1
|
Zhao X, Liu J, Shao Z, Liu X, Wang Z, Yuan K, Zhang B, Li Y, Sheng X, Zhu Y, Guo Y. Restoration of abnormal sleep EEG power in patients with insomnia disorder after 1Hz rTMS over left DLPFC. Front Psychiatry 2024; 15:1431837. [PMID: 39319359 PMCID: PMC11419987 DOI: 10.3389/fpsyt.2024.1431837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/23/2024] [Indexed: 09/26/2024] Open
Abstract
Introduction Hyperarousal has been a significant pathophysiological theory related to insomnia disorder (ID), characterized by excessive cortical activation and abnormal electroencephalogram (EEG) power during daytime or sleep. However, there is currently insufficient attention to the EEG power during rapid eye movement (REM) sleep and different stages of non-rapid eye movement (NREM) sleep. Additionally, whether the abnormal sleep EEG power in ID patients can be restored by repetitive transcranial magnetic stimulation (rTMS) remains unclear. Methods> Data of 26 ID patients and 26 healthy controls (HCs) were included in the current observational study. The comparisons of relative power between patients and HCs at baseline in each band of each sleep stage and the changes in patients before and after rTMS treatment were performed. The correlations between relative power and behavioral measures of the patients were also investigated. Results Abnormalities in sleep EEG relative power in the delta, beta and gamma bands of the patients were observed in NREM2, NREM3 and REM sleep. Correlations were identified between relative power and behavioral measures in ID group, primarily encompassing sleep efficiency, sleep onset latency and depression scores. Post-treatment improvements in relative power of the delta and beta band were observed in NREM2 sleep. Discussion The relative power of sleep EEG exhibited a significant correlation with sleep measures in ID patients, and demonstrated notable differences from HCs across the delta, beta, and gamma frequency bands. Furthermore, our findings suggest that rTMS treatment may partially ameliorate relative power abnormalities in patients with ID.
Collapse
Affiliation(s)
- Xumeng Zhao
- Department of Psychosomatic Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jiayi Liu
- Department of Psychosomatic Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ziqiang Shao
- School of Life Science and Technology, Xidian University, Xi'an, Shanxi, China
| | - Xiaoyang Liu
- School of Life Science and Technology, Xidian University, Xi'an, Shanxi, China
| | - Zhen Wang
- School of Life Science and Technology, Xidian University, Xi'an, Shanxi, China
| | - Kai Yuan
- School of Life Science and Technology, Xidian University, Xi'an, Shanxi, China
| | - Bingqian Zhang
- Department of Psychosomatic Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yan Li
- Department of Psychosomatic Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaona Sheng
- Department of Psychosomatic Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yifei Zhu
- Department of Psychosomatic Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yansu Guo
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
2
|
Zhang Z, Zhang W, Fang Y, Wang N, Liu G, Zou N, Song Z, Liu H, Wang L, Xiao Q, Zhao J, Wang Y, Lei T, Zhang C, Liu X, Zhang B, Luo F, Xia J, He C, Hu Z, Ren S, Zhao H. A potentiation of REM sleep-active neurons in the lateral habenula may be responsible for the sleep disturbance in depression. Curr Biol 2024; 34:3287-3300.e6. [PMID: 38944036 DOI: 10.1016/j.cub.2024.05.075] [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: 06/01/2023] [Revised: 03/25/2024] [Accepted: 05/31/2024] [Indexed: 07/01/2024]
Abstract
Psychiatric disorders with dysfunction of the lateral habenula (LHb) show sleep disturbance, especially a disinhibition of rapid eye movement (REM) sleep in major depression. However, the role of LHb in physiological sleep control and how LHb contributes to sleep disturbance in major depression remain elusive. Here, we found that functional manipulations of LHb glutamatergic neurons bidirectionally modulated both non-REM (NREM) sleep and REM sleep. Activity recording revealed heterogeneous activity patterns of LHb neurons across sleep/wakefulness cycles, but LHb neurons were preferentially active during REM sleep. Using an activity-dependent tagging method, we selectively labeled a population of REM sleep-active LHb neurons and demonstrated that these neurons specifically promoted REM sleep. Neural circuit studies showed that LHb neurons regulated REM sleep via projections to the ventral tegmental area but not to the rostromedial tegmental nucleus. Furthermore, we found that the increased REM sleep in a depression mouse model was associated with a potentiation of REM sleep-active LHb neurons, including an increased proportion, elevated spike firing, and altered activity mode. Importantly, inhibition of REM sleep-active LHb neurons not only attenuated the increased REM sleep but also alleviated depressive-like behaviors in a depression mouse model. Thus, our results demonstrated that REM sleep-active LHb neurons selectively promoted REM sleep, and a potentiation of these neurons contributed to depression-associated sleep disturbance.
Collapse
Affiliation(s)
- Zehui Zhang
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Wei Zhang
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Yuanyuan Fang
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China; Department of Anaesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, China
| | - Na Wang
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Guoying Liu
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing 400064, China
| | - Nan Zou
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing 400064, China
| | - Zhenbo Song
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Hanshu Liu
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China; Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Longshuo Wang
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Qin Xiao
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Juanjuan Zhao
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Yaling Wang
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Ting Lei
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Cai Zhang
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Xiaofeng Liu
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Beilin Zhang
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Fenlan Luo
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Jianxia Xia
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Chao He
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Zhian Hu
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China; Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing 400064, China.
| | - Shuancheng Ren
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China.
| | - Hua Zhao
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
3
|
Ba W, Nollet M, Yin C, Yu X, Wong S, Miao A, Beckwith EJ, Harding EC, Ma Y, Yustos R, Vyssotski AL, Wisden W, Franks NP. A REM-active basal ganglia circuit that regulates anxiety. Curr Biol 2024; 34:3301-3314.e4. [PMID: 38944034 DOI: 10.1016/j.cub.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 04/22/2024] [Accepted: 06/05/2024] [Indexed: 07/01/2024]
Abstract
Rapid eye movement (REM) sleep has been hypothesized to promote emotional resilience, but any neuronal circuits mediating this have not been identified. We find that in mice, somatostatin (Som) neurons in the entopeduncular nucleus (EPSom)/internal globus pallidus are predominantly active during REM sleep. This unique REM activity is both necessary and sufficient for maintaining normal REM sleep. Inhibiting or exciting EPSom neurons reduced or increased REM sleep duration, respectively. Activation of the sole downstream target of EPSom neurons, Vglut2 cells in the lateral habenula (LHb), increased sleep via the ventral tegmental area (VTA). A simple chemogenetic scheme to periodically inhibit the LHb over 4 days selectively removed a significant amount of cumulative REM sleep. Chronic, but not acute, REM reduction correlated with mice becoming anxious and more sensitive to aversive stimuli. Therefore, we suggest that cumulative REM sleep, in part generated by the EP → LHb → VTA circuit identified here, could contribute to stabilizing reactions to habitual aversive stimuli.
Collapse
Affiliation(s)
- Wei Ba
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Mathieu Nollet
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK; UK Dementia Research Institute, Imperial College London, London SW7 2AZ, UK
| | - Chunyu Yin
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK; Department of Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Xiao Yu
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Sara Wong
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK; UK Dementia Research Institute, Imperial College London, London SW7 2AZ, UK
| | - Andawei Miao
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK; UK Dementia Research Institute, Imperial College London, London SW7 2AZ, UK
| | - Esteban J Beckwith
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Edward C Harding
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Ying Ma
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Raquel Yustos
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Alexei L Vyssotski
- Institute of Neuroinformatics, University of Zurich and ETH Zurich, Zurich 8057, Switzerland
| | - William Wisden
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK; UK Dementia Research Institute, Imperial College London, London SW7 2AZ, UK.
| | - Nicholas P Franks
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK; UK Dementia Research Institute, Imperial College London, London SW7 2AZ, UK.
| |
Collapse
|
4
|
Ku SP, Atucha E, Alavi N, Mulla-Osman H, Kayumova R, Yoshida M, Csicsvari J, Sauvage MM. Phase locking of hippocampal CA3 neurons to distal CA1 theta oscillations selectively predicts memory performance. Cell Rep 2024; 43:114276. [PMID: 38814781 DOI: 10.1016/j.celrep.2024.114276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 01/09/2024] [Accepted: 05/09/2024] [Indexed: 06/01/2024] Open
Abstract
How the coordination of neuronal spiking and brain rhythms between hippocampal subregions supports memory function remains elusive. We studied the interregional coordination of CA3 neuronal spiking with CA1 theta oscillations by recording electrophysiological signals along the proximodistal axis of the hippocampus in rats that were performing a high-memory-demand recognition memory task adapted from humans. We found that CA3 population spiking occurs preferentially at the peak of distal CA1 theta oscillations when memory was tested but only when previously encountered stimuli were presented. In addition, decoding analyses revealed that only population cell firing of proximal CA3 together with that of distal CA1 can predict performance at test in the present non-spatial task. Overall, our work demonstrates an important role for the synchronization of CA3 neuronal activity with CA1 theta oscillations during memory testing.
Collapse
Affiliation(s)
- Shih-Pi Ku
- Leibniz Institute for Neurobiology, Functional Architecture of Memory Department, Magdeburg, Germany.
| | - Erika Atucha
- Leibniz Institute for Neurobiology, Functional Architecture of Memory Department, Magdeburg, Germany
| | - Nico Alavi
- Leibniz Institute for Neurobiology, Functional Architecture of Memory Department, Magdeburg, Germany
| | - Halla Mulla-Osman
- Leibniz Institute for Neurobiology, Functional Architecture of Memory Department, Magdeburg, Germany
| | - Rukhshona Kayumova
- Leibniz Institute for Neurobiology, Functional Architecture of Memory Department, Magdeburg, Germany
| | - Motoharu Yoshida
- Leibniz Institute for Neurobiology, Functional Architecture of Memory Department, Magdeburg, Germany; German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Jozsef Csicsvari
- Institute of Science and Technology (IST), Klosterneuburg, Austria
| | - Magdalena M Sauvage
- Leibniz Institute for Neurobiology, Functional Architecture of Memory Department, Magdeburg, Germany; Otto von Guericke University, Medical Faculty, Functional Neuroplasticity Department, Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany.
| |
Collapse
|
5
|
Hashim Z, Gupta M, Neyaz Z, Srivastava S, Mani V, Nath A, Khan AR. Biophysical modeling and diffusion kurtosis imaging reveal microstructural alterations in normal-appearing white-matter regions of the brain in obstructive sleep apnea. SLEEP ADVANCES : A JOURNAL OF THE SLEEP RESEARCH SOCIETY 2024; 5:zpae031. [PMID: 38903701 PMCID: PMC11187986 DOI: 10.1093/sleepadvances/zpae031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 05/17/2024] [Indexed: 06/22/2024]
Abstract
Study Objectives Studies have indicated that sleep abnormalities are a strong risk factor for developing cognitive impairment, cardiomyopathies, and neurodegenerative disorders. However, neuroimaging modalities are unable to show any consistent markers in obstructive sleep apnea (OSA) patients. We hypothesized that, compared with those of the control cohort, advanced diffusion MRI metrics could show subtle microstructural alterations in the brains of patients with OSA. Methods Sixteen newly diagnosed patients with moderate to severe OSA and 15 healthy volunteers of the same age and sex were considered healthy controls. Multishell diffusion MRI data of the brain, along with anatomical data (T1 and T2 images), were obtained on a 3T MRI system (Siemens, Germany) after a polysomnography (PSG) test for sleep abnormalities and a behavioral test battery to evaluate cognitive and executive brain functions. Diffusion MRI data were used to compute diffusion tensor imaging and diffusion kurtosis imaging (DKI) parameters along with white-matter tract integrity (WMTI) metrics for only parallel white-matter fibers. Results OSA was diagnosed when the patient's apnea-hypopnea index was ≥ 15. No significant changes in cognitive or executive functions were observed in the OSA cohort. DKI parameters can show significant microstructural alterations in the white-matter region, while the WMTI metric, the axonal-water-fraction (fp), reveals a significant decrease in OSA patients concerning the control cohort. Conclusions Advanced diffusion MRI-based microstructural alterations in the white-matter region of the brain suggest that white-matter tracts are more sensitive to OSA-induced intermittent hypoxia.
Collapse
Affiliation(s)
- Zia Hashim
- Department of Pulmonary Medicine, SGPGIMS, Lucknow, India
| | - Mansi Gupta
- Department of Pulmonary Medicine, SGPGIMS, Lucknow, India
| | - Zafar Neyaz
- Department of Radio-diagnosis, SGPGIMS, Lucknow, India
| | | | - Vinita Mani
- Department of Neurology, SGPGIMS, Lucknow, India
| | - Alok Nath
- Department of Pulmonary Medicine, SGPGIMS, Lucknow, India
| | - Ahmad Raza Khan
- Department of Advanced Spectroscopy and Imaging, CBMR, SGPGIMS Campus, Lucknow, India
| |
Collapse
|
6
|
White AG, Elias E, Orozco A, Robinson SA, Manners MT. Chronic Stress-Induced Neuroinflammation: Relevance of Rodent Models to Human Disease. Int J Mol Sci 2024; 25:5085. [PMID: 38791125 PMCID: PMC11121038 DOI: 10.3390/ijms25105085] [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: 04/09/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
The brain is the central organ of adaptation to stress because it perceives and determines threats that induce behavioral, physiological, and molecular responses. In humans, chronic stress manifests as an enduring consistent feeling of pressure and being overwhelmed for an extended duration. This can result in a persistent proinflammatory response in the peripheral and central nervous system (CNS), resulting in cellular, physiological, and behavioral effects. Compounding stressors may increase the risk of chronic-stress-induced inflammation, which can yield serious health consequences, including mental health disorders. This review summarizes the current knowledge surrounding the neuroinflammatory response in rodent models of chronic stress-a relationship that is continually being defined. Many studies investigating the effects of chronic stress on neuroinflammation in rodent models have identified significant changes in inflammatory modulators, including nuclear factor-κB (NF-κB) and toll-like receptors (TLRs), and cytokines, including tumor necrosis factor-alpha (TNF-α), interleukin (IL)-1β, and IL-6. This suggests that these are key inflammatory factors in the chronic stress response, which may contribute to the establishment of anxiety and depression-like symptoms. The behavioral and neurological effects of modulating inflammatory factors through gene knockdown (KD) and knockout (KO), and conventional and alternative medicine approaches, are discussed.
Collapse
Affiliation(s)
- Abigail G. White
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA
| | - Elias Elias
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA
| | - Andrea Orozco
- Department of Psychology, Williams College, Williamstown, MA 01267, USA
| | | | - Melissa T. Manners
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA
| |
Collapse
|
7
|
Wang R, Hu X, Liu S, Wang J, Xiong F, Zhang X, Ye W, Wang H. Kaempferol-3-O-sophoroside (PCS-1) contributes to modulation of depressive-like behaviour in C57BL/6J mice by activating AMPK. Br J Pharmacol 2024; 181:1182-1202. [PMID: 37949672 DOI: 10.1111/bph.16283] [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: 03/25/2023] [Revised: 10/18/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND AND PURPOSE Kaempferol-3-O-sophoroside (PCS-1) is the main component in Crocus sativus (Saffron), a herb with mood-enhancing properties. AMP-activated protein kinase (AMPK) is a potential therapeutic target for depression. This study explores the antidepressive-like properties of PCS-1 and its AMPK activation to confirm AMPK as a target for antidepression. EXPERIMENTAL APPROACH Corticosterone (CORT)-induced PC12 cell injury served as an in vitro model to evaluate the neuroprotective effect of PCS-1. Neuro-2a cells and primary neurons were utilized to evaluate the synaptogenesis role of PCS-1. CORT-induced mouse depression model and chronic unpredictable mild stress (CUMS) model were used to assess the antidepressive-like properties of PCS-1 through behavioural tests, magnetic resonance imaging, and biochemical index measurements. Western blot and immunofluorescence assays were used to study the mechanisms of PCS-1. Cellular thermal shift assay was used to confirm the binding target. KEY RESULTS PCS-1 (12.5-50 μM) ameliorated CORT-induced PC12 cell damage, oxidative stress and inflammation. PCS-1 alone promoted an increase in synapses in Neuro-2a cells and primary neurons. Oral administration of PCS-1 (10 and 20 mg·kg-1 ) ameliorated weight loss, dyskinesia, and hippocampal volume reduction induced by CORT and CUMS. PCS-1 bound to AMPK to improve the expression of brain-derived neurotrophic factor (BDNF) and induce autophagy. CONCLUSION AND IMPLICATIONS PCS-1 binds to AMPK to promote BDNF production and autophagy enhancement, ultimately achieving antidepressant effects. This study provides support for the clinical application of saffron petals and provides further evidence for AMPK as a potential target for antidepression.
Collapse
Affiliation(s)
- Rong Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Xiaolong Hu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Shumeng Liu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Jingjin Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Fei Xiong
- State Key Laboratory of Bioelectronics, Jiangsu Laboratory for Biomaterials and Devices, Southeast University, Nanjing, People's Republic of China
| | - Xiaoqi Zhang
- Institute of Traditional Chinese Medicine & Natural Products, Jinan University, Guangzhou, People's Republic of China
| | - Wencai Ye
- Institute of Traditional Chinese Medicine & Natural Products, Jinan University, Guangzhou, People's Republic of China
| | - Hao Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|
8
|
Zhang Q, Chen F. Impact of single-trial avoidance learning on subsequent sleep. Eur J Neurosci 2024; 59:739-751. [PMID: 38342099 DOI: 10.1111/ejn.16274] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 02/13/2024]
Abstract
Both non-rapid eye movement (NonREM) sleep and rapid eye movement (REM) sleep, as well as sleep spindle and ripple oscillations, are important for memory formation. Through cortical EEG recordings of prefrontal cortex and hippocampus during and after an inhibitory avoidance task, we analysed the dynamic changes in the amounts of sleep, spindle and ripple oscillations related to memory formation. The total amount of NonREM sleep was reduced during the first hour after learning. Moreover, significant decrease of the total spindle and ripple counts was observed at the first hour after learning as well. In addition, foot shock alone, with no associated learning, produced little effect on the dynamics of sleep oscillations, indicating that the learning experience is necessary for these changes to occur.
Collapse
Affiliation(s)
- Qianwen Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Fujun Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
9
|
Jiao H, Fan Y, Gong A, Li T, Fu X, Yan Z. Xiaoyaosan ameliorates CUMS-induced depressive-like and anorexia behaviors in mice via necroptosis related cellular senescence in hypothalamus. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116938. [PMID: 37495029 DOI: 10.1016/j.jep.2023.116938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/28/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Depression and anorexia often co-occur and share symptoms such as low mood, lack of energy, and weight loss. Xiaoyaosan is a classic formula comprising of a combination of eight herbs, possessing definitive therapeutic effects, minimal side effects, and economical benefits. It has been extensively employed in clinical treatment of ailments and symptoms such as depression, anxiety, and appetite problems. Nonetheless, its exact pharmacological mechanism with necroptosis remains incompletely explicit. AIM OF THE STUDY The aim of this study is to explore the potential mechanisms of anti-depressive and appetite-regulating effects of the active ingredients in Xiaoyaosan, and to investigate whether there is a correlation with necroptosis. MATERIALS AND METHODS The network pharmacology method was conducted to identify active ingredients, which were used to predict the possible targets of Xiaoyaosan and explore the potential targets in treating depression and anorexia by overlapping with differentially expressed genes (DEGs) screened from GEO datasets (GSE125441, GSE198597, and GSE69151). Afterwards, the protein-protein interaction (PPI) network, enrichment analyses, hub gene identification, co-expression study and molecular docking were used to study the potential mechanism of Xiaoyaosan. Then, a mice model of depression was established by chronic unpredictable mild stress (CUMS) and the incidence of necroptosis in the hypothalamus of CUMS mice was investigated, while verifying the key therapeutic target of Xiaoyaosan. RESULTS Through network pharmacology research, it had been discovered that the 145 active ingredients of the 8 herbs in the Xiaoyaosan could regulate 198 disease targets. Through PPI network analysis and functional enrichment analysis, it had been found that the pharmacological mechanism of Xiaoyaosan mainly involved biological processes such as oxidative stress, kinase activity, and DNA metabolism. It is related to various pathways such as cellular senescence, immune inflammation, and the cell cycle, and 9 hub targets had been identified. Further analysis of the 9 hub targets and the key PPI network clusters clarified the key mechanisms by which Xiaoyaosan exerts anti-depressant and appetite regulating effects, possibly related to necroptosis-mediated cellular senescence. Molecular docking of the key indicators of cellular senescence screened by bioinformatics, SIRT1, ABL1, and MYC, revealed that the key component regulating SIRT1 is 2-[3,4-dihydroxyphenyl]-5,7-dihydroxy-6-[3-methylbut-2-enyl]chromone in licorice root, Glabridin in licorice root regulates ABL1, and β-sitosterol found in Chinese angelica, debark peony root, and fresh ginger regulates MYC. Finally, through in vivo experiments, the expression of necroptosis in the hypothalamus of CUMS mice was verified. The regulatory effects of Xiaoyaosan on key substances RIPK1, RIPK3, MLKL, and p-MLKL were determined, while regulating effects on SIRT1, ABL1, and MYC were also observed. CONCLUSION The present study have revealed the common mechanism of Xiaoyaosan in treating depression and anorexia, indicating that the active ingredients of Xiaoyaosan may alleviate the symptoms of depression and anorexia by intervening in the pathways related to necroptosis and cellular senescence. The hub genes and common pathways identified by the study also provide new insights into the therapeutic targets of depression and anorexia, as well as the exploration of pharmacological mechanism of Xiaoyaosan.
Collapse
Affiliation(s)
- Haiyan Jiao
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou 571199, China.
| | - Yingli Fan
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou 571199, China.
| | - Aimin Gong
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou 571199, China.
| | - Tian Li
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou 571199, China.
| | - Xing Fu
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou 571199, China.
| | - Zhiyi Yan
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou 571199, China; Dongfang Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100078, China.
| |
Collapse
|
10
|
Mamelak M. Depression and the Glutamate/GABA-Glutamine Cycle. Curr Neuropharmacol 2024; 23:75-84. [PMID: 39150032 PMCID: PMC11519819 DOI: 10.2174/1570159x22666240815120244] [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: 01/22/2024] [Revised: 02/07/2024] [Accepted: 02/23/2024] [Indexed: 08/17/2024] Open
Abstract
Many features of major depressive disorder are mirrored in rodent models of psychological stress. These models have been used to examine the relationship between the activation of the hypothalamic- pituitary axis in response to stress, the development of oxidative stress and neuroinflammation, the dominance of cholinergic neurotransmission and the associated increase in REM sleep pressure. Rodent models have also provided valuable insights into the impairment of glycolysis and brain glucose utilization by the brain under stress, the resulting decrease in brain energy production and the reduction in glutamate/GABA-glutamine cycling. The rapidly acting antidepressants, scopolamine, ketamine and ECT, all raise extracellular glutamate and scopolamine and ketamine have specifically been shown to increase glutamate/GABA-glutamine cycling in men and rodents with corresponding short-term relief of depression. The nightly use of gammahydroxybutyrate (GHB) may achieve more permanent results and may even act prophylactically to prevent the development or recurrence of depression. GHB is a GABAB agonist and restores the normal balance between cholinergic and monoaminergic neurotransmission by inhibiting cholinergic neurotransmission. It relieves REM sleep pressure. GHB's metabolism generates NADPH, a key antioxidant cofactor. Its metabolism also generates succinate, the tricarboxylic acid cycle intermediate, to provide energy to the cell and to synthesize glutamate. In both animals and man, GHB increases the level of brain glutamate.
Collapse
Affiliation(s)
- Mortimer Mamelak
- Department of Psychiatry, Baycrest Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
11
|
Holloway AL, Lerner TN. Hidden variables in stress neurobiology research. Trends Neurosci 2024; 47:9-17. [PMID: 37985263 PMCID: PMC10842876 DOI: 10.1016/j.tins.2023.10.006] [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: 07/03/2023] [Revised: 10/11/2023] [Accepted: 10/31/2023] [Indexed: 11/22/2023]
Abstract
Among the central goals of stress neurobiology research is to understand the mechanisms by which stressors change neural circuit function to precipitate or exacerbate psychiatric symptoms. Yet despite decades of effort, psychiatric medications that target the biological substrates of the stress response are largely lacking. We propose that the clinical advancement of stress response-based therapeutics for psychiatric disorders may be hindered by 'hidden variables' in stress research, including considerations of behavioral study design (stressors and outcome measures), individual variability, sex differences, and the interaction of the body's stress hormone system with endogenous circadian and ultradian rhythms. We highlight key issues and suggest ways forward in stress neurobiology research that may improve the ability to assess stress mechanisms and translate preclinical findings.
Collapse
Affiliation(s)
- Ashley L Holloway
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Northwestern University Interdepartmental Neuroscience Program (NUIN), Evanston, IL, USA
| | - Talia N Lerner
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Northwestern University Interdepartmental Neuroscience Program (NUIN), Evanston, IL, USA.
| |
Collapse
|
12
|
Yao D, Li R, Hao J, Huang H, Wang X, Ran L, Fang Y, He Y, Wang W, Liu X, Wang M. Melatonin alleviates depression-like behaviors and cognitive dysfunction in mice by regulating the circadian rhythm of AQP4 polarization. Transl Psychiatry 2023; 13:310. [PMID: 37802998 PMCID: PMC10558463 DOI: 10.1038/s41398-023-02614-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/17/2023] [Accepted: 09/25/2023] [Indexed: 10/08/2023] Open
Abstract
Depression is a common chronic psychiatric illness, which is resistant to medical treatments. While melatonin may alleviate certain depression symptoms, evidence for its efficacy against core symptoms is lacking. Here, we tested a mechanism whereby melatonin rescues the behavioral outcomes of the chronic unpredictable mild stress (CUMS) mouse model of depression. CUMS mice showed depressive behaviors to tail suspension, open field behavior, and sucrose preference test, and cognitive dysfunction in the Morris water maze. Impairments in these measures were relieved by melatonin treatment. Moreover, CUMS mice had impaired glymphatic function across the sleep-wake cycle due to the astrocytic loss and disturbance of circadian regulation of the polarized expression of aquaporin-4 (AQP4) water channels in perivascular astrocytes. EEG results in CUMS mice showed a reduced total sleep time and non-rapid eye movement (NREM) sleep, due to sleep fragmentation in the light phase. CUMS mice lost the normal rhythmic expressions of circadian proteins Per2, Cry2, Bmal1, Clock, and Per1. However, the melatonin treatment restored glymphatic system function and the polarization of AQP4, while improving sleep structure, and rectifying the abnormal expression of Per2, Bmal1, Clock, and Per1 in CUMS mice. Interestingly, Per2 expression correlated negatively with the polarization of AQP4. Further studies demonstrated that Per2 directed the location of AQP4 expression via interactions with the α-dystrobrevin (Dtna) subunit of AQP4 in primary cultured astrocytes. In conclusion, we report a new mechanism whereby melatonin improves depression outcomes by regulating the expression of the circadian protein Per2, maintaining the circadian rhythm of astrocytic AQP4 polarization, and restoring glymphatic function.
Collapse
Affiliation(s)
- Di Yao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Rong Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jiahuan Hao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hongqing Huang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xubiao Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lusen Ran
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuanyuan Fang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuqin He
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xinghua Liu
- Trauma Centre/ Department of Emergency and Trauma Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Minghuan Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
13
|
Adcock SJJ, Downey BC, Owens C, Tucker CB. Behavioral changes in the first 3 weeks after disbudding in dairy calves. J Dairy Sci 2023; 106:6365-6374. [PMID: 37500438 DOI: 10.3168/jds.2023-23237] [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: 01/05/2023] [Accepted: 03/27/2023] [Indexed: 07/29/2023]
Abstract
Hot-iron disbudding, the practice of cauterizing horn bud tissue to prevent horn growth in dairy calves, results in behavioral changes indicative of pain in the first few days after the procedure. However, few studies have quantified behavioral changes in the following weeks, while the burn wounds are still healing. Female Holstein calves were disbudded with a heated iron and pain relief (5.5 mL lidocaine cornual nerve block and 1 mg/kg oral meloxicam) at 4 to 10 d of age (n = 19) or not disbudded (n = 19). Calves wore ear tag accelerometers that reported the dominant behavior being performed at 1-min intervals from 3 to 21 d after disbudding. Compared with age-matched controls, disbudded calves tended to spend more time inactive throughout the observation period, ruminated less in the first 3 to 11 d after disbudding, and sucked more from a milk bottle beginning 5 d after disbudding until the end of the 21-d observation period. In addition to the accelerometer data, live observations of sleeping (using a behavioral proxy), lying, and ruminating were collected using instantaneous sampling at 5-min intervals for 24-h periods 3, 10, and 17 d after disbudding. Disbudded calves slept with their head down more on all live observation days and spent more time lying on the 17th d after disbudding, but ruminating did not differ compared with controls, in contrast to the accelerometer results. More time spent inactive, sleeping, and lying, and less time spent ruminating (as indicated by the accelerometer) can be interpreted as attempts to reduce painful stimulation of the disbudding wounds and allocate energy to healing. It is unclear whether the greater amount of sucking in the disbudded calves is nutritive (milk present) or non-nutritive (milk absent), as the algorithm did not distinguish the type of sucking, and further research is needed to explore the factors underlying this effect. We conclude that disbudding alters daily behavior patterns for at least 3 wk, far beyond the duration of recommended pain medication, raising additional welfare concerns about the procedure.
Collapse
Affiliation(s)
- Sarah J J Adcock
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706; Center for Animal Welfare, Department of Animal Science, University of California, Davis, Davis, CA 95616
| | - Blair C Downey
- Center for Animal Welfare, Department of Animal Science, University of California, Davis, Davis, CA 95616
| | - Chela Owens
- Center for Animal Welfare, Department of Animal Science, University of California, Davis, Davis, CA 95616
| | - Cassandra B Tucker
- Center for Animal Welfare, Department of Animal Science, University of California, Davis, Davis, CA 95616.
| |
Collapse
|
14
|
Jeanneteau F. Stress and the risk of Alzheimer dementia: Can deconstructed engrams be rebuilt? J Neuroendocrinol 2023; 35:e13235. [PMID: 36775895 DOI: 10.1111/jne.13235] [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: 11/28/2022] [Revised: 01/20/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023]
Abstract
The exact neuropathological mechanism by which the dementia process unfolds is under intense scrutiny. The disease affects about 38 million people worldwide, 70% of which are clinically diagnosed with Alzheimer's disease (AD). If the destruction of synapses essential for learning, planning and decision-making is part of the problem, must the restoration of previously lost synapses be part of the solution? It is plausible that neuronal capacity to restitute information corresponds with the adaptive capacity of its connectivity reserve. A challenge will be to promote the functional connectivity that can compensate for the lost one. This will require better clarification of the remodeling of functional connectivity during the progression of AD dementia and its reversal upon experimental treatment. A major difficulty is to promote the neural pathways that are atrophied in AD dementia while suppressing others that are bolstered. Therapeutic strategies should aim at scaling functional connectivity to a just balance between the atrophic and hypertrophic systems. However, the exact factors that can help reach this objective are still unclear. Similarities between the effects of chronic stress and some neuropathological mechanisms underlying AD dementia support the idea that common components deserve prime attention as therapeutic targets.
Collapse
Affiliation(s)
- Freddy Jeanneteau
- Institut de génomique fonctionnelle, Université de Montpellier, INSERM, CNRS, Montpellier, France
| |
Collapse
|
15
|
Liu X, He J, Jiang W, Wen S, Xiao Z. The Roles of Periaqueductal Gray and Dorsal Raphe Nucleus Dopaminergic Systems in the Mechanisms of Thermal Hypersensitivity and Depression in Mice. THE JOURNAL OF PAIN 2023; 24:1213-1228. [PMID: 36796500 DOI: 10.1016/j.jpain.2023.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 01/05/2023] [Accepted: 02/05/2023] [Indexed: 02/16/2023]
Abstract
Depression and thermal hypersensitivity share pathogenic features and symptomology, but their pathophysiologic interactions have not been fully elucidated. Dopaminergic systems in the ventrolateral periaqueductal gray (vlPAG) and dorsal raphe nucleus have been implicated in these conditions due to their antinociception and antidepression effects, although their specific roles and underlying mechanisms remain obscure. In this study, chronic unpredictable mild stress (CMS) was used to induce depression-like behaviors and thermal hypersensitivity in C57BL/6J (wild-type) or dopamine transporter promoter mice to establish a mouse model of pain and depression comorbidity. Microinjections of quinpirole, a dopamine D2 receptor agonist, up-regulated D2 receptor expression in dorsal raphe nucleus and reduced depressive behaviors and thermal hypersensitivity with CMS, while dorsal raphe nucleus injections of JNJ-37822681, an antagonist of D2 receptors, had the reciprocal effect on dopamine D2 receptor expression and behaviors. Moreover, using a chemical genetics approach to activate or inhibit dopaminergic neurons in vlPAG ameliorated or exacerbated depression-like behaviors and thermal hypersensitivity, respectively, in dopamine transporter promoter-Cre CMS mice. Collectively these results demonstrated the specific role of vlPAG and dorsal raphe nucleus dopaminergic systems in the regulation of pain and depression comorbidity in mice. PERSPECTIVE: The current study provides insights into the complex mechanisms underlying thermal hypersensitivity induced by depression, and the findings suggest that pharmacological and chemogenetic modulation of dopaminergic systems in the vlPAG and dorsal raphe nucleus may be a promising therapeutic strategy to simultaneously mitigate pain and depression.
Collapse
Affiliation(s)
- Xingfeng Liu
- Guizhou Key Laboratory of Brain Science, Zunyi Medical University, Zunyi, Guizhou, China; Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jingxin He
- Graduate School, Zunyi Medical University, Zunyi, Guizhou, China
| | - Wei Jiang
- Graduate School, Zunyi Medical University, Zunyi, Guizhou, China
| | - Song Wen
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Zhi Xiao
- Guizhou Key Laboratory of Brain Science, Zunyi Medical University, Zunyi, Guizhou, China; Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, Guizhou, China.
| |
Collapse
|
16
|
Xia TJ, Wang Z, Jin SW, Liu XM, Liu YG, Zhang SS, Pan RL, Jiang N, Liao YH, Yan MZ, Du LD, Chang Q. Melatonin-related dysfunction in chronic restraint stress triggers sleep disorders in mice. Front Pharmacol 2023; 14:1210393. [PMID: 37408758 PMCID: PMC10318904 DOI: 10.3389/fphar.2023.1210393] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 06/09/2023] [Indexed: 07/07/2023] Open
Abstract
Stress may trigger sleep disorders and are also risk factors for depression. The study explored the melatonin-related mechanisms of stress-associated sleep disorders on a mouse model of chronic stress by exploring the alteration in sleep architecture, melatonin, and related small molecule levels, transcription and expression of melatonin-related genes as well as proteins. Mice undergoing chronic restraint stress modeling for 28 days showed body weight loss and reduced locomotor activity. Sleep fragmentation, circadian rhythm disorders, and insomnia exhibited in CRS-treated mice formed sleep disorders. Tryptophan and 5-hydroxytryptamine levels were increased in the hypothalamus, while melatonin level was decreased. The transcription and expression of melatonin receptors were reduced, and circadian rhythm related genes were altered. Expression of downstream effectors to melatonin receptors was also affected. These results identified sleep disorders in a mice model of chronic stress. The alteration of melatonin-related pathways was shown to trigger sleep disorders.
Collapse
Affiliation(s)
- Tian-Ji Xia
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhi Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Su-Wei Jin
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin-Min Liu
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, China
| | - Yong-Guang Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shan-Shan Zhang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rui-Le Pan
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ning Jiang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yong-Hong Liao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ming-Zhu Yan
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li-Da Du
- Institute of Molecular Medicine and Innovative Pharmaceutics, Qingdao University, Qingdao, China
- Department of Surgery, University of Toronto, Toronto, TO, Canada
| | - Qi Chang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
17
|
陶 然, 丁 胜, 陈 洁, 朱 学, 倪 照, 胡 令, 张 杨, 许 燕, 孙 洪. [Identifying Depressive Disorder With Sleep Electroencephalogram Data: A Study Based on Deep Learning]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2023; 54:287-292. [PMID: 36949687 PMCID: PMC10409169 DOI: 10.12182/20230360212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Indexed: 03/24/2023]
Abstract
Objective To explore the effectiveness of using deep learning network combined Vision Transformer (ViT) and Transformer to identify patients with depressive disorder on the basis of their sleep electroencephalogram (EEG) signals. Methods The sleep EEG signals of 28 patients with depressive disorder and 37 normal controls were preprocessed. Then, the signals were converted into image format and the feature information on frequency domain and spatial domain was retained. After that, the images were transmitted to the ViT-Transformer coding network for deep learning of the EEG signal characteristics of the rapid eye movement (REM) sleep and non-rapid eye movement (NREM) sleep in patients with depressive disorder and those in normal controls, respectively, and to identify patients with depressive disorder. Results Based on the ViT-Transformer network, after examining different EEG frequencies, we found that the combination of delta, theta, and beta waves produced better results in identifying depressive disorder. Among the different EEG frequencies, EEG signal features of delta-theta-beta combination waves in REM sleep achieved 92.8% accuracy and 93.8% precision for identifying depression, with the recall rate of patients with depression being 84.7%, and the F0.5 value being 0.917±0.074. When using the delta-theta-beta combination EEG signal features in NREM sleep to identify depressive disorder, the accuracy was 91.7%, the precision was 90.8%, the recall rate was 85.2%, and the F0.5 value was 0.914±0.062. In addition, through visualization of the sleep EEG of different sleep stages for the whole night, it was found that classification errors usually occurred during transition to a different sleep stage. Conclusion Using the deep learning ViT-Transformer network, we found that the EEG signal features in REM sleep based on delta-theta-beta combination waves showed better effect in identifying depressive disorder.
Collapse
Affiliation(s)
- 然 陶
- 北京大学第六医院医技科,北京大学精神卫生研究所,国家卫生健康委员会精神卫生学重点实验室(北京大学) (北京 100191)Department of Medical Technology, Peking University Sixth Hospital, Peking University Institute of Mental Health, and Key Laboratory of Mental Health of the National Health Commission (Peking University), Beijing 100191, China
| | - 胜男 丁
- 北京大学第六医院医技科,北京大学精神卫生研究所,国家卫生健康委员会精神卫生学重点实验室(北京大学) (北京 100191)Department of Medical Technology, Peking University Sixth Hospital, Peking University Institute of Mental Health, and Key Laboratory of Mental Health of the National Health Commission (Peking University), Beijing 100191, China
| | - 洁 陈
- 北京大学第六医院医技科,北京大学精神卫生研究所,国家卫生健康委员会精神卫生学重点实验室(北京大学) (北京 100191)Department of Medical Technology, Peking University Sixth Hospital, Peking University Institute of Mental Health, and Key Laboratory of Mental Health of the National Health Commission (Peking University), Beijing 100191, China
| | - 学敏 朱
- 北京大学第六医院医技科,北京大学精神卫生研究所,国家卫生健康委员会精神卫生学重点实验室(北京大学) (北京 100191)Department of Medical Technology, Peking University Sixth Hospital, Peking University Institute of Mental Health, and Key Laboratory of Mental Health of the National Health Commission (Peking University), Beijing 100191, China
| | - 照军 倪
- 北京大学第六医院医技科,北京大学精神卫生研究所,国家卫生健康委员会精神卫生学重点实验室(北京大学) (北京 100191)Department of Medical Technology, Peking University Sixth Hospital, Peking University Institute of Mental Health, and Key Laboratory of Mental Health of the National Health Commission (Peking University), Beijing 100191, China
| | - 令明 胡
- 北京大学第六医院医技科,北京大学精神卫生研究所,国家卫生健康委员会精神卫生学重点实验室(北京大学) (北京 100191)Department of Medical Technology, Peking University Sixth Hospital, Peking University Institute of Mental Health, and Key Laboratory of Mental Health of the National Health Commission (Peking University), Beijing 100191, China
| | - 杨 张
- 北京大学第六医院医技科,北京大学精神卫生研究所,国家卫生健康委员会精神卫生学重点实验室(北京大学) (北京 100191)Department of Medical Technology, Peking University Sixth Hospital, Peking University Institute of Mental Health, and Key Laboratory of Mental Health of the National Health Commission (Peking University), Beijing 100191, China
| | - 燕 许
- 北京大学第六医院医技科,北京大学精神卫生研究所,国家卫生健康委员会精神卫生学重点实验室(北京大学) (北京 100191)Department of Medical Technology, Peking University Sixth Hospital, Peking University Institute of Mental Health, and Key Laboratory of Mental Health of the National Health Commission (Peking University), Beijing 100191, China
| | - 洪强 孙
- 北京大学第六医院医技科,北京大学精神卫生研究所,国家卫生健康委员会精神卫生学重点实验室(北京大学) (北京 100191)Department of Medical Technology, Peking University Sixth Hospital, Peking University Institute of Mental Health, and Key Laboratory of Mental Health of the National Health Commission (Peking University), Beijing 100191, China
| |
Collapse
|
18
|
Xu YX, Liu GY, Ji ZZ, Li YY, Wang YL, Wu XY, Liu JL, Ma DX, Zhong MK, Gao CB, Xu Q. Restraint stress induced anxiety and sleep in mice. Front Psychiatry 2023; 14:1090420. [PMID: 37124267 PMCID: PMC10130584 DOI: 10.3389/fpsyt.2023.1090420] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 03/20/2023] [Indexed: 05/02/2023] Open
Abstract
In humans and animals, exposure to changes in internal or external environments causes acute stress, which changes sleep and enhances neurochemical, neuroendocrine, and sympathetic activities. Repeated stress responses play an essential role in the pathogenesis of psychiatric diseases and sleep disorders. However, the underlying mechanism of sleep changes and anxiety disorders in response to acute stress is not well established. In the current study, the effects of restraint stress (RS) on anxiety and sleep-wake cycles in mice were investigated. We found that after RS, the mice showed anxiety-like behavior after RS manipulation and increased the amounts of both non-rapid eye movement (NREM) and rapid eye movement (REM) sleep in the dark period. The increase in sleep time was mainly due to the increased number of episodes of NREM and REM sleep during the dark period. In addition, the mice showed an elevation of the EEG power spectrum of both NREM and REM sleep 2 h after RS manipulation. There was a significant reduction in the EEG power spectrum of both NREM and REM sleep during the darkperiod in the RS condition. The expression of the c-Fos protein was significantly increased in the parabrachial nucleus, bed nucleus of the stria terminalis, central amygdala, and paraventricular hypothalamus by RS manipulation. Altogether, the findings from the present study indicated that neural circuits from the parabrachial nucleus might regulate anxiety and sleep responses to acute stress, and suggest a potential therapeutic target for RS induced anxiety and sleep alterations.
Collapse
Affiliation(s)
- Yong-Xia Xu
- Department of Geriatric Endocrinology, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Guo-Ying Liu
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Zhang-Zhang Ji
- Department of Stomatology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yue-Yun Li
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yan-Li Wang
- Department of Geriatric Endocrinology, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xue-Yan Wu
- Department of Human Anatomy, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Jun-Lin Liu
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Dan-Xia Ma
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Ming-Kui Zhong
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
- *Correspondence: Ming-Kui Zhong,
| | - Chao-Bing Gao
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Chao-Bing Gao,
| | - Qi Xu
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
- Qi Xu,
| |
Collapse
|
19
|
Nollet M, Franks NP, Wisden W. Understanding Sleep Regulation in Normal and Pathological Conditions, and Why It Matters. J Huntingtons Dis 2023; 12:105-119. [PMID: 37302038 PMCID: PMC10473105 DOI: 10.3233/jhd-230564] [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] [Accepted: 05/22/2023] [Indexed: 06/12/2023]
Abstract
Sleep occupies a peculiar place in our lives and in science, being both eminently familiar and profoundly enigmatic. Historically, philosophers, scientists and artists questioned the meaning and purpose of sleep. If Shakespeare's verses from MacBeth depicting "Sleep that soothes away all our worries" and "relieves the weary laborer and heals hurt minds" perfectly epitomize the alleviating benefits of sleep, it is only during the last two decades that the growing understanding of the sophisticated sleep regulatory mechanisms allows us to glimpse putative biological functions of sleep. Sleep control brings into play various brain-wide processes occurring at the molecular, cellular, circuit, and system levels, some of them overlapping with a number of disease-signaling pathways. Pathogenic processes, including mood disorders (e.g., major depression) and neurodegenerative illnesses such Huntington's or Alzheimer's diseases, can therefore affect sleep-modulating networks which disrupt the sleep-wake architecture, whereas sleep disturbances may also trigger various brain disorders. In this review, we describe the mechanisms underlying sleep regulation and the main hypotheses drawn about its functions. Comprehending sleep physiological orchestration and functions could ultimately help deliver better treatments for people living with neurodegenerative diseases.
Collapse
Affiliation(s)
- Mathieu Nollet
- UK Dementia Research Institute and Department of Life Sciences, Imperial College London, London, UK
| | - Nicholas P. Franks
- UK Dementia Research Institute and Department of Life Sciences, Imperial College London, London, UK
| | - William Wisden
- UK Dementia Research Institute and Department of Life Sciences, Imperial College London, London, UK
| |
Collapse
|
20
|
Liu D, Hu H, Hong Y, Xiao Q, Tu J. Sugar Beverage Habitation Relieves Chronic Stress-Induced Anxiety-like Behavior but Elicits Compulsive Eating Phenotype via vLS GAD2 Neurons. Int J Mol Sci 2022; 24:ijms24010661. [PMID: 36614104 PMCID: PMC9820526 DOI: 10.3390/ijms24010661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/10/2022] [Accepted: 12/13/2022] [Indexed: 01/03/2023] Open
Abstract
Chronically stressed individuals are reported to overconsume tasty, palatable foods like sucrose to blunt the psychological and physiological impacts of stress. Negative consequences of high-sugar intake on feeding behavior include increased metabolic disease burdens like obesity. However, the neural basis underlying long-term high-sugar intake-induced overeating during stress is not fully understood. To investigate this question, we used the two-bottle sucrose choice paradigm in mice exposed to chronic unpredictable mild stressors (CUMS) that mimic those of daily life stressors. After 21 days of CUMS paralleled by consecutive sucrose drinking, we explored anxiety-like behavior using the elevated plus maze and open field tests. The normal water-drinking stressed mice displayed more anxiety than the sucrose-drinking stressed mice. Although sucrose-drinking displayed anxiolytic effects, the sucrose-drinking mice exhibited binge eating (chow) and a compulsive eating phenotype. The sucrose-drinking mice also showed a significant body-weight gain compared to the water-drinking control mice during stress. We further found that c-Fos expression was significantly increased in the ventral part of the lateral septum (vLS) of the sucrose-treated stressed mice after compulsive eating. Pharmacogenetic activation of the vLS glutamate decarboxylase 2(GAD2) neurons maintained plain chow intake but induced a compulsive eating phenotype in the naïve GAD2-Cre mice when mice feeding was challenged by flash stimulus, mimicking the negative consequences of excessive sucrose drinking during chronic stress. Further, pharmacogenetic activation of the vLSGAD2 neurons aggravated anxiety of the stressed GAD2-Cre mice but did not alter the basal anxiety level of the naïve ones. These findings indicate the GABAergic neurons within the vLS may be a potential intervention target for anxiety comorbid eating disorders during stress.
Collapse
Affiliation(s)
- Dan Liu
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Haohao Hu
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yuchuan Hong
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qian Xiao
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jie Tu
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Correspondence:
| |
Collapse
|
21
|
Functional roles of REM sleep. Neurosci Res 2022; 189:44-53. [PMID: 36572254 DOI: 10.1016/j.neures.2022.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 12/01/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
Rapid eye movement (REM) sleep is an enigmatic and intriguing sleep state. REM sleep differs from non-REM sleep by its characteristic brain activity and from wakefulness by a reduced anti-gravity muscle tone. In addition to these key traits, diverse physiological phenomena appear across the whole body during REM sleep. However, it remains unclear whether these phenomena are the causes or the consequences of REM sleep. Experimental approaches using humans and animal models have gradually revealed the functional roles of REM sleep. Extensive efforts have been made to interpret the characteristic brain activity in the context of memory functions. Numerous physical and psychological functions of REM sleep have also been proposed. Moreover, REM sleep has been implicated in aspects of brain development. Here, we review the variety of functional roles of REM sleep, mainly as revealed by animal models. In addition, we discuss controversies regarding the functional roles of REM sleep.
Collapse
|
22
|
Stankiewicz AM, Jaszczyk A, Goscik J, Juszczak GR. Stress and the brain transcriptome: Identifying commonalities and clusters in standardized data from published experiments. Prog Neuropsychopharmacol Biol Psychiatry 2022; 119:110558. [PMID: 35405299 DOI: 10.1016/j.pnpbp.2022.110558] [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: 07/06/2021] [Revised: 03/17/2022] [Accepted: 04/04/2022] [Indexed: 12/28/2022]
Abstract
Interpretation of transcriptomic experiments is hindered by many problems including false positives/negatives inherent to big-data methods and changes in gene nomenclature. To find the most consistent effect of stress on brain transcriptome, we retrieved data from 79 studies applying animal models and 3 human studies investigating post-traumatic stress disorder (PTSD). The analyzed data were obtained either with microarrays or RNA sequencing applied to samples collected from more than 1887 laboratory animals and from 121 human subjects. Based on the initial database containing a quarter million differential expression effect sizes representing transcripts in three species, we identified the most frequently reported genes in 223 stress-control comparisons. Additionally, the analysis considers sex, individual vulnerability and contribution of glucocorticoids. We also found an overlap between gene expression in PTSD patients and animals which indicates relevance of laboratory models for human stress response. Our analysis points to genes that, as far as we know, were not specifically tested for their role in stress response (Pllp, Arrdc2, Midn, Mfsd2a, Ccn1, Htra1, Csrnp1, Tenm4, Tnfrsf25, Sema3b, Fmo2, Adamts4, Gjb1, Errfi1, Fgf18, Galnt6, Slc25a42, Ifi30, Slc4a1, Cemip, Klf10, Tom1, Dcdc2c, Fancd2, Luzp2, Trpm1, Abcc12, Osbpl1a, Ptp4a2). Provided transcriptomic resource will be useful for guiding the new research.
Collapse
Affiliation(s)
- Adrian M Stankiewicz
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzebiec, Poland
| | - Aneta Jaszczyk
- Department of Animal Behavior and Welfare, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzebiec, Poland
| | - Joanna Goscik
- Faculty of Computer Science, Bialystok University of Technology, Bialystok, Poland
| | - Grzegorz R Juszczak
- Department of Animal Behavior and Welfare, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzebiec, Poland.
| |
Collapse
|
23
|
Mahony C, O'Ryan C. A molecular framework for autistic experiences: Mitochondrial allostatic load as a mediator between autism and psychopathology. Front Psychiatry 2022; 13:985713. [PMID: 36506457 PMCID: PMC9732262 DOI: 10.3389/fpsyt.2022.985713] [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: 07/04/2022] [Accepted: 11/07/2022] [Indexed: 11/27/2022] Open
Abstract
Molecular autism research is evolving toward a biopsychosocial framework that is more informed by autistic experiences. In this context, research aims are moving away from correcting external autistic behaviors and toward alleviating internal distress. Autism Spectrum Conditions (ASCs) are associated with high rates of depression, suicidality and other comorbid psychopathologies, but this relationship is poorly understood. Here, we integrate emerging characterizations of internal autistic experiences within a molecular framework to yield insight into the prevalence of psychopathology in ASC. We demonstrate that descriptions of social camouflaging and autistic burnout resonate closely with the accepted definitions for early life stress (ELS) and chronic adolescent stress (CAS). We propose that social camouflaging could be considered a distinct form of CAS that contributes to allostatic overload, culminating in a pathophysiological state that is experienced as autistic burnout. Autistic burnout is thought to contribute to psychopathology via psychological and physiological mechanisms, but these remain largely unexplored by molecular researchers. Building on converging fields in molecular neuroscience, we discuss the substantial evidence implicating mitochondrial dysfunction in ASC to propose a novel role for mitochondrial allostatic load in the relationship between autism and psychopathology. An interplay between mitochondrial, neuroimmune and neuroendocrine signaling is increasingly implicated in stress-related psychopathologies, and these molecular players are also associated with neurodevelopmental, neurophysiological and neurochemical aspects of ASC. Together, this suggests an increased exposure and underlying molecular susceptibility to ELS that increases the risk of psychopathology in ASC. This article describes an integrative framework shaped by autistic experiences that highlights novel avenues for molecular research into mechanisms that directly affect the quality of life and wellbeing of autistic individuals. Moreover, this framework emphasizes the need for increased access to diagnoses, accommodations, and resources to improve mental health outcomes in autism.
Collapse
Affiliation(s)
| | - Colleen O'Ryan
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
24
|
Zhang H, Wei M, Sun N, Wang H, Fan H. Melatonin attenuates chronic stress-induced hippocampal inflammatory response and apoptosis by inhibiting ADAM17/TNF-α axis. Food Chem Toxicol 2022; 169:113441. [PMID: 36162616 DOI: 10.1016/j.fct.2022.113441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/14/2022] [Accepted: 09/19/2022] [Indexed: 12/22/2022]
Abstract
Melatonin, as a dietary supplement, has a potent neuroprotective effect and exerts a certain antidepressant effect. This study explored the molecular mechanisms and targets of melatonin on chronic stress-induced hippocampal damage from the perspective of inhibiting inflammatory cytokines release. Our results indicated that melatonin alleviated chronic restraint stress (CRS)-induced inflammatory response and apoptosis, thus improving hippocampal structural damage and subsequent depression-like behaviors in rats. The radar map displayed that the change of TNF-α content was the most significant. Meanwhile, correlation analysis showed that TNF-α content was highly positively correlated with apoptosis. Molecular autodocking studies suggested that TNF-α converting enzyme ADAM17 as a potential target has a priority in docking with melatonin. Molecular mechanism studies indicated that melatonin inhibited CRS-induced activation of the ADAM17/TNF-α axis and its downstream proteins p38 and p53 phosphorylation in the hippocampus. Analogously, Both ADAM17 inhibitor TMI-1 and TNF-α inhibitor thalidomide relieved the effects of CRS on ADAM17/TNF-α axis and its downstream proteins phosphorylation, hippocampal apoptosis, hippocampal inflammatory response, and depression-like behaviors in rats. Altogether, these findings reveal that melatonin relieves CRS-induced inflammatory response and apoptosis, and subsequent depression-like behaviors by inhibiting ADAM17/TNF-α axis.
Collapse
Affiliation(s)
- Haiyang Zhang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Mian Wei
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Ning Sun
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Hui Wang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Honggang Fan
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
25
|
Heritability of REM sleep neurophysiology in adolescence. Transl Psychiatry 2022; 12:399. [PMID: 36130941 PMCID: PMC9492899 DOI: 10.1038/s41398-022-02106-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/20/2022] [Accepted: 08/04/2022] [Indexed: 12/04/2022] Open
Abstract
Alterations of rapid eye movement (REM) sleep have long been observed in patients with psychiatric disorders and proposed as an endophenotype-a link between behavior and genes. Recent experimental work has shown that REM sleep plays an important role in the emotional processing of memories, emotion regulation, and is altered in the presence of stress, suggesting a mechanism by which REM sleep may impact psychiatric illness. REM sleep shows a developmental progression and increases during adolescence-a period of rapid maturation of the emotional centers of the brain. This study uses a behavioral genetics approach to understand the relative contribution of genes, shared environmental and unique environmental factors to REM sleep neurophysiology in adolescents. Eighteen monozygotic (MZ; n = 36; 18 females) and 12 dizygotic (DZ; n = 24; 12 females) same-sex twin pairs (mean age = 12.46; SD = 1.36) underwent whole-night high-density sleep EEG recordings. We find a significant genetic contribution to REM sleep EEG power across frequency bands, explaining, on average, between 75 to 88% of the variance in power, dependent on the frequency band. In the lower frequency bands between delta and sigma, however, we find an additional impact of shared environmental factors over prescribed regions. We hypothesize that these regions may reflect the contribution of familial and environmental stress shared amongst the twins. The observed strong genetic contribution to REM sleep EEG power in early adolescence establish REM sleep neurophysiology as a potentially strong endophenotype, even in adolescence-a period marked by significant brain maturation.
Collapse
|
26
|
Vrontou S, Bédécarrats A, Wei X, Ayodeji M, Brassai A, Molnár L, Mody I. Altered brain rhythms and behaviour in the accelerated ovarian failure mouse model of human menopause. Brain Commun 2022; 4:fcac166. [PMID: 35794872 PMCID: PMC9253886 DOI: 10.1093/braincomms/fcac166] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 03/30/2022] [Accepted: 06/20/2022] [Indexed: 11/14/2022] Open
Abstract
To date, potential mechanisms of menopause-related memory and cognitive deficits have not been elucidated. Therefore, we studied brain oscillations, their phase–amplitude coupling, sleep and vigilance state patterns, running wheel use and other behavioural measures in a translationally valid mouse model of menopause, the 4-vinylcyclohexene-diepoxide-induced accelerated ovarian failure. After accelerated ovarian failure, female mice show significant alterations in brain rhythms, including changes in the frequencies of θ (5–12 Hz) and γ (30–120 Hz) oscillations, a reversed phase–amplitude coupling, altered coupling of hippocampal sharp-wave ripples to medial prefrontal cortical sleep spindles and reduced δ oscillation (0.5–4 Hz) synchrony between the two regions during non-rapid eye movement sleep. In addition, we report on significant circadian variations in the frequencies of θ and γ oscillations, and massive synchronous δ oscillations during wheel running. Our results reveal novel and specific network alterations and feasible signs for diminished brain connectivity in the accelerated ovarian failure mouse model of menopause. Taken together, our results may have identified changes possibly responsible for some of the memory and cognitive deficits previously described in this model. Corresponding future studies in menopausal women could shed light on fundamental mechanisms underlying the neurological and psychiatric comorbidities present during this important transitional phase in women’s lives.
Collapse
Affiliation(s)
- Sophia Vrontou
- Department of Neurology, The David Geffen School of Medicine at UCLA , Los Angeles, CA 90095 , USA
| | - Alexis Bédécarrats
- Department of Neurology, The David Geffen School of Medicine at UCLA , Los Angeles, CA 90095 , USA
| | - Xiaofei Wei
- Department of Neurology, The David Geffen School of Medicine at UCLA , Los Angeles, CA 90095 , USA
| | | | - Attila Brassai
- Department of Pharmacology, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology , Târgu Mureş 540139 , Romania
| | - László Molnár
- Department of Electrical Engineering, Sapientia Hungarian University of Transylvania , Târgu Mureş 540485 , Romania
| | - Istvan Mody
- Department of Neurology, The David Geffen School of Medicine at UCLA , Los Angeles, CA 90095 , USA
- Department of Physiology, The David Geffen School of Medicine at UCLA , Los Angeles, CA 90095 , USA
| |
Collapse
|
27
|
Yu X, Zhao G, Wang D, Wang S, Li R, Li A, Wang H, Nollet M, Chun YY, Zhao T, Yustos R, Li H, Zhao J, Li J, Cai M, Vyssotski AL, Li Y, Dong H, Franks NP, Wisden W. A specific circuit in the midbrain detects stress and induces restorative sleep. Science 2022; 377:63-72. [PMID: 35771921 PMCID: PMC7612951 DOI: 10.1126/science.abn0853] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In mice, social defeat stress (SDS), an ethological model for psychosocial stress, induces sleep. Such sleep could enable resilience, but how stress promotes sleep is unclear. Activity-dependent tagging revealed a subset of ventral tegmental area γ-aminobutyric acid (GABA)-somatostatin (VTAVgat-Sst) cells that sense stress and drive non-rapid eye movement (NREM) and REM sleep through the lateral hypothalamus and also inhibit corticotropin-releasing factor (CRF) release in the paraventricular hypothalamus. Transient stress enhances the activity of VTAVgat-Sst cells for several hours, allowing them to exert their sleep effects persistently. Lesioning of VTAVgat-Sst cells abolished SDS-induced sleep; without it, anxiety and corticosterone concentrations remained increased after stress. Thus, a specific circuit allows animals to restore mental and body functions by sleeping, potentially providing a refined route for treating anxiety disorders.
Collapse
Affiliation(s)
- Xiao Yu
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Guangchao Zhao
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Dan Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Sa Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Rui Li
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ao Li
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Huan Wang
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Mathieu Nollet
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
- UK Dementia Research Institute, Imperial College London, London SW7 2AZ, UK
| | - You Young Chun
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Tianyuan Zhao
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Raquel Yustos
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Huiming Li
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jianshuai Zhao
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jiannan Li
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Min Cai
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Alexei L Vyssotski
- Institute of Neuroinformatics, University of Zürich/ETH Zürich, Zürich, Switzerland
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Hailong Dong
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Nicholas P Franks
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
- UK Dementia Research Institute, Imperial College London, London SW7 2AZ, UK
| | - William Wisden
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
- UK Dementia Research Institute, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
28
|
Casaglia E, Luppi PH. Is paradoxical sleep setting up innate and acquired complex sensorimotor and adaptive behaviours?: A proposed function based on literature review. J Sleep Res 2022; 31:e13633. [PMID: 35596591 DOI: 10.1111/jsr.13633] [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: 04/18/2022] [Revised: 04/25/2022] [Accepted: 04/25/2022] [Indexed: 11/30/2022]
Abstract
We summarize here the progress in identifying the neuronal network as well as the function of paradoxical sleep and the gaps of knowledge that should be filled in priority. The core system generating paradoxical sleep localized in the brainstem is now well identified, and the next step is to clarify the role of the forebrain in particular that of the hypothalamus including the melanin-concentrating hormone neurons and of the basolateral amygdala. We discuss these two options, and also the discovery that cortical activation during paradoxical sleep is restricted to a few limbic cortices activated by the lateral supramammillary nucleus and the claustrum. Such activation nicely supports the findings recently obtained showing that neuronal reactivation occurs during paradoxical sleep in these structures, and induces both memory consolidation of important memory and forgetting of less relevant ones. The question that still remains to be answered is whether paradoxical sleep is playing more crucial roles in processing emotional and procedural than other types of memories. One attractive hypothesis is that paradoxical sleep is responsible for erasing negative emotional memories, and that this function is not properly functioning in depressed patients. On the other hand, the presence of a muscle atonia during paradoxical sleep is in favour of a role in procedural memory as new types of motor behaviours can be tried without harm during the state. In a way, it also fits with the proposed role of paradoxical sleep in setting up the sensorimotor system during development.
Collapse
Affiliation(s)
- Elisa Casaglia
- INSERM, U1028, CNRS UMR5292, Lyon Neuroscience Research Center, Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France.,University Lyon 1, Lyon, France.,University of Cagliari, Cagliari, Italy
| | - Pierre-Hervé Luppi
- INSERM, U1028, CNRS UMR5292, Lyon Neuroscience Research Center, Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France.,University Lyon 1, Lyon, France
| |
Collapse
|
29
|
Osorio-Forero A, Cherrad N, Banterle L, Fernandez LMJ, Lüthi A. When the Locus Coeruleus Speaks Up in Sleep: Recent Insights, Emerging Perspectives. Int J Mol Sci 2022; 23:ijms23095028. [PMID: 35563419 PMCID: PMC9099715 DOI: 10.3390/ijms23095028] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 12/03/2022] Open
Abstract
For decades, numerous seminal studies have built our understanding of the locus coeruleus (LC), the vertebrate brain’s principal noradrenergic system. Containing a numerically small but broadly efferent cell population, the LC provides brain-wide noradrenergic modulation that optimizes network function in the context of attentive and flexible interaction with the sensory environment. This review turns attention to the LC’s roles during sleep. We show that these roles go beyond down-scaled versions of the ones in wakefulness. Novel dynamic assessments of noradrenaline signaling and LC activity uncover a rich diversity of activity patterns that establish the LC as an integral portion of sleep regulation and function. The LC could be involved in beneficial functions for the sleeping brain, and even minute alterations in its functionality may prove quintessential in sleep disorders.
Collapse
|
30
|
Eacret D, Lemchi C, Caulfield JI, Cavigelli SA, Veasey SC, Blendy JA. Chronic Sleep Deprivation Blocks Voluntary Morphine Consumption but Not Conditioned Place Preference in Mice. Front Neurosci 2022; 16:836693. [PMID: 35250468 PMCID: PMC8892254 DOI: 10.3389/fnins.2022.836693] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/24/2022] [Indexed: 12/26/2022] Open
Abstract
The opioid epidemic remains a significant healthcare problem and is attributable to over 100,000 deaths per year. Poor sleep increases sensitivity to pain, impulsivity, inattention, and negative affect, all of which might perpetuate drug use. Opioid users have disrupted sleep during drug use and withdrawal and report poor sleep as a reason for relapse. However, preclinical studies investigating the relationship between sleep loss and substance use and the associated underlying neurobiological mechanisms of potential interactions are lacking. One of the most common forms of sleep loss in modern society is chronic short sleep (CSS) (<7 h/nightly for adults). Here, we used an established model of CSS to investigate the influence of disrupted sleep on opioid reward in male mice. The CSS paradigm did not increase corticosterone levels or depressive-like behavior after a single sleep deprivation session but did increase expression of Iba1, which typically reflects microglial activation, in the hypothalamus after 4 weeks of CSS. Rested control mice developed a morphine preference in a 2-bottle choice test, while mice exposed to CSS did not develop a morphine preference. Both groups demonstrated morphine conditioned place preference (mCPP), but there were no differences in conditioned preference between rested and CSS mice. Taken together, our results show that recovery sleep after chronic sleep disruption lessens voluntary opioid intake, without impacting conditioned reward associated with morphine.
Collapse
Affiliation(s)
- Darrell Eacret
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Crystal Lemchi
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jasmine I. Caulfield
- Huck Institute for Life Sciences, Pennsylvania State University, University Park, PA, United States
| | - Sonia A. Cavigelli
- Huck Institute for Life Sciences, Pennsylvania State University, University Park, PA, United States
| | - Sigrid C. Veasey
- Department of Medicine, Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Julie A. Blendy
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- *Correspondence: Julie A. Blendy,
| |
Collapse
|
31
|
Tseng YT, Zhao B, Chen S, Ye J, Liu J, Liang L, Ding H, Schaefke B, Yang Q, Wang L, Wang F, Wang L. The subthalamic corticotropin-releasing hormone neurons mediate adaptive REM-sleep responses to threat. Neuron 2022; 110:1223-1239.e8. [PMID: 35065715 DOI: 10.1016/j.neuron.2021.12.033] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 09/10/2021] [Accepted: 12/23/2021] [Indexed: 01/25/2023]
Abstract
When an animal faces a threatening situation while asleep, rapid arousal is the essential prerequisite for an adequate response. Here, we find that predator stimuli induce immediate arousal from REM sleep compared with NREM sleep. Using in vivo neural activity recording and cell-type-specific manipulations, we identify neurons in the medial subthalamic nucleus (mSTN) expressing corticotropin-releasing hormone (CRH) that mediate arousal and defensive responses to acute predator threats received through multiple sensory modalities across REM sleep and wakefulness. We observe involvement of the same neurons in the normal regulation of REM sleep and the adaptive increase in REM sleep induced by sustained predator stress. Projections to the lateral globus pallidus (LGP) are the effector pathway for the threat-coping responses and REM-sleep expression. Together, our findings suggest adaptive REM-sleep responses could be protective against threats and uncover a critical component of the neural circuitry at their basis.
Collapse
Affiliation(s)
- Yu-Ting Tseng
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Binghao Zhao
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Shanping Chen
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jialin Ye
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingjing Liu
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lisha Liang
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hui Ding
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Bernhard Schaefke
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Qin Yang
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Lina Wang
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Feng Wang
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Liping Wang
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China.
| |
Collapse
|
32
|
Jun JS, Sunwoo JS, Byun JI, Shin JW, Kim TJ, Schenck CH, Jung KY. Emotional and Environmental Factors Aggravating Dream Enactment Behaviors in Patients with Isolated REM Sleep Behavior Disorder. Nat Sci Sleep 2022; 14:1713-1720. [PMID: 36187325 PMCID: PMC9519124 DOI: 10.2147/nss.s372823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/14/2022] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE To identify emotional and environmental factors that aggravate dream enactment behaviors (DEBs) in isolated rapid eye movement (REM) sleep behavior disorder (iRBD). METHODS In this cross-sectional study, a total of 96 polysomnography-confirmed iRBD patients (mean age, 68.5 years; men, 68%) and their caregivers completed questionnaires regarding potential aggravating factors related to DEBs, including emotion/feelings (stress, anger, anxiety, depressive mood, fatigue, pain), food (alcohol, caffeine, overeating in the evening, fasting/hunger), activities and sleep patterns (strenuous exercise, sex before bed, conflict/fighting, sleep deprivation, oversleeping, sleeping away from home, watching TV before bed), weather/environmental factors (cloudy or rainy weather, heat, cold, noise) and medication (skipping medication, taking hypnotics). RESULTS The patients reported that stress (61%) was the most aggravating factor for DEBs, followed by anxiety (56%), anger (51%), fatigue (49%), and watching TV before bed (46%). Similarly, the caregivers reported that these factors were most relevant to the aggravation of DEBs in the patients, although some factors were ranked differently. In the subgroup analyses, aggravating factors for DEBs did not differ by RBD symptom severity. Interestingly, the proportion of patients experiencing DEB aggravation by stress, anxiety and depressive mood was significantly higher in women than in men. Furthermore, depressed patients reported that stress and cloudy or rainy weather made DEBs worse than nondepressed patients. CONCLUSION Our results suggest that DEBs in iRBD patients may be mainly aggravated by emotional factors. These negative effects appeared to be more prominent in female and depressed patients.
Collapse
Affiliation(s)
- Jin-Sun Jun
- Department of Neurology, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Jun-Sang Sunwoo
- Department of Neurology, Kangbuk Samsung Hospital, Seoul, Republic of Korea
| | - Jung-Ick Byun
- Department of Neurology, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| | - Jung-Won Shin
- Department of Neurology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Tae-Joon Kim
- Department of Neurology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Carlos H Schenck
- Minnesota Regional Sleep Disorders Center, and Department of Psychiatry, Hennepin County Medical Center and University of Minnesota Medical School, Minneapolis, MN, USA
| | - Ki-Young Jung
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
33
|
Ibarguen-Vargas Y, Leman S, Palme R, Belzung C, Surget A. CRF-R1 Antagonist Treatment Exacerbates Circadian Corticosterone Secretion under Chronic Stress, but Preserves HPA Feedback Sensitivity. Pharmaceutics 2021; 13:pharmaceutics13122114. [PMID: 34959395 PMCID: PMC8707167 DOI: 10.3390/pharmaceutics13122114] [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: 11/03/2021] [Revised: 12/01/2021] [Accepted: 12/04/2021] [Indexed: 11/16/2022] Open
Abstract
Despite promising initial reports, corticotropin-releasing factor receptor type-1 (CRF-R1) antagonists have mostly failed to display efficacy in clinical trials for anxiety or depression. Rather than broad-spectrum antidepressant/anxiolytic-like drugs, they may represent an ‘antistress’ solution for single stressful situations or for patients with chronic stress conditions. However, the impact of prolonged CRF-R1 antagonist treatments on the hypothalamic–pituitary–adrenal (HPA) axis under chronic stress conditions remained to be characterized. Hence, our study investigated whether a chronic CRF-R1 antagonist (crinecerfont, formerly known as SSR125543, 20 mg·kg−1·day−1 ip, 5 weeks) would alter HPA axis basal circadian activity and negative feedback sensitivity in mice exposed to either control or chronic stress conditions (unpredictable chronic mild stress, UCMS, 7 weeks), through measures of fecal corticosterone metabolites, plasma corticosterone, and dexamethasone suppression test. Despite preserving HPA axis parameters in control non-stressed mice, the 5-week crinercerfont treatment improved the negative feedback sensitivity in chronically stressed mice, but paradoxically exacerbated their basal corticosterone secretion nearly all along the circadian cycle. The capacity of chronic CRF-R1 antagonists to improve the HPA negative feedback in UCMS argues in favor of a potential therapeutic benefit against stress-related conditions. However, the treatment-related overactivation of HPA circadian activity in UCMS raise questions about possible physiological outcomes with long-standing treatments under ongoing chronic stress.
Collapse
Affiliation(s)
- Yadira Ibarguen-Vargas
- UMR1253, iBrain, Université de Tours, Inserm, 37200 Tours, France; (Y.I.-V.); (S.L.)
- EUK-CVL, Université d’Orléans, 45100 Orléans, France
| | - Samuel Leman
- UMR1253, iBrain, Université de Tours, Inserm, 37200 Tours, France; (Y.I.-V.); (S.L.)
| | - Rupert Palme
- Department of Biomedical Sciences/Biochemistry, University of Veterinary Medicine, 1210 Vienna, Austria;
| | - Catherine Belzung
- UMR1253, iBrain, Université de Tours, Inserm, 37200 Tours, France; (Y.I.-V.); (S.L.)
- Correspondence: (C.B.); (A.S.); Tel.: +33-2-47366994 (C.B.); +33-2-47367305 (A.S.)
| | - Alexandre Surget
- UMR1253, iBrain, Université de Tours, Inserm, 37200 Tours, France; (Y.I.-V.); (S.L.)
- Correspondence: (C.B.); (A.S.); Tel.: +33-2-47366994 (C.B.); +33-2-47367305 (A.S.)
| |
Collapse
|
34
|
O’Regan D, Jackson ML, Young AH, Rosenzweig I. Understanding the Impact of the COVID-19 Pandemic, Lockdowns and Social Isolation on Sleep Quality. Nat Sci Sleep 2021; 13:2053-2064. [PMID: 34795545 PMCID: PMC8593898 DOI: 10.2147/nss.s266240] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 10/17/2021] [Indexed: 12/15/2022] Open
Abstract
The uncertain, ever-changing and an ongoing nature of the COVID-19 pandemic means that it may take some time before we can fully appreciate the negative effect of the pandemic and lockdown on our sleep and mental health. It is increasingly recognised that in the aftermath of pandemic, several persistent sleep, neuropsychiatric and physical sequelae may continue long after the pandemic is over. A body of evidence to date also highlights a significant disparity in sleep and mental health difficulties in specific vulnerable groups in the community, with different temporal profiles and sleep issues that are reported. In this perspective, we argue for a possible mechanistic impact of the COVID-19 pandemic, with its imposed restrictions and social isolation on sleep quality. We similarly discuss some of the potential international differences, as well as similarities, behind reported idiosyncratic biological vulnerabilities that may have contributed to the genesis of sleep issues. Lastly, we propose some possible implementations and innovations that may be needed in restructuring of sleep disorders services in order to benefit recovering COVID-19 patients.
Collapse
Affiliation(s)
- David O’Regan
- Sleep Disorders Centre, Guy’s and St Thomas’ Hospital, GSTT NHS, London, UK
- Faculty of Life and Sciences Medicine, King’s College London, London, UK
| | - Melinda L Jackson
- Turner Institute for Brain and Mental Health, Monash University, Melbourne, Australia
| | - Allan H Young
- School of Academic Psychiatry, IoPPN, King’s College London, London, UK
| | - Ivana Rosenzweig
- Sleep Disorders Centre, Guy’s and St Thomas’ Hospital, GSTT NHS, London, UK
- Sleep and Brain Plasticity Centre, CNS, IoPPN, King’s College London, London, UK
| |
Collapse
|
35
|
Pesonen AK, Makkonen T, Elovainio M, Halonen R, Räikkönen K, Kuula L. Presleep physiological stress is associated with a higher cortical arousal in sleep and more consolidated REM sleep. Stress 2021; 24:667-675. [PMID: 33461366 DOI: 10.1080/10253890.2020.1869936] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
How sleep regulates physiological stress in healthy individuals is not well understood. We explored the associations between naturally occurring pre-sleep physiological arousal and EEG power spectral density together with rapid eye movement sleep (REMS) continuity. One hundred and fifty-four individuals (mean age 16.9, SD 0.1 years) collected five samples of saliva between the evening (mean time 18:20) and bedtime (mean 23:00) by using swabs, and underwent an overnight in-home polysomnography. We calculated spectral density for REMS and non-rapid eye movement sleep (non-REMS), and the number and duration of REMS arousals (<15 s) during sleep. An observational design allowed for measurement of natural variation in physiological and sleep arousal. Increasing cortisol levels toward bedtime were associated with higher EEG power spectral density at all frequency ranges in frontal locations, the highest association being for the beta1 frequency band. In central locations, the associations were pronounced for beta1 and beta2 bands. Higher overall cortisol levels in the evening were associated with less fragmented REMS. Presleep arousal was not associated with sleep staging. Physiological arousal toward bedtime was associated with EEG power spectral density values during sleep specifically at high EEG frequencies. This may represent a compensatory mechanism that serves as an adaptation to stress, since the REMS was more continuous along a higher physiological arousal level in the evening. Although causality cannot be inferred, a design with nonmanipulated physiological stress followed by naturally timed sleep at home provides new insights into stress regulation homeostasis.
Collapse
Affiliation(s)
- Anu-Katriina Pesonen
- Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tommi Makkonen
- Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Marko Elovainio
- Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Risto Halonen
- Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Katri Räikkönen
- Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Liisa Kuula
- Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
36
|
Gui S, Liu Y, Pu J, Song X, Chen X, Chen W, Zhong X, Wang H, Liu L, Xie P. Comparative analysis of hippocampal transcriptional features between major depressive disorder patients and animal models. J Affect Disord 2021; 293:19-28. [PMID: 34161882 DOI: 10.1016/j.jad.2021.06.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/03/2021] [Accepted: 06/07/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Major depressive disorder (MDD) is a psychiatric disorder caused by various etiologies. Chronic stress models are used to simulate the heterogeneous pathogenic processes of depression. However, few studies have compared transcriptional features between stress models and MDD patients. METHODS We generated hippocampal transcriptional profiles of the chronic social defeat model by RNA sequencing and downloaded raw data of the same brain region from public databases of the chronic unpredictable mild stress model, the learned helplessness model, and MDD patients. Differential expression and gene co-expression analyses were integrated to compare transcriptional features between stress models and MDD patients. RESULTS Each stress model shared 11.4% to 16.3% of differentially expressed genes with MDD patients. Functional analysis at the gene expression level identified altered ensheathment of neurons in both stress models and MDD patients. At the gene network level, each stress model shared 20.9% to 41.6% of co-expressed genes with MDD patients. Functional analysis based on these genes found that axon guidance signaling is the most significantly enriched pathway that was shared by all stress models and MDD patients. LIMITATIONS This study was limited by considering only a single brain region and a single sex of stress model animals. CONCLUSIONS Our results show that hippocampal transcriptional features of stress models partially overlap with those of MDD patients. The canonical pathways of MDD patients, including ensheathment of neurons, PTEN signaling, and axonal guidance signaling, were shared with all stress models. Our findings provide further clues to understand the molecular mechanisms of depression.
Collapse
Affiliation(s)
- Siwen Gui
- College of Biomedical Engineering, Chongqing Medical University, Chongqing 40016, China; State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing 40016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yiyun Liu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Juncai Pu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xuemian Song
- College of Biomedical Engineering, Chongqing Medical University, Chongqing 40016, China; State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing 40016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiaopeng Chen
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Weiyi Chen
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiaogang Zhong
- College of Stomatology and Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Haiyang Wang
- College of Stomatology and Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Lanxiang Liu
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Peng Xie
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
37
|
Bjørkum AA, Carrasco Duran A, Frode B, Sinha Roy D, Rosendahl K, Birkeland E, Stuhr L. Human blood serum proteome changes after 6 hours of sleep deprivation at night. SLEEP SCIENCE AND PRACTICE 2021. [DOI: 10.1186/s41606-021-00066-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Abstract
Background
The aim of this study was to discover significantly changed proteins in human blood serum after loss of 6 h sleep at night. Furthermore, to reveal affected biological process- and molecular function categories that might be clinically relevant, by exploring systems biological databases.
Methods
Eight females were recruited by volunteer request. Peripheral venous whole blood was sampled at 04:00 am, after 6 h of sleep and after 6 h of sleep deprivation. We used within-subjects design (all subjects were their own control). Blood serum from each subject was depleted before protein digestion by trypsin and iTRAQ labeling. Labled peptides were analyzed by mass spectrometry (LTQ OritrapVelos Elite) connected to a LC system (Dionex Ultimate NCR-3000RS).
Results
We identified 725 proteins in human blood serum. 34 proteins were significantly differentially expressed after 6 h of sleep deprivation at night. Out of 34 proteins, 14 proteins were up-regulated, and 20 proteins were down-regulated. We emphasized the functionality of the 16 proteins commonly differentiated in all 8 subjects and the relation to pathological conditions. In addition, we discussed Histone H4 (H4) and protein S100-A6/Calcyclin (S10A6) that were upregulated more than 1.5-fold. Finally, we discussed affected biological process- and molecular function categories.
Conclusions
Overall, our study suggest that acute sleep deprivation, at least in females, affects several known biological processes- and molecular function categories and associates to proteins that also are changed under pathological conditions like impaired coagulation, oxidative stress, immune suppression, neurodegenerative related disorder, and cancer. Data are available via ProteomeXchange with identifier PXD021004.
Collapse
|
38
|
Abstract
Major depression is a complex psychiatric disorder characterized by affective, cognitive, and physiological impairments that lead to maladaptive behavior. The high lifetime prevalence of this disabling condition, coupled with limitations of existing medications, make necessary the development of improved therapeutics. This requires animal models that allow investigation of key biological correlates of the disorder. Described in this article is the unpredictable chronic mild stress mouse model that can be used to screen for antidepressant drug candidates. Originally designed for rats, this model has been adapted for mice to capitalize on the advantages of this species as an experimental model, including inter-strain variability, which permits an exploration of the contribution of genetic background; the ability to create transgenic animals; and lower cost. Thus, because it combines genetic features and socio-environmental chronic stressful events, the unpredictable chronic mild stress model in mice is a relevant and valuable paradigm to gain insight into the etiological and developmental components of major depression, as well as to identify novel treatments for this condition. © 2021 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Unpredictable Chronic Mild Stress (UCMS) Test in Mice Basic Protocol 2: Assessment Of Self-Directed Activity And Anhedonia in Mice.
Collapse
Affiliation(s)
- Mathieu Nollet
- Department of Life Sciences, Imperial College London, London, United Kingdom
- UK Dementia Research Institute at Imperial College London, London, United Kingdom
| |
Collapse
|
39
|
Cardis R, Lecci S, Fernandez LM, Osorio-Forero A, Chu Sin Chung P, Fulda S, Decosterd I, Lüthi A. Cortico-autonomic local arousals and heightened somatosensory arousability during NREMS of mice in neuropathic pain. eLife 2021; 10:65835. [PMID: 34227936 PMCID: PMC8291975 DOI: 10.7554/elife.65835] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 07/02/2021] [Indexed: 12/22/2022] Open
Abstract
Frequent nightly arousals typical for sleep disorders cause daytime fatigue and present health risks. As such arousals are often short, partial, or occur locally within the brain, reliable characterization in rodent models of sleep disorders and in human patients is challenging. We found that the EEG spectral composition of non-rapid eye movement sleep (NREMS) in healthy mice shows an infraslow (~50 s) interval over which microarousals appear preferentially. NREMS could hence be vulnerable to abnormal arousals on this time scale. Chronic pain is well-known to disrupt sleep. In the spared nerve injury (SNI) mouse model of chronic neuropathic pain, we found more numerous local cortical arousals accompanied by heart rate increases in hindlimb primary somatosensory, but not in prelimbic, cortices, although sleep macroarchitecture appeared unaltered. Closed-loop mechanovibrational stimulation further revealed higher sensory arousability. Chronic pain thus preserved conventional sleep measures but resulted in elevated spontaneous and evoked arousability. We develop a novel moment-to-moment probing of NREMS vulnerability and propose that chronic pain-induced sleep complaints arise from perturbed arousability.
Collapse
Affiliation(s)
- Romain Cardis
- Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.,Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Sandro Lecci
- Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Laura Mj Fernandez
- Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Alejandro Osorio-Forero
- Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Paul Chu Sin Chung
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Stephany Fulda
- Sleep Medicine Unit, Neurocenter of Southern Switzerland, Civic Hospital (EOC) of Lugano, Lugano, Switzerland
| | - Isabelle Decosterd
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Anita Lüthi
- Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
40
|
Bowers SJ, Lambert S, He S, Lowry CA, Fleshner M, Wright KP, Turek FW, Vitaterna MH. Immunization with a heat-killed bacterium, Mycobacterium vaccae NCTC 11659, prevents the development of cortical hyperarousal and a PTSD-like sleep phenotype after sleep disruption and acute stress in mice. Sleep 2021; 44:6025170. [PMID: 33283862 DOI: 10.1093/sleep/zsaa271] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 11/20/2020] [Indexed: 12/27/2022] Open
Abstract
STUDY OBJECTIVES Sleep deprivation induces systemic inflammation that may contribute to stress vulnerability and other pathologies. We tested the hypothesis that immunization with heat-killed Mycobacterium vaccae NCTC 11659 (MV), an environmental bacterium with immunoregulatory and anti-inflammatory properties, prevents the negative impacts of 5 days of sleep disruption on stress-induced changes in sleep, behavior, and physiology in mice. METHODS In a 2 × 2 × 2 experimental design, male C57BL/6N mice were given injections of either MV or vehicle on days -17, -10, and -3. On days 1-5, mice were exposed to intermittent sleep disruption, whereby sleep was disrupted for 20 h per day. Immediately following sleep disruption, mice were exposed to 1-h social defeat stress or novel cage (control) conditions. Object location memory (OLM) testing was conducted 24 h after social defeat, and tissues were collected 6 days later to measure inflammatory markers. Sleep was recorded using electroencephalography (EEG) and electromyography (EMG) throughout the experiment. RESULTS In vehicle-treated mice, only the combination of sleep disruption followed by social defeat (double hit): (1) increased brief arousals and NREM beta (15-30 Hz) EEG power in sleep immediately post-social defeat compared to baseline; (2) induced an increase in the proportion of rapid-eye-movement (REM) sleep and number of state shifts for at least 5 days post-social defeat; and (3) induced hyperlocomotion and lack of habituation in the OLM task. Immunization with MV prevented most of these sleep and behavioral changes. CONCLUSIONS Immunization with MV ameliorates a stress-induced sleep and behavioral phenotype that shares features with human posttraumatic stress disorder.
Collapse
Affiliation(s)
- Samuel J Bowers
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL.,Department of Neurobiology, Northwestern University, Evanston, IL
| | - Sophie Lambert
- Department of Neurobiology, Northwestern University, Evanston, IL
| | - Shannon He
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL.,Department of Neurobiology, Northwestern University, Evanston, IL
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO
| | - Monika Fleshner
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO
| | - Kenneth P Wright
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO.,Sleep and Chronobiology Laboratory, University of Colorado Boulder, Boulder, CO
| | - Fred W Turek
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL.,Department of Neurobiology, Northwestern University, Evanston, IL.,The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Martha H Vitaterna
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL.,Department of Neurobiology, Northwestern University, Evanston, IL
| |
Collapse
|
41
|
Wang P, Gao X, Zhao F, Gao Y, Wang K, Tian JS, Li Z, Qin XM. Study of the Neurotransmitter Changes Adjusted by Circadian Rhythm in Depression Based on Liver Transcriptomics and Correlation Analysis. ACS Chem Neurosci 2021; 12:2151-2166. [PMID: 34060807 DOI: 10.1021/acschemneuro.1c00115] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Depression has drawn increasing attention from the public around the world in recent years. Studies have shown that liver injury caused by chronic stress is relevant to depression and neurotransmitter changes. It is essential to clarify the relationship between neurotransmitter changes and hepatic gene expression in depression. In this study, we used the chronic unpredictable mild stress (CUMS) model combined with UHPLC-MS to explore the changes of neurotransmitters in serum and hippocampus and to decipher the differential gene expression in the liver by using the RNA-Seq combined with multivariate statistical analysis. Compared with the control group, the levels of neurotransmitters including 5-hydroxytryptamine (5-HT), acetylcholine, glutamate (Glu), and dopamine (DA) in the hippocampus and 5-HT, norepinephrine, γ-aminobutyric acid (GABA), and 5-hydroxyindoleacetic acid in serum were significantly changed in the CUMS rats. The results of liver transcriptomic analysis and correlation analysis showed that the Glu, DA, 5-HT, and GABA were impacted by 68 liver genes which were mainly enriched in three pathways including circadian rhythm, serotonergic synapse, and p53 signaling pathway. The expressive levels of clock genes and serotonergic synapse genes were validated by using q-PCR, and the diurnal rhythms of neurotransmitters were validated by in vivo hippocampus microdialysis. The CUMS stressors might cause phase advance of Glu and GABA by adjusting clock genes. The transcriptomic technique combined with correlation analysis and in vivo microdialysis could be used to discover comprehensive pathways of depression. It provides a new strategy for the rational assessment of the mechanism of disease.
Collapse
Affiliation(s)
- Peng Wang
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China
| | - Xiaoxia Gao
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry Education of Shanxi University, Taiyuan 030006, China
| | - Fang Zhao
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China
- College of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China
| | - Yao Gao
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China
| | - Kexin Wang
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China
| | - Jun-Sheng Tian
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry Education of Shanxi University, Taiyuan 030006, China
| | - Zhenyu Li
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry Education of Shanxi University, Taiyuan 030006, China
| | - Xue-Mei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry Education of Shanxi University, Taiyuan 030006, China
| |
Collapse
|
42
|
Drews HJ, Drews A. Couple Relationships Are Associated With Increased REM Sleep-A Proof-of-Concept Analysis of a Large Dataset Using Ambulatory Polysomnography. Front Psychiatry 2021; 12:641102. [PMID: 34040553 PMCID: PMC8141649 DOI: 10.3389/fpsyt.2021.641102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 04/07/2021] [Indexed: 11/24/2022] Open
Abstract
Background/Objectives: Rapid Eye Movement (REM) sleep is associated with memory consolidation and several health effects including stress response, mental health, and longevity. Recently, it has been shown that regularly co-sleeping couples have increased and stabilized REM sleep when co-sleeping as compared to sleeping individually. However, it remained unclear whether this is due to a specific effect of altering the usual sleeping environment by partner deprivation or due to a generalizable REM-sleep promoting effect of couple relationships. The present study aims to clarify this ambiguity. Methods: Married or never married individuals were taken from the Sleep Heart Health Study (n = 5,804) and matched regarding sociodemographic and health parameters. Matching was done using propensity score matching (1:1, nearest neighbor) and resulted in two groups of n = 69 each (married vs. never married). After confirmation of successful matching, samples were compared regarding REM sleep and other polysomnographic parameters (paired Students t-tests or Wilcoxon signed-rank tests). Results: Married individuals showed significantly higher levels of total and relative REM sleep as compared to never married individuals (all p's ≤ 0.003). Neither other sleep stages nor REM-sleep fragmentation differed between groups (all p's ≥ 0.29). Results regarding number of sleep cycles were ambiguous. Conclusion: This is the first between-subjects study to show that couple relationships are associated with increased REM sleep. This finding represents a necessary (but not sufficient) condition for the previously hypothesized self-enhancing feedback loop of REM sleep and sociality as well as for REM-sleep promotion as a mechanism through which couple relationships prevent mental illness.
Collapse
Affiliation(s)
- Henning Johannes Drews
- Department of Mental Health, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Psychiatry and Psychotherapy, Christian-Albrechts-University, Kiel, Germany
| | - Annika Drews
- Department of Climate and Environment, SINTEF Ocean AS, Trondheim, Norway
| |
Collapse
|
43
|
Mamelak M. Sleep, Narcolepsy, and Sodium Oxybate. Curr Neuropharmacol 2021; 20:272-291. [PMID: 33827411 PMCID: PMC9413790 DOI: 10.2174/1570159x19666210407151227] [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: 01/17/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 11/23/2022] Open
Abstract
Sodium oxybate (SO) has been in use for many decades to treat narcolepsy with cataplexy. It functions as a weak GABAB agonist but also as an energy source for the brain as a result of its metabolism to succinate and as a powerful antioxidant because of its capacity to induce the formation of NADPH. Its actions at thalamic GABAB receptors can induce slow-wave activity, while its actions at GABAB receptors on monoaminergic neurons can induce or delay REM sleep. By altering the balance between monoaminergic and cholinergic neuronal activity, SO uniquely can induce and prevent cataplexy. The formation of NADPH may enhance sleep’s restorative process by accelerating the removal of the reactive oxygen species (ROS), which accumulate during wakefulness. SO improves alertness in normal subjects and in patients with narcolepsy. SO may allay severe psychological stress - an inflammatory state triggered by increased levels of ROS and characterized by cholinergic supersensitivity and monoaminergic deficiency. SO may be able to eliminate the inflammatory state and correct the cholinergic/ monoaminergic imbalance.
Collapse
Affiliation(s)
- Mortimer Mamelak
- Department of Psychiatry, Baycrest Hospital, University of Toronto, Toronto, Ontario. Canada
| |
Collapse
|
44
|
Simmons JM, Winsky L, Zehr JL, Gordon JA. Priorities in stress research: a view from the U.S. National Institute of Mental Health. Stress 2021; 24:123-129. [PMID: 32608314 DOI: 10.1080/10253890.2020.1781084] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The mission of the National Institute of Mental Health is to transform the understanding and treatment of mental illnesses through basic and clinical research, paving the way for prevention, recovery, and cure. In consultation with a broad range of experts, the NIMH has identified a set of priorities for stress biology research aimed squarely at creating the basic and clinical knowledge bases for reducing and alleviating mental health burden across the lifespan. Here, we discuss these priority areas in stress biology research, which include: understanding the heterogeneity of stressors and outcomes; refining and expanding the experimental systems used to study stress and its effects; embracing and exploiting the complexity of the stress response; and prioritizing translational studies that seek to test mechanistic hypotheses in human beings. We emphasize the challenge of establishing mechanistic links across levels of analysis to explain how and when specific and diverse stressors lead to enduring changes in neural systems and produce lasting functional deficits in mental health relevant behaviors. An improved understanding of mechanisms underlying stress responses and the functional consequences of stress can and will speed translation from basic research to predictive markers of risk and to improved, personalized interventions for mental illness.
Collapse
Affiliation(s)
| | - Lois Winsky
- National Institute of Mental Health, Bethesda, MD, USA
| | - Julia L Zehr
- National Institute of Mental Health, Bethesda, MD, USA
| | | |
Collapse
|
45
|
Bruce HA, Kochunov P, Chiappelli J, Savransky A, Carino K, Sewell J, Marshall W, Kvarta M, McMahon FJ, Ament SA, Postolache TT, O'Connell J, Shuldiner A, Mitchell B, Hong LE. Genetic versus stress and mood determinants of sleep in the Amish. Am J Med Genet B Neuropsychiatr Genet 2021; 186:113-121. [PMID: 33650257 PMCID: PMC8994156 DOI: 10.1002/ajmg.b.32840] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/28/2021] [Accepted: 02/10/2021] [Indexed: 12/26/2022]
Abstract
Sleep is essential to the human brain and is regulated by genetics with many features conserved across species. Sleep is also influenced by health and environmental factors; identifying replicable genetic variants contributing to sleep may require accounting for these factors. We examined how stress and mood disorder contribute to sleep and impact its heritability. Our sample included 326 Amish/Mennonite individuals with a lifestyle with limited technological interferences with sleep. Sleep measures included Pittsburgh Sleep Quality Index (PSQI), bedtime, wake time, and time to sleep onset. Current stress level, cumulative life stressors, and mood disorder were also evaluated. We estimated the heritability of sleep features and examined the impact of current stress, lifetime stress, mood diagnosis on sleep quality. The results showed current stress, lifetime stress, and mood disorder were independently associated with PSQI score (p < .05). Heritability of PSQI was low (0-0.23) before and after accounting for stress and mood. Bedtime, wake time, and minutes to sleep time did show significant heritability at 0.44, 0.42, and 0.29. However, after adjusting for shared environment, only heritability of wake time remained significant. Sleep is affected by environmental stress and mental health factors even in a society with limited technological interference with sleep. Wake time may be a more biological marker of sleep as compared to the evening measures which are more influenced by other household members. Accounting for nongenetic and partially genetic determinants of sleep particularly stress and mood disorder is likely important for improving the precision of genetic studies of sleep.
Collapse
Affiliation(s)
- Heather A. Bruce
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Peter Kochunov
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Joshua Chiappelli
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Anya Savransky
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Kathleen Carino
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jessica Sewell
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Wyatt Marshall
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Mark Kvarta
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Francis J. McMahon
- Human Genetics Branch, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland
| | - Seth A. Ament
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland,Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Teodor T. Postolache
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland,Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC) for Suicide Prevention, Colorado, Aurora,Capitol MIRECC, Baltimore, Maryland
| | - Jeff O'Connell
- Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland,Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Alan Shuldiner
- Regeneron Genetics Center, Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | - Braxton Mitchell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland,Geriatrics Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, Baltimore, Maryland
| | - L. Elliot Hong
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
46
|
Song J, Kim YK. Animal models for the study of depressive disorder. CNS Neurosci Ther 2021; 27:633-642. [PMID: 33650178 PMCID: PMC8111503 DOI: 10.1111/cns.13622] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 01/01/2023] Open
Abstract
Depressive disorder is one of the most widespread forms of psychiatric pathology, worldwide. According to a report by the World Health Organization, the number of people with depression, globally, is increasing dramatically with each year. Previous studies have demonstrated that various factors, including genetics and environmental stress, contribute to the risk of depression. As such, it is crucial to develop a detailed understanding of the pathogenesis of depressive disorder and animal studies are essential for identifying the mechanisms and genetic disorders underlying depression. Recently, many researchers have reported on the pathology of depression via various models of depressive disorder. Given that different animal models of depression show differences in terms of patterns of depressive behavior and pathology, the comparison between depressive animal models is necessary for progress in the field of the depression study. However, the various animal models of depression have not been fully compared or evaluated until now. In this paper, we reviewed the pathophysiology of the depressive disorder and its current animal models with the analysis of their transcriptomic profiles. We provide insights for selecting different animal models for the study of depression.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun, Korea
| | - Young-Kook Kim
- Department of Biochemistry, Chonnam National University Medical School, Hwasun, Korea
| |
Collapse
|
47
|
Eliwa H, Brizard B, Le Guisquet AM, Hen R, Belzung C, Surget A. Adult neurogenesis augmentation attenuates anhedonia and HPA axis dysregulation in a mouse model of chronic stress and depression. Psychoneuroendocrinology 2021; 124:105097. [PMID: 33302237 PMCID: PMC8715720 DOI: 10.1016/j.psyneuen.2020.105097] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/06/2020] [Accepted: 11/26/2020] [Indexed: 11/26/2022]
Abstract
Major depressive disorder is a common debilitating mental health problem that represents one of the leading causes of disability. Up to date, the therapeutic targets and approaches are still limited. Adult hippocampal neurogenesis (AHN) has been proposed as a critical contributor to the pathophysiology and treatment of depression, altering the hippocampal control over stress response at network, neuroendocrine and behavioral levels. These findings together have suggested that manipulating AHN may be a promising therapeutic strategy for depression. To investigate this question, we assessed whether increasing adult neurogenesis would be sufficient to produce antidepressant-like effects at behavioral and neuroendocrine levels in a mouse model of depression; the unpredictable chronic mild stress (UCMS). For this purpose, we used a bi-transgenic mouse line (iBax) in which AHN increase was induced by deletion of the pro-apoptotic gene Bax from the neural progenitors following the tamoxifen-dependent action of CreERT2 recombinases. UCMS induced a syndrome that is reminiscent of depression-like states, including anhedonia (cookie test), physical changes (coat deterioration, reduced weight gain), anxiety-like behaviors (higher latency in the novelty-supressed feeding -NSF- test), passive stress-coping behaviors (immobility in the forced swim test -FST-) and a blunted hypothalamo-pituitary-adrenal (HPA) axis reactivity to acute stress in addition to AHN decrease. Tamoxifen injection reversed the AHN decrease as well as partly counteracted UCMS effects on the cookie test and HPA axis but not for the coat state, weight gain, NSF test and FST. Taken together, our results suggest that a strategy directing at increasing AHN may be able to alleviate some depression-related behavioral and neuroendocrine dimensions of UCMS, such as anhedonia and HPA axis reactivity deficits, but may be hardly sufficient to produce a complete recovery.
Collapse
Affiliation(s)
- Hoda Eliwa
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France; Department of Cell Biology, Medical Research Institute, Alexandria University, Egypt
| | - Bruno Brizard
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | | | - René Hen
- Departments of Neuroscience, Psychiatry, & Pharmacology, Columbia University, New York, NY, USA; Division of Integrative Neuroscience, Department of Psychiatry, New York State Psychiatric Institute, New York, NY, USA; Kavli Institute for Brain Sciences, Columbia University, New York, NY, USA
| | | | | |
Collapse
|
48
|
Silva TM, Borniger JC, Alves MJ, Alzate Correa D, Zhao J, Fadda P, Toland AE, Takakura AC, Moreira TS, Czeisler CM, Otero JJ. Machine learning approaches reveal subtle differences in breathing and sleep fragmentation in Phox2b-derived astrocytes ablated mice. J Neurophysiol 2021; 125:1164-1179. [PMID: 33502943 DOI: 10.1152/jn.00155.2020] [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] [Indexed: 02/06/2023] Open
Abstract
Modern neurophysiology research requires the interrogation of high-dimensionality data sets. Machine learning and artificial intelligence (ML/AI) workflows have permeated into nearly all aspects of daily life in the developed world but have not been implemented routinely in neurophysiological analyses. The power of these workflows includes the speed at which they can be deployed, their availability of open-source programming languages, and the objectivity permitted in their data analysis. We used classification-based algorithms, including random forest, gradient boosted machines, support vector machines, and neural networks, to test the hypothesis that the animal genotypes could be separated into their genotype based on interpretation of neurophysiological recordings. We then interrogate the models to identify what were the major features utilized by the algorithms to designate genotype classification. By using raw EEG and respiratory plethysmography data, we were able to predict which recordings came from genotype class with accuracies that were significantly improved relative to the no information rate, although EEG analyses showed more overlap between groups than respiratory plethysmography. In comparison, conventional methods where single features between animal classes were analyzed, differences between the genotypes tested using baseline neurophysiology measurements showed no statistical difference. However, ML/AI workflows successfully were capable of providing successful classification, indicating that interactions between features were different in these genotypes. ML/AI workflows provide new methodologies to interrogate neurophysiology data. However, their implementation must be done with care so as to provide high rigor and reproducibility between laboratories. We provide a series of recommendations on how to report the utilization of ML/AI workflows for the neurophysiology community.NEW & NOTEWORTHY ML/AI classification workflows are capable of providing insight into differences between genotypes for neurophysiology research. Analytical techniques utilized in the neurophysiology community can be augmented by implementing ML/AI workflows. Random forest is a robust classification algorithm for respiratory plethysmography data. Utilization of ML/AI workflows in neurophysiology research requires heightened transparency and improved community research standards.
Collapse
Affiliation(s)
- Talita M Silva
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine.,Department of Physiology and Biophysics, Institute of Biomedical Science, University of São Paulo
| | | | - Michele Joana Alves
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine
| | - Diego Alzate Correa
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine
| | - Jing Zhao
- Department of Biomedical Informatics, The Ohio State University College of Dentistry
| | - Paolo Fadda
- Genomics Shared Resource-Comprehensive Cancer Center, The Ohio State University
| | - Amanda Ewart Toland
- Genomics Shared Resource-Comprehensive Cancer Center, The Ohio State University.,Department of Cancer Biology and Genetics, The Ohio State University College of Medicine
| | - Ana C Takakura
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo
| | - Thiago S Moreira
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of São Paulo
| | - Catherine M Czeisler
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine
| | - José Javier Otero
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine
| |
Collapse
|
49
|
Fitzgerald PJ, Hale PJ, Ghimire A, Watson BO. Repurposing Cholinesterase Inhibitors as Antidepressants? Dose and Stress-Sensitivity May Be Critical to Opening Possibilities. Front Behav Neurosci 2021; 14:620119. [PMID: 33519395 PMCID: PMC7840590 DOI: 10.3389/fnbeh.2020.620119] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022] Open
Abstract
When stress becomes chronic it can trigger lasting brain and behavioral changes including Major Depressive Disorder (MDD). There is conflicting evidence regarding whether acetylcholinesterase inhibitors (AChEIs) may have antidepressant properties. In a recent publication, we demonstrated a strong dose-dependency of the effect of AChEIs on antidepressant-related behavior in the mouse forced swim test: whereas the AChEI donepezil indeed promotes depression-like behavior at a high dose, it has antidepressant-like properties at lower doses in the same experiment. Our data therefore suggest a Janus-faced dose-response curve for donepezil in depression-related behavior. In this review, we investigate the mood-related properties of AChEIs in greater detail, focusing on both human and rodent studies. In fact, while there have been many studies showing pro-depressant activity by AChEIs and this is a major concept in the field, a variety of other studies in both humans and rodents show antidepressant effects. Our study was one of the first to systematically vary dose to include very low concentrations while measuring behavioral effects, potentially explaining the apparent disparate findings in the field. The possibility of antidepressant roles for AChEIs in rodents may provide hope for new depression treatments. Importantly, MDD is a psychosocial stress-linked disorder, and in rodents, stress is a major experimental manipulation for studying depression mechanisms, so an important future direction will be to determine the extent to which these depression-related effects are stress-sensitive. In sum, gaining a greater understanding of the potentially therapeutic mood-related effects of low dose AChEIs, both in rodent models and in human subjects, should be a prioritized topic in ongoing translational research.
Collapse
Affiliation(s)
- Paul J Fitzgerald
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, United States
| | - Pho J Hale
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, United States
| | - Anjesh Ghimire
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, United States
| | - Brendon O Watson
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
50
|
A gene expression atlas for different kinds of stress in the mouse brain. Sci Data 2020; 7:437. [PMID: 33328476 PMCID: PMC7744580 DOI: 10.1038/s41597-020-00772-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 11/25/2020] [Indexed: 12/17/2022] Open
Abstract
Stressful experiences are part of everyday life and animals have evolved physiological and behavioral responses aimed at coping with stress and maintaining homeostasis. However, repeated or intense stress can induce maladaptive reactions leading to behavioral disorders. Adaptations in the brain, mediated by changes in gene expression, have a crucial role in the stress response. Recent years have seen a tremendous increase in studies on the transcriptional effects of stress. The input raw data are freely available from public repositories and represent a wealth of information for further global and integrative retrospective analyses. We downloaded from the Sequence Read Archive 751 samples (SRA-experiments), from 18 independent BioProjects studying the effects of different stressors on the brain transcriptome in mice. We performed a massive bioinformatics re-analysis applying a single, standardized pipeline for computing differential gene expression. This data mining allowed the identification of novel candidate stress-related genes and specific signatures associated with different stress conditions. The large amount of computational results produced was systematized in the interactive “Stress Mice Portal”.
Collapse
|