps. Systematic searches were conducted in three major online databases for experimental studies performed on rat models with oxidative stress measurements, published between 2015 and 2022. A total of 54 studies were included in the review. Most results seem to point to changes in oxidative stress parameters after sleep deprivation, further suggesting an antioxidant role of sleep. Alterations in these parameters were observed in both paradoxical and total sleep deprivation protocols and in multiple rat strains. Furthermore, the effects of sleep deprivation seem to extend beyond the central nervous system, affecting multiple other body sites in the periphery. Sleep recovery seems to be characterized by an increased variability, with the presence of both normalizations in some parameters and long-lasting changes after sleep deprivation. Surprisingly, most studies revealed the presence of a stress response following sleep deprivation. However, the origin and the impact of the stress response during sleep deprivation remain somewhat unclear. While a definitive exclusion of the influence of the sleep deprivation protocol on the stress response is not possible, the available data seem to suggest that the observed stress response may be determined by sleep deprivation itself as opposed to the experimental conditions. Due to this fact, the observed oxidative changes could be attributed directly to sleep deprivation.
Collapse
Affiliation(s)
- Vlad Sever Neculicioiu
- Department of Microbiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Ioana Alina Colosi
- Department of Microbiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Carmen Costache
- Department of Microbiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Dan Alexandru Toc
- Department of Microbiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Alexandra Sevastre-Berghian
- Department of Physiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Horațiu Alexandru Colosi
- Division of Medical Informatics and Biostatistics, Department of Medical Education, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Simona Clichici
- Department of Physiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| |
Collapse
|
16
|
Gros A, Wang SH. Cognitive rescue in aging through prior training in rats. Aging (Albany NY) 2023; 15:5990-6010. [PMID: 37338529 PMCID: PMC10373978 DOI: 10.18632/aging.204808] [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: 02/22/2023] [Accepted: 05/23/2023] [Indexed: 06/21/2023]
Abstract
Cognitive decline in spatial memory is seen in aging. Understanding affected processes in aging is vital for developing methods to improve wellbeing. Daily memory persistence can be influenced by events around the time of learning or by prior experiences in early life. Fading memories in young can last longer if a novel event is introduced around encoding, a process called behavioral tagging. Based on this principle, we asked what processes are affected in aging and if prior training can rescue them. Two groups of aged rats received training in an appetitive delayed matching-to-place task. One of the groups additionally received prior training of the same task in young and in mid-life, constituting a longitudinal study. The results showed long-term memory decline in late aging without prior training. This would reflect affected encoding and consolidation. On the other hand, short-term memory was preserved and novelty at memory reactivation and reconsolidation enabled memory maintenance in aging. Prior training improved cognition through facilitating task performance, strengthening short-term memory and intermediate memory, and enabling encoding-boosted long-term memory. Implication of these findings in understanding brain mechanisms in cognitive aging and in beneficial effects of prior training is discussed.
Collapse
Affiliation(s)
- Alexandra Gros
- Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor’s Building, Edinburgh, Scotland, UK
| | - Szu-Han Wang
- Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor’s Building, Edinburgh, Scotland, UK
| |
Collapse
|
17
|
Hu Y, Bringmann H. Tfap2b acts in GABAergic neurons to control sleep in mice. Sci Rep 2023; 13:8026. [PMID: 37198238 DOI: 10.1038/s41598-023-34772-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 05/07/2023] [Indexed: 05/19/2023] Open
Abstract
Sleep is a universal state of behavioral quiescence in both vertebrates and invertebrates that is controlled by conserved genes. We previously found that AP2 transcription factors control sleep in C. elegans, Drosophila, and mice. Heterozygous deletion of Tfap2b, one of the mammalian AP2 paralogs, reduces sleep in mice. The cell types and mechanisms through which Tfap2b controls sleep in mammals are, however, not known. In mice, Tfap2b acts during early embryonic stages. In this study, we used RNA-seq to measure the gene expression changes in brains of Tfap2b-/- embryos. Our results indicated that genes related to brain development and patterning were differentially regulated. As many sleep-promoting neurons are known to be GABAergic, we measured the expression of GAD1, GAD2 and Vgat genes in different brain areas of adult Tfap2b+/- mice using qPCR. These experiments suggested that GABAergic genes are downregulated in the cortex, brainstem and cerebellum areas, but upregulated in the striatum. To investigate whether Tfap2b controls sleep through GABAergic neurons, we specifically deleted Tfap2b in GABAergic neurons. We recorded the EEG and EMG before and after a 6-h period of sleep deprivation and extracted the time spent in NREM and in REM sleep as well as delta and theta power to assess NREM and REM sleep, respectively. During baseline conditions, Vgat-tfap2b-/- mice exhibited both shortened NREM and REM sleep time and reduced delta and theta power. Consistently, weaker delta and theta power were observed during rebound sleep in the Vgat-tfap2b-/- mice after sleep deprivation. Taken together, the results indicate that Tfap2b in GABAergic neurons is required for normal sleep.
Collapse
Affiliation(s)
- Yang Hu
- Max Planck Research Group "Sleep and Waking", Max Planck Institute for Biophysical Chemistry, 37077, Göttingen, Germany
| | - Henrik Bringmann
- Max Planck Research Group "Sleep and Waking", Max Planck Institute for Biophysical Chemistry, 37077, Göttingen, Germany.
- Cellular Circuits and Systems, Biotechnology Center (BIOTEC), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, 01307, Dresden, Germany.
| |
Collapse
|
18
|
Wright CJ, Milosavljevic S, Pocivavsek A. The stress of losing sleep: Sex-specific neurobiological outcomes. Neurobiol Stress 2023; 24:100543. [PMID: 37252645 PMCID: PMC10209346 DOI: 10.1016/j.ynstr.2023.100543] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/20/2023] [Accepted: 05/06/2023] [Indexed: 05/31/2023] Open
Abstract
Sleep is a vital and evolutionarily conserved process, critical to daily functioning and homeostatic balance. Losing sleep is inherently stressful and leads to numerous detrimental physiological outcomes. Despite sleep disturbances affecting everyone, women and female rodents are often excluded or underrepresented in clinical and pre-clinical studies. Advancing our understanding of the role of biological sex in the responses to sleep loss stands to greatly improve our ability to understand and treat health consequences of insufficient sleep. As such, this review discusses sex differences in response to sleep deprivation, with a focus on the sympathetic nervous system stress response and activation of the hypothalamic-pituitary-adrenal (HPA) axis. We review sex differences in several stress-related consequences of sleep loss, including inflammation, learning and memory deficits, and mood related changes. Focusing on women's health, we discuss the effects of sleep deprivation during the peripartum period. In closing, we present neurobiological mechanisms, including the contribution of sex hormones, orexins, circadian timing systems, and astrocytic neuromodulation, that may underlie potential sex differences in sleep deprivation responses.
Collapse
Affiliation(s)
- Courtney J. Wright
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Snezana Milosavljevic
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Ana Pocivavsek
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| |
Collapse
|
19
|
Morrone CD, Raghuraman R, Hussaini SA, Yu WH. Proteostasis failure exacerbates neuronal circuit dysfunction and sleep impairments in Alzheimer's disease. Mol Neurodegener 2023; 18:27. [PMID: 37085942 PMCID: PMC10119020 DOI: 10.1186/s13024-023-00617-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/29/2023] [Indexed: 04/23/2023] Open
Abstract
Failed proteostasis is a well-documented feature of Alzheimer's disease, particularly, reduced protein degradation and clearance. However, the contribution of failed proteostasis to neuronal circuit dysfunction is an emerging concept in neurodegenerative research and will prove critical in understanding cognitive decline. Our objective is to convey Alzheimer's disease progression with the growing evidence for a bidirectional relationship of sleep disruption and proteostasis failure. Proteostasis dysfunction and tauopathy in Alzheimer's disease disrupts neurons that regulate the sleep-wake cycle, which presents behavior as impaired slow wave and rapid eye movement sleep patterns. Subsequent sleep loss further impairs protein clearance. Sleep loss is a defined feature seen early in many neurodegenerative disorders and contributes to memory impairments in Alzheimer's disease. Canonical pathological hallmarks, β-amyloid, and tau, directly disrupt sleep, and neurodegeneration of locus coeruleus, hippocampal and hypothalamic neurons from tau proteinopathy causes disruption of the neuronal circuitry of sleep. Acting in a positive-feedback-loop, sleep loss and circadian rhythm disruption then increase spread of β-amyloid and tau, through impairments of proteasome, autophagy, unfolded protein response and glymphatic clearance. This phenomenon extends beyond β-amyloid and tau, with interactions of sleep impairment with the homeostasis of TDP-43, α-synuclein, FUS, and huntingtin proteins, implicating sleep loss as an important consideration in an array of neurodegenerative diseases and in cases of mixed neuropathology. Critically, the dynamics of this interaction in the neurodegenerative environment are not fully elucidated and are deserving of further discussion and research. Finally, we propose sleep-enhancing therapeutics as potential interventions for promoting healthy proteostasis, including β-amyloid and tau clearance, mechanistically linking these processes. With further clinical and preclinical research, we propose this dynamic interaction as a diagnostic and therapeutic framework, informing precise single- and combinatorial-treatments for Alzheimer's disease and other brain disorders.
Collapse
Affiliation(s)
- Christopher Daniel Morrone
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON, M5T 1R8, Canada.
| | - Radha Raghuraman
- Taub Institute, Columbia University Irving Medical Center, 630W 168th Street, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 630W 168th Street, New York, NY, 10032, USA
| | - S Abid Hussaini
- Taub Institute, Columbia University Irving Medical Center, 630W 168th Street, New York, NY, 10032, USA.
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 630W 168th Street, New York, NY, 10032, USA.
| | - Wai Haung Yu
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON, M5T 1R8, Canada.
- Geriatric Mental Health Research Services, Centre for Addiction and Mental Health, 250 College St., Toronto, ON, M5T 1R8, Canada.
- Department of Pharmacology and Toxicology, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
20
|
Zhang F, Niu L, Zhong R, Li S, Le W. Chronic Sleep Disturbances Alters Sleep Structure and Tau Phosphorylation in AβPP/PS1 AD Mice and Their Wild-Type Littermates. J Alzheimers Dis 2023; 92:1341-1355. [PMID: 37038814 DOI: 10.3233/jad-221048] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Abstract
Background: Emerging evidence indicates that sleep disorders are the common non-cognitive symptoms of Alzheimer’s disease (AD), and they may contribute to the pathogenesis of this disease. Objective: In this study, we aim to investigate the effect of chronic sleep deprivation (CSD) on AD-related pathologies with a focus on tau phosphorylation and the underlying DNA methylation regulation. Methods: AβPPswe/PS1ΔE9 AD mice and their wild-type (WT) littermates were subjected to a two-month CSD followed by electroencephalography and electromyography recording. The mice were examined for learning and memory evaluation, then pathological, biochemical, and epigenetic assessments including western blotting, immunofluorescence, dot blotting, and bisulfite sequencing. Results: The results show that CSD caused sleep disorders shown as sleep pattern change, poor sleep maintenance, and increased sleep fragmentation. CSD increased tau phosphorylation at different sites and increased the level of tau kinases in AD and WT mice. The increased expression of cyclin-dependent kinase 5 (CDK5) may result from decreased DNA methylation of CpG sites in the promoter region of CDK5 gene, which might be associated with the downregulation of DNA methyltransferase 3A and 3B. Conclusion: CSD altered AD-related tau phosphorylation through epigenetic modification of tau kinase gene. The findings in this study may give insights into the mechanisms underlying the effects of sleep disorders on AD pathology and provide new therapeutic targets for the treatment of this disease.
Collapse
Affiliation(s)
- Feng Zhang
- Center for Clinical and Translational Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Long Niu
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Rujia Zhong
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Song Li
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Weidong Le
- Center for Clinical and Translational Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
21
|
Griffin SM, Lehtinen MJ, Meunier J, Ceolin L, Roman FJ, Patterson E. Restorative effects of probiotics on memory impairment in sleep-deprived mice. Nutr Neurosci 2023; 26:254-264. [PMID: 35236257 DOI: 10.1080/1028415x.2022.2042915] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Insufficient sleep is a serious public health epidemic in modern society, impairing memory and other cognitive functions. In this study, partial sleep deprivation (SD) was used to induce cognitive impairment in mice to determine the effects of probiotics on subsequent cognitive deficits. METHODS Lactiplantibacillus plantarum Lp-115 (Lp-115), Lacticaseibacillus paracasei Lpc-37 (Lpc-37), Bifidobacterium animalis subsp. lactis 420 (B420) and their combination were administered to mice subjected to partial SD and compared with non-SD and SD vehicle groups. Mice were administered a daily oral gavage containing either 1 × 109 colony forming units (CFU) of single-strain, 1.5 × 109 CFU of multi-strain (5 × 108 CFU/strain), or vehicle for thirty days prior to and for nine days during a behavioural test paradigm. The novel object recognition (NOR) test, spontaneous alternation Y-maze (Y-maze), and the step-through passive avoidance (STPA) task were applied to evaluate learning and memory performance following partial SD. RESULTS Partial SD had a significant impact on cognitive function in vehicle mice. Intervention with Lpc-37 significantly improved recognition memory deficits in the NOR test, spatial working memory deficits in the Y-maze, and contextual long-term memory impairments in the STPA task, in mice subjected to partial SD compared to the SD vehicle group. The multi-strain significantly improved recognition memory deficits in the NOR test and spatial working memory deficits in the Y-maze in mice subjected to partial SD compared to the SD vehicle group. CONCLUSIONS These findings demonstrate that Lpc-37 and the multi-strain may play a role in alleviating memory impairments and improve cognitive function in partially sleep-deprived mice.
Collapse
Affiliation(s)
- Síle M Griffin
- IFF Health & Biosciences, Danisco Sweeteners Oy, Kantvik, Finland
| | | | | | | | | | - Elaine Patterson
- IFF Health & Biosciences, Danisco Sweeteners Oy, Kantvik, Finland
| |
Collapse
|
22
|
Bian WJ, De Lecea L. Automated Sleep Deprivation Setup Using a Shaking Platform in Mice. Bio Protoc 2023; 13:e4620. [PMID: 36845529 PMCID: PMC9947545 DOI: 10.21769/bioprotoc.4620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/20/2022] [Accepted: 01/29/2023] [Indexed: 02/17/2023] Open
Abstract
The functions of sleep remain largely unclear, and even less is known about its role in development. A general strategy to tackle these questions is to disrupt sleep and measure the outcomes. However, some existing sleep deprivation methods may not be suitable for studying the effects of chronic sleep disruption, due to their lack of effectiveness and/or robustness, substantial stress caused by the deprivation method, or consuming a large quantity of time and manpower. More problems may be encountered when applying these existing protocols to young, developing animals, because of their likely heightened vulnerability to stressors, and difficulties in precisely monitoring sleep at young ages. Here, we report a protocol of automated sleep disruption in mice using a commercially available, shaking platform-based deprivation system. We show that this protocol effectively and robustly deprives both non-rapid-eye-movement (NREM) sleep and rapid-eye-movement (REM) sleep without causing a significant stress response, and does not require human supervision. This protocol uses adolescent mice, but the method also works with adult mice. Graphical abstract Automated sleep deprivation system. The platform of the deprivation chamber was programmed to shake in a given frequency and intensity to keep the animal awake while its brain and muscle activities were continuously monitored by electroencephalography and electromyography.
Collapse
Affiliation(s)
- Wen-Jie Bian
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA,*For correspondence: ;
| | - Luis De Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA,*For correspondence: ;
| |
Collapse
|
23
|
Giri B, Kaya U, Maboudi K, Abel T, Diba K. Sleep loss diminishes hippocampal reactivation and replay. RESEARCH SQUARE 2023:rs.3.rs-2540186. [PMID: 36824950 PMCID: PMC9949250 DOI: 10.21203/rs.3.rs-2540186/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Memories benefit from sleep, and sleep loss immediately following learning has a negative impact on subsequent memory storage. Several prominent hypotheses ascribe a central role to hippocampal sharp-wave ripples (SWRs), and the concurrent reactivation and replay of neuronal patterns from waking experience, in the offline memory consolidation process that occurs during sleep. However, little is known about how SWRs, reactivation, and replay are affected when animals are subjected to sleep deprivation. We performed long duration (~12 h), high-density silicon probe recordings from rat hippocampal CA1 neurons, in animals that were either sleeping or sleep deprived following exposure to a novel maze environment. We found that SWRs showed a sustained rate of activity during sleep deprivation, similar to or higher than in natural sleep, but with decreased amplitudes for the sharp-waves combined with higher frequencies for the ripples. Furthermore, while hippocampal pyramidal cells showed a log-normal distribution of firing rates during sleep, these distributions were negatively skewed with a higher mean firing rate in both pyramidal cells and interneurons during sleep deprivation. During SWRs, however, firing rates were remarkably similar between both groups. Despite the abundant quantity of SWRs and the robust firing activity during these events in both groups, we found that reactivation of neurons was either completely abolished or significantly diminished during sleep deprivation compared to sleep. Interestingly, reactivation partially rebounded upon recovery sleep, but failed to reach the levels characteristic of natural sleep. Similarly, the number of replays were significantly lower during sleep deprivation and recovery sleep compared to natural sleep. These results provide a network-level account for the negative impact of sleep loss on hippocampal function and demonstrate that sleep loss impacts memory storage by causing a dissociation between the amount of SWRs and the replays and reactivations that take place during these events.
Collapse
Affiliation(s)
- Bapun Giri
- Dept of Anesthesiology and Neuroscience Graduate Program, 1150 W Medical Center Dr, University of Michigan Medical School, Ann Arbor, MI 48109
- Dept of Psychology, University of Wisconsin-Milwaukee, PO Box 413, Milwaukee, WI 53201
| | - Utku Kaya
- Dept of Anesthesiology and Neuroscience Graduate Program, 1150 W Medical Center Dr, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Kourosh Maboudi
- Dept of Anesthesiology and Neuroscience Graduate Program, 1150 W Medical Center Dr, University of Michigan Medical School, Ann Arbor, MI 48109
- Dept of Psychology, University of Wisconsin-Milwaukee, PO Box 413, Milwaukee, WI 53201
| | - Ted Abel
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA
| | - Kamran Diba
- Dept of Anesthesiology and Neuroscience Graduate Program, 1150 W Medical Center Dr, University of Michigan Medical School, Ann Arbor, MI 48109
| |
Collapse
|
24
|
Morrone CD, Tsang AA, Giorshev SM, Craig EE, Yu WH. Concurrent behavioral and electrophysiological longitudinal recordings for in vivo assessment of aging. Front Aging Neurosci 2023; 14:952101. [PMID: 36742209 PMCID: PMC9891465 DOI: 10.3389/fnagi.2022.952101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 12/12/2022] [Indexed: 01/19/2023] Open
Abstract
Electrophysiological and behavioral alterations, including sleep and cognitive impairments, are critical components of age-related decline and neurodegenerative diseases. In preclinical investigation, many refined techniques are employed to probe these phenotypes, but they are often conducted separately. Herein, we provide a protocol for one-time surgical implantation of EMG wires in the nuchal muscle and a skull-surface EEG headcap in mice, capable of 9-to-12-month recording longevity. All data acquisitions are wireless, making them compatible with simultaneous EEG recording coupled to multiple behavioral tasks, as we demonstrate with locomotion/sleep staging during home-cage video assessments, cognitive testing in the Barnes maze, and sleep disruption. Time-course EEG and EMG data can be accurately mapped to the behavioral phenotype and synchronized with neuronal frequencies for movement and the location to target in the Barnes maze. We discuss critical steps for optimizing headcap surgery and alternative approaches, including increasing the number of EEG channels or utilizing depth electrodes with the system. Combining electrophysiological and behavioral measurements in preclinical models of aging and neurodegeneration has great potential for improving mechanistic and therapeutic assessments and determining early markers of brain disorders.
Collapse
Affiliation(s)
- Christopher Daniel Morrone
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada,*Correspondence: Christopher Daniel Morrone,
| | - Arielle A. Tsang
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada,Department of Biological Sciences, University of Toronto Scarborough, Toronto, ON, Canada
| | - Sarah M. Giorshev
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada,Department of Psychology, University of Toronto Scarborough, Toronto, ON, Canada
| | - Emily E. Craig
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Wai Haung Yu
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada,Geriatric Mental Health Research Services, Centre for Addiction and Mental Health, Toronto, ON, Canada,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada,Wai Haung Yu,
| |
Collapse
|
25
|
Gabova AV, Sarkisova KY. Maternal Methyl-Enriched Diet Normalizes Characteristics of the Sleep–Wake Cycle and Sleep Spindles in Adult Offspring of WAG/Rij Rats with Genetic Absence Epilepsy. J EVOL BIOCHEM PHYS+ 2023. [DOI: 10.1134/s0022093023010143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
|
26
|
Ugalde-Muñiz P, Hernández-Luna MG, García-Velasco S, Lugo-Huitrón R, Murcia-Ramírez J, Martínez-Tapia RJ, Noriega-Navarro R, Navarro L. Activation of dopamine D2 receptors attenuates neuroinflammation and ameliorates the memory impairment induced by rapid eye movement sleep deprivation in a murine model. Front Neurosci 2022; 16:988167. [PMID: 36278007 PMCID: PMC9579422 DOI: 10.3389/fnins.2022.988167] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
The proinflammatory state, which may be induced by sleep deprivation, seems to be a determining factor in the development of neurodegenerative processes. Investigations of mechanisms that help to mitigate the inflammatory effects of sleep disorders are important. A new proposal involves the neurotransmitter dopamine, which may modulate the progression of the immune response by activating receptors expressed on immune cells. This study aimed to determine whether dopamine D2 receptor (D2DR) activation attenuates the proinflammatory response derived from rapid eye movement (REM) sleep deprivation in mice. REM sleep deprivation (RSD) was induced in 2-month-old male CD1 mice using the multiple platform model for three consecutive days; during this period, the D2DR receptor agonist quinpirole (QUIN) was administered (2 mg/kg/day i.p.). Proinflammatory cytokine levels were assessed in serum and homogenates of the brain cortex, hippocampus, and striatum using ELISAs. Long-term memory deficits were identified using the Morris water maze (MWM) and novel object recognition (NOR) tests. Animals were trained until learning criteria were achieved; then, they were subjected to RSD and treated with QUIN for 3 days. Memory evocation was determined afterward. Moreover, we found RSD induced anhedonia, as measured by the sucrose consumption test, which is commonly related to the dopaminergic system. Our data revealed increased levels of proinflammatory cytokines (TNFα and IL-1β) in both the hippocampus and serum from RSD mice. However, QUIN attenuated the increased levels of these cytokines. Furthermore, RSD caused a long-term memory evocation deficit in both the MWM and NOR tests. In contrast, QUIN coadministration during the RSD period significantly improved the performance of the animals. On the other hand, QUIN prevented the anhedonic condition induced by RSD. Based on our results, D2DR receptor activation protects against memory impairment induced by disturbed REM sleep by inhibiting neuroinflammation.
Collapse
Affiliation(s)
- Perla Ugalde-Muñiz
- Laboratory of Neuroendocrinology, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - María Guadalupe Hernández-Luna
- Laboratory of Neuroendocrinology, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Stephany García-Velasco
- Laboratory of Neuroendocrinology, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Rafael Lugo-Huitrón
- Laboratory of Behavioral Neurobiology, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Jimena Murcia-Ramírez
- Laboratory of Neuroendocrinology, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Ricardo Jesus Martínez-Tapia
- Laboratory of Neuroendocrinology, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Roxana Noriega-Navarro
- Laboratory of Neuroendocrinology, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Luz Navarro
- Laboratory of Neuroendocrinology, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
- *Correspondence: Luz Navarro,
| |
Collapse
|
27
|
Gentry NW, McMahon T, Yamazaki M, Webb J, Arnold TD, Rosi S, Ptáček LJ, Fu YH. Microglia are involved in the protection of memories formed during sleep deprivation. Neurobiol Sleep Circadian Rhythms 2022; 12:100073. [PMID: 35028489 PMCID: PMC8741522 DOI: 10.1016/j.nbscr.2021.100073] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 12/10/2021] [Accepted: 12/15/2021] [Indexed: 01/09/2023] Open
Abstract
Sleep deprivation can generate inflammatory responses in the central nervous system. In turn, this inflammation increases sleep drive, leading to a rebound in sleep duration. Microglia, the innate immune cells found exclusively in the CNS, have previously been found to release inflammatory signals and exhibit altered characteristics in response to sleep deprivation. Together, this suggests that microglia may be partially responsible for the brain's response to sleep deprivation through their inflammatory activity. In this study, we ablated microglia from the mouse brain and assessed resulting sleep, circadian, and sleep deprivation phenotypes. We find that microglia are dispensable for both homeostatic sleep and circadian function and the sleep rebound response to sleep deprivation. However, we uncover a phenomenon by which microglia appear to be essential for the protection of fear-conditioning memories formed during the recovery sleep period following a period of sleep deprivation. This phenomenon occurs potentially through the upregulation of synaptic-homeostasis related genes to protect nascent dendritic spines that may be otherwise removed or downscaled during recovery sleep. These findings further expand the list of known functions for microglia in synaptic modulation.
Collapse
Affiliation(s)
- Nicholas W. Gentry
- Department of Neurology, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Thomas McMahon
- Department of Neurology, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Maya Yamazaki
- Department of Neurology, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - John Webb
- Department of Neurology, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Thomas D. Arnold
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, 94143, USA
- Department of Physical Rehabilitation Science, University of California, San Francisco, San Francisco, CA, 94143, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Susanna Rosi
- Department of Physical Rehabilitation Science, University of California, San Francisco, San Francisco, CA, 94143, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, 94143, USA
- Weill Institute for Neuroscience, University of California, San Francisco, San Francisco, CA, 94143, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Louis J. Ptáček
- Department of Neurology, University of California, San Francisco, San Francisco, CA, 94143, USA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, 94143, USA
- Weill Institute for Neuroscience, University of California, San Francisco, San Francisco, CA, 94143, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Ying-Hui Fu
- Department of Neurology, University of California, San Francisco, San Francisco, CA, 94143, USA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, 94143, USA
- Weill Institute for Neuroscience, University of California, San Francisco, San Francisco, CA, 94143, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, 94143, USA
| |
Collapse
|
28
|
Interaction of lithium and sleep deprivation on memory performance and anxiety-like behavior in male Wistar rats. Behav Brain Res 2022; 428:113890. [DOI: 10.1016/j.bbr.2022.113890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/12/2022] [Accepted: 04/07/2022] [Indexed: 12/28/2022]
|
29
|
Brodin ATS, Gabulya S, Wellfelt K, Karlsson TE. Five Hours Total Sleep Deprivation Does Not Affect CA1 Dendritic Length or Spine Density. Front Synaptic Neurosci 2022; 14:854160. [PMID: 35359703 PMCID: PMC8964138 DOI: 10.3389/fnsyn.2022.854160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Sleep is essential for long term memory function. However, the neuroanatomical consequences of sleep loss are disputed. Sleep deprivation has been reported to cause both decreases and increases of dendritic spine density. Here we use Thy1-GFP expressing transgenic mice to investigate the effects of acute sleep deprivation on the dendritic architecture of hippocampal CA1 pyramidal neurons. We found that 5 h of sleep deprivation had no effect on either dendritic length or dendritic spine density. Our work suggests that no major neuroanatomical changes result from a single episode of sleep deprivation.
Collapse
Affiliation(s)
| | - Sarolta Gabulya
- Institute of Neuroinformatics, University of Zurich and ETH, Zürich, Switzerland
| | - Katrin Wellfelt
- Department of Neuroscience, Karolinska Institutet, Solna, Sweden
| | - Tobias E. Karlsson
- Department of Neuroscience, Karolinska Institutet, Solna, Sweden
- *Correspondence: Tobias E. Karlsson,
| |
Collapse
|
30
|
Moore JL, Brook EI. Using Home-Cage Monitoring to Determine the Impact of Timed Mating on Male Mouse Welfare. Front Neurosci 2022; 16:786652. [PMID: 35281486 PMCID: PMC8904386 DOI: 10.3389/fnins.2022.786652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/04/2022] [Indexed: 11/30/2022] Open
Abstract
Some of our breeding programs include the use of Prm1 male Homozygous mice which are naturally sterile. This removes the need to use vasectomized males to induce pseudopregnancy in female mice. These males can be kept for up to 9 months and are housed with a companion female. During the timed mating period the companion female is replaced with a new female. This procedure can occur at regular intervals causing a significant increase in cage activity; one of our objectives was to determine whether this was as a result of timed mating. We wanted to investigate the disruption caused to mice during the day of the swap and how long it would take for the cage activity to return to pre-replacement baseline levels. We hypothesized that this impact would be reflected as a significant increase in cage activity, which in itself may not be a result of a negative experience but the potential of repeated disruption to their activity pattern should be considered. We used a well-known home-cage monitoring system to assess changes to the activity pattern in cages when a companion female is replaced. Data from our initial study showed that in the 2-h period after the female is replaced there is a significant increase in cage activity compared to the same time frame on the previous day. In the subsequent study, where no cage change occurred, an increase in activity was also observed when females were replaced; this returned to baseline after approximately 4 h. Prolonged activity during the rest period of mice (over 2 h) could lead to them being fatigued during their active period; therefore, as a refinement we propose that timed matings be performed later in the day, at a time when the animals are active.
Collapse
|
31
|
DiNuzzo M, Mangia S, Giove F. Manipulations of sleep‐like slow‐wave activity by noninvasive brain stimulation. J Neurosci Res 2022; 100:1218-1225. [DOI: 10.1002/jnr.25029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 01/18/2022] [Accepted: 01/29/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Mauro DiNuzzo
- Magnetic Resonance for Brain Investigation Laboratory Museo Storico della Fisica e Centro di Studi e Ricerche Enrico Fermi Rome Italy
| | - Silvia Mangia
- Center for Magnetic Resonance Research, Department of Radiology University of Minnesota Minneapolis Minnesota USA
| | - Federico Giove
- Magnetic Resonance for Brain Investigation Laboratory Museo Storico della Fisica e Centro di Studi e Ricerche Enrico Fermi Rome Italy
- Laboratory of Neurophysics and Neuroimaging Fondazione Santa Lucia IRCCS Rome Italy
| |
Collapse
|
32
|
Chittora R, Jain A, Shukla SD, Bhatnagar M. Cytomorphological Analysis and Interpretation of Nitric Oxide-Mediated Neurotoxicity in Sleep-Deprived Mice Model. Ann Neurosci 2022; 29:7-15. [PMID: 35875423 PMCID: PMC9305911 DOI: 10.1177/09727531211059925] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 10/07/2021] [Indexed: 12/02/2022] Open
Abstract
Background: Sleep deprivation (SD) is a biological stress condition for the brain, and the pathogenesis of SD is closely related to elevated oxidative stress, mitochondrial dysfunction, a major cause of neurodegeneration. This oxidative stress-mediated cell death is attributed to rise in calcium ion influx which further excites or alters the neurotransmitters level by activating neuronal nitric oxide (NO) synthase (nNOS) release of NO in mouse SD model. This study indicates that the nitrergic neurons are possible therapeutic targets for the amelioration of SD-induced cognitive dysfunction and behavioral alterations. Purpose: SD is considered as a risk factor for various neurodegenerative diseases. SD leads to biochemical, behavioral, and neurochemical alterations in animals. This study was designed to explore the possible involvement of a nitrergic neuron system in six days SD-induced morphological and neurodegenerative changes in mice. Methods: Using nNOS immunohistochemistry, we have investigated the effects of SD on nNOS positive neurons. Immunohistochemical study for the distribution of nNOS positive neuronal cell bodies was carried out in the hippocampus, prefrontal cortex (PFC), and amygdaloid nuclei of mice brain. Results: Sleep-deprived animals showed a significantly increased number of nNOS positive neurons and altered neuronal cytomorphology as compared with the control group. Conclusion: These results indicate that total SD may induce morphological changes in nNOS positive neurons in the brain, thus increasing NO synthesis, which is implicated in SD-induced neuronal cell death.
Collapse
Affiliation(s)
- Reena Chittora
- Department of Physiology, Neurophysiology Laboratory, All India Institute of Medical Sciences, New Delhi, Delhi, India
- Department of Zoology, Animal Biotechnology and Molecular Neuroscience Laboratory, University College of Science, Mohan Lal Sukhadia University, Udaipur, Rajasthan, India
| | - Ayushi Jain
- Department of Zoology, Animal Biotechnology and Molecular Neuroscience Laboratory, University College of Science, Mohan Lal Sukhadia University, Udaipur, Rajasthan, India
- Department of Biochemistry, King George Medical University, Lucknow, Uttar Pradesh, India
| | - Sunil Dutt Shukla
- Department of Zoology, Government Meera Girls College, Udaipur, Rajasthan, India
| | - Maheep Bhatnagar
- Department of Zoology, Animal Biotechnology and Molecular Neuroscience Laboratory, University College of Science, Mohan Lal Sukhadia University, Udaipur, Rajasthan, India
| |
Collapse
|
33
|
Tripathi S, Jha SK. REM Sleep Deprivation Alters Learning-Induced Cell Proliferation and Generation of Newborn Young Neurons in the Dentate Gyrus of the Dorsal Hippocampus. ACS Chem Neurosci 2022; 13:194-206. [PMID: 34990120 DOI: 10.1021/acschemneuro.1c00465] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The hippocampus-dependent "trace-appetitive conditioning task" increases cell proliferation and the generation of newborn young neurons. Evidence suggests that adult hippocampal neurogenesis and rapid eye movement (REM) sleep play an essential role in memory consolidation. On the other hand, REM sleep deprivation (REM-SD) induces detrimental effects on training-induced cell proliferation in the hippocampus's dentate gyrus (DG). Nonetheless, the role of REM sleep in the trace-appetitive memory and fate determination of the newly proliferated cells is not known. Here, we have studied the following: (I) the effects of 24 h of REM-SD (soon after training) on trace- and delay-appetitive memory and cell proliferation in the adult DG and (II) the effects of chronic (96 h) REM-SD (3 days after the training, the period in which newly generated cells progressed toward the neuronal lineage) on trace-appetitive memory and the generation of newborn young neurons. We used a modified multiple platform method for the selective REM-SD without altering non-REM (NREM) sleep. We found that 24 h of REM-SD, soon after trace-conditioning, impaired the trace-appetitive memory and the training-induced cell proliferation. Nevertheless, 96 h of REM-SD (3 days after the training) did not impair trace memory. Interestingly, 96 h of REM-SD altered the generation of newborn young neurons. These results suggest that REM sleep plays an essential role in training-induced cell proliferation and the fate determination of the newly generated cells toward the neuronal lineage.
Collapse
Affiliation(s)
- Shweta Tripathi
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Sushil K. Jha
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| |
Collapse
|
34
|
Effect of chronic sleep deprivation and sleep recovery on hippocampal CA3 neurons, spatial memory and anxiety-like behavior in rats. Neurobiol Learn Mem 2021; 187:107559. [PMID: 34808338 DOI: 10.1016/j.nlm.2021.107559] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 11/12/2021] [Accepted: 11/14/2021] [Indexed: 11/24/2022]
Abstract
Sleep deprivation-induced degenerative changes in the brain lead to the impairment of memory, anxiety, and quality of life. Several studies have reported the effects of sleep deprivation on CA1 and dentate gyrus regions of the hippocampus; in contrast, there is less known about the impact of chronic sleep deprivation (CSD) and sleep recovery on CA3 neurons and behavior. Hence, the present study aimed to understand the effect of CSD and sleep recovery on hippocampal CA3 neurons and spatial memory, and anxiety-like behavior in rats. Sixty male rats (Sprague Dawley) were grouped as control, environmental control (EC), CSD, 5 days sleep recovery (CSD + 5D SR), and 21 days sleep recovery (CSD + 21D SR). CSD, CSD + 5D SR and, CSD + 21D SR group rats were sleep deprived for 21 days (18 h/day). After CSD, the CSD + 5D SR and CSD + 21D SR rats were sleep recovered for 5- and 21-days respectively. Oxidative stress, dendritic arborization of CA3 neurons, spatial memory, and anxiety-like behavior was assessed. Spatial memory, basal, and apical dendritic branching points/intersections in hippocampal CA3 neurons were reduced, and anxiety-like behavior and oxidative stress increased significantly in the CSD group compared to control (p < 0.001). The CSD + 21D SR showed a significant improvement in spatial memory, reduction in anxiety-like behavior, and oxidative stress when compared to the CSD group (p < 0.05). The basal and apical dendritic branching points/intersections in hippocampal CA3 neurons were increased after CSD + 21D SR, however, it was not significant (p > 0.05). Even though the CSD + 21D SR showed a significant improvement in all the parameters, it did not reach the control level. There was an improvement in all the parameters after CSD + 5D SR but this was not significant compared to the CSD group (p > 0.05). Overall results indicate that the CSD-induced impairment of spatial memory and anxiety-like behavior was associated with oxidative stress and reduced dendritic arborization of hippocampal CA3 neurons. The CSD + 21D SR significantly reduced the damage caused by CSD, but it was not sufficient to reach the control level.
Collapse
|
35
|
Piber D. The role of sleep disturbance and inflammation for spatial memory. Brain Behav Immun Health 2021; 17:100333. [PMID: 34589818 PMCID: PMC8474561 DOI: 10.1016/j.bbih.2021.100333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 11/09/2022] Open
Abstract
Spatial memory is a brain function involved in multiple behaviors such as planning a route or recalling an object's location. The formation of spatial memory relies on the homeostasis of various biological systems, including healthy sleep and a well-functioning immune system. While sleep is thought to promote the stabilization and storage of spatial memories, considerable evidence shows that the immune system modulates neuronal processes underlying spatial memory such as hippocampal neuroplasticity, long-term potentiation, and neurogenesis. Conversely, when sleep is disturbed and/or states of heightened immune activation occur, hippocampal regulatory pathways are altered, which - on a behavioral level - may result in spatial memory impairments. In this Brief Review, I summarize how sleep and the immune system contribute to spatial memory processes. In addition, I present emerging evidence suggesting that sleep disturbance and inflammation might jointly impair spatial memory. Finally, potentials of integrated strategies that target sleep disturbance and inflammation to possibly mitigate risk for spatial memory impairment are discussed.
Collapse
Affiliation(s)
- Dominique Piber
- Department of Psychiatry, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Hindenburgdamm 30, 12203, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, USA
| |
Collapse
|
36
|
Arkell D, Groves I, Wood ER, Hardt O. The Black Box effect: sensory stimulation after learning interferes with the retention of long-term object location memory in rats. ACTA ACUST UNITED AC 2021; 28:390-399. [PMID: 34526383 PMCID: PMC8456983 DOI: 10.1101/lm.053256.120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 07/06/2021] [Indexed: 11/29/2022]
Abstract
Reducing sensory experiences during the period that immediately follows learning improves long-term memory retention in healthy humans, and even preserves memory in patients with amnesia. To date, it is entirely unclear why this is the case, and identifying the neurobiological mechanisms underpinning this effect requires suitable animal models, which are currently lacking. Here, we describe a straightforward experimental procedure in rats that future studies can use to directly address this issue. Using this method, we replicated the central findings on quiet wakefulness obtained in humans: We show that rats that spent 1 h alone in a familiar dark and quiet chamber (the Black Box) after exploring two objects in an open field expressed long-term memory for the object locations 6 h later, while rats that instead directly went back into their home cage with their cage mates did not. We discovered that both visual stimulation and being together with conspecifics contributed to the memory loss in the home cage, as exposing rats either to light or to a cage mate in the Black Box was sufficient to disrupt memory for object locations. Our results suggest that in both rats and humans, everyday sensory experiences that normally follow learning in natural settings can interfere with processes that promote long-term memory retention, thereby causing forgetting in form of retroactive interference. The processes involved in this effect are not sleep-dependent because we prevented sleep in periods of reduced sensory experience. Our findings, which also have implications for research practices, describe a potentially useful method to study the neurobiological mechanisms that might explain why normal sensory processing after learning impairs memory both in healthy humans and in patients suffering from amnesia.
Collapse
Affiliation(s)
- Daisy Arkell
- Centre for Discovery Brain Science, School of Medicine, The University of Edinburgh, Edingurgh, Scotland EH8 9XD, United Kingdom.,The Simons Initiative for the Developing Brain, The Patrick Wild Centre, The University of Edinburgh, Edingurgh, Scotland EH8 9XD, United Kingdom
| | - Isabelle Groves
- Department of Psychology, McGill University, Montréal, Quebec H3A 1G1, Canada
| | - Emma R Wood
- Centre for Discovery Brain Science, School of Medicine, The University of Edinburgh, Edingurgh, Scotland EH8 9XD, United Kingdom.,The Simons Initiative for the Developing Brain, The Patrick Wild Centre, The University of Edinburgh, Edingurgh, Scotland EH8 9XD, United Kingdom
| | - Oliver Hardt
- Centre for Discovery Brain Science, School of Medicine, The University of Edinburgh, Edingurgh, Scotland EH8 9XD, United Kingdom.,The Simons Initiative for the Developing Brain, The Patrick Wild Centre, The University of Edinburgh, Edingurgh, Scotland EH8 9XD, United Kingdom.,Department of Psychology, McGill University, Montréal, Quebec H3A 1G1, Canada
| |
Collapse
|
37
|
Maher A, El Sayed N, Nafea H, Gad M. Rolipram rescues memory consolidation deficits caused by sleep deprivation: Implication of the cAMP/PKA and cAMP/Epac pathways. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 21:631-639. [PMID: 34397335 DOI: 10.2174/1871527320666210816105144] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Over the last few years, the number of people suffering from sleeping disorders has increased significantly despite negative effects on cognition and an association with brain inflammation. OBJECTIVES We assessed memory deficits caused by sleep deprivation (SD) to determine the therapeutic effect of phosphodiesterase 4 (PDE4) inhibitors on SD-induced memory deficits and to investigate whether the modulation of memory deficits by PDE4 inhibitors is mediated by a protein kinase A (PKA)-independent pathway in conjunction with a PKA-dependent pathway. METHODS Adult male mice were divided into four groups. Three SD groups were deprived of Rapid eye movement (REM) sleep for 12 h a day for six consecutive days. They were tested daily in the Morris water maze to evaluate learning and memory. One of the SD groups was injected with a PDE4 inhibitor, rolipram (1 mg/kg ip), whereas another had rolipram co-administered with chlorogenic acid (CHA, 20 mg/kg ip), an inhibitor of PKA. After 6 days, the mice were sacrificed, and the hippocampi were evaluated for cyclic AMP (cAMP) and nuclear factor Nrf-2 levels. The hippocampal expression of PKA, phosphorylated cAMP response element-binding protein (CREB), and phosphorylated glycogen synthase 3β (Ser389) were also evaluated. RESULTS SD caused a significant decrease in cAMP levels in the brain and had a detrimental effect on learning and memory. The administration of rolipram or rolipram+CHA resulted in an improvement in cognitive function. CONCLUSION The present study provides evidence that restoration of memory with PDE4 inhibitors occurs through a dual mechanism involving the PKA and Epac pathways.
Collapse
Affiliation(s)
- Ahmed Maher
- Biochemistry Department, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Cairo. Egypt
| | - Nesrine El Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University. Egypt
| | - Heba Nafea
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), Cairo. Egypt
| | - Mohamed Gad
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), Cairo. Egypt
| |
Collapse
|
38
|
Zhou HR, Wu JR, Bei L, Wang BX, Xu H, Wang JT, Ma SX. Hydroalcoholic extract from Abelmoschus manihot (Linn.) Medicus flower reverses sleep deprivation-evoked learning and memory deficit. Food Funct 2021; 11:8978-8986. [PMID: 33001073 DOI: 10.1039/d0fo02158j] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Previous researches have indicated that sleep plays a vital role in cognitive functions. Sleep deprivation (SD) causes learning and memory damage, which is associated with oxidative stress. This study was performed to investigate the neuroprotective effects of an extract of Abelmoschus manihot flower (EAM) against memory deficit induced by SD in mice. The SD model was evoked by multiple platform method for 5 days, successively. The learning and memory-improving effects of EAM were assessed by behavioral trials and the underlying mechanism was investigated by measuring the oxidative stress alteration. Our findings indicated that the SD-induced memory deficit and the EAM treatment improved the cognitive functions of mice in the object location recognition test and passive avoidance task. In addition, EAM effectively improved the activities of the antioxidant enzyme, decreased the content of malondialdehyde (MDA), and restored the protein expression of the brain-derived neurotrophic factor (BDNF), tyrosine kinase B (TrkB) and glutamate receptor 1 (GluR1) in brain tissues. In conclusion, EAM could improve the SD-evoked learning and memory impairments. The possible underlying mechanisms of EAM may be related to its antioxidant capacity and enhanced BDNF/TrkB/GluR1 levels in the hippocampal memory.
Collapse
Affiliation(s)
- Hai-Rui Zhou
- Basic medical college of Jiamusi University, Heilongjiang 154007, China.
| | - Jing-Ru Wu
- Basic medical college of Jiamusi University, Heilongjiang 154007, China.
| | - Lei Bei
- Basic medical college of Jiamusi University, Heilongjiang 154007, China.
| | - Bai-Xin Wang
- Basic medical college of Jiamusi University, Heilongjiang 154007, China.
| | - Hui Xu
- Basic medical college of Jiamusi University, Heilongjiang 154007, China.
| | - Jing-Tao Wang
- Basic medical college of Jiamusi University, Heilongjiang 154007, China.
| | - Shu-Xia Ma
- Basic medical college of Jiamusi University, Heilongjiang 154007, China.
| |
Collapse
|
39
|
Wang J, Li Q, Huang Q, Lv M, Li P, Dai J, Zhou M, Xu J, Zhang F, Gao J. Washed Microbiota Transplantation Accelerates the Recovery of Abnormal Changes by Light-Induced Stress in Tree Shrews. Front Cell Infect Microbiol 2021; 11:685019. [PMID: 34249778 PMCID: PMC8262326 DOI: 10.3389/fcimb.2021.685019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/21/2021] [Indexed: 12/12/2022] Open
Abstract
The gut and brain interact constantly in a complex fashion. Its intricacy and intrigue is progressively being revealed in the study of the "gut-brain axis". Among many factors, abnormal light exposure is a potential powerful stressor, which is becoming ever more pervasive in our modern society. However, little is known about how stress, induced by staying up late by light, affects the gut-brain axis. We addressed this question by extending the normal circadian light for four hours at night in fifteen male tree shrews to simulate the pattern of staying up late in humans. The behavior, biochemical tests, microbiota dynamics, and brain structure of tree shrews were evaluated. The simple prolongation of light in the environment resulted in substantial changes of body weight loss, behavioral differences, total sleep time reduction, and an increased level of urine cortisol. These alterations were rescued by the treatment of either ketamine or washed microbiota transplantation (WMT). Importantly, the sustainability of WMT effect was better than that of ketamine. Magnetic Resonance Imaging analysis indicated that ketamine acted on the hippocampus and thalamus, and WMT mainly affected the piriform cortex and lateral geniculate nucleus. In conclusion, long-term light stimulation could change the behaviors, composition of gut microbiota and brain structure in tree shrews. Targeting microbiota thus certainly holds promise as a treatment for neuropsychiatric disorders, including but not limited to stress-related diseases.
Collapse
Affiliation(s)
- Jing Wang
- Department of Neurobiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China.,Visual Cognition Laboratory, Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Qianqian Li
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Lab of Holistic Integrative Enterology, Nanjing Medical University, Nanjing, China
| | - Qi Huang
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Meng Lv
- Animal Core Facility of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Pan Li
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Lab of Holistic Integrative Enterology, Nanjing Medical University, Nanjing, China
| | - Jing Dai
- Department of Neurobiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Minjie Zhou
- Department of Neurobiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Jialu Xu
- Department of Neurobiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Faming Zhang
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Lab of Holistic Integrative Enterology, Nanjing Medical University, Nanjing, China
| | - Jun Gao
- Department of Neurobiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China.,Department of Rehabilitation Medicine, Jiangsu Shengze Hospital Affiliated to Nanjing Medical University, Nanjing Medical University, Nanjing, China
| |
Collapse
|
40
|
Sharma B, Sengupta T, Chandra Vishwakarma L, Akhtar N, Mallick HN. Muscle temperature is least altered during total sleep deprivation in rats. J Therm Biol 2021; 98:102910. [PMID: 34016337 DOI: 10.1016/j.jtherbio.2021.102910] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/11/2021] [Accepted: 03/11/2021] [Indexed: 11/27/2022]
Abstract
It has often been said that the brain is mostly benefitted from sleep. To understand the importance of sleep, extensive studies on other organs are too required. One such unexplored area is the understanding of muscle physiology during the sleep-wake cycle. Changes in muscle tone with different sleep phases are evident from the rapid eye movement sleep muscle atonia. There is variation in brain and body temperature during sleep stages, the brain temperature being higher during rapid eye movement sleep than slow-wave sleep. However, the change in muscle temperature with different sleep stages is not known. In this study, we have implanted pre-calibrated K-type thermocouples in the hypothalamus and the dorsal nuchal muscle, and a peritoneal transmitter to monitor the hypothalamic, muscle, and body temperature respectively in rats during 24 h sleep-wake cycle. The changes in muscle, body, and hypothalamic temperature during total sleep deprivation were also monitored. During normal sleep-wake stages, the temperature in the decreasing order was that of the hypothalamus, body, and muscle. Total sleep deprivation by gentle handling caused a significant increase in hypothalamic and body temperature, while there was least change in the muscle temperature. The circadian rhythm of the hypothalamic and body temperature in the sleep-deprived rats was disrupted, while the same was preserved in the muscle temperature. The results of our study show that muscle atonia during rapid eye movement sleep is a physiologically regulated thermally quiescent muscle state offering a conducive environment for muscle rest and repair.
Collapse
Affiliation(s)
- Binney Sharma
- Department of Physiology, All India Institute of Medical Sciences, New Delhi, 110029, India.
| | - Trina Sengupta
- Department of Physiology, All India Institute of Medical Sciences, New Delhi, 110029, India; Department of Physiology, All India Institute of Medical Sciences, Jodhpur, 342005, India.
| | - Lal Chandra Vishwakarma
- Department of Physiology, All India Institute of Medical Sciences, New Delhi, 110029, India.
| | - Nasreen Akhtar
- Department of Physiology, All India Institute of Medical Sciences, New Delhi, 110029, India.
| | - Hruda Nanda Mallick
- Department of Physiology, All India Institute of Medical Sciences, New Delhi, 110029, India; Department of Physiology, Faculty of Medicine & Health Sciences, SGT University, Gurgaon, Haryana, 122505, India.
| |
Collapse
|
41
|
Rynes ML, Surinach DA, Linn S, Laroque M, Rajendran V, Dominguez J, Hadjistamoulou O, Navabi ZS, Ghanbari L, Johnson GW, Nazari M, Mohajerani MH, Kodandaramaiah SB. Miniaturized head-mounted microscope for whole-cortex mesoscale imaging in freely behaving mice. Nat Methods 2021; 18:417-425. [PMID: 33820987 PMCID: PMC8034419 DOI: 10.1038/s41592-021-01104-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 02/18/2021] [Accepted: 02/26/2021] [Indexed: 12/28/2022]
Abstract
The advent of genetically encoded calcium indicators, along with surgical preparations such as thinned skulls or refractive index matched skulls, have enabled mesoscale cortical activity imaging in head-fixed mice. However, neural activity during unrestrained behavior substantially differs from neural activity in head-fixed animals. For whole-cortex imaging in freely behaving mice, we here present the “mini-mScope,” a wide-field, miniaturized, and head-mounted fluorescence microscope compatible with transparent polymer skull preparations. With a field of view of 8 mm x 10 mm and weighing less than 4 g, the mini-mScope can image most of the mouse dorsal cortex with resolution ranging from 39 to 56 μm. We have used the mini-mScope to record mesoscale calcium activity across the dorsal cortex during sensory-evoked stimuli, open field behaviors, social interactions, and transitions from wakefulness to sleep.
Collapse
Affiliation(s)
- Mathew L Rynes
- Department of Biomedical Engineering, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Daniel A Surinach
- Department of Mechanical Engineering, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Samantha Linn
- Department of Mechanical Engineering, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Michael Laroque
- Schools of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Vijay Rajendran
- Department of Mechanical Engineering, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Judith Dominguez
- Department of Mechanical Engineering, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Orestes Hadjistamoulou
- Department of Mechanical Engineering, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Zahra S Navabi
- Department of Mechanical Engineering, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Leila Ghanbari
- Department of Mechanical Engineering, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Gregory W Johnson
- Department of Mechanical Engineering, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Mojtaba Nazari
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Majid H Mohajerani
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Suhasa B Kodandaramaiah
- Department of Biomedical Engineering, University of Minnesota, Twin Cities, Minneapolis, MN, USA. .,Department of Mechanical Engineering, University of Minnesota, Twin Cities, Minneapolis, MN, USA. .,Department of Neuroscience, University of Minnesota, Twin Cities, Minneapolis, MN, USA.
| |
Collapse
|
42
|
Ahmadi-Soleimani SM, Azizi H, Abbasi-Mazar A. Intermittent REM sleep deprivation attenuates the development of morphine tolerance and dependence in male rats. Neurosci Lett 2021; 748:135735. [PMID: 33592307 DOI: 10.1016/j.neulet.2021.135735] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 02/04/2023]
Abstract
Opioid agonists are used in clinic for pain management, however this application is challenged by development of tolerance and dependence following prolonged exposure. Various approaches have been suggested to address this concern, however, there is still no consensus among the researchers. Neural processing of sleep and nociception are co-regulated through shared brain regions having bidirectional interplays. Thus, we aimed to investigate whether application of REM sleep deprivation (REM-SD) could affect morphine analgesic tolerance and dependence. To this end, adult male rats underwent sleep deprivation during light and dark phases (LSD and DSD, respectively) using the inverted flower pot method and then tolerance and dependence was induced by repeated injection of morphine for 7 days (10 mg/kg, daily, i.p.). Results indicated that REM-SD delays the development of tolerance to morphine during both phases; however this effect was more potent following LSD. Moreover, LSD decreased the baseline thermal threshold and total withdrawal score. One possible hypothesis for our observations is REM-SD-induced attenuation of orexin system which is still controversial among the researchers. Other stronger possibilities might be down-regulation of opioid receptors in response to sleep loss experience. Finally, it seems that modification of sleep periods may assist to decrease the severity of opioid tolerance and dependence.
Collapse
Affiliation(s)
- S Mohammad Ahmadi-Soleimani
- Deparment of Physiology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran.
| | - Hossein Azizi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Alireza Abbasi-Mazar
- Student Research Committee, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| |
Collapse
|
43
|
Jung T, Noh J. Alteration of fear behaviors in sleep-deprived adolescent rats: increased fear expression and delayed fear extinction. Anim Cells Syst (Seoul) 2021; 25:83-92. [PMID: 34234889 PMCID: PMC8118405 DOI: 10.1080/19768354.2021.1902854] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Disruption of sleep due to acute or chronic stress can lead to changes in emotional memory processing. Sleep disturbances are highly prevalent in post-traumatic stress disorder (PTSD), but still, the contribution of sleep deprivation on the susceptibility to PTSD has received little attention. To determine whether rapid eye movement sleep deprivation (SD) alters the development of fear expression or fear-associated memory impairment in adolescent rats, we performed animal emotional behavior tests using an SD animal model with the flowerpot technique. SD rats showed an increase in locomotor activity frequency and a decrease in sucrose consumption compared to control rats. An increase in freezing behavior during shock trials was observed in SD rats. Noticeably, it was observed that when applying the SD condition after fear stimuli exposure, fear extinction was delayed more in SD rats than in control rats. Overall, these results indicate that SD in adolescent rats leads to increased locomotor activity and anhedonic behavior, as well as increased fear expression and delayed fear extinction, suggesting that SD would lead to increased severity of PTSD-like phenotype.
Collapse
Affiliation(s)
- Taesub Jung
- Department of Science Education, Dankook University, Yongin-si, Republic of Korea
| | - Jihyun Noh
- Department of Science Education, Dankook University, Yongin-si, Republic of Korea
| |
Collapse
|
44
|
Chronic sleep fragmentation enhances habenula cholinergic neural activity. Mol Psychiatry 2021; 26:941-954. [PMID: 30980042 PMCID: PMC6790161 DOI: 10.1038/s41380-019-0419-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 02/21/2019] [Accepted: 03/26/2019] [Indexed: 01/25/2023]
Abstract
Sleep is essential to emotional health. Sleep disturbance, particularly REM sleep disturbance, profoundly impacts emotion regulation, but the underlying neural mechanisms remain elusive. Here we show that chronic REM sleep disturbance, achieved in mice by chronic sleep fragmentation (SF), enhanced neural activity in the medial habenula (mHb), a brain region increasingly implicated in negative affect. Specifically, after a 5-day SF procedure that selectively fragmented REM sleep, cholinergic output neurons (ChNs) in the mHb exhibited increased spontaneous firing rate and enhanced firing regularity in brain slices. The SF-induced firing changes remained intact upon inhibition of glutamate, GABA, acetylcholine, and histamine receptors, suggesting cell-autonomous mechanisms independent of synaptic transmissions. Moreover, the SF-induced hyperactivity was not because of enhanced intrinsic membrane excitability, but was accompanied by depolarized resting membrane potential in mHb ChNs. Furthermore, inhibition of TASK-3 (KCNK9) channels, a subtype of two-pore domain K+ channels, mimicked the SF effects by increasing the firing rate and regularity, as well as depolarizing the resting membrane potential in mHb ChNs in control-sleep mice. These effects of TASK-3 inhibition were absent in SF mice, suggesting reduced TASK-3 activity following SF. By contrast, inhibition of small-conductance Ca2+-activated K+ (SK) channels did not produce similar effects. Thus, SF compromised TASK-3 function in mHb ChNs, which likely led to depolarized resting membrane potential and increased spontaneous firing. These results not only demonstrate that selective REM sleep disturbance leads to hyperactivity of mHb ChNs, but also identify a key molecular substrate through which REM sleep disturbance may alter affect regulation.
Collapse
|
45
|
Tuan LH, Tsao CY, Lee LJH, Lee LJ. Voluntary exercise ameliorates synaptic pruning deficits in sleep-deprived adolescent mice. Brain Behav Immun 2021; 93:96-110. [PMID: 33358980 DOI: 10.1016/j.bbi.2020.12.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/17/2020] [Accepted: 12/16/2020] [Indexed: 11/26/2022] Open
Abstract
Adolescence is a critical period for brain development and adequate sleep during this period is essential for physical function and mental health. Emerging evidence has detailed the neurological impacts of sleep insufficiency on adolescents, as was unveiled by our previous study, microglia, one of the crucial contributors to synaptic pruning, is functionally disrupted by lack of sleep. Here, we provided evidence featuring the protective effect and the underlying mechanisms of voluntary exercise (VE) on microglial functions in an adolescent 72 h sleep deprivation (SD) model. We identified that the aberrant hippocampal neuronal activity and impaired short-term memory performance in sleep-deprived mice were prevented by 11 days of VE. VE significantly normalized the SD-induced dendritic spine increment and maintained the microglial phagocytic ability in sleep-deprived mice. Moreover, we found that the amendment of the noradrenergic signal in the central nervous system may explain the preventative effects of VE on the abnormalities of microglial and neuronal functions caused by SD. These data suggested that VE may confer protection to the microglia-mediated synaptic pruning in the sleep-deprived adolescent brains. Therefore, physical exercise could be a beneficial health practice for the adolescents that copes the adverse influence of inevitable sleep insufficiency.
Collapse
Affiliation(s)
- Li-Heng Tuan
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University, Taipei, Taiwan, ROC
| | - Chih-Yu Tsao
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University, Taipei, Taiwan, ROC
| | - Lukas Jyuhn-Hsiarn Lee
- Division of Environmental Health and Occupational Medicine, National Health Research Institutes, Miaoli, Taiwan, ROC
| | - Li-Jen Lee
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University, Taipei, Taiwan, ROC; Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan, ROC; Institute of Brain and Mind Sciences, National Taiwan University, Taipei, Taiwan, ROC.
| |
Collapse
|
46
|
Contributions of animal models of cognitive disorders to neuropsychopharmacology. Therapie 2021; 76:87-99. [PMID: 33589315 DOI: 10.1016/j.therap.2021.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 01/30/2021] [Indexed: 12/18/2022]
Abstract
Cognitive disorders and symptoms are key features of many mental and neurological diseases, with a large spectrum of impaired domains. Because of their possible evolution and detrimental functioning impact, they are a major pharmacological target for both symptomatic and disease-modifier drugs, while few cognitive enhancers have been marketed with an insufficient efficiency. It explains the need to model these cognitive disorders beyond the modelization of mental or neurological diseases themselves. According to the experimental strategy used to induce cognitive impairment, three categories of models have been identified: neurotransmission-driven models; pathophysiology-driven models; environment-driven models. These three categories of models reflect different levels of integration of endogenous and exogenous mechanisms underlying cognitive disorders in humans. Their comprehensive knowledge and illustration of their pharmacological modulation could help to propose a renewing strategy of drug development in central nervous system (CNS) field at a time when the academic and industrial invest seems to be declining despite the medical and social burden of brain diseases.
Collapse
|
47
|
Jiang N, Li YJ, Wang MD, Huang H, Chen S, Li Y, Qu L, Wang F, Liu X, Wang Q. The Cognitive-Enhancing Effects of Dendrobium nobile Lindl Extract in Sleep Deprivation-Induced Amnesic Mice. Front Psychiatry 2021; 12:596017. [PMID: 35126189 PMCID: PMC8808596 DOI: 10.3389/fpsyt.2021.596017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/27/2021] [Indexed: 11/24/2022] Open
Abstract
Chronic sleep deprivation (SD) causes neurological and neurodegenerative dysfunction including learning and memory deficit. The orchid Dendrobium nobile Lindl (DNL), is widely used as a Yin tonic and medicinal food throughout Asia, and has many reported pharmacological effects. This study focused on the cognitive-enhancing effects of DNL in sleep deprivation-induced amnesia in mice and its biochemical mechanisms. Our results showed that the mice displayed significant cognitive deficits after 2-week SD while treatment with the extract of DNL prevented these impairments. In the novel object recognition and object location recognition tasks, a significant increase in the discrimination index was observed in DNL-treated (200 and 400 mg/kg) mice. In the MWM test, DNL (200 and 400 mg/kg) treatment shorten the prolongation of latency and increased the crossing numbers compared with SD mice. The biochemical analysis of brain tissue showed a decrease in NE, dismutase (T-SOD) and catalase (CAT) activity and an increase in 5-HT and malondialdehyde (MDA) concentration after the treatment with DNL in mice. Our findings indicated that DNL exerted a positive effect in preventing and improving cognitive impairment induced by SD, which may be mediated via the regulation of neurotransmitters and alleviation of oxidative stress.
Collapse
Affiliation(s)
- Ning Jiang
- Sino-Portugal TCM International Cooperation Center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China.,Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu-Jiao Li
- Sino-Portugal TCM International Cooperation Center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Meng-di Wang
- Sino-Portugal TCM International Cooperation Center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Hong Huang
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shanguang Chen
- National Key Laboratory of Human Factors Engineering/State Key Laboratory of Space Medicine Fundamentals and Application, Chinese Astronaut Research and Training Center, Beijing, China
| | - Yinghui Li
- National Key Laboratory of Human Factors Engineering/State Key Laboratory of Space Medicine Fundamentals and Application, Chinese Astronaut Research and Training Center, Beijing, China
| | - Lina Qu
- National Key Laboratory of Human Factors Engineering/State Key Laboratory of Space Medicine Fundamentals and Application, Chinese Astronaut Research and Training Center, Beijing, China
| | - Fengzhong Wang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Xinmin Liu
- Sino-Portugal TCM International Cooperation Center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China.,Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiong Wang
- Sino-Portugal TCM International Cooperation Center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China.,Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| |
Collapse
|
48
|
Sleep loss mediates the effect of stress on nitrergic signaling in female mice. Neurosci Lett 2020; 740:135362. [PMID: 33166635 PMCID: PMC10084941 DOI: 10.1016/j.neulet.2020.135362] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 08/12/2020] [Accepted: 09/03/2020] [Indexed: 01/04/2023]
Abstract
Nitric oxide (NO) has been implicated as an important neurotransmitter in stress responses and sleep regulatory processes. However, the role of NO in the relationship between stress and sleep remains unclear. The medial septum (MS) and vertical diagonal band (VDB), regions of the basal forebrain involved in sleep regulation, contain nitric oxide synthase (NOS) producing neurons. Additionally, NOS neurons in the dorsal raphe nucleus (DRN) encode information about stress duration. The role of nitrergic neurons in these regions in subserving sex-specific responses to stress and sleep loss has yet to be elucidated. In this study, NADPH-d, an index of NOS activity, was used to examine the effects of acute restraint stress and sleep loss on NOS activity in the MS, VDB, and DRN. We show that NOS activity in response to restraint stress, total sleep deprivation (TSD), and partial sleep restriction (PSR) differs based on sex and region. Initial analysis showed no effect of restraint stress or TSD on NOS activity in the basal forebrain. However, investigation of each sex separately revealed that restraint stress and TSD significantly decrease NOS activity in the MS of females, but not males. Interestingly, the difference in NOS activity between restraint stress and TSD in females was not significant. Furthermore, PSR was not sufficient to affect NOS activity in males or females. These data suggest that restraint stress and sleep loss regulate NOS activation in a sex-dependent manner, and that the NOS stress response in females may be mediated by sleep loss.
Collapse
|
49
|
Brice KN, Hagen CW, Peterman JL, Figg JW, Braden PN, Chumley MJ, Boehm GW. Chronic sleep restriction increases soluble hippocampal Aβ-42 and impairs cognitive performance. Physiol Behav 2020; 226:113128. [PMID: 32791178 DOI: 10.1016/j.physbeh.2020.113128] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 11/17/2022]
Abstract
Currently, over 44 million people worldwide suffer from Alzheimer's disease (AD). A common feature of AD is disrupted sleep. Sleep is essential for many psychological and physiological functions, though 35.3% of adults report getting less than 7 hours per night. The present research examined whether chronic sleep restriction would elevate hippocampal amyloid-beta1-42 expression or alter cognitive ability in adult C57BL/6 mice. Chronic sleep restriction was associated with cognitive impairment and increased hippocampal amyloid-beta. Thus, chronic sleep loss may have a detrimental effect upon cognitive function, in part, via increasing amyloid-beta levels in the hippocampus, even in non-genetically modified mice.
Collapse
Affiliation(s)
- Kelly N Brice
- Texas Christian University, Department of Psychology, 2955 South University Drive, Fort Worth, TX 76109, USA
| | - Christopher W Hagen
- Texas Christian University, Department of Biology, 2955 South University Drive, Fort Worth, TX 76109, USA
| | - Julia L Peterman
- Texas Christian University, Department of Psychology, 2955 South University Drive, Fort Worth, TX 76109, USA
| | - John W Figg
- Texas Christian University, Department of Biology, 2955 South University Drive, Fort Worth, TX 76109, USA
| | - Paige N Braden
- Texas Christian University, Department of Psychology, 2955 South University Drive, Fort Worth, TX 76109, USA
| | - Michael J Chumley
- Texas Christian University, Department of Biology, 2955 South University Drive, Fort Worth, TX 76109, USA
| | - Gary W Boehm
- Texas Christian University, Department of Psychology, 2955 South University Drive, Fort Worth, TX 76109, USA.
| |
Collapse
|
50
|
Hu Y, Korovaichuk A, Astiz M, Schroeder H, Islam R, Barrenetxea J, Fischer A, Oster H, Bringmann H. Functional Divergence of Mammalian TFAP2a and TFAP2b Transcription Factors for Bidirectional Sleep Control. Genetics 2020; 216:735-752. [PMID: 32769099 PMCID: PMC7648577 DOI: 10.1534/genetics.120.303533] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/20/2020] [Indexed: 11/18/2022] Open
Abstract
Sleep is a conserved behavioral state. Invertebrates typically show quiet sleep, whereas in mammals, sleep consists of periods of nonrapid-eye-movement sleep (NREMS) and REM sleep (REMS). We previously found that the transcription factor AP-2 promotes sleep in Caenorhabditiselegans and Drosophila In mammals, several paralogous AP-2 transcription factors exist. Sleep-controlling genes are often conserved. However, little is known about how sleep genes evolved from controlling simpler types of sleep to govern complex mammalian sleep. Here, we studied the roles of Tfap2a and Tfap2b in sleep control in mice. Consistent with our results from C. elegans and Drosophila, the AP-2 transcription factors Tfap2a and Tfap2b also control sleep in mice. Surprisingly, however, the two AP-2 paralogs play contrary roles in sleep control. Tfap2a reduction of function causes stronger delta and theta power in both baseline and homeostasis analysis, thus indicating increased sleep quality, but did not affect sleep quantity. By contrast, Tfap2b reduction of function decreased NREM sleep time specifically during the dark phase, reduced NREMS and REMS power, and caused a weaker response to sleep deprivation. Consistent with the observed signatures of decreased sleep quality, stress resistance and memory were impaired in Tfap2b mutant animals. Also, the circadian period was slightly shortened. Taken together, AP-2 transcription factors control sleep behavior also in mice, but the role of the AP-2 genes functionally diversified to allow for a bidirectional control of sleep quality. Divergence of AP-2 transcription factors might perhaps have supported the evolution of more complex types of sleep.
Collapse
Affiliation(s)
- Yang Hu
- Max Planck Research Group "Sleep and Waking", Max Planck Institute for Biophysical Chemistry, Göttingen 37077, Germany
| | - Alejandra Korovaichuk
- Max Planck Research Group "Sleep and Waking", Max Planck Institute for Biophysical Chemistry, Göttingen 37077, Germany
| | - Mariana Astiz
- Institute of Neurobiology, University of Lübeck, 23562, Germany
| | - Henning Schroeder
- German Center for Neurodegenerative Diseases, Göttingen 37075, Germany
| | - Rezaul Islam
- German Center for Neurodegenerative Diseases, Göttingen 37075, Germany
| | - Jon Barrenetxea
- Max Planck Research Group "Sleep and Waking", Max Planck Institute for Biophysical Chemistry, Göttingen 37077, Germany
| | - Andre Fischer
- German Center for Neurodegenerative Diseases, Göttingen 37075, Germany
- Department for Psychiatry and Psychotherapy, University Medical Center, Göttingen 37075, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, 37073, Germany
| | - Henrik Oster
- Institute of Neurobiology, University of Lübeck, 23562, Germany
| | - Henrik Bringmann
- Max Planck Research Group "Sleep and Waking", Max Planck Institute for Biophysical Chemistry, Göttingen 37077, Germany
- Department of Animal Physiology/Neurophysiology, Philipps University Marburg, Marburg 35043, Germany
- BIOTEC of the Technical University Dresden, Dresden 01307, Germany
| |
Collapse
|