1
|
Zhao Y, Fang R, Bian H, Zhang K, Yu S, Wang Y, Huang L. Comparative analysis of sleep deprivation models: Impacts on sleep architecture, emotional state, cognitive function, and biochemical indicators in male rats. Behav Brain Res 2025; 482:115451. [PMID: 39889831 DOI: 10.1016/j.bbr.2025.115451] [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: 10/21/2024] [Revised: 01/05/2025] [Accepted: 01/22/2025] [Indexed: 02/03/2025]
Abstract
Sleep deprivation significantly affects both physiological and psychological health, with various experimental models used to study these impacts. This study compares three sleep deprivation models-Modified Multiple Platform Method (MMPM), treadmill method, and p-chlorophenylalanine (PCPA) method-on key physiological, cognitive, and emotional parameters in male Sprague-Dawley rats. The rats were subjected to 72 hours of sleep deprivation using these methods, followed by behavioral, cognitive, physiological, and biochemical assessments. Results indicated that the treadmill and PCPA methods led to significant reductions in both NREM and REM sleep (P < 0.05), with the PCPA method showing the most severe emotional effects, including heightened anxiety and depressive behaviors (P < 0.001). Cognitive impairments were most pronounced in the MMPM and treadmill groups (P < 0.01). All sleep deprivation models showed signs of autonomic nervous system dysfunction, as reflected by elevated LF/HF ratios in heart rate variability assessments (P < 0.05). Neurochemical analysis revealed reductions in hypothalamic 5-HT, Glu, and GABA levels, with the MMPM and treadmill methods causing more pronounced decreases (P < 0.05). Additionally, IL-2 levels significantly decreased while TNF-α levels increased in sleep-deprived rats compared to controls (P < 0.05). These findings highlight the distinct physiological, emotional, and cognitive impacts of different sleep deprivation models, providing a basis for model selection in future studies.
Collapse
Affiliation(s)
- Yiyang Zhao
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Runchen Fang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Hongsheng Bian
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | | | - Shuang Yu
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yanyan Wang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lili Huang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China.
| |
Collapse
|
2
|
Sun Q, Zhu J, Zhao X, Huang X, Qu W, Tang X, Ma D, Shu Q, Li X. Mettl3-m 6A-NPY axis governing neuron-microglia interaction regulates sleep amount of mice. Cell Discov 2025; 11:10. [PMID: 39905012 PMCID: PMC11794856 DOI: 10.1038/s41421-024-00756-y] [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: 05/14/2024] [Accepted: 11/13/2024] [Indexed: 02/06/2025] Open
Abstract
Sleep behavior is regulated by diverse mechanisms including genetics, neuromodulation and environmental signals. However, it remains completely unknown regarding the roles of epitranscriptomics in regulating sleep behavior. In the present study, we showed that the deficiency of RNA m6A methyltransferase Mettl3 in excitatory neurons specifically induces microglia activation, neuroinflammation and neuronal loss in thalamus of mice. Mettl3 deficiency remarkably disrupts sleep rhythm and reduces the amount of non-rapid eye movement sleep. We also showed that Mettl3 regulates neuropeptide Y (NPY) via m6A modification and Mettl3 conditional knockout (cKO) mice displayed significantly decreased expression of NPY in thalamus. In addition, the dynamic distribution pattern of NPY is observed during wake-sleep cycle in cKO mice. Ectopic expression of Mettl3 and NPY significantly inhibits microglia activation and neuronal loss in thalamus, and restores the disrupted sleep behavior of cKO mice. Collectively, our study has revealed the critical function of Mettl3-m6A-NPY axis in regulating sleep behavior.
Collapse
Affiliation(s)
- Qihang Sun
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jinpiao Zhu
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
- Department of Rehabilitation, Perioperative and Systems Medicine Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
| | - Xingsen Zhao
- Institute of Biotechnology, Xianghu Laboratory, Hangzhou, Zhejiang, China
| | - Xiaoli Huang
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Wenzheng Qu
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Xia Tang
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Daqing Ma
- Department of Rehabilitation, Perioperative and Systems Medicine Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
- Division of Anesthetics, Pain Medicine & Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK.
| | - Qiang Shu
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
| | - Xuekun Li
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
- Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Wang W, Wang Z, Cao J, Dong Y, Chen Y. Melatonin ameliorates chronic sleep deprivation against memory encoding vulnerability: Involvement of synapse regulation via the mitochondrial-dependent redox homeostasis-induced autophagy inhibition. Free Radic Biol Med 2024; 225:398-414. [PMID: 39396581 DOI: 10.1016/j.freeradbiomed.2024.10.279] [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: 07/03/2024] [Revised: 08/13/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Voluntary sleep curtailment is increasingly more rampant in modern society and compromises healthy cognition, including memory, to varying degrees. However, whether memory encoding is impaired after chronic sleep deprivation (CSD) and the underlying molecular mechanisms involved remain unclear. Here, using the mice, we tested the impact of CSD on the encoding abilities of social recognition-dependent memory and object recognition-dependent memory. We found that memory encoding was indeed vulnerable to CSD, while memory retrieval remained unaffected. The hippocampal neurons of mice with memory encoding deficits exhibited significant synapse damage and hyperactive autophagy, which dissipates during regular sleep cycles. This excessive autophagy appeared to be triggered by damage to mitochondrial DNA (mtDNA), resulting from oxidative stress within the mitochondria. The relief at the behavioral and molecular biological levels can be achieved with intraperitoneal injections of the antioxidant compound melatonin. Moreover, our in vitro experiments using HT-22 cells demonstrated that oxidative stress induced by hydrogen peroxide led to oxidative damage, including mtDNA damage, and activation of autophagy. Melatonin treatment effectively countered these effects, restoring redox homeostasis and reducing excessive autophagic activity. Notably, this protective effect was not observed when melatonin was administered as a pre-treatment. Together, our findings reveal the vulnerability of memory encoding during chronic sleep curtailment, which is caused by oxidative stress and consequent enhancement of autophagy, suggest a potential therapeutic strategy for addressing these effects following prolonged wakefulness through melatonin intervention, and reiterate the significance of adequate sleep for memory formation and retention.
Collapse
Affiliation(s)
- Wei Wang
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; National Key Laboratory of Veterinary Public Health and Safety, China Agricultural University, Beijing, 100193, China
| | - Zixu Wang
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; National Key Laboratory of Veterinary Public Health and Safety, China Agricultural University, Beijing, 100193, China
| | - Jing Cao
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; National Key Laboratory of Veterinary Public Health and Safety, China Agricultural University, Beijing, 100193, China
| | - Yulan Dong
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; National Key Laboratory of Veterinary Public Health and Safety, China Agricultural University, Beijing, 100193, China
| | - Yaoxing Chen
- Neurobiology Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; National Key Laboratory of Veterinary Public Health and Safety, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
4
|
Cowen MH, Raizen DM, Hart MP. Structural neuroplasticity after sleep loss modifies behavior and requires neurexin and neuroligin. iScience 2024; 27:109477. [PMID: 38551003 PMCID: PMC10973677 DOI: 10.1016/j.isci.2024.109477] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/05/2023] [Accepted: 03/08/2024] [Indexed: 02/08/2025] Open
Abstract
Structural neuroplasticity (changes in the size, strength, number, and targets of synaptic connections) can be modified by sleep and sleep disruption. However, the causal relationships between genetic perturbations, sleep loss, neuroplasticity, and behavior remain unclear. The C. elegans GABAergic DVB neuron undergoes structural plasticity in adult males in response to adolescent stress, which rewires synaptic connections, alters behavior, and is dependent on conserved autism-associated genes NRXN1/nrx-1 and NLGN3/nlg-1. We find that four methods of sleep deprivation transiently induce DVB neurite extension in day 1 adults and increase the time to spicule protraction, which is the functional and behavioral output of the DVB neuron. Loss of nrx-1 and nlg-1 prevent DVB structural plasticity and behavioral changes at day 1 caused by adolescent sleep loss. Therefore, nrx-1 and nlg-1 mediate the morphologic and behavioral consequences of sleep loss, providing insight into the relationship between sleep, neuroplasticity, behavior, and neurologic disease.
Collapse
Affiliation(s)
- Mara H. Cowen
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David M. Raizen
- Department of Neurology and the Chronobiology and Sleep Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Michael P. Hart
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
5
|
Guo F, Zhang T, Wang C, Xu Z, Chang Y, Zheng M, Fang P, Zhu Y. White matter structural topologic efficiency predicts individual resistance to sleep deprivation. CNS Neurosci Ther 2024; 30:e14349. [PMID: 37408437 PMCID: PMC10848061 DOI: 10.1111/cns.14349] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 06/05/2023] [Accepted: 06/24/2023] [Indexed: 07/07/2023] Open
Abstract
BACKGROUND Sleep deprivation (SD) is commonplace in modern society and there are large individual differences in the vulnerability to SD. We aim to identify the structural network differences based on diffusion tensor imaging (DTI) that contribute to the individual different vulnerability to SD. METHODS The number of psychomotor vigilance task (PVT) lapses was used to classify 49 healthy subjects on the basis of whether they were vulnerable or resistant to SD. DTI and graph theory approaches were used to investigate the topologic organization differences of the brain structural connectome between SD-vulnerable and -resistant individuals. We measured the level of global efficiency and clustering in rich club and non-rich club organizations. RESULTS We demonstrated that participants vulnerable to SD had less global efficiency, network strength, and local efficiency but longer shortest path length compared with participants resistant to SD. Lower efficiency was mainly distributed in the right insula, bilateral thalamus, bilateral frontal, temporal, and temporal lobes. Furthermore, a disrupted subnetwork was observed that consisted of widespread connections. Moreover, the vulnerable group showed significantly decreased strength of the rich club compared with the resistant group. The strength of rich club connectivity was found to be correlated negatively with PVT performance (r = -0.395, p = 0.005). We further tested the reliability of the results. CONCLUSION The findings revealed that individual differences in resistance to SD are related to disrupted topologic efficiency connectome pattern, and our study may provide potential connectome-based biomarkers for the early detection of the vulnerable degree to SD.
Collapse
Affiliation(s)
- Fan Guo
- Department of Radiology, Xijing HospitalAir Force Medical UniversityXi'anChina
| | - Tian Zhang
- Department of Military Medical PsychologyAir Force Medical UniversityXi'anChina
| | - Chen Wang
- Department of Radiology, Xijing HospitalAir Force Medical UniversityXi'anChina
| | - Ziliang Xu
- Department of Radiology, Xijing HospitalAir Force Medical UniversityXi'anChina
| | - Yingjuan Chang
- Department of Radiology, Xijing HospitalAir Force Medical UniversityXi'anChina
| | - Minwen Zheng
- Department of Radiology, Xijing HospitalAir Force Medical UniversityXi'anChina
| | - Peng Fang
- Department of Military Medical PsychologyAir Force Medical UniversityXi'anChina
| | - Yuanqiang Zhu
- Department of Radiology, Xijing HospitalAir Force Medical UniversityXi'anChina
| |
Collapse
|
6
|
Shi S, Zhang M, Xie W, Ju P, Chen N, Wang F, Lyu D, Wang M, Hong W. Sleep deprivation alleviates depression-like behaviors in mice via inhibiting immune and inflammatory pathways and improving neuroplasticity. J Affect Disord 2023; 340:100-112. [PMID: 37543111 DOI: 10.1016/j.jad.2023.07.119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 08/07/2023]
Abstract
BACKGROUND Sleep deprivation (SD) has been suggested to have a rapid antidepressant effect. There is substantial evidence that neuroinflammation and neuroplasticity play critical roles in the pathophysiology and treatment of depression. Here, we investigated the mechanisms of SD to alleviate depression-like behaviors of mice, and the role of neuroinflammation and neuroplasticity in it. METHODS Adult male C57BL/6 J mice were subjected to chronic restraint stress (CRS) for 6 weeks, and 6 h of SD were administrated. Behavioral tests were performed to measure depression-like behaviors. RNA-sequencing and bioinformatic analysis were performed in the anterior cingulate cortex (ACC). The differentially expressed genes were confirmed by quantitative real-time polymerase chain reaction (RT-qPCR). Neuroinflammation and neuroplasticity were measured by western blotting and immunofluorescence staining. RESULTS Behavioral tests demonstrated that SD swiftly attenuated the depression-like behaviors induced by CRS. RNA-sequencing identified the upregulated immune and inflammatory pathways after CRS exposure were downregulated by SD. Furthermore, SD reversed the levels of immune and inflammation-related mRNA, pro-inflammatory factors and microglia activation in ACC. Additionally, the impaired neuroplasticity elicited by CRS in the prefrontal cortex (PFC) and ACC were improved by SD. LIMITATIONS More in-depth studies are required to determine the role of different SD protocols in depressive symptoms and their underlying mechanisms. CONCLUSIONS Our study revealed the rapid antidepressant effect of SD on CRS mice through the reduction of the neuroinflammatory response in ACC and the improvement of neuroplasticity in PFC and ACC, providing a theoretical basis for the clinical application of SD as a rapid antidepressant treatment.
Collapse
Affiliation(s)
- Shuxiang Shi
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai 201108, China
| | - Mengke Zhang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai 201108, China
| | - Weijie Xie
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai 201108, China
| | - Peijun Ju
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai 201108, China
| | - Ningning Chen
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai 201108, China
| | - Fan Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai 201108, China
| | - Dongbin Lyu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai 201108, China
| | - Meiti Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai 201108, China.
| | - Wu Hong
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai 201108, China; Mental Health Branch, China Hospital Development Institute, Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|
7
|
Bolsius YG, Heckman PRA, Paraciani C, Wilhelm S, Raven F, Meijer EL, Kas MJH, Ramirez S, Meerlo P, Havekes R. Recovering object-location memories after sleep deprivation-induced amnesia. Curr Biol 2023; 33:298-308.e5. [PMID: 36577400 DOI: 10.1016/j.cub.2022.12.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 10/19/2022] [Accepted: 12/02/2022] [Indexed: 12/29/2022]
Abstract
It is well established that sleep deprivation after learning impairs hippocampal memory processes and can cause amnesia. It is unknown, however, whether sleep deprivation leads to the loss of information or merely the suboptimal storage of information that is difficult to retrieve. Here, we show that hippocampal object-location memories formed under sleep deprivation conditions can be successfully retrieved multiple days following training, using optogenetic dentate gyrus (DG) memory engram activation or treatment with the clinically approved phosphodiesterase 4 (PDE4) inhibitor roflumilast. Moreover, the combination of optogenetic DG memory engram activation and roflumilast treatment, 2 days following training and sleep deprivation, made the memory more persistently accessible for retrieval even several days later (i.e., without further optogenetic or pharmacological manipulation). Altogether, our studies in mice demonstrate that sleep deprivation does not necessarily cause memory loss but instead leads to the suboptimal storage of information that cannot be retrieved without drug treatment or optogenetic stimulation. Furthermore, our findings suggest that object-location memories, consolidated under sleep deprivation conditions and thought to be lost, can be made accessible again several days after the learning and sleep deprivation episode, using the clinically approved PDE4 inhibitor roflumilast.
Collapse
Affiliation(s)
- Youri G Bolsius
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands
| | - Pim R A Heckman
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands; Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, the Netherlands
| | - Camilla Paraciani
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands
| | - Sophia Wilhelm
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands
| | - Frank Raven
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands
| | - Elroy L Meijer
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands
| | - Martien J H Kas
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands
| | - Steve Ramirez
- Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Boston University, Boston, MA 02215, USA
| | - Peter Meerlo
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands
| | - Robbert Havekes
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands.
| |
Collapse
|
8
|
Jiang C, Cai S, Zhang L. Functional Connectivity of White Matter and Its Association with Sleep Quality. Nat Sci Sleep 2023; 15:287-300. [PMID: 37123094 PMCID: PMC10132294 DOI: 10.2147/nss.s406120] [Citation(s) in RCA: 2] [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: 02/06/2023] [Accepted: 04/04/2023] [Indexed: 05/02/2023] Open
Abstract
Purpose Functional magnetic resonance imaging (fMRI) has been widely adopted to investigate the neural activity in gray matter (GM) in the field of sleep research, but the neural activity in white matter (WM) has received much less attention. The current study set out to test our hypothesis that WM functional abnormality is associated with poor sleep quality. Participants and Methods K-means clustering analysis was performed on 78 healthy adults drawn from the Human Connectome Project dataset to extract stable WM functional networks (WM-FNs) and GM-FNs. The differences in functional connectivity within WM-FNs and between WM- and GM-FNs, as well as the power spectrum between good sleep quality group (Pittsburgh Sleep Quality Index (PSQI) <6, daytime dysfunction = 0) and poor sleep quality group (PSQI >6, daytime dysfunction >0) were examined between groups with good and poor sleep quality. Additionally, linear relationships between sleep quality and altered functional characteristics of WM-FNs were evaluated. Results Functional connectivity between middle and superficial WM-FNs, short- and long-range functional connectivity between WM- and GM-FNs were decreased in poor sleepers and negatively correlated with PSQI score. The mean amplitudes of right sensorimotor WM networks at whole, high and low frequency bands were higher in poor sleepers and were positively correlated with PSQI score. Conclusion WM functional abnormality is associated with poor sleep quality. The neurobiological mechanisms that underlie the functional alterations of WM-FNs in poor sleepers need to be investigated in future studies.
Collapse
Affiliation(s)
- Chunxiang Jiang
- Paul. C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Siqi Cai
- Paul. C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Lijuan Zhang
- Paul. C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
- Correspondence: Lijuan Zhang, Paul. C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen, 518055, People’s Republic of China, Tel +86 0755 86392247, Fax +86 0755 86392299, Email
| |
Collapse
|
9
|
A short-term memory trace persists for days in the mouse hippocampus. Commun Biol 2022; 5:1168. [PMID: 36329137 PMCID: PMC9633825 DOI: 10.1038/s42003-022-04167-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
Active recall of short-term memory (STM) is known to last for a few hours, but whether STM has long-term functions is unknown. Here we show that STM can be optogenetically retrieved at a time point during which natural recall is not possible, uncovering the long-term existence of an STM engram. Moreover, re-training within 3 days led to natural long-term recall, indicating facilitated consolidation. Inhibiting offline CA1 activity during non-rapid eye movement (NREM) sleep, N-methyl-D-aspartate receptor (NMDAR) activity, or protein synthesis after first exposure to the STM-forming event impaired the future re-exposure-facilitated consolidation, which highlights a role of protein synthesis, NMDAR and NREM sleep in the long-term storage of an STM trace. These results provide evidence that STM is not completely lost within hours and demonstrates a possible two-step STM consolidation, first long-term storage as a behaviorally inactive engram, then transformation into an active state by recurrence within 3 days. Short-term memory (STM) forms a protein synthesis-, NMDAR- and NREM sleep-dependent engram which lasts at least 3 days in the mouse hippocampus following a novel object location task, suggesting that STM is not completely lost within hours.
Collapse
|
10
|
Deurveilher S, Golovin T, Hall S, Semba K. Microglia dynamics in sleep/wake states and in response to sleep loss. Neurochem Int 2020; 143:104944. [PMID: 33359188 DOI: 10.1016/j.neuint.2020.104944] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/15/2020] [Accepted: 12/20/2020] [Indexed: 12/22/2022]
Abstract
Sleep has an essential role for optimal brain function, but the cellular substrates for sleep regulation are not fully understood. Microglia, the immune cells of the brain, have gained increasingly more attention over the last two decades for their important roles in various brain functions that extend beyond their well-known immune function, including brain development, neuronal protection, and synaptic plasticity. Here we review recent advances in understanding: i) morphological and phenotypic dynamics of microglia including process motility/growth and gene/protein expression, and ii) microglia-neuron interactions including phagocytosis and contact at synapses which alters neuronal circuit activity, both under physiological state in the adult brain. We discuss how the microglia-neuron interactions particularly at synapses could influence microglia and neuronal activities across circadian cycles and sleep/wake states. We also review recent findings on how microglia respond to sleep loss. We conclude by pointing out key questions and proposing suggestions for future research to better understand the role of microglia in sleep regulation, sleep homeostasis, and the function of sleep.
Collapse
Affiliation(s)
- Samuel Deurveilher
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Tatjana Golovin
- Department of Physiology & Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Shannon Hall
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kazue Semba
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Psychology & Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
11
|
Blum ID, Keleş MF, Baz ES, Han E, Park K, Luu S, Issa H, Brown M, Ho MCW, Tabuchi M, Liu S, Wu MN. Astroglial Calcium Signaling Encodes Sleep Need in Drosophila. Curr Biol 2020; 31:150-162.e7. [PMID: 33186550 DOI: 10.1016/j.cub.2020.10.012] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 09/17/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022]
Abstract
Sleep is under homeostatic control, whereby increasing wakefulness generates sleep need and triggers sleep drive. However, the molecular and cellular pathways by which sleep need is encoded are poorly understood. In addition, the mechanisms underlying both how and when sleep need is transformed to sleep drive are unknown. Here, using ex vivo and in vivo imaging, we show in Drosophila that astroglial Ca2+ signaling increases with sleep need. We demonstrate that this signaling is dependent on a specific L-type Ca2+ channel and is necessary for homeostatic sleep rebound. Thermogenetically increasing Ca2+ in astrocytes induces persistent sleep behavior, and we exploit this phenotype to conduct a genetic screen for genes required for the homeostatic regulation of sleep. From this large-scale screen, we identify TyrRII, a monoaminergic receptor required in astrocytes for sleep homeostasis. TyrRII levels rise following sleep deprivation in a Ca2+-dependent manner, promoting further increases in astrocytic Ca2+ and resulting in a positive-feedback loop. Moreover, our findings suggest that astrocytes then transmit this sleep need to a sleep drive circuit by upregulating and releasing the interleukin-1 analog Spätzle, which then acts on Toll receptors on R5 neurons. These findings define astroglial Ca2+ signaling mechanisms encoding sleep need and reveal dynamic properties of the sleep homeostatic control system.
Collapse
Affiliation(s)
- Ian D Blum
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Mehmet F Keleş
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - El-Sayed Baz
- VIB Center for Brain and Disease Research and Department of Neurosciences, KU Leuven, Leuven 3000, Belgium
| | - Emily Han
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Kristen Park
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Skylar Luu
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Habon Issa
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Matt Brown
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Margaret C W Ho
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Masashi Tabuchi
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Sha Liu
- VIB Center for Brain and Disease Research and Department of Neurosciences, KU Leuven, Leuven 3000, Belgium.
| | - Mark N Wu
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
12
|
Khlghatyan J, Evstratova A, Bozoyan L, Chamberland S, Chatterjee D, Marakhovskaia A, Soares Silva T, Toth K, Mongrain V, Beaulieu J. Fxr1 regulates sleep and synaptic homeostasis. EMBO J 2020; 39:e103864. [PMID: 32893934 PMCID: PMC7604579 DOI: 10.15252/embj.2019103864] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 08/01/2020] [Accepted: 08/06/2020] [Indexed: 12/22/2022] Open
Abstract
The fragile X autosomal homolog 1 (Fxr1) is regulated by lithium and has been GWAS-associated with schizophrenia and insomnia. Homeostatic regulation of synaptic strength is essential for the maintenance of brain functions and involves both cell-autonomous and system-level processes such as sleep. We examined the contribution of Fxr1 to cell-autonomous homeostatic synaptic scaling and neuronal responses to sleep loss, using a combination of gene overexpression and Crispr/Cas9-mediated somatic knockouts to modulate gene expression. Our findings indicate that Fxr1 is downregulated during both scaling and sleep deprivation via a glycogen synthase kinase 3 beta (GSK3β)-dependent mechanism. In both conditions, downregulation of Fxr1 is essential for the homeostatic modulation of surface AMPA receptors and synaptic strength. Preventing the downregulation of Fxr1 during sleep deprivation results in altered EEG signatures. Furthermore, sequencing of neuronal translatomes revealed the contribution of Fxr1 to changes induced by sleep deprivation. These findings uncover a role of Fxr1 as a shared signaling hub between cell-autonomous homeostatic plasticity and system-level responses to sleep loss, with potential implications for neuropsychiatric illnesses and treatments.
Collapse
Affiliation(s)
- Jivan Khlghatyan
- Department of Pharmacology & ToxicologyUniversity of TorontoTorontoONCanada
- Department of Psychiatry and NeuroscienceFaculty of MedicineUniversité LavalQuébec‐CityQCCanada
- Present address:
Department of NeuroscienceNovartis Institutes for Biomedical ResearchCambridgeMAUSA
| | - Alesya Evstratova
- Department of Pharmacology & ToxicologyUniversity of TorontoTorontoONCanada
| | - Lusine Bozoyan
- Department of Pharmacology & ToxicologyUniversity of TorontoTorontoONCanada
| | - Simon Chamberland
- Department of Psychiatry and NeuroscienceFaculty of MedicineUniversité LavalQuébec‐CityQCCanada
- Present address:
NYU Neuroscience InstituteLangone Medical CenterNew York UniversityNew YorkNYUSA
| | | | | | - Tiago Soares Silva
- Department of Pharmacology & ToxicologyUniversity of TorontoTorontoONCanada
| | - Katalin Toth
- Department of Cellular and Molecular MedicineFaculty of MedicineUniversity of OttawaOttawaONCanada
| | - Valerie Mongrain
- Department of NeuroscienceUniversité de Montréal and Center for Advanced Research in Sleep MedicineHôpital du Sacré‐Coeur de Montréal (CIUSSS‐NIM)MontrealQCCanada
| | | |
Collapse
|
13
|
Jasinska M, Woznicka O, Jasek-Gajda E, Lis GJ, Pyza E, Litwin JA. Circadian Changes of Dendritic Spine Geometry in Mouse Barrel Cortex. Front Neurosci 2020; 14:578881. [PMID: 33117123 PMCID: PMC7550732 DOI: 10.3389/fnins.2020.578881] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/26/2020] [Indexed: 11/13/2022] Open
Abstract
The circadian rhythmicity changes the density and shape of dendritic spines in mouse somatosensory barrel cortex, influencing their stability and maturation. In this study, we analyzed the main geometric parameters of dendritic spines reflecting the strength of synapses located on these spines under light/dark (12:12) and constant darkness conditions, in order to distinguish between endogenously regulated and light-driven parameters. Using morphological analysis of serial electron micrographs, as well as three-dimensional reconstructions, we found that the light induces elongation of single-synapse spine necks and increases in the diameter of double-synapse spine necks, increasing and decreasing the isolation of synapses from the parent dendrite, respectively. During the subjective night of constant darkness, we observed an enlargement of postsynaptic density area in inhibitory synapses and an increase in the number of polyribosomes inside double-synapse spines. The results show that both endogenous effect (circadian clock/locomotor activity) and light affect the morphological parameters of single- and double-synapse spines in the somatosensory cortex: light reduces the efficiency of excitatory synapses on single-synapse spines, increases the effect of synaptic transmission in double-synapse spines, and additionally masks the endogenous clock-driven enlargement of inhibitory synapses located on double-synapse spines. This indicates a special role of double-synapse spines and their inhibitory synapses in the regulation of synaptic transmission during both circadian and diurnal cycles in the mouse somatosensory cortex.
Collapse
Affiliation(s)
- Malgorzata Jasinska
- Department of Histology, Jagiellonian University Medical College, Krakow, Poland
| | - Olga Woznicka
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Ewa Jasek-Gajda
- Department of Histology, Jagiellonian University Medical College, Krakow, Poland
| | - Grzegorz J Lis
- Department of Histology, Jagiellonian University Medical College, Krakow, Poland
| | - Elzbieta Pyza
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Jan A Litwin
- Department of Histology, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
14
|
Brown AMC, Gervais NJ. Role of Ovarian Hormones in the Modulation of Sleep in Females Across the Adult Lifespan. Endocrinology 2020; 161:5879359. [PMID: 32735650 PMCID: PMC7450669 DOI: 10.1210/endocr/bqaa128] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/22/2020] [Indexed: 12/13/2022]
Abstract
Ovarian hormones, including 17β-estradiol, are implicated in numerous physiological processes, including sleep. Beginning at puberty, girls report more sleep complaints than boys, which is maintained throughout the reproductive life stage. Sleep problems are exacerbated during the menopausal transition, evidenced by greater risk for sleep disorders. There is emerging evidence that menopause-associated hormone loss contributes to this elevated risk, but age is also an important factor. The extent to which menopause-associated sleep disturbance persists into postmenopause above and beyond the effects of age remains unknown. Untreated sleep disturbances have important implications for cognitive health, as they are emerging as risk factors for dementia. Given that sleep loss impairs memory, an important knowledge gap concerns the role played by menopause-associated hormone loss in exacerbating sleep disturbance and, ultimately, cognitive function in aging women. In this review, we take a translational approach to illustrate the contribution of ovarian hormones in maintaining the sleep-wake cycle in younger and middle-aged females, with evidence implicating 17β-estradiol in supporting the memory-promoting effects of sleep. Sleep physiology is briefly reviewed before turning to behavioral and neural evidence from young females linking 17β-estradiol to sleep-wake cycle maintenance. Implications of menopause-associated 17β-estradiol loss is also reviewed before discussing how ovarian hormones may support the memory-promoting effects of sleep, and why menopause may exacerbate pathological aging via effects on sleep. While still in its infancy, this research area offers a new sex-based perspective on aging research, with a focus on a modifiable risk factor for pathological aging.
Collapse
Affiliation(s)
- Alana M C Brown
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Nicole J Gervais
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
- Correspondence: Nicole J. Gervais, University of Toronto, Department of Psychology, 100 St. George Street, Toronto, ON, Canada M5S 3G3. E-mail:
| |
Collapse
|
15
|
Sun J, Zhao R, Yang X, Deng H, Zhu Y, Chen Y, Yuan K, Xi Y, Yin H, Qin W. Alteration of Brain Gray Matter Density After 24 h of Sleep Deprivation in Healthy Adults. Front Neurosci 2020; 14:754. [PMID: 32903801 PMCID: PMC7438917 DOI: 10.3389/fnins.2020.00754] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 06/26/2020] [Indexed: 12/19/2022] Open
Abstract
It has been reported that one night of acute sleep deprivation (SD) could induce brain structural changes at the synaptic and neuronal levels in animal studies, and could lead to white matter microstructure and cortical thickness change in human neuroimaging studies. In this study, we focused on changes of brain gray matter density (GMD) after one night of acute SD, which has not been explored previously. Twenty-three normal young participants completed the experiment. Each participant underwent twice T1-weighted structural image scanning with one at 08:00 after normal sleep [resting wakeful (RW)] and the other at 08:00 after 24 h of SD. Using voxel-based morphometry (VBM) analysis by FSL-VBM software, we compared GMD between RW and SD. In addition, the gray matter volume (GMV) and cortical thickness (CT) were also calculated based on volumetric and surface measures with FreeSurfer software. The psychomotor vigilance test (PVT) and the Karolinska Sleepiness Scale (KSS) were performed and evaluated for correlation analysis with GMD, GMV, and CT of the significant regions. Our results showed that the GMD in the right frontal pole (FP), right superior frontal gyrus (SFG), and right middle frontal gyrus significantly increased and GMV and CT in the right temporal pole (TP) significantly decreased after 24 h of acute SD. SD-induced changes in GMD in the right middle frontal gyrus were positively correlated with the changes of KSS scores (Spearman’s correlation r = 0.625, p = 0.0014, Bonferroni correction with p < 0.05/25). Taken together, our findings suggested that one night of acute SD could induce substantial brain structure changes and the alterations in GMD in the right middle frontal gyrus (MFG) might be implicated in sleepiness after SD.
Collapse
Affiliation(s)
- Jinbo Sun
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Sciences and Technology, Xidian University, Xi'an, China
| | - Rui Zhao
- School of Electronics and Information, Xi'an Polytechnic University, Xi'an, China
| | - Xuejuan Yang
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Sciences and Technology, Xidian University, Xi'an, China
| | - Hui Deng
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Sciences and Technology, Xidian University, Xi'an, China
| | - Yuanqiang Zhu
- Department of Radiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yao Chen
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Sciences and Technology, Xidian University, Xi'an, China
| | - Kai Yuan
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Sciences and Technology, Xidian University, Xi'an, China
| | - Yibin Xi
- Department of Radiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Hong Yin
- Department of Radiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wei Qin
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Sciences and Technology, Xidian University, Xi'an, China
| |
Collapse
|
16
|
Rosenthal SJ, Josephs T, Kovtun O, McCarty R. Seasonal effects on bipolar disorder: A closer look. Neurosci Biobehav Rev 2020; 115:199-219. [DOI: 10.1016/j.neubiorev.2020.05.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 11/15/2022]
|
17
|
Areal CC, Cao R, Sonenberg N, Mongrain V. Wakefulness/sleep architecture and electroencephalographic activity in mice lacking the translational repressor 4E-BP1 or 4E-BP2. Sleep 2020; 43:5573651. [PMID: 31553042 DOI: 10.1093/sleep/zsz210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 08/05/2019] [Indexed: 12/26/2022] Open
Abstract
Sleep and sleep loss are affecting protein synthesis in the brain, but the contribution of translational control to wakefulness and sleep regulation remains poorly understood. Here, we studied the role of two suppressors of protein synthesis, the eukaryotic translation initiation factor 4E-binding proteins 1 and 2 (4E-BP1 and 4E-BP2), in sleep architecture and electroencephalographic (EEG) activity as well as in the EEG and molecular responses to acute sleep loss. The EEG of mice mutant for the genes encoding 4E-BP1 or 4E-BP2 (Eif4ebp1 and Eif4ebp2 knockout [KO] mice) was recorded under undisturbed conditions and following a 6-hour sleep deprivation (SD). The effect of SD on the expression of genes known to respond to SD was also measured in the prefrontal cortex of Eif4ebp1 and Eif4ebp2 KO mice. Eif4ebp1 KO mice differed from wild-type mice in parameters of wakefulness and sleep quantity and quality, and more subtly in the gene expression response to SD. For instance, Eif4ebp1 KO mice spent more time in slow-wave sleep (SWS) and showed altered baseline 24-h time courses of SWS delta (1-4 Hz) activity and sigma (10-13 Hz) activity. Eif4ebp2 KO mice differed from wild-type mice only for wakefulness and sleep quality, expressing changes in EEG spectral activity generally revealed during and after SD. These findings suggest different roles of effectors of translational control in the regulation of wakefulness and sleep and of synchronized cortical activity.
Collapse
Affiliation(s)
- Cassandra C Areal
- Research Center and Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal (CIUSSS-NIM), Montreal, Québec, Canada.,Department of Neuroscience, Université de Montréal, Montreal, Québec, Canada
| | - Ruifeng Cao
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN.,Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN
| | - Nahum Sonenberg
- Department of Biochemistry and Goodman Cancer Research Center, McGill University, Montreal, Québec, Canada
| | - Valérie Mongrain
- Research Center and Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal (CIUSSS-NIM), Montreal, Québec, Canada.,Department of Neuroscience, Université de Montréal, Montreal, Québec, Canada
| |
Collapse
|
18
|
Wang T, Niu K, Fan A, Bi N, Tao H, Chen XT, Wang HL. Dietary intake of polyunsaturated fatty acids alleviates cognition deficits and depression-like behaviour via cannabinoid system in sleep deprivation rats. Behav Brain Res 2020; 384:112545. [PMID: 32035867 DOI: 10.1016/j.bbr.2020.112545] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/02/2020] [Accepted: 02/05/2020] [Indexed: 01/01/2023]
Abstract
Sleep deprivation (SD) is a common feature in modern society. Prolonged sleep deprivation causes cognition deficits and depression-like behavior in the model of animal experiments. Endocannabinoid system are key modulators of synaptic function, which were related to memory and mood. Although the underlying mechanism remains unknown, several studies indicated the benefits of polyunsaturated fatty acids (PUFAs, linolenic acid, 39.7 %; linoleic acid, 28 %; and oleic acid, 22 %) on brain function through the endocannabinoid system. The present study aimed to evaluate the influence of dietary PUFAs on cognition deficits induced by sleep deprivation in Sprague Dawley rats. The rats were sleep deprivation continuously for 7 days and fed with PUFAs at three different dosages (2, 4 and 8 μl/g body weight) at the meantime. The effect of PUFAs on cognition was investigated by object recognition test while depressive-like behavior were detected using sucrose preference test and forced swim test. The mechanism of PUFAs was elucidated by hippocampal synaptic transmission analyses. The resluts revealed that SD led to the disorder of cognition and mood which was improved by the supplement of PUFAs. SD significantly increased the mEPSC frequency, and decreased the protein level of cannabinoid type-1 receptors (CB1R). These changes were restored by supplement of PUFAs, which showed a similar level to the control group. Behaviour tests showed that the positive effects on repairing cognition and anxiety disorders were almost completely abolished when the CB1R receptor antagonist rimonabant was applied to the SD rats. These findings indicated that PUFAs are a factor regulating cognition deficits and depression induced by SD via cannabinoid type-1 receptors.
Collapse
Affiliation(s)
- Tiandong Wang
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, 193 Tunxi Road, Hefei, Anhui 230009, PR China; School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Kang Niu
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Anni Fan
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Nanxi Bi
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Han Tao
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Xiang-Tao Chen
- School of Pharmacy, Anhui Medical University, Hefei, Anhui 230031, PR China.
| | - Hui-Li Wang
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, 193 Tunxi Road, Hefei, Anhui 230009, PR China; School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China.
| |
Collapse
|
19
|
Serchov T, Schwarz I, Theiss A, Sun L, Holz A, Döbrössy MD, Schwarz MK, Normann C, Biber K, van Calker D. Enhanced adenosine A 1 receptor and Homer1a expression in hippocampus modulates the resilience to stress-induced depression-like behavior. Neuropharmacology 2019; 162:107834. [PMID: 31682853 DOI: 10.1016/j.neuropharm.2019.107834] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 10/19/2019] [Accepted: 10/29/2019] [Indexed: 11/30/2022]
Abstract
Resilience to stress is critical for the development of depression. Enhanced adenosine A1 receptor (A1R) signaling mediates the antidepressant effects of acute sleep deprivation (SD). However, chronic SD causes long-lasting upregulation of brain A1R and increases the risk of depression. To investigate the effects of A1R on mood, we utilized two transgenic mouse lines with inducible A1R overexpression in forebrain neurons. These two lines have identical levels of A1R increase in the cortex, but differ in the transgenic A1R expression in the hippocampus. Switching on the transgene promotes robust antidepressant and anxiolytic effects in both lines. The mice of the line without transgenic A1R overexpression in the hippocampus (A1Hipp-) show very strong resistance towards development of stress-induced chronic depression-like behavior. In contrast, the mice of the line in which A1R upregulation extends to the hippocampus (A1Hipp+), exhibit decreased resilience to depression as compared to A1Hipp-. Similarly, automatic analysis of reward behavior of the two lines reveals that depression resistant A1Hipp-transgenic mice exhibit high sucrose preference, while mice of the vulnerable A1Hipp + line developed stress-induced anhedonic phenotype. The A1Hipp + mice have increased Homer1a expression in hippocampus, correlating with impaired long-term potentiation in the CA1 region, mimicking the stressed mice. Furthermore, virus-mediated overexpression of Homer1a in the hippocampus decreases stress resilience. Taken together our data indicate for first time that increased expression of A1R and Homer1a in the hippocampus modulates the resilience to stress-induced depression and thus might potentially mediate the detrimental effects of chronic sleep restriction on mood.
Collapse
Affiliation(s)
- Tsvetan Serchov
- Department of Stereotactic and Functional Neurosurgery, Medical Center - University Freiburg, Faculty of Medicine, University of Freiburg, Breisacher Str. 64, 79106, Freiburg, Germany.
| | - Inna Schwarz
- Functional Neuroconnectomics Group, Department of Experimental Epileptology and Cognition Research, Life and Brain Centre, University of Bonn, Medical School, 53105, Bonn, Germany
| | - Alice Theiss
- Department for Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hauptstrasse 5, 79104, Freiburg, Germany
| | - Lu Sun
- Department for Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hauptstrasse 5, 79104, Freiburg, Germany; Department Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, 9713, AV Groningen, the Netherlands
| | - Amrei Holz
- Department for Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hauptstrasse 5, 79104, Freiburg, Germany; Faculty of Biology, University of Freiburg, Schaenzlestr. 1, 79104, Freiburg, Germany
| | - Mate D Döbrössy
- Department of Stereotactic and Functional Neurosurgery, Medical Center - University Freiburg, Faculty of Medicine, University of Freiburg, Breisacher Str. 64, 79106, Freiburg, Germany
| | - Martin K Schwarz
- Functional Neuroconnectomics Group, Department of Experimental Epileptology and Cognition Research, Life and Brain Centre, University of Bonn, Medical School, 53105, Bonn, Germany
| | - Claus Normann
- Department for Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hauptstrasse 5, 79104, Freiburg, Germany
| | - Knut Biber
- Department for Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hauptstrasse 5, 79104, Freiburg, Germany; Department Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, 9713, AV Groningen, the Netherlands
| | - Dietrich van Calker
- Department for Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hauptstrasse 5, 79104, Freiburg, Germany
| |
Collapse
|
20
|
Wirz-Justice A, Benedetti F. Perspectives in affective disorders: Clocks and sleep. Eur J Neurosci 2019; 51:346-365. [PMID: 30702783 DOI: 10.1111/ejn.14362] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 12/30/2018] [Accepted: 01/22/2019] [Indexed: 12/17/2022]
Abstract
Mood disorders are often characterised by alterations in circadian rhythms, sleep disturbances and seasonal exacerbation. Conversely, chronobiological treatments utilise zeitgebers for circadian rhythms such as light to improve mood and stabilise sleep, and manipulations of sleep timing and duration as rapid antidepressant modalities. Although sleep deprivation ("wake therapy") can act within hours, and its mood-elevating effects be maintained by regular morning light administration/medication/earlier sleep, it has not entered the regular guidelines for treating affective disorders as a first-line treatment. The hindrances to using chronotherapeutics may lie in their lack of patentability, few sponsors to carry out large multi-centre trials, non-reimbursement by medical insurance and their perceived difficulty or exotic "alternative" nature. Future use can be promoted by new technology (single-sample phase measurements, phone apps, movement and sleep trackers) that provides ambulatory documentation over long periods and feedback to therapist and patient. Light combinations with cognitive behavioural therapy and sleep hygiene practice may speed up and also maintain response. The urgent need for new antidepressants should hopefully lead to reconsideration and implementation of these non-pharmacological methods, as well as further clinical trials. We review the putative neurochemical mechanisms underlying the antidepressant effect of sleep deprivation and light therapy, and current knowledge linking clocks and sleep with affective disorders: neurotransmitter switching, stress and cortico-limbic reactivity, clock genes, cortical neuroplasticity, connectomics and neuroinflammation. Despite the complexity of multi-system mechanisms, more insight will lead to fine tuning and better application of circadian and sleep-related treatments of depression.
Collapse
Affiliation(s)
- Anna Wirz-Justice
- Centre for Chronobiology, Transfaculty Research Platform Molecular and Cognitive Neurosciences, Psychiatric Hospital of the University of Basel, Basel, Switzerland
| | - Francesco Benedetti
- University Vita-Salute San Raffaele, Milano, Italy.,Psychiatry & Clinical Psychobiology, Division of Neuroscience, San Raffaele Scientific Institute, Milano, Italy
| |
Collapse
|
21
|
Abstract
Sleep is a highly conserved phenomenon in endotherms, and therefore it must serve at least one basic function across this wide range of species. What that function is remains one of the biggest mysteries in neurobiology. By using the word neurobiology, we do not mean to exclude possible non-neural functions of sleep, but it is difficult to imagine why the brain must be taken offline if the basic function of sleep did not involve the nervous system. In this chapter we discuss several current hypotheses about sleep function. We divide these hypotheses into two categories: ones that propose higher-order cognitive functions and ones that focus on housekeeping or restorative processes. We also pose four aspects of sleep that any successful functional hypothesis has to account for: why do the properties of sleep change across the life span? Why and how is sleep homeostatically regulated? Why must the brain be taken offline to accomplish the proposed function? And, why are there two radically different stages of sleep?The higher-order cognitive function hypotheses we discuss are essential mechanisms of learning and memory and synaptic plasticity. These are not mutually exclusive hypotheses. Each focuses on specific mechanistic aspects of sleep, and higher-order cognitive processes are likely to involve components of all of these mechanisms. The restorative hypotheses are maintenance of brain energy metabolism, macromolecular biosynthesis, and removal of metabolic waste. Although these three hypotheses seem more different than those related to higher cognitive function, they may each contribute important components to a basic sleep function. Any sleep function will involve specific gene expression and macromolecular biosynthesis, and as we explain there may be important connections between brain energy metabolism and the need to remove metabolic wastes.A deeper understanding of sleep functions in endotherms will enable us to answer whether or not rest behaviors in species other than endotherms are homologous with mammalian and avian sleep. Currently comparisons across the animal kingdom depend on superficial and phenomenological features of rest states and sleep, but investigations of sleep functions would provide more insight into the evolutionary relationships between EEG-defined sleep in endotherms and rest states in ectotherms.
Collapse
Affiliation(s)
- Marcos G Frank
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University Spokane, Spokane, WA, USA
| | - H Craig Heller
- Department of Biology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
22
|
Cooper JM, Halter KA, Prosser RA. Circadian rhythm and sleep-wake systems share the dynamic extracellular synaptic milieu. Neurobiol Sleep Circadian Rhythms 2018; 5:15-36. [PMID: 31236509 PMCID: PMC6584685 DOI: 10.1016/j.nbscr.2018.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 03/06/2018] [Accepted: 04/10/2018] [Indexed: 01/23/2023] Open
Abstract
The mammalian circadian and sleep-wake systems are closely aligned through their coordinated regulation of daily activity patterns. Although they differ in their anatomical organization and physiological processes, they utilize overlapping regulatory mechanisms that include an assortment of proteins and molecules interacting within the extracellular space. These extracellular factors include proteases that interact with soluble proteins, membrane-attached receptors and the extracellular matrix; and cell adhesion molecules that can form complex scaffolds connecting adjacent neurons, astrocytes and their respective intracellular cytoskeletal elements. Astrocytes also participate in the dynamic regulation of both systems through modulating neuronal appositions, the extracellular space and/or through release of gliotransmitters that can further contribute to the extracellular signaling processes. Together, these extracellular elements create a system that integrates rapid neurotransmitter signaling across longer time scales and thereby adjust neuronal signaling to reflect the daily fluctuations fundamental to both systems. Here we review what is known about these extracellular processes, focusing specifically on areas of overlap between the two systems. We also highlight questions that still need to be addressed. Although we know many of the extracellular players, far more research is needed to understand the mechanisms through which they modulate the circadian and sleep-wake systems.
Collapse
Key Words
- ADAM, A disintegrin and metalloproteinase
- AMPAR, AMPA receptor
- Astrocytes
- BDNF, brain-derived neurotrophic factor
- BMAL1, Brain and muscle Arnt-like-1 protein
- Bmal1, Brain and muscle Arnt-like-1 gene
- CAM, cell adhesion molecules
- CRY, cryptochrome protein
- Cell adhesion molecules
- Circadian rhythms
- Cry, cryptochrome gene
- DD, dark-dark
- ECM, extracellular matrix
- ECS, extracellular space
- EEG, electroencephalogram
- Endo N, endoneuraminidase N
- Extracellular proteases
- GFAP, glial fibrillary acidic protein
- IL, interleukin
- Ig, immunoglobulin
- LC, locus coeruleus
- LD, light-dark
- LH, lateral hypothalamus
- LRP-1, low density lipoprotein receptor-related protein 1
- LTP, long-term potentiation
- MMP, matrix metalloproteinases
- NCAM, neural cell adhesion molecule protein
- NMDAR, NMDA receptor
- NO, nitric oxide
- NST, nucleus of the solitary tract
- Ncam, neural cell adhesion molecule gene
- Nrl, neuroligin gene
- Nrx, neurexin gene
- P2, purine type 2 receptor
- PAI-1, plasminogen activator inhibitor-1
- PER, period protein
- PPT, peduculopontine tegmental nucleus
- PSA, polysialic acid
- Per, period gene
- REMS, rapid eye movement sleep
- RSD, REM sleep disruption
- SCN, suprachiasmatic nucleus
- SWS, slow wave sleep
- Sleep-wake system
- Suprachiasmatic nucleus
- TNF, tumor necrosis factor
- TTFL, transcriptional-translational negative feedback loop
- VIP, vasoactive intestinal polypeptide
- VLPO, ventrolateral preoptic
- VP, vasopressin
- VTA, ventral tegmental area
- dNlg4, drosophila neuroligin-4 gene
- nNOS, neuronal nitric oxide synthase gene
- nNOS, neuronal nitric oxide synthase protein
- tPA, tissue-type plasminogen activator
- uPA, urokinase-type plasminogen activator
- uPAR, uPA receptor
Collapse
|
23
|
Frank MG, Seibt J. Sleep and plasticity: Waking from a fevered dream. Sleep Med Rev 2018; 39:1-2. [DOI: 10.1016/j.smrv.2017.12.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 12/19/2017] [Indexed: 12/15/2022]
|
24
|
A brief period of sleep deprivation causes spine loss in the dentate gyrus of mice. Neurobiol Learn Mem 2018; 160:83-90. [PMID: 29588221 DOI: 10.1016/j.nlm.2018.03.018] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/05/2018] [Accepted: 03/23/2018] [Indexed: 11/22/2022]
Abstract
Sleep and sleep loss have a profound impact on hippocampal function, leading to memory impairments. Modifications in the strength of synaptic connections directly influences neuronal communication, which is vital for normal brain function, as well as the processing and storage of information. In a recently published study, we found that as little as five hours of sleep deprivation impaired hippocampus-dependent memory consolidation, which was accompanied by a reduction in dendritic spine numbers in hippocampal area CA1. Surprisingly, loss of sleep did not alter the spine density of CA3 neurons. Although sleep deprivation has been reported to affect the function of the dentate gyrus, it is unclear whether a brief period of sleep deprivation impacts spine density in this region. Here, we investigated the impact of a brief period of sleep deprivation on dendritic structure in the dentate gyrus of the dorsal hippocampus. We found that five hours of sleep loss reduces spine density in the dentate gyrus with a prominent effect on branched spines. Interestingly, the inferior blade of the dentate gyrus seems to be more vulnerable in terms of spine loss than the superior blade. This decrease in spine density predominantly in the inferior blade of the dentate gyrus may contribute to the memory deficits observed after sleep loss, as structural reorganization of synaptic networks in this subregion is fundamental for cognitive processes.
Collapse
|
25
|
Noorafshan A, Karimi F, Kamali AM, Karbalay-Doust S, Nami M. Could curcumin protect the dendritic trees of the CA1 neurons from shortening and shedding induced by chronic sleep restriction in rats? Life Sci 2018; 198:65-70. [PMID: 29455005 DOI: 10.1016/j.lfs.2018.02.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 02/05/2018] [Accepted: 02/14/2018] [Indexed: 01/04/2023]
Abstract
BACKGROUND AND OBJECTIVE This study evaluated the effect of chronic sleep restriction (CSR) with or without curcumin (CUR) treatment on dendritic lengths and spines of the CA1 hippocampus using the virtual space-ball method. MATERIALS AND METHODS Male rats were randomly submitted to nine groups, including distilled water, CUR (100 mg/kg/day), olive oil, CSR plus distilled water, CSR plus CUR, CSR plus olive oil, grid-floor plus distilled water, grid-floor plus CUR, and grid-floor plus olive oil groups. Sleep deficiency was imposed using the multi-platform box containing water for 18 h/day. In 21 days, animal's brains were prepared for stereological studies. RESULTS The mean dendrite length in CA1 neurons was reduced by 39% (p < 0.05) while the density of stubby, thin, and mushroom spines reduced by 38%, 33% and 32%, respectively (p < 0.01), in the CSR + distilled water group compared to the distilled water group. Yet, CUR treatment in CSR-rats was found to protect the declined dendritic length as well as loss of stubby and mushroom but not thin spines. CONCLUSION The estimated dendritic length using the virtual space-ball method revealed that chronic sleep restriction for 18 h/day over 21 days could induce shortening and shedding of the CA1 dendritic trees which could notably be protected by CUR.
Collapse
Affiliation(s)
- Ali Noorafshan
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Karimi
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali-Mohammad Kamali
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Neuroscience Laboratory, NSL (Brain, Cognition and Behavior), Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Students' research committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saied Karbalay-Doust
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Nami
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Neuroscience Laboratory, NSL (Brain, Cognition and Behavior), Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|