51
|
Zhao Y, Yang X, Cheng S, Li C, He D, Cai Q, Wei W, Qin X, Zhang N, Shi S, Chu X, Meng P, Zhang F. Assessing the effect of interaction between lifestyle and longitudinal changes in brain structure on sleep phenotypes. Cereb Cortex 2023:7030864. [PMID: 36750265 DOI: 10.1093/cercor/bhac526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 02/09/2023] Open
Abstract
Longitudinal changes in brain structure and lifestyle can affect sleep phenotypes. However, the influence of the interaction between longitudinal changes in brain structure and lifestyle on sleep phenotypes remains unclear. Genome-wide association study dataset of longitudinal changes in brain structure was obtained from published study. Phenotypic data of lifestyles and sleep phenotypes were obtained from UK Biobank cohort. Using genotype data from UK Biobank, we calculated polygenetic risk scores of longitudinal changes in brain structure phenotypes. Linear/logistic regression analysis was conducted to evaluate interactions between longitudinal changes in brain structure and lifestyles on sleep duration, chronotype, insomnia, snoring and daytime dozing. Multiple lifestyle × longitudinal changes in brain structure interactions were detected for 5 sleep phenotypes, such as physical activity×caudate_age2 for daytime dozing (OR = 1.0389, P = 8.84 × 10-3) in total samples, coffee intake×cerebellar white matter volume_age2 for daytime dozing (OR = 0.9652, P = 1.13 × 10-4) in females. Besides, we found 4 overlapping interactions in different sleep phenotypes. We conducted sex stratification analysis and identified one overlapping interaction between female and male. Our results support the moderate effects of interaction between lifestyle and longitudinal changes in brain structure on sleep phenotypes, and deepen our understanding of the pathogenesis of sleep disorders.
Collapse
Affiliation(s)
- Yijing Zhao
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xuena Yang
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Shiqiang Cheng
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Chun'e Li
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Dan He
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Qingqing Cai
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Wenming Wei
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaoyue Qin
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Na Zhang
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Sirong Shi
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaoge Chu
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Peilin Meng
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Feng Zhang
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, No. 76 Yan Ta West Road, Xi'an 710061, China
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
52
|
Gómez LJ, Dooley JC, Blumberg MS. Activity in developing prefrontal cortex is shaped by sleep and sensory experience. eLife 2023; 12:e82103. [PMID: 36745108 PMCID: PMC9901933 DOI: 10.7554/elife.82103] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 01/12/2023] [Indexed: 02/07/2023] Open
Abstract
In developing rats, behavioral state exerts a profound modulatory influence on neural activity throughout the sensorimotor system, including primary motor cortex (M1). We hypothesized that similar state-dependent modulation occurs in prefrontal cortical areas with which M1 forms functional connections. Here, using 8- and 12-day-old rats cycling freely between sleep and wake, we record neural activity in M1, secondary motor cortex (M2), and medial prefrontal cortex (mPFC). At both ages in all three areas, neural activity increased during active sleep (AS) compared with wake. Also, regardless of behavioral state, neural activity in all three areas increased during periods when limbs were moving. The movement-related activity in M2 and mPFC, like that in M1, is driven by sensory feedback. Our results, which diverge from those of previous studies using anesthetized pups, demonstrate that AS-dependent modulation and sensory responsivity extend to prefrontal cortex. These findings expand the range of possible factors shaping the activity-dependent development of higher-order cortical areas.
Collapse
Affiliation(s)
- Lex J Gómez
- Interdisciplinary Graduate Program in Neuroscience, University of IowaIowa CityUnited States
| | - James C Dooley
- Department of Psychological and Brain Sciences, University of IowaIowa CityUnited States
- DeLTA Center, University of IowaIowa CityUnited States
| | - Mark S Blumberg
- Interdisciplinary Graduate Program in Neuroscience, University of IowaIowa CityUnited States
- Department of Psychological and Brain Sciences, University of IowaIowa CityUnited States
- DeLTA Center, University of IowaIowa CityUnited States
- Iowa Neuroscience Institute, University of IowaIowa CityUnited States
| |
Collapse
|
53
|
Li T, Gao Y, He M, Gui Z, Zhao B, Cao Y, Chen T, Zhu J, Wang J, Zhong Q, Zhang Z. P2X7 receptor-activated microglia in cortex is critical for sleep disorder under neuropathic pain. Front Neurosci 2023; 17:1095718. [PMID: 36816134 PMCID: PMC9936193 DOI: 10.3389/fnins.2023.1095718] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Neuropathic pain (NP) is associated with sleep disturbances, which may substantially influence the quality of life. Clinical and animal studies demonstrated that neurotransmitter is one of the main contributors to cause sleep disturbances induced by NP. Recently, it was reported that P2X7 receptors (P2X7R) are widely expressed in microglia, which serves crucial role in neuronal activity in the pain and sleep-awake cycle. In this study, we adopted the chronic constriction injury (CCI) model to establish the progress of chronic pain and investigated whether P2X7R of microglia in cortex played a critical role in sleep disturbance induced by NP. At electroencephalogram (EEG) level, sleep disturbance was observed in mice treated with CCI as they exhibited mechanical and thermal hypersensitivity, and inhibition of P2X7R ameliorated these changes. We showed a dramatic high level of P2X7R and Iba-1 co-expression in the cortical region, and the inhibition of P2X7R also adversely affected it. Furthermore, the power of LFPs in ventral posterior nucleus (VP) and primary somatosensory cortex (S1) which changed in the CCI group was adverse after the inhibition of P2X7R. Furthermore, inhibition of P2X7R also decreased the VP-S1 coherence which increased in CCI group. Nuclear magnetic resonance demonstrated inhibition of P2X7R decreased glutamate (Glu) levels in thalamic and cortical regions which were significantly increased in the CCI mice. Our findings provide evidence that NP has a critical effect on neuronal activity linked to sleep and may built up a new target for the development of sleep disturbances under chronic pain conditions.
Collapse
Affiliation(s)
- Tingting Li
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Yunling Gao
- Xiangyang Central Hospital, Institute of Neuroscience and Brain Diseases, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Mengying He
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China,Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, China
| | - Zhu Gui
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, China,University of Chinese Academy of Sciences, Beijing, China
| | - Bingchu Zhao
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, China,School of Computer Science, Wuhan University, Wuhan, Hubei, China
| | - Yue Cao
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Ting Chen
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Jinpiao Zhu
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China,Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, China
| | - Jie Wang
- Xiangyang Central Hospital, Institute of Neuroscience and Brain Diseases, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China,Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, China,University of Chinese Academy of Sciences, Beijing, China
| | - Qi Zhong
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China,*Correspondence: Qi Zhong,
| | - Zongze Zhang
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China,Zongze Zhang,
| |
Collapse
|
54
|
Melatonin alleviates PTSD-like behaviors and restores serum GABA and cortisol levels in mice. Psychopharmacology (Berl) 2023; 240:259-269. [PMID: 36642730 DOI: 10.1007/s00213-023-06312-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 01/04/2023] [Indexed: 01/17/2023]
Abstract
RATIONALE Melatonin is an endogenous hormone which modulates sleep-wake cycles. Previous studies have found a close correlation between melatonin and post-traumatic stress disorder (PTSD), a trauma- and stress-related psychiatric disorder with symptoms of sleep disturbance. However, it is still unclear if melatonin can have a therapeutic effect on PTSD. OBJECTIVE This study aimed to investigate the effects of melatonin on foot shocks induced PTSD-like behaviors and abnormal neuroendocrine levels in mice. RESULTS As compared to no-shock controls, PTSD-like mice spent significantly more time freezing and displayed less rearing in a contextual fear test, spent significantly less time in and had fewer entries into open arms in an elevated maze test, and spent significantly less time in and had fewer entries into a light box in a light-dark transition task. In addition, serum GABA and cortisol levels were both found to be significantly decreased, whereas epinephrine levels were significantly increased in the PTSD-like mice. Our results showed that intraperitoneal injections of melatonin (2 mM, but not 0.2 nor 20 mM, 0.1 ml/day for two consecutive weeks) alleviated PTSD-like behaviors and restored serum GABA and cortisol levels. Further, it was found that melatonin receptor 1/2 antagonist luzindole significantly blocked the beneficial effects of melatonin for PTSD-like behaviors and serum GABA and cortisol levels, whereas melatonin receptor 2 antagonist 4-P-PDOT slightly blocked these effects. CONCLUSIONS These results indicate that melatonin has a potential therapeutic effect on PTSD-like symptoms in mice, and melatonin receptor 1 mediated the effect.
Collapse
|
55
|
Steffens SK, Stenberg TH, Wigren HKM. Alterations in microglial morphology concentrate in the habitual sleeping period of the mouse. Glia 2023; 71:366-376. [PMID: 36196985 PMCID: PMC10092278 DOI: 10.1002/glia.24279] [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: 11/10/2021] [Revised: 09/10/2022] [Accepted: 09/20/2022] [Indexed: 11/05/2022]
Abstract
In nocturnal animals, waking appears during the dark period while maximal non-rapid-eye-movement sleep (NREMS) with electroencephalographic slow-wave-activity (SWA) takes place at the beginning of the light period. Vigilance states associate with variable levels of neuronal activity: waking with high-frequency activity patterns while during NREMS, SWA influences neuronal activity in many brain areas. On a glial level, sleep deprivation modifies microglial morphology, but only few studies have investigated microglia through the physiological sleep-wake cycle. To quantify microglial morphology (territory, volume, ramification) throughout the 24 h light-dark cycle, we collected brain samples from inbred C57BL male mice (n = 51) every 3 h and applied a 3D-reconstruction method for microglial cells on the acquired confocal microscopy images. As microglia express regional heterogeneity and are influenced by local neuronal activity, we chose to investigate three interconnected and functionally well-characterized brain areas: the somatosensory cortex (SC), the dorsal hippocampus (HC), and the basal forebrain (BF). To temporally associate microglial morphology with vigilance stages, we performed a 24 h polysomnography in a separate group of animals (n = 6). In line with previous findings, microglia displayed de-ramification in the 12 h light- and hyper-ramification in the 12 h dark period. Notably, we found that the decrease in microglial features was most prominent within the early hours of the light period, co-occurring with maximal sleep SWA. By the end of the light period, all features reached maximum levels and remained steadily elevated throughout the dark period with minor regional differences. We propose that vigilance-stage specific neuronal activity, and SWA, could modify microglial morphology.
Collapse
Affiliation(s)
| | - Tarja Helena Stenberg
- SleepWell Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | |
Collapse
|
56
|
Romier A, Maruani J, Lopez-Castroman J, Palagini L, Serafini G, Lejoyeux M, d'Ortho MP, Geoffroy PA. Objective sleep markers of suicidal behaviors in patients with psychiatric disorders: A systematic review and meta-analysis. Sleep Med Rev 2023; 68:101760. [PMID: 36706699 DOI: 10.1016/j.smrv.2023.101760] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/29/2022] [Accepted: 01/10/2023] [Indexed: 01/19/2023]
Abstract
Close relationships have been reported between sleep alterations and suicidal behaviors, nevertheless few studies used objective measures of sleep. Such objective markers would be interesting in clinical practice to better screen and prevent suicide. We conducted a systematic review and meta-analysis of published studies examining the relationship between sleep markers and suicidal behaviors using PubMed, Cochrane Library, and Web of Science databases. Actigraphy, polysomnography, and nocturnal EEG were considered. The qualitative analysis retained 15 original studies, including 1179 participants (939 with a psychiatric disorder), and 11 studies were included for the meta-analysis. Current suicidal behaviors were associated with a decreased total sleep time (TST) (SMD = -0.35, [95% CI: -0.66 to -0.04], p = 0.026, I2 = 39.8%). The evaluation of possible moderators shows that age, gender, and depression scores had no effects on the random effect model. No significant differences were observed regarding sleep efficiency, REM latency, or percentage of REM sleep. In conclusion, among candidate objective markers, decreased total sleep time seems associated with suicidal behaviors and could be easily used to assess suicide risk. Alterations of regular sleep duration should invite healthcare professionals to screen the cause and propose sleep interventions to prevent suicide.
Collapse
Affiliation(s)
- Alix Romier
- Département de psychiatrie et d'addictologie, AP-HP, GHU Paris Nord, DMU Neurosciences, Hopital Bichat - Claude Bernard, F-75018, Paris, France; Université Paris Cité, NeuroDiderot, Inserm, FHU I2-D2, F-75019, Paris, France.
| | - Julia Maruani
- Département de psychiatrie et d'addictologie, AP-HP, GHU Paris Nord, DMU Neurosciences, Hopital Bichat - Claude Bernard, F-75018, Paris, France; Université Paris Cité, NeuroDiderot, Inserm, FHU I2-D2, F-75019, Paris, France; GHU Paris - Psychiatry & Neurosciences, 1 rue Cabanis, 75014, Paris, France
| | - Jorge Lopez-Castroman
- Department of Psychiatry, CHU Nîmes & IGF, CNRS-INSERM, University of Montpellier, France
| | - Laura Palagini
- Department of Clinical Experimental Medicine, Psychiatric Unit, University of Pisa, School of Medicine, Pisa, Italy
| | - Gianluca Serafini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Maternal and Child Health, Psychiatry Section, University of Genoa, IRCCS San Martino, Genoa, Italy
| | - Michel Lejoyeux
- Département de psychiatrie et d'addictologie, AP-HP, GHU Paris Nord, DMU Neurosciences, Hopital Bichat - Claude Bernard, F-75018, Paris, France; Université Paris Cité, NeuroDiderot, Inserm, FHU I2-D2, F-75019, Paris, France; GHU Paris - Psychiatry & Neurosciences, 1 rue Cabanis, 75014, Paris, France
| | - Marie-Pia d'Ortho
- Université Paris Cité, NeuroDiderot, Inserm, FHU I2-D2, F-75019, Paris, France; Centre du Sommeil, Service de Physiologie - Explorations Fonctionnelles, AP-HP, Hôpital Bichat, F-75018, Paris, France
| | - Pierre A Geoffroy
- Département de psychiatrie et d'addictologie, AP-HP, GHU Paris Nord, DMU Neurosciences, Hopital Bichat - Claude Bernard, F-75018, Paris, France; Université Paris Cité, NeuroDiderot, Inserm, FHU I2-D2, F-75019, Paris, France; GHU Paris - Psychiatry & Neurosciences, 1 rue Cabanis, 75014, Paris, France; CNRS UPR 3212, Institute for Cellular and Integrative Neurosciences, F-67000, Strasbourg, France.
| |
Collapse
|
57
|
Holter KM, Pierce BE, Gould RW. Metabotropic glutamate receptor function and regulation of sleep-wake cycles. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 168:93-175. [PMID: 36868636 PMCID: PMC10973983 DOI: 10.1016/bs.irn.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Metabotropic glutamate (mGlu) receptors are the most abundant family of G-protein coupled receptors and are widely expressed throughout the central nervous system (CNS). Alterations in glutamate homeostasis, including dysregulations in mGlu receptor function, have been indicated as key contributors to multiple CNS disorders. Fluctuations in mGlu receptor expression and function also occur across diurnal sleep-wake cycles. Sleep disturbances including insomnia are frequently comorbid with neuropsychiatric, neurodevelopmental, and neurodegenerative conditions. These often precede behavioral symptoms and/or correlate with symptom severity and relapse. Chronic sleep disturbances may also be a consequence of primary symptom progression and can exacerbate neurodegeneration in disorders including Alzheimer's disease (AD). Thus, there is a bidirectional relationship between sleep disturbances and CNS disorders; disrupted sleep may serve as both a cause and a consequence of the disorder. Importantly, comorbid sleep disturbances are rarely a direct target of primary pharmacological treatments for neuropsychiatric disorders even though improving sleep can positively impact other symptom clusters. This chapter details known roles of mGlu receptor subtypes in both sleep-wake regulation and CNS disorders focusing on schizophrenia, major depressive disorder, post-traumatic stress disorder, AD, and substance use disorder (cocaine and opioid). In this chapter, preclinical electrophysiological, genetic, and pharmacological studies are described, and, when possible, human genetic, imaging, and post-mortem studies are also discussed. In addition to reviewing the important relationships between sleep, mGlu receptors, and CNS disorders, this chapter highlights the development of selective mGlu receptor ligands that hold promise for improving both primary symptoms and sleep disturbances.
Collapse
Affiliation(s)
- Kimberly M Holter
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Bethany E Pierce
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Robert W Gould
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States.
| |
Collapse
|
58
|
Rosales-Lagarde A, Cubero-Rego L, Menéndez-Conde F, Rodríguez-Torres EE, Itzá-Ortiz B, Martínez-Alcalá C, Vázquez-Tagle G, Vázquez-Mendoza E, Eraña Díaz ML. Dissociation of Arousal Index Between REM and NREM Sleep in Elderly Adults with Cognitive Impairment, No Dementia: A Pilot Study. J Alzheimers Dis 2023; 95:477-491. [PMID: 37574730 DOI: 10.3233/jad-230101] [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] [Indexed: 08/15/2023]
Abstract
BACKGROUND Sleep disruption in elderly has been associated with an increased risk of cognitive impairment and its transition into Alzheimer's disease (AD). High arousal indices (AIs) during sleep may serve as an early-stage biomarker of cognitive impairment non-dementia (CIND). OBJECTIVE Using full-night polysomnography (PSG), we investigated whether CIND is related to different AIs between NREM and REM sleep stages. METHODS Fourteen older adults voluntarily participated in this population-based study that included Mini-Mental State Examination, Neuropsi battery, Katz Index of Independence in Activities of Daily Living, and single-night PSG. Subjects were divided into two groups (n = 7 each) according to their results in Neuropsi memory and attention subtests: cognitively unimpaired (CU), with normal results; and CIND, with -2.5 standard deviations in memory and/or attention subtests. AIs per hour of sleep during N1, N2, N3, and REM stages were obtained and correlated with Neuropsi total score (NTS). RESULTS AI (REM) was significantly higher in CU group than in CIND group. For the total sample, a positive correlation between AI (REM) and NTS was found (r = 0.68, p = 0.006), which remained significant when controlling for the effect of age and education. In CIND group, the AI (N2) was significantly higher than the AI (REM) . CONCLUSION In CIND older adults, this attenuation of normal arousal mechanisms in REM sleep are dissociated from the relative excess of arousals observed in stage N2. We propose as probable etiology an early hypoactivity at the locus coeruleus noradrenergic system, associated to its early pathological damage, present in the AD continuum.
Collapse
Affiliation(s)
- Alejandra Rosales-Lagarde
- CONACyT Chairs, National Council of Science and Technology, Mexico
- National Institute of Psychiatry Ramón de la Fuente Muñiz, Mexico
| | - Lourdes Cubero-Rego
- Neurodevelopmental Research Unit, Institute of Neurobiology, National Autonomous University of Mexico, Campus Juriquilla-Queretaro, Querétaro, México
| | | | | | - Benjamín Itzá-Ortiz
- Mathematics Research Center, Autonomous University of the State of Hidalgo, Mexico
| | - Claudia Martínez-Alcalá
- CONACyT Chairs, National Council of Science and Technology, Mexico
- Institute of Health Sciences, Autonomous University of the State of Hidalgo, Mexico
| | | | | | - Marta L Eraña Díaz
- Center for Research in Engineering and Applied Sciences, Autonomous University of the State of Morelos, Mexico
| |
Collapse
|
59
|
Kuo CC, Chan H, Hung WC, Chen RF, Yang HW, Min MY. Carbachol increases locus coeruleus activation by targeting noradrenergic neurons, inhibitory interneurons and inhibitory synaptic transmission. Eur J Neurosci 2023; 57:32-53. [PMID: 36382388 DOI: 10.1111/ejn.15866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/11/2022] [Accepted: 11/10/2022] [Indexed: 11/18/2022]
Abstract
The locus coeruleus (LC) consists of noradrenergic (NA) neurons and plays an important role in controlling behaviours. Although much of the knowledge regarding LC functions comes from studying behavioural outcomes upon administration of muscarinic acetylcholine receptor (mAChR) agonists into the nucleus, the exact mechanisms remain unclear. Here, we report that the application of carbachol (CCh), an mAChR agonist, increased the spontaneous action potentials (sAPs) of both LC-NA neurons and local inhibitory interneurons (LC I-INs) in acute brain slices by activating M1/M3 mAChRs (m1/3 AChRs). Optogenetic activation of LC I-INs evoked inhibitory postsynaptic currents (IPSCs) in LC-NA neurons that were mediated by γ-aminobutyric acid type A (GABAA ) and glycine receptors, and CCh application decreased the IPSC amplitude through a presynaptic mechanism by activating M4 mAChRs (m4 AChRs). LC-NA neurons also exhibited spontaneous phasic-like activity (sPLA); CCh application increased the incidence of this activity. This effect of CCh application was not observed with blockade of GABAA and glycine receptors, suggesting that the sPLA enhancement occurred likely because of the decreased synaptic transmission of LC I-INs onto LC-NA neurons by the m4 AChR activation and/or increased spiking rate of LC I-INs by the m1/3 AChR activation, which could lead to fatigue of the synaptic transmission. In conclusion, we report that CCh application, while inhibiting their synaptic transmission, increases sAP rates of LC-NA neurons and LC I-INs. Collectively, these effects provide insight into the cellular mechanisms underlying the behaviour modulations following the administration of muscarinic receptor agonists into the LC reported by the previous studies.
Collapse
Affiliation(s)
- Chao-Cheng Kuo
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Hao Chan
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Wei-Chen Hung
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Ruei-Feng Chen
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan.,Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan
| | - Hsiu-Wen Yang
- Department of Biomedical Sciences, Chung-Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung-Shan Medical University Hospital, Taichung, Taiwan
| | - Ming-Yuan Min
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan.,Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
60
|
Agrawal S, Kumar V, Singh V, Singh C, Singh A. A Review on Pathophysiological Aspects of Sleep Deprivation. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:1194-1208. [PMID: 35549867 DOI: 10.2174/1871527321666220512092718] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 12/03/2021] [Accepted: 01/16/2022] [Indexed: 11/22/2022]
Abstract
Sleep deprivation (SD) (also referred as insomnia) is a condition in which individuals fail to get enough sleep due to excessive yawning, facing difficulty to learn new concepts, experiencing forgetfulness as well as depressed mood. This could occur due to several possible reasons, including medications and stress (caused by shift work). Despite the fact that sleep is important for normal physiology, it currently affects millions of people around the world, especially the US (70 million) and Europe (45 million). Due to increased work demand nowadays, lots of people are experiencing sleep deprivation hence, this could be the reason for several car accidents followed by death and morbidity. This review highlighted the impact of SD on neurotransmitter release and functions, theories (Flip-flop theory, oxidative stress theory, neuroinflammation theory, neurotransmitter theory, and hormonal theory) associated with SD pathogenesis; apart from this, it also demonstrates the molecular pathways underlying SD (PI3K and Akt, NF-κB, Nrf2, and adenosine pathway. However, this study also elaborates on the SD-induced changes in the level of neurotransmitters, hormonal, and mitochondrial functions. Along with this, it also covers several molecular aspects associated with SD as well. Through this study, a link is made between SD and associated causes, which will further help to develop a potential therapeutic strategy against SD.
Collapse
Affiliation(s)
- Shelly Agrawal
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Vishal Kumar
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Vishesh Singh
- Department of Pharmaceutics, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Charan Singh
- Department of Pharmaceutics, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Arti Singh
- Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab, India
| |
Collapse
|
61
|
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
|
62
|
Doppler CEJ, Smit J, Hommelsen M, Seger A, Okkels N, Horsager J, Kinnerup M, Hansen AK, Fedorova TD, Knudsen K, Otto M, Nahimi A, Fink GR, Borghammer P, Sommerauer M. Disruption of Sleep Microarchitecture Is a Sensitive and Early Marker of Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2022; 12:2555-2560. [PMID: 36189604 DOI: 10.3233/jpd-223442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Although sleep disturbances are highly prevalent in patients with Parkinson's disease, sleep macroarchitecture metrics show only minor changes. OBJECTIVE To assess alterations of the cyclic alternating pattern (CAP) as a critical feature of sleep microarchitecture in patients with prodromal, recent, and established Parkinson's disease. METHODS We evaluated overnight polysomnography for classic sleep macroarchitecture and CAP metrics in 68 patients at various disease stages and compared results to 22 age- and sex-matched controls. RESULTS Already at the prodromal stage, patients showed a significantly reduced CAP rate as a central characteristic of sleep microarchitecture. Temporal characteristics of CAP showed a gradual change over disease stages and correlated with motor performance. In contrast, the sleep macroarchitecture metrics did not differ between groups. CONCLUSION Data suggest that alterations of sleep microarchitecture are an early and more sensitive characteristic of Parkinson's disease than changes in sleep macroarchitecture.
Collapse
Affiliation(s)
- Christopher E J Doppler
- Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich, Jülich, Germany.,Department of Neurology, University Hospital Cologne, Faculty of Medicine, University of Cologne, Köln, Germany
| | - Julia Smit
- Department of Neurology, University Hospital Cologne, Faculty of Medicine, University of Cologne, Köln, Germany
| | - Maximilian Hommelsen
- Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich, Jülich, Germany
| | - Aline Seger
- Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich, Jülich, Germany.,Department of Neurology, University Hospital Cologne, Faculty of Medicine, University of Cologne, Köln, Germany
| | - Niels Okkels
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus, Denmark.,Department of Neurology, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jacob Horsager
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Martin Kinnerup
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Allan K Hansen
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Tatyana D Fedorova
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Karoline Knudsen
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Marit Otto
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Neurophysiology, Aarhus University Hospital, Aarhus, Denmark
| | - Adjmal Nahimi
- Department of Neurology, Rehabilitation Medicine, Memory Disorders, and Geriatrics, Skåne University Hospital, Lund, Sweden
| | - Gereon R Fink
- Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich, Jülich, Germany.,Department of Neurology, University Hospital Cologne, Faculty of Medicine, University of Cologne, Köln, Germany
| | - Per Borghammer
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Michael Sommerauer
- Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich, Jülich, Germany.,Department of Neurology, University Hospital Cologne, Faculty of Medicine, University of Cologne, Köln, Germany.,Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
63
|
He Z, Wang X, Ma K, Zheng L, Zhang Y, Liu C, Sun T, Wang P, Rong W, Niu J. Selective activation of the hypothalamic orexinergic but not melanin-concentrating hormone neurons following pilocarpine-induced seizures in rats. Front Neurosci 2022; 16:1056706. [DOI: 10.3389/fnins.2022.1056706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/11/2022] [Indexed: 12/05/2022] Open
Abstract
IntroductionSleep disorders are common comorbidities in patients with temporal lobe epilepsy (TLE), but the underlying mechanisms remain poorly understood. Since the lateral hypothalamic (LH) and the perifornical orexinergic (ORX) and melanin-concentrating hormone (MCH) neurons are known to play opposing roles in the regulation of sleep and arousal, dysregulation of ORX and MCH neurons might contribute to the disturbance of sleep-wakefulness following epileptic seizures.MethodsTo test this hypothesis, rats were treated with lithium chloride and pilocarpine to induce status epilepticus (SE). Electroencephalogram (EEG) and electromyograph (EMG) were recorded for analysis of sleep-wake states before and 24 h after SE. Double-labeling immunohistochemistry of c-Fos and ORX or MCH was performed on brain sections from the epileptic and control rats. In addition, anterograde and retrograde tracers in combination with c-Fos immunohistochemistry were used to analyze the possible activation of the amygdala to ORX neural pathways following seizures.ResultsIt was found that epileptic rats displayed prolonged wake phase and decreased non-rapid eye movement (NREM) and rapid eye movement (REM) phase compared to the control rats. Prominent neuronal activation was observed in the amygdala and the hypothalamus following seizures. Interestingly, in the LH and the perifornical nucleus, ORX but not MCH neurons were significantly activated (c-Fos+). Neural tracing showed that seizure-activated (c-Fos+) ORX neurons were closely contacted by axon terminals originating from neurons in the medial amygdala.DiscussionThese findings suggest that the spread of epileptic activity from amygdala to the hypothalamus causes selective activation of the wake-promoting ORX neurons but not sleep-promoting MCH neurons, which might contribute to the disturbance of sleep-wakefulness in TLE.
Collapse
|
64
|
Sun J, Chen Y, Sun Y, Yang B, Zhou J. Short sleep duration associated with increased risk for new-onset cardiovascular diseases in individuals with metabolic syndromes: Evidence from the China Health and Retirement Longitudinal Study. Front Cardiovasc Med 2022; 9:1010941. [PMID: 36419500 PMCID: PMC9678247 DOI: 10.3389/fcvm.2022.1010941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/13/2022] [Indexed: 11/09/2022] Open
Abstract
To explore the impact and risk of short sleep duration (sleep duration < 6 h/night) on new-onset cardiovascular and cerebrovascular diseases (CVDs) in people with metabolic syndromes (Mets), this study used the 2011 baseline and 2015 follow-up data from the China Longitudinal Study of Health and Retirement (CHARLS) to conduct a prospective study of people aged ≥ 45 years in China. A total of 5,530 individuals without pre-existing CVDs in baseline were included. Mets were defined according to the harmonized criteria. We applied the Logistic Regression (LR), the Deep Neural Networks (DNN), and the Adaptive Boosting (AdaBoost), to evaluate the association between Mets components, short sleep, and the risk of new-onset CVDs, and the importance of multiple variates for new-onset CVDs. During the 4-year follow-up period, 512 individuals developed CVDs, and short sleep increased the risk of CVD in individuals with Mets. The odds ratio for prevalent CVD in Mets with short sleep group was 3.73 (95%CI 2.95–4.71; P < 0.001) compared to the normal group, and 1.99 (95% CI 1.58–2.51; P < 0.001) compared to the Mets without short sleep group. The DNN method reached the highest precision of 92.24% and f1-score of 95.86%, and the Adaboost method reached the highest recall of 99.92%. Both DNN and Adaboost have better predictive performance than LR and revealed short sleep duration and components of Mets are all the strongest predictors of CVD onset.
Collapse
Affiliation(s)
- Jiaxin Sun
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yizhou Chen
- School of Computer Science, Wuhan University, Wuhan, China
| | - Yazhou Sun
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
- *Correspondence: Bo Yang
| | - Jining Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Jining Zhou
| |
Collapse
|
65
|
Tong C, Liu C, Zhang K, Bo B, Xia Y, Yang H, Feng Y, Liang Z. Multimodal analysis demonstrating the shaping of functional gradients in the marmoset brain. Nat Commun 2022; 13:6584. [PMID: 36329036 PMCID: PMC9633775 DOI: 10.1038/s41467-022-34371-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022] Open
Abstract
The discovery of functional gradients introduce a new perspective in understanding the cortical spectrum of intrinsic dynamics, as it captures major axes of functional connectivity in low-dimensional space. However, how functional gradients arise and dynamically vary remains poorly understood. In this study, we investigated the biological basis of functional gradients using awake resting-state fMRI, retrograde tracing and gene expression datasets in marmosets. We found functional gradients in marmosets showed a sensorimotor-to-visual principal gradient followed by a unimodal-to-multimodal gradient, resembling functional gradients in human children. Although strongly constrained by structural wirings, functional gradients were dynamically modulated by arousal levels. Utilizing a reduced model, we uncovered opposing effects on gradient dynamics by structural connectivity (inverted U-shape) and neuromodulatory input (U-shape) with arousal fluctuations, and dissected the contribution of individual neuromodulatory receptors. This study provides insights into biological basis of functional gradients by revealing the interaction between structural connectivity and ascending neuromodulatory system.
Collapse
Affiliation(s)
- Chuanjun Tong
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
- Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- Guangdong Provincial Key Laboratory of Medical Image Processing & Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, China
| | - Cirong Liu
- Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Kaiwei Zhang
- Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Binshi Bo
- Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Ying Xia
- Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Hao Yang
- Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yanqiu Feng
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Medical Image Processing & Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, China.
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence & Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, China.
| | - Zhifeng Liang
- Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China.
| |
Collapse
|
66
|
Bibineyshvili Y, Schiff ND, Calderon DP. Dexmedetomidine-mediated sleep phase modulation ameliorates motor and cognitive performance in a chronic blast-injured mouse model. Front Neurol 2022; 13:1040975. [PMID: 36388181 PMCID: PMC9663850 DOI: 10.3389/fneur.2022.1040975] [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: 09/09/2022] [Accepted: 10/17/2022] [Indexed: 10/22/2024] Open
Abstract
Multiple studies have shown that blast injury is followed by sleep disruption linked to functional sequelae. It is well established that improving sleep ameliorates such functional deficits. However, little is known about longitudinal brain activity changes after blast injury. In addition, the effects of directly modulating the sleep/wake cycle on learning task performance after blast injury remain unclear. We hypothesized that modulation of the sleep phase cycle in our injured mice would improve post-injury task performance. Here, we have demonstrated that excessive sleep electroencephalographic (EEG) patterns are accompanied by prominent motor and cognitive impairment during acute stage after secondary blast injury (SBI) in a mouse model. Over time we observed a transition to more moderate and prolonged sleep/wake cycle disturbances, including changes in theta and alpha power. However, persistent disruptions of the non-rapid eye movement (NREM) spindle amplitude and intra-spindle frequency were associated with lasting motor and cognitive deficits. We, therefore, modulated the sleep phase of injured mice using subcutaneous (SC) dexmedetomidine (Dex), a common, clinically used sedative. Dex acutely improved intra-spindle frequency, theta and alpha power, and motor task execution in chronically injured mice. Moreover, dexmedetomidine ameliorated cognitive deficits a week after injection. Our results suggest that SC Dex might potentially improve impaired motor and cognitive behavior during daily tasks in patients that are chronically impaired by blast-induced injuries.
Collapse
Affiliation(s)
- Yelena Bibineyshvili
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY, United States
| | - Nicholas D. Schiff
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, United States
| | - Diany P. Calderon
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY, United States
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
67
|
Blumberg MS, Dooley JC, Tiriac A. Sleep, plasticity, and sensory neurodevelopment. Neuron 2022; 110:3230-3242. [PMID: 36084653 PMCID: PMC9588561 DOI: 10.1016/j.neuron.2022.08.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 07/04/2022] [Accepted: 08/11/2022] [Indexed: 11/18/2022]
Abstract
A defining feature of early infancy is the immense neural plasticity that enables animals to develop a brain that is functionally integrated with a growing body. Early infancy is also defined as a period dominated by sleep. Here, we describe three conceptual frameworks that vary in terms of whether and how they incorporate sleep as a factor in the activity-dependent development of sensory and sensorimotor systems. The most widely accepted framework is exemplified by the visual system where retinal waves seemingly occur independent of sleep-wake states. An alternative framework is exemplified by the sensorimotor system where sensory feedback from sleep-specific movements activates the brain. We prefer a third framework that encompasses the first two but also captures the diverse ways in which sleep modulates activity-dependent development throughout the nervous system. Appreciation of the third framework will spur progress toward a more comprehensive and cohesive understanding of both typical and atypical neurodevelopment.
Collapse
Affiliation(s)
- Mark S Blumberg
- Department of Psychological & Brain Sciences, University of Iowa, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA.
| | - James C Dooley
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA.
| | - Alexandre Tiriac
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA.
| |
Collapse
|
68
|
Jiang H. Hypothalamic GABAergic neurocircuitry in the regulation of energy homeostasis and sleep/wake control. MEDICAL REVIEW (BERLIN, GERMANY) 2022; 2:531-540. [PMID: 37724165 PMCID: PMC10388747 DOI: 10.1515/mr-2022-0022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 08/10/2022] [Indexed: 09/20/2023]
Abstract
Gamma-aminobutyric acid (GABAergic) neuron, as one of important cell types in synaptic transmission, has been widely involved in central nervous system (CNS) regulation of organismal physiologies including cognition, emotion, arousal and reward. However, upon their distribution in various brain regions, effects of GABAergic neurons in the brain are very diverse. In current report, we will present an overview of the role of GABAergic mediated inhibitory neurocircuitry in the hypothalamus, underlying mechanism of feeding and sleep homeostasis as well as the characteristics of latest transcriptome profile in order to call attention to the GABAergic system as potentially a promising pharmaceutical intervention or a deep brain stimulation target in eating and sleep disorders.
Collapse
Affiliation(s)
- Hong Jiang
- Department of Neurobiology, School of Basic Medical Sciences, Neuroscience Research Institute, Peking University, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of China, Peking University, Beijing, China
| |
Collapse
|
69
|
Maness EB, Burk JA, McKenna JT, Schiffino FL, Strecker RE, McCoy JG. Role of the locus coeruleus and basal forebrain in arousal and attention. Brain Res Bull 2022; 188:47-58. [PMID: 35878679 PMCID: PMC9514025 DOI: 10.1016/j.brainresbull.2022.07.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 07/11/2022] [Accepted: 07/20/2022] [Indexed: 12/11/2022]
Abstract
Experimental evidence has implicated multiple neurotransmitter systems in either the direct or indirect modulation of cortical arousal and attention circuitry. In this review, we selectively focus on three such systems: 1) norepinephrine (NE)-containing neurons of the locus coeruleus (LC), 2) acetylcholine (ACh)-containing neurons of the basal forebrain (BF), and 3) parvalbumin (PV)-containing gamma-aminobutyric acid neurons of the BF. Whereas BF-PV neurons serve as a rapid and transient arousal system, LC-NE and BF-ACh neuromodulation are typically activated on slower but longer-lasting timescales. Recent findings suggest that the BF-PV system serves to rapidly respond to even subtle sensory stimuli with a microarousal. We posit that salient sensory stimuli, such as those that are threatening or predict the need for a response, will quickly activate the BF-PV system and subsequently activate both the BF-ACh and LC-NE systems if the circumstances require longer periods of arousal and vigilance. We suggest that NE and ACh have overlapping psychological functions with the main difference being the precise internal/environmental sensory situations/contexts that recruit each neurotransmitter system - a goal for future research to determine. Implications of dysfunction of each of these three attentional systems for our understanding of neuropsychiatric conditions are considered. Finally, the contemporary availability of research tools to selectively manipulate and measure the activity of these distinctive neuronal populations promises to answer longstanding questions, such as how various arousal systems influence downstream decision-making and motor responding.
Collapse
Affiliation(s)
- Eden B Maness
- VA Boston Healthcare System and Department of Psychiatry, Harvard Medical School, West Roxbury, MA 02132, USA.
| | - Joshua A Burk
- Department of Psychological Sciences, College of William and Mary, Williamsburg, VA 23187, USA
| | - James T McKenna
- VA Boston Healthcare System and Department of Psychiatry, Harvard Medical School, West Roxbury, MA 02132, USA
| | - Felipe L Schiffino
- VA Boston Healthcare System and Department of Psychiatry, Harvard Medical School, West Roxbury, MA 02132, USA; Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Robert E Strecker
- VA Boston Healthcare System and Department of Psychiatry, Harvard Medical School, West Roxbury, MA 02132, USA.
| | - John G McCoy
- Department of Psychology, Stonehill College, Easton, MA 02357, USA.
| |
Collapse
|
70
|
Setzer B, Fultz NE, Gomez DEP, Williams SD, Bonmassar G, Polimeni JR, Lewis LD. A temporal sequence of thalamic activity unfolds at transitions in behavioral arousal state. Nat Commun 2022; 13:5442. [PMID: 36114170 PMCID: PMC9481532 DOI: 10.1038/s41467-022-33010-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 08/29/2022] [Indexed: 11/30/2022] Open
Abstract
Awakening from sleep reflects a profound transformation in neural activity and behavior. The thalamus is a key controller of arousal state, but whether its diverse nuclei exhibit coordinated or distinct activity at transitions in behavioral arousal state is unknown. Using fast fMRI at ultra-high field (7 Tesla), we measured sub-second activity across thalamocortical networks and within nine thalamic nuclei to delineate these dynamics during spontaneous transitions in behavioral arousal state. We discovered a stereotyped sequence of activity across thalamic nuclei and cingulate cortex that preceded behavioral arousal after a period of inactivity, followed by widespread deactivation. These thalamic dynamics were linked to whether participants subsequently fell back into unresponsiveness, with unified thalamic activation reflecting maintenance of behavior. These results provide an outline of the complex interactions across thalamocortical circuits that orchestrate behavioral arousal state transitions, and additionally, demonstrate that fast fMRI can resolve sub-second subcortical dynamics in the human brain.
Collapse
Affiliation(s)
- Beverly Setzer
- Graduate Program for Neuroscience, Boston University, Boston, MA, 02215, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Nina E Fultz
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, 02129, USA
| | - Daniel E P Gomez
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, 02129, USA
- Department of Radiology, Harvard Medical School, Boston, MA, 02115, USA
| | | | - Giorgio Bonmassar
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, 02129, USA
- Department of Radiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Jonathan R Polimeni
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, 02129, USA
- Department of Radiology, Harvard Medical School, Boston, MA, 02115, USA
- Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Laura D Lewis
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA.
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, 02129, USA.
| |
Collapse
|
71
|
Darchia N, Campbell IG, Basishvili T, Eliozishvili M, Tchintcharauli T, Oniani N, Sakhelashvili I, Feinberg I. Sleep electroencephalogram evidence of delayed brain maturation in attention deficit hyperactivity disorder: a longitudinal study. Sleep 2022; 45:6648473. [DOI: 10.1093/sleep/zsac163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/25/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
Study Objectives
This study investigates whether longitudinally measured changes in adolescent brain electrophysiology corroborate the maturational lag associated with attention deficit hyperactivity disorder (ADHD) reported in magnetic resonance imaging (MRI) studies and cross-sectional sleep electroencephalogram (EEG) data.
Methods
Semiannually nine adolescents diagnosed with ADHD (combined presentation, DSM-V criteria, mean age 12.39 ± 0.61 years at first time-point, two females) and nine typically developing controls (12.08 ± 0.35 years, four females) underwent all-night laboratory polysomnography, yielding four recordings.
Results
Sleep macrostructure was similar between groups. A quadratic model of the age change in non-rapid eye movement (NREM) delta (1.07–4 Hz) power, with sex effects accounted for, found that delta power peaked 0.92 ± 0.37 years later in the ADHD group. A Gompertz function fit to the same data showed that the age of most rapid delta power decline occurred 0.93 ± 0.41 years later in the ADHD group (p = 0.037), but this group difference was not significant (p = 0.38) with sex effects accounted for. For very low frequency (0.29–1.07 Hz) EEG, the ADHD lag (1.07 ± 0.42 years later, p = 0.019) was significant for a Gompertz model with sex effects accounted for (p = 0.044). Theta (4–7.91 Hz) showed a trend (p = 0.064) toward higher power in the ADHD group. Analysis of the EEG decline across the night found that standardized delta and theta power in NREMP1 were significantly (p < 0.05 for both) lower in adolescents with ADHD.
Conclusions
This is the first longitudinal study to reveal electrophysiological evidence of a maturational lag associated with ADHD. In addition, our findings revealed basically unaltered sleep macrostructure but altered sleep homeostasis associated with ADHD.
Collapse
Affiliation(s)
- Nato Darchia
- Tengiz Oniani Laboratory of Sleep-Wakefulness Cycle Study, Ilia State University , Tbilisi , Georgia
| | - Ian G Campbell
- Department of Psychiatry and Behavioral Sciences, University of California Davis , Davis, CA , USA
| | - Tamar Basishvili
- Tengiz Oniani Laboratory of Sleep-Wakefulness Cycle Study, Ilia State University , Tbilisi , Georgia
| | - Marine Eliozishvili
- Tengiz Oniani Laboratory of Sleep-Wakefulness Cycle Study, Ilia State University , Tbilisi , Georgia
| | | | - Nikoloz Oniani
- Tengiz Oniani Laboratory of Sleep-Wakefulness Cycle Study, Ilia State University , Tbilisi , Georgia
| | - Irine Sakhelashvili
- Tengiz Oniani Laboratory of Sleep-Wakefulness Cycle Study, Ilia State University , Tbilisi , Georgia
| | - Irwin Feinberg
- Department of Psychiatry and Behavioral Sciences, University of California Davis , Davis, CA , USA
| |
Collapse
|
72
|
Moscarello JM, Penzo MA. The central nucleus of the amygdala and the construction of defensive modes across the threat-imminence continuum. Nat Neurosci 2022; 25:999-1008. [PMID: 35915178 DOI: 10.1038/s41593-022-01130-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/23/2022] [Indexed: 11/09/2022]
Abstract
In nature, animals display defensive behaviors that reflect the spatiotemporal distance of threats. Laboratory-based paradigms that elicit specific defensive responses in rodents have provided valuable insight into the brain mechanisms that mediate the construction of defensive modes with varying degrees of threat imminence. In this Review, we discuss accumulating evidence that the central nucleus of the amygdala (CeA) plays a key role in this process. Specifically, we propose that the mutually inhibitory circuits of the CeA use a winner-takes-all strategy that supports transitioning across defensive modes and the execution of specific defensive behaviors to previously formed threat associations. Our proposal provides a conceptual framework in which seemingly divergent observations regarding CeA function can be interpreted and identifies various areas of priority for future research.
Collapse
Affiliation(s)
- Justin M Moscarello
- Department of Psychological & Brain Sciences, Institute for Neuroscience, Texas A&M University, College Station, TX, USA.
| | - Mario A Penzo
- Unit on the Neurobiology of Affective Memory, National Institute of Mental Health, Bethesda, MD, USA.
| |
Collapse
|
73
|
Kirouac GJ, Li S, Li S. Convergence of monosynaptic inputs from neurons in the brainstem and forebrain on parabrachial neurons that project to the paraventricular nucleus of the thalamus. Brain Struct Funct 2022; 227:2409-2437. [PMID: 35838792 PMCID: PMC9418111 DOI: 10.1007/s00429-022-02534-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 06/30/2022] [Indexed: 11/28/2022]
Abstract
The paraventricular nucleus of the thalamus (PVT) projects to areas of the forebrain involved in regulating behavior. Homeostatic challenges and salient cues activate the PVT and evidence shows that the PVT regulates appetitive and aversive responses. The brainstem is a source of afferents to the PVT and the present study was done to determine if the lateral parabrachial nucleus (LPB) is a relay for inputs to the PVT. Retrograde tracing experiments with cholera toxin B (CTB) demonstrate that the LPB contains more PVT projecting neurons than other regions of the brainstem including the catecholamine cell groups. The hypothesis that the LPB is a relay for signals to the PVT was assessed using an intersectional monosynaptic rabies tracing approach. Sources of inputs to LPB included the reticular formation; periaqueductal gray (PAG); nucleus cuneiformis; and superior and inferior colliculi. Distinctive clusters of input cells to LPB-PVT projecting neurons were also found in the dorsolateral bed nucleus of the stria terminalis (BSTDL) and the lateral central nucleus of the amygdala (CeL). Anterograde viral tracing demonstrates that LPB-PVT neurons densely innervate all regions of the PVT in addition to providing collateral innervation to the preoptic area, lateral hypothalamus, zona incerta and PAG but not the BSTDL and CeL. The paper discusses the anatomical evidence that suggests that the PVT is part of a network of interconnected neurons involved in arousal, homeostasis, and the regulation of behavioral states with forebrain regions potentially providing descending modulation or gating of signals relayed from the LPB to the PVT.
Collapse
Affiliation(s)
- Gilbert J Kirouac
- Department of Oral Biology, Dr. Gerald Niznick College of Dentistry, Rady Faculty of Health Sciences, University of Manitoba, 780 Bannatyne Avenue, Winnipeg, MB, R3E 0W2, Canada. .,Departments of Psychiatry and Human Anatomy and Cell Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, R3E 0W2, Canada.
| | - Sa Li
- Department of Oral Biology, Dr. Gerald Niznick College of Dentistry, Rady Faculty of Health Sciences, University of Manitoba, 780 Bannatyne Avenue, Winnipeg, MB, R3E 0W2, Canada
| | - Shuanghong Li
- Department of Oral Biology, Dr. Gerald Niznick College of Dentistry, Rady Faculty of Health Sciences, University of Manitoba, 780 Bannatyne Avenue, Winnipeg, MB, R3E 0W2, Canada
| |
Collapse
|
74
|
Leung LS, Ma J. Medial Septum Modulates Consciousness and Psychosis-Related Behaviors Through Hippocampal Gamma Activity. Front Neural Circuits 2022; 16:895000. [PMID: 35874429 PMCID: PMC9301478 DOI: 10.3389/fncir.2022.895000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
Abnormally high-amplitude hippocampal gamma activity (30–100 Hz) in behaving animals is seen after a hippocampal seizure, following injection of phencyclidine (PCP) or ketamine, and transiently in a delirium stage during induction of general anesthesia. High-amplitude hippocampal gamma activity in behaving rats is associated with hyperactive behavior and impairment in sensorimotor gating and sensory gating. The medial septum is necessary for the high-amplitude gamma activity and abnormal behaviors observed following a hippocampal seizure or injection of PCP/ketamine. Glutamatergic projection of the hippocampus to the nucleus accumbens (NAC) and dopaminergic transmission in NAC is necessary for abnormal behaviors. Large hippocampal gamma waves are suggested to contribute to seizure-induced automatism following temporal lobe seizures, and the schizophrenia-like symptoms induced by PCP/ketamine. Low-amplitude gamma activity is found during general anesthesia, associated with loss of consciousness in humans and loss of righting reflex in animals. Local inactivation or lesion of the medial septum, NAC, and brain areas connected to the septohippocampal-NAC system attenuates the increase in hippocampal gamma and associated behavioral disruptions induced by hippocampal seizure or PCP/ketamine. Inactivation or lesion of the septohippocampal-NAC system decreases the dose of anesthetic necessary for gamma decrease and loss of consciousness in animals. Thus, it is proposed that the septohippocampal-NAC system serves to control consciousness and the behavioral hyperactivity and neural dysfunctions during psychosis.
Collapse
|
75
|
Pandi-Perumal SR, Cardinali DP, Zaki NFW, Karthikeyan R, Spence DW, Reiter RJ, Brown GM. Timing is everything: Circadian rhythms and their role in the control of sleep. Front Neuroendocrinol 2022; 66:100978. [PMID: 35033557 DOI: 10.1016/j.yfrne.2022.100978] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/12/2021] [Accepted: 01/08/2022] [Indexed: 01/16/2023]
Abstract
Sleep and the circadian clock are intertwined and have persisted throughout history. The suprachiasmatic nucleus (SCN) orchestrates sleep by controlling circadian (Process C) and homeostatic (Process S) activities. As a "hand" on the endogenous circadian clock, melatonin is critical for sleep regulation. Light serves as a cue for sleep/wake control by activating retino-recipient cells in the SCN and subsequently suppressing melatonin. Clock genes are the molecular timekeepers that keep the 24 h cycle in place. Two main sleep and behavioural disorder diagnostic manuals have now officially recognised the importance of these processes for human health and well-being. The body's ability to respond to daily demands with the least amount of effort is maximised by carefully timing and integrating all components of sleep and waking. In the brain, the organization of timing is essential for optimal brain physiology.
Collapse
Affiliation(s)
- Seithikurippu R Pandi-Perumal
- Somnogen Canada Inc, College Street, Toronto, ON, Canada; Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| | - Daniel P Cardinali
- Faculty of Medical Sciences, Pontificia Universidad Católica Argentina, 1107 Buenos Aires, Argentina
| | - Nevin F W Zaki
- Department of Psychiatry, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | | | | | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Gregory M Brown
- Centre for Addiction and Mental Health, Molecular Brain Sciences, University of Toronto, 250 College St. Toronto, ON, Canada
| |
Collapse
|
76
|
Murillo-Rodríguez E, Coronado-Álvarez A, López-Muciño LA, Pastrana-Trejo JC, Viana-Torre G, Barberena JJ, Soriano-Nava DM, García-García F. Neurobiology of dream activity and effects of stimulants on dreams. Curr Top Med Chem 2022; 22:1280-1295. [PMID: 35761491 DOI: 10.2174/1568026622666220627162032] [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: 10/24/2021] [Revised: 03/18/2022] [Accepted: 04/11/2022] [Indexed: 11/22/2022]
Abstract
The sleep-wake cycle is the result of the activity of a multiple neurobiological network interaction. Dreaming feature is one interesting sleep phenomena that represents sensorial components, mostly visual perceptions, accompanied with intense emotions. Further complexity has been added to the topic of the neurobiological mechanism of dreams generation by the current data that suggests the influence of drugs on dream generation. Here, we discuss the review on some of the neurobiological mechanism of the regulation of dream activity, with special emphasis on the effects of stimulants on dreaming.
Collapse
Affiliation(s)
- Eric Murillo-Rodríguez
- Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud. Universidad Anáhuac Mayab. Mérida, Yucatán. México.,Intercontinental Neuroscience Research Group
| | - Astrid Coronado-Álvarez
- Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud. Universidad Anáhuac Mayab. Mérida, Yucatán. México.,Intercontinental Neuroscience Research Group
| | - Luis Angel López-Muciño
- Health Sciences Program. Health Sciences Institute. Veracruzana University. Xalapa. Veracruz. Mexico
| | - José Carlos Pastrana-Trejo
- Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud. Universidad Anáhuac Mayab. Mérida, Yucatán. México.,Intercontinental Neuroscience Research Group
| | - Gerardo Viana-Torre
- Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud. Universidad Anáhuac Mayab. Mérida, Yucatán. México.,Intercontinental Neuroscience Research Group
| | - Juan José Barberena
- Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud. Universidad Anáhuac Mayab. Mérida, Yucatán. México.,Intercontinental Neuroscience Research Group.,Escuela de Psicología, División Ciencias de la Salud. Universidad Anáhuac Mayab. Mérida, Yucatán. México
| | - Daniela Marcia Soriano-Nava
- Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud. Universidad Anáhuac Mayab. Mérida, Yucatán. México.,Intercontinental Neuroscience Research Group
| | - Fabio García-García
- Intercontinental Neuroscience Research Group.,Health Sciences Program. Health Sciences Institute. Veracruzana University. Xalapa. Veracruz. Mexico
| |
Collapse
|
77
|
Hartman AG, McKendry S, Soehner A, Bodison S, Akcakaya M, DeAlmeida D, Bendixen R. Characterizing Sleep Differences in Children With and Without Sensory Sensitivities. Front Psychol 2022; 13:875766. [PMID: 35814144 PMCID: PMC9257069 DOI: 10.3389/fpsyg.2022.875766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/03/2022] [Indexed: 01/28/2023] Open
Abstract
Objectives Individuals register and react to daily sensory stimuli differently, which influences participation in occupations. Sleep is a foundational nightly occupation that impacts overall health and development in children. Emerging research suggests that certain sensory processing patterns, specifically sensory sensitivities, may have a negative impact on sleep health in children. In this study, we aimed to (i) characterize sleep in children with and without sensory sensitivities and (ii) examine the relationship between sensory processing patterns (using the Sensory Profile-2) and sleep using validated parent- and child-reported questionnaires. We hypothesized that children with sensory sensitivities will exhibit more difficulties with sleep. Methods We recruited 22 children (ages 6-10) with sensory sensitivities (SS) and 33 children without sensory sensitivities (NSS) to complete validated sleep and sensory processing questionnaires: the Children's Sleep Habits Questionnaire (CSHQ), Sleep Self-Report (SSR), and Sensory Profile-2. Results Children with SS had significantly more sleep behaviors reported by both parents (p < 0.001, g = 1.11) and children (p < 0.001, g = 1.17) compared to children with NSS. Specifically, children with SS had higher frequencies of sleep anxiety (p = 0.004, g = 0.79), bedtime resistance (p = 0.001, g = 0.83), and sleep onset delay (p = 0.003, g = 0.95). Spearman's ρ correlations indicated significant positive correlations between parent- and child-reported sleep. Children with SS showed a larger association and greater variability between sleep and sensory processing compared to their peers. Significant positive correlations between parent-reported sleep behaviors and sensory sensitive and avoiding patterns were identified for both children with SS and NSS. Child-reported sleep behaviors were most strongly associated with sensitive and avoiding patterns for children with NSS and seeking patterns for children with SS. Conclusion We present evidence that sleep is impacted for children with SS to a greater extent than children with NSS. We also identified that a child's sensory processing pattern may be an important contributor to sleep problems in children with and without sensory sensitivities. Sleep concerns should be addressed within routine care for children with sensory sensitivities. Future studies will inform specific sleep intervention targets most salient for children with SS and other sensory processing patterns.
Collapse
Affiliation(s)
- Amy G. Hartman
- Department of Occupational Therapy, University of Pittsburgh, Pittsburgh, PA, United States
| | - Sarah McKendry
- Department of Occupational Therapy, University of Pittsburgh, Pittsburgh, PA, United States
| | - Adriane Soehner
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Stefanie Bodison
- Department of Occupational Therapy, University of Florida, Gainesville, FL, United States
| | - Murat Akcakaya
- Department of Electrical and Computer Engineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Dilhari DeAlmeida
- Department of Health Information Management, University of Pittsburgh, Pittsburgh, PA, United States
| | - Roxanna Bendixen
- Department of Occupational Therapy, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
78
|
Spatiotemporal dynamics of noradrenaline during learned behaviour. Nature 2022; 606:732-738. [PMID: 35650441 PMCID: PMC9837982 DOI: 10.1038/s41586-022-04782-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 04/20/2022] [Indexed: 01/17/2023]
Abstract
Noradrenaline released from the locus coeruleus (LC) is a ubiquitous neuromodulator1-4 that has been linked to multiple functions including arousal5-8, action and sensory gain9-11, and learning12-16. Whether and how activation of noradrenaline-expressing neurons in the LC (LC-NA) facilitates different components of specific behaviours is unknown. Here we show that LC-NA activity displays distinct spatiotemporal dynamics to enable two functions during learned behaviour: facilitating task execution and encoding reinforcement to improve performance accuracy. To examine these functions, we used a behavioural task in mice with graded auditory stimulus detection and task performance. Optogenetic inactivation of the LC demonstrated that LC-NA activity was causal for both task execution and optimization. Targeted recordings of LC-NA neurons using photo-tagging, two-photon micro-endoscopy and two-photon output monitoring showed that transient LC-NA activation preceded behavioural execution and followed reinforcement. These two components of phasic activity were heterogeneously represented in LC-NA cortical outputs, such that the behavioural response signal was higher in the motor cortex and facilitated task execution, whereas the negative reinforcement signal was widely distributed among cortical regions and improved response sensitivity on the subsequent trial. Modular targeting of LC outputs thus enables diverse functions, whereby some noradrenaline signals are segregated among targets, whereas others are broadly distributed.
Collapse
|
79
|
Chaturvedi R, Stork T, Yuan C, Freeman MR, Emery P. Astrocytic GABA transporter controls sleep by modulating GABAergic signaling in Drosophila circadian neurons. Curr Biol 2022; 32:1895-1908.e5. [PMID: 35303417 PMCID: PMC9090989 DOI: 10.1016/j.cub.2022.02.066] [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: 04/27/2021] [Revised: 01/11/2022] [Accepted: 02/23/2022] [Indexed: 11/16/2022]
Abstract
A precise balance between sleep and wakefulness is essential to sustain a good quality of life and optimal brain function. GABA is known to play a key and conserved role in sleep control, and GABAergic tone should, therefore, be tightly controlled in sleep circuits. Here, we examined the role of the astrocytic GABA transporter (GAT) in sleep regulation using Drosophila melanogaster. We found that a hypomorphic gat mutation (gat33-1) increased sleep amount, decreased sleep latency, and increased sleep consolidation at night. Interestingly, sleep defects were suppressed when gat33-1 was combined with a mutation disrupting wide-awake (wake), a gene that regulates the cell-surface levels of the GABAA receptor resistance to dieldrin (RDL) in the wake-promoting large ventral lateral neurons (l-LNvs). Moreover, RNAi knockdown of rdl and its modulators dnlg4 and wake in these circadian neurons also suppressed gat33-1 sleep phenotypes. Brain immunohistochemistry showed that GAT-expressing astrocytes were located near RDL-positive l-LNv cell bodies and dendritic processes. We concluded that astrocytic GAT decreases GABAergic tone and RDL activation in arousal-promoting LNvs, thus determining proper sleep amount and quality in Drosophila.
Collapse
Affiliation(s)
- Ratna Chaturvedi
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Tobias Stork
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Chunyan Yuan
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Marc R Freeman
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Patrick Emery
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
80
|
Meneghini S, Modena D, Colombo G, Coatti A, Milani N, Madaschi L, Amadeo A, Becchetti A. The β2V287L nicotinic subunit linked to sleep-related epilepsy differently affects fast-spiking and regular spiking somatostatin-expressing neurons in murine prefrontal cortex. Prog Neurobiol 2022; 214:102279. [DOI: 10.1016/j.pneurobio.2022.102279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 04/02/2022] [Accepted: 04/28/2022] [Indexed: 12/01/2022]
|
81
|
Wainstein G, Müller EJ, Taylor N, Munn B, Shine JM. The role of the locus coeruleus in shaping adaptive cortical melodies. Trends Cogn Sci 2022; 26:527-538. [DOI: 10.1016/j.tics.2022.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/03/2022] [Accepted: 03/17/2022] [Indexed: 10/18/2022]
|
82
|
Ågmo A, Laan E. Sexual incentive motivation, sexual behavior, and general arousal: Do rats and humans tell the same story? Neurosci Biobehav Rev 2022; 135:104595. [PMID: 35231490 DOI: 10.1016/j.neubiorev.2022.104595] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/28/2022] [Accepted: 02/24/2022] [Indexed: 12/13/2022]
Abstract
Sexual incentive stimuli activate sexual motivation and heighten the level of general arousal. The sexual motive may induce the individual to approach the incentive, and eventually to initiate sexual acts. Both approach and the ensuing copulatory interaction further enhance general arousal. We present data from rodents and humans in support of these assertions. We then suggest that orgasm is experienced when the combined level of excitation surpasses a threshold. In order to analyze the neurobiological bases of sexual motivation, we employ the concept of a central motive state. We then discuss the mechanisms involved in the long- and short-term control of that state as well as those mediating the momentaneous actions of sexual incentive stimuli. This leads to an analysis of the neurobiology behind the interindividual differences in responsivity of the sexual central motive state. Knowledge is still fragmentary, and many contradictory observations have been made. Nevertheless, we conclude that the basic mechanisms of sexual motivation and the role of general arousal are similar in rodents and humans.
Collapse
Affiliation(s)
- Anders Ågmo
- Department of Psychology, University of Tromsø, Norway.
| | - Ellen Laan
- Department of Sexology and Psychosomatic Gynaecology, Amsterdam UMC, University of Amsterdam, The Netherlands
| |
Collapse
|
83
|
Surman CBH, Walsh DM. Understanding the Impact of Stimulants on Sleep in ADHD: Evidence from Systematic Assessment of Sleep in Adults. CNS Drugs 2022; 36:253-260. [PMID: 35246824 DOI: 10.1007/s40263-022-00905-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/29/2022] [Indexed: 11/03/2022]
Abstract
Stimulants are widely prescribed to manage attention-deficit/hyperactivity disorder (ADHD) in adults. Stimulants promote wakefulness and can produce insomnia side effects. We hypothesized that systematic studies of sleep effects would reveal patterns of sleep impairment that may be important for clinicians to monitor and manage. We conducted a review and analysis of studies that measured sleep systematically during stimulant treatment in adults. We identified nine studies that met our search criteria, including four double-blind placebo-controlled studies. All studies recorded self-report subjective sleep quality data, three studies collected actigraphy data, and three studies collected polysomnography data. One study found better subjective sleep quality under open-label treatment conditions. Both polysomnography studies found improvement in aspects of sleep patterns. Two of the actigraphy studies suggested that adults receiving stimulant treatment may have less movement during sleep, and one showed reduction in amount of sleep. Further research could inform best practices for maintaining sleep quality during stimulant treatment.
Collapse
Affiliation(s)
- Craig B H Surman
- Clinical and Research Program in ADHD and Related Disorders, Massachusetts General Hospital, Boston, MA, USA. .,Harvard Medical School, Boston, MA, USA.
| | - Daniel M Walsh
- Clinical and Research Program in ADHD and Related Disorders, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
84
|
Zhang J, Li J, Liu C, Gui H, Yuan C, Zhang Y. The role of intracerebral dopamine D1 and D2 receptors in sleep-wake cycles and general anesthesia. IBRAIN 2022; 8:48-54. [PMID: 37786416 PMCID: PMC10528804 DOI: 10.1002/ibra.12024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/25/2022] [Accepted: 01/25/2022] [Indexed: 10/04/2023]
Abstract
Dopamine (DA), a monoamine neurotransmitter, is synthesized and released mainly by neurons in the ventral tegmental area and the substantia nigra (SN) pars compacta of the midbrain. DA and its receptors are essential for the regulation of arousal, movement, cognition, reward, and other neurobiological behaviors. Arousal, locomotion, cognition, reward, and other neurobiological functions are all regulated by dopamine and its receptors. Dopamine receptors can be divided into D1-like receptors (including D1 and D5) or D2-like receptors (containing D2, D3, and D4), with D1 and D2 receptors (D1Rs, and D2Rs) being the most important. Currently, studies indicated that D1Rs and D2Rs are tightly involved with the process of sleep-wake and general anesthesia, but the specific mechanism remains unclear. In this review, we compiled the most recent findings, mainly focusing on the structure, distribution, and signal pathway of D1Rs and D2Rs in the central nervous system, as well as the involvement of D1Rs and D2Rs in sleep-wake and general anesthesia. Thus, the investigations of the D1Rs and D2Rs will benefit not only better knowledge for how sleep-wake control works but also the mechanism of general anesthesia.
Collapse
Affiliation(s)
- Jie Zhang
- The Second Affiliated Hospital of Zunyi Medical UniversityZunyiChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
- School of AnesthesiologyZunyi Medical UniversityZunyiChina
| | - Jia Li
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
- School of AnesthesiologyZunyi Medical UniversityZunyiChina
| | - Cheng‐Xi Liu
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
- School of AnesthesiologyZunyi Medical UniversityZunyiChina
| | - Huan Gui
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
- School of AnesthesiologyZunyi Medical UniversityZunyiChina
| | - Cheng‐Dong Yuan
- The Second Affiliated Hospital of Zunyi Medical UniversityZunyiChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
- School of AnesthesiologyZunyi Medical UniversityZunyiChina
| | - Yi Zhang
- The Second Affiliated Hospital of Zunyi Medical UniversityZunyiChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
- School of AnesthesiologyZunyi Medical UniversityZunyiChina
| |
Collapse
|
85
|
Lefter R, Cojocariu RO, Ciobica A, Balmus IM, Mavroudis I, Kis A. Interactions between Sleep and Emotions in Humans and Animal Models. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:274. [PMID: 35208598 PMCID: PMC8877042 DOI: 10.3390/medicina58020274] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/31/2022] [Accepted: 02/09/2022] [Indexed: 12/12/2022]
Abstract
Recently, increased interest and efforts were observed in describing the possible interaction between sleep and emotions. Human and animal model studies addressed the implication of both sleep patterns and emotional processing in neurophysiology and neuropathology in suggesting a bidirectional interaction intimately modulated by complex mechanisms and factors. In this context, we aimed to discuss recent evidence and possible mechanisms implicated in this interaction, as provided by both human and animal models in studies. In addition, considering the affective component of brain physiological patterns, we aimed to find reasonable evidence in describing the two-way association between comorbid sleep impairments and psychiatric disorders. The main scientific literature databases (PubMed/Medline, Web of Science) were screened with keyword combinations for relevant content taking into consideration only English written papers and the inclusion and exclusion criteria, according to PRISMA guidelines. We found that a strong modulatory interaction between sleep processes and emotional states resides on the activity of several key brain structures, such as the amygdala, prefrontal cortex, hippocampus, and brainstem nuclei. In addition, evidence suggested that physiologically and behaviorally related mechanisms of sleep are intimately interacting with emotional perception and processing which could advise the key role of sleep in the unconscious character of emotional processes. However, further studies are needed to explain and correlate the functional analysis with causative and protective factors of sleep impairments and negative emotional modulation on neurophysiologic processing, mental health, and clinical contexts.
Collapse
Affiliation(s)
- Radu Lefter
- Center of Biomedical Research, Romanian Academy, Iasi Branch, B dul Carol I, no. 8, 700506 Iasi, Romania;
| | - Roxana Oana Cojocariu
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University, B dul Carol I, no 11, 700506 Iasi, Romania;
| | - Alin Ciobica
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University, B dul Carol I, no 11, 700506 Iasi, Romania;
- Center of Biomedical Research, Romanian Academy, B dul Carol I, no 8, 700505 Iasi, Romania
- Academy of Romanian Scientists, Splaiul Independentei nr. 54, Sector 5, 050094 Bucuresti, Romania
| | - Ioana-Miruna Balmus
- Department of Exact Sciences and Natural Sciences, Institute of Interdisciplinary Research, Alexandru Ioan Cuza University of Iasi, Alexandru Lapusneanu Street, no. 26, 700057 Iasi, Romania
| | - Ioannis Mavroudis
- Department of Neurology, Leeds Teaching Hospitals NHS Trust, Leeds LS2 9JT, UK;
| | - Anna Kis
- Institute of Cognitive Neuroscience and Psychology, Hungarian Academy of Sciences, 1117 Budapest, Hungary;
| |
Collapse
|
86
|
Neuroethology: Regulation of pre-sleep behaviors. Curr Biol 2022; 32:R160-R162. [DOI: 10.1016/j.cub.2022.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
87
|
Li IC, Chang FC, Kuo CC, Chu HT, Li TJ, Chen CC. Pilot Study: Nutritional and Preclinical Safety Investigation of Fermented Hispidin-Enriched Sanghuangporus sanghuang Mycelia: A Promising Functional Food Material to Improve Sleep. Front Nutr 2022; 8:788965. [PMID: 35111796 PMCID: PMC8801445 DOI: 10.3389/fnut.2021.788965] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/27/2021] [Indexed: 12/16/2022] Open
Abstract
Sleep disturbances have been the hallmark of the recent coronavirus disease 2019 pandemic. Studies have shown that once sleep is disrupted, it can lead to psychological and physical health issues which can, in turn, disrupt circadian rhythm and induce further sleep disruption. As consumers are trying to establish healthy routines, nutritional and preclinical safety investigation of fermented hispidin-enriched Sanghuangporus sanghuang mycelia (GKSS) as a novel food material for spontaneous sleep in Sprague-Dawley rats is conducted for the first time. Results showed that the nutritional analysis of GKSS including moisture, ash, crude lipid, crude protein, carbohydrate, and energy were found to be 2.4 ± 0.3%, 8.0 ± 2.5%, 1.7 ± 0.3%, 22.9 ± 1.2%, 65.1 ± 3.1%, and 367.1 ± 10.2 kcal/100 g respectively. In the 28-day repeated-dose oral toxicity study, only Sprague-Dawley male rats receiving 5 g/kg showed a slight decrease in feed consumption at week 3, but no associated clinical signs of toxicity or significant weight loss were observed. Although a significant reduction of the platelet count was found in mid- and high-dose GKSS treated male groups, such changes were noted to be within the normal range and were not correlated with relative spleen weight changes. Hence, the no observed adverse effect level (NOAEL) of GKSS was identified to be higher than 5 g/kg in rats. After the safety of GKSS is confirmed, the sleep-promoting effect of GKSS ethanolic extract enriched with hispidin was further assessed. Despite 75 mg/kg of GKSS ethanolic extract does not affect wakefulness, rapid eye movement (REM) sleep and non-REM (NREM) sleep, GKSS ethanolic extract at 150 mg/kg significantly decreased wakefulness and enhanced NREM and REM sleep. Interestingly, such effects seem to be mediated through anti-inflammatory activities via NF-E2-related factor-2 (Nrf2) signaling pathway. Taken together, these findings provide the preliminary evidence to studies support the claims suggesting that GKSS contained useful phytochemical hispidin could be considered as and is safe to use as a functional food agent or nutraceutical for relieving sleep problems mediated by Nrf2 pathway, which the results are useful for future clinical pilot study.
Collapse
Affiliation(s)
- I-Chen Li
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan, Taiwan
| | - Fang-Chia Chang
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Ching-Chuan Kuo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Hsin-Tung Chu
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan, Taiwan
| | - Tsung-Ju Li
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan, Taiwan
- *Correspondence: Tsung-Ju Li
| | - Chin-Chu Chen
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan, Taiwan
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan, Taiwan
- Department of Food Science, Nutrition and Nutraceutical Biotechnology, Shih Chien University, Taipei, Taiwan
- Chin-Chu Chen
| |
Collapse
|
88
|
Murillo-Rodríguez E, Carreón C, Acosta-Hernández ME, García-García F. Stimulants and Depressor Drugs in the Sleep-Wake Cycle Modulation: The case of alcohol and cannabinoids. Curr Top Med Chem 2022; 22:1270-1279. [PMID: 34986773 DOI: 10.2174/1568026622666220105105054] [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: 08/10/2021] [Revised: 11/26/2021] [Accepted: 12/05/2021] [Indexed: 11/22/2022]
Abstract
A complex neurobiological network drives the sleep-wake cycle. In addition, external stimuli, including stimulants or depressor drugs, also influence the control of sleep. Here we review the recent advances that contribute to the comprehensive understanding of the actions of stimulants and depressor compounds, such as alcohol and cannabis, in sleep regulation. The objective of this review is to highlight the neurobiological mechanism engaged by alcohol and cannabis in sleep control.
Collapse
Affiliation(s)
- Eric Murillo-Rodríguez
- Laboratorio de Neurociencias Moleculares e Integrativas. Escuela de Medicina, División Ciencias de la Salud Universidad Anáhuac Mayab. Mérida, Yucatán. México
| | - Cristina Carreón
- Laboratorio de Neurociencias Moleculares e Integrativas. Escuela de Medicina, División Ciencias de la Salud Universidad Anáhuac Mayab. Mérida, Yucatán. México
| | | | - Fabio García-García
- Biomedicine Department, Health Science Institute, Veracruzana University. Xalapa, Veracruz. México
| |
Collapse
|
89
|
Jiao J, Tan L, Zhang Y, Li T, Tang X. Repetitive transcranial magnetic stimulation for insomnia in patients with autism spectrum disorder: Study protocol for a randomized, double-blind, and sham-controlled clinical trial. Front Psychiatry 2022; 13:977341. [PMID: 36245883 PMCID: PMC9554245 DOI: 10.3389/fpsyt.2022.977341] [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: 06/24/2022] [Accepted: 09/06/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Insomnia is the most common comorbidity in children with autism spectrum disorder (ASD) and seriously affects their rehabilitation and prognosis. Thus, an intervention targeting insomnia in ASD seems warranted. Repetitive transcranial magnetic stimulation (rTMS), a potentially effective treatment for improving sleep quality and optimizing sleep structure, has already been demonstrated to alleviate insomnia symptoms and sleep disturbance in different neurological and neuropsychiatric conditions. This trial aims to investigate the effects of rTMS on insomnia in patients with ASD. METHOD This study is designed to be a double-blind, randomized, and sham-controlled trial with a target sample size of 30 participants (aged 3-13 years) diagnosed with ASD comorbid with insomnia. The intervention phase will comprise 20 sessions of rTMS or sham rTMS applied over the right dorsolateral prefrontal cortex (DLPFC) within four consecutive weeks. The effect of rTMS on insomnia and other symptoms of ASD will be investigated through home-PSG (two consecutive overnights), sleep diary, CSHQ, CARS, ABC, SRS, RBS-R, and metabolomics analysis at baseline and posttreatment. A follow-up assessment 1 month after the intervention will examine the long-term effects. DISCUSSION The results of this study may address an important knowledge gap and may provide evidence for the use of rTMS to treat insomnia in ASD. Furthermore, it will elucidate the potential mechanism and link between sleep disorders and clinical symptoms. CLINICAL TRIAL REGISTRATION The study is ongoing and has been registered at the Chinese Clinical Trial Registry (ChiCTR2100049266) on 28/07/2021.
Collapse
Affiliation(s)
- Jian Jiao
- Sleep Medicine Center, Mental Health Center, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Tan
- Sleep Medicine Center, Mental Health Center, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ye Zhang
- Sleep Medicine Center, Mental Health Center, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Taomei Li
- Sleep Medicine Center, Mental Health Center, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiangdong Tang
- Sleep Medicine Center, Mental Health Center, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
90
|
Jiang G, Feng Y, Li M, Wen H, Wang T, Shen Y, Chen Z, Li S. Distinct alterations of functional connectivity of the basal forebrain subregions in insomnia disorder. Front Psychiatry 2022; 13:1036997. [PMID: 36311494 PMCID: PMC9606586 DOI: 10.3389/fpsyt.2022.1036997] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 09/22/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Cholinergic basal forebrain (BF) plays an important role in sleep-wake regulation and is implicated in cortical arousal and activation. However, less is known currently regarding the abnormal BF-related neuronal circuit in human patients with insomnia disorder (ID). In this study, we aimed to explore alterations of functional connectivity (FC) in subregions of the BF and the relationships between FC alterations and sleep and mood measures in ID. MATERIALS AND METHODS One hundred and two ID patients and ninety-six healthy controls (HC) were included in this study. Each subject underwent both resting-state fMRI and high-resolution anatomical scanning. All participants completed the sleep and mood questionnaires in ID patients. Voxel-based resting-state FC in each BF subregion (Ch_123 and Ch_4) were computed. For the voxel-wise FC differences between groups, a two-sample t-test was performed on the individual maps in a voxel-by-voxel manner. To examine linear relationships with sleep and mood measures, Pearson correlations were calculated between FC alterations and sleep and mood measures, respectively. RESULTS The ID group showed significantly decreased FC between the medial superior frontal gyrus and Ch_123 compared to HC. However, increased FC between the midbrain and Ch_4 was found in ID based on the voxel-wise analysis. The correlation analysis only revealed that the altered FC between the midbrain with Ch_4 was significantly negatively correlated with the self-rating anxiety scale. CONCLUSION Our findings of decreased FC between Ch_123 and medial superior frontal gyrus and increased FC between midbrain and Ch4 suggest distinct roles of subregions of BF underlying the neurobiology of ID.
Collapse
Affiliation(s)
- Guihua Jiang
- Department of Medical Imaging, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Ying Feng
- Department of Radiology, Affiliated Hospital of Chengdu University, Chengdu, China
| | - Meng Li
- Department of Medical Imaging, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Hua Wen
- Department of Medical Imaging, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Tianyue Wang
- Department of Medical Imaging, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Yanan Shen
- The First School of Clinical Medicine, Guangdong Medical University, Zhanjiang, China
| | - Ziwei Chen
- Department of Medical Imaging, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, China
| | - Shumei Li
- Department of Medical Imaging, Guangdong Second Provincial General Hospital, Guangzhou, China
| |
Collapse
|
91
|
Mutti C, Misirocchi F, Zilioli A, Rausa F, Pizzarotti S, Spallazzi M, Parrino L. Sleep and brain evolution across the human lifespan: A mutual embrace. FRONTIERS IN NETWORK PHYSIOLOGY 2022; 2:938012. [PMID: 36926070 PMCID: PMC10013002 DOI: 10.3389/fnetp.2022.938012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022]
Abstract
Sleep can be considered a window to ascertain brain wellness: it dynamically changes with brain maturation and can even indicate the occurrence of concealed pathological processes. Starting from prenatal life, brain and sleep undergo an impressive developmental journey that accompanies human life throughout all its steps. A complex mutual influence rules this fascinating course and cannot be ignored while analysing its evolution. Basic knowledge on the significance and evolution of brain and sleep ontogenesis can improve the clinical understanding of patient's wellbeing in a more holistic perspective. In this review we summarized the main notions on the intermingled relationship between sleep and brain evolutionary processes across human lifespan, with a focus on sleep microstructure dynamics.
Collapse
Affiliation(s)
- Carlotta Mutti
- Department of General and Specialized Medicine, Parma University Hospital, Parma, Italy
| | - Francesco Misirocchi
- Department of General and Specialized Medicine, Parma University Hospital, Parma, Italy
| | - Alessandro Zilioli
- Department of General and Specialized Medicine, Parma University Hospital, Parma, Italy
| | - Francesco Rausa
- Department of General and Specialized Medicine, Parma University Hospital, Parma, Italy
| | - Silvia Pizzarotti
- Department of General and Specialized Medicine, Parma University Hospital, Parma, Italy
| | - Marco Spallazzi
- Department of General and Specialized Medicine, Parma University Hospital, Parma, Italy
| | - Liborio Parrino
- Department of General and Specialized Medicine, Parma University Hospital, Parma, Italy
| |
Collapse
|
92
|
Yang Y, Chen M, Zhai Z, Dai Y, Gu H, Zhou X, Hong J. Long Non-coding RNAs Gabarapl2 and Chrnb2 Positively Regulate Inflammatory Signaling in a Mouse Model of Dry Eye. Front Med (Lausanne) 2021; 8:808940. [PMID: 34957168 PMCID: PMC8703135 DOI: 10.3389/fmed.2021.808940] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 11/22/2021] [Indexed: 12/26/2022] Open
Abstract
Purpose: To elucidate the expression profile and the potential role of long non-coding ribonucleic acids (RNAs) (lncRNAs) in a dry eye disease (DED) model. Methods: A DED model was established in C57BL/6J mice with 0.2% benzalkonium chloride (BAC) twice a day for 14 days. The differentially expressed lncRNAs were detected by RNA-seq technology (Gene Expression Omnibus, GEO GSE186450) and the aberrantly expressed lncRNAs were further verified by RT-qPCR. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted to predicate the related candidate genes and potential pathological pathways. Cells from a human corneal epithelial cell line (HCECs) were cultured under hyperosmolarity. The regulation of inflammatory factors by silencing potential targeted lncRNAs was verified in vitro in HCECs. Results: In our study, a significant increase in corneal fluorescence staining and a reduction in tear production were observed in DED mice at all follow-ups compared with the controls, and the differences were increasing over time. In total, 2,649 upregulated and 704 downregulated lncRNAs were identified in DED mice. We selected six aberrantly expressed and most abundant lncRNAs and performed RT-qPCR using the samples for RNA-seq. Chrnb2, Gabarapl2, and Usp31 were thereby confirmed as the most significantly altered lncRNAs. Pathway analysis revealed that the neuroactive ligand–receptor interaction signaling pathway was the most enriched, followed by the calcium signaling pathway and cytokine–cytokine receptor interaction. Following treatment of Gabarapl2 siRNA and Chrnb2 siRNA, tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, and IL-6 were significantly downregulated in the HCECs. Conclusion: Our study suggests that Chrnb2 and Gabarapl2 may be involved in the inflammation response by regulating TNF-α, IL-1β, and IL-6 in DED. These candidate lncRNAs may be both potential biomarkers and therapeutic targets for DED.
Collapse
Affiliation(s)
- Yuhan Yang
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China.,Department of Ophthalmology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Minjie Chen
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Zimeng Zhai
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Yiqin Dai
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Hao Gu
- Department of Ophthalmology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xujiao Zhou
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Jiaxu Hong
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, China.,Department of Ophthalmology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
93
|
Ammar N, Attarian H, Wilson A. Daytime sleepiness and circadian rhythm disturbance due to pineal and pontine damage. Sleep Med 2021; 89:90-92. [PMID: 34936934 DOI: 10.1016/j.sleep.2021.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/24/2021] [Accepted: 12/01/2021] [Indexed: 10/19/2022]
Affiliation(s)
- Nady Ammar
- Université Catholique de Louvain, Belgium
| | | | | |
Collapse
|
94
|
Xie J, Wang L, Xiao C, Ying S, Ren J, Chen Z, Yu Y, Xu D, Yao D, Wu B, Liu T. Low Frequency Transcranial Alternating Current Stimulation Accelerates Sleep Onset Process. IEEE Trans Neural Syst Rehabil Eng 2021; 29:2540-2549. [PMID: 34851828 DOI: 10.1109/tnsre.2021.3131728] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
GOAL The aim of this study is to find a kind of low frequency oscillation transcranial alternating current stimulation, which is directly applied to the scalp epidermal, to stimulate the cerebral cortex with a large spatial range of electric field oscillation across the brain hemisphere, and then trigger the start of the Top-Down processing of sleep homeostasis, in the daytime nap. METHODS Thirty healthy subjects, to take naps, underwent an intervention of electrical stimulation at 5 Hz, applied to the dorsal lateral prefrontal cortex. The subjects in the experiments were strictly controlled, and opened their eyes when stimulation was transmitted. Subsequently, after 15 min transcranial alternating current stimulation, subjects entered the experimental procedure of sleep. Electroencephalograph was taken at baseline and during sleep. Behavioral indicators were also added to the experiment. RESULTS We found that the total power of Electroencephalograph activity in the theta band, as well as low-frequency power at 1-7 Hz, was significantly entrained and increased, and that alpha activity was attenuated faster and spindle activity active earlier. Even more, the transition from awake to Non-rapid eye movement stages occurs earlier. Alertness also decreased when the subjects woke up after brief sleep. CONCLUSION The intervention of low frequency brain rhythmic transcranial alternating current stimulation may induce accelerated effect on sleep onset process, thereby possibly alleviating the problems related to sleep disorders such as difficulty to reach the real sleep state quickly after lying down.
Collapse
|
95
|
Yves D, Barateau L, Middleton B, Van Der Veen D, Skene DJ. Metabolomic Signature of Patients With Narcolepsy. Neurology 2021; 98:e493-e505. [PMID: 34845055 DOI: 10.1212/wnl.0000000000013128] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 11/12/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Narcolepsy type 1 (NT1) is an orphan brain disorder caused by the irreversible destruction of orexin neurons. Metabolic disturbances are common in patients with NT1 who have a body mass index (BMI) 10-20% higher than the general population, with one third being obese (BMI>30 kg/m2). Besides the destruction of orexin neurons in NT1, the metabolic alterations in obese and non-obese patients with narcolepsy type 1 remain unknown. The aim of the study was to identify possible differences in plasma metabolic profiles between patients with NT1 and controls as a function of their BMI status. METHODS We used a targeted liquid chromatography-mass spectrometry metabolomics approach to measure 141 circulating, low molecular weight metabolites in drug-free fasted plasma samples from 117 NT1 patients (including 41 obese subjects) compared with 116 BMI-matched controls (including 57 obese subjects). RESULTS Common metabolites driving the difference between NT1 and controls, irrespective of BMI, were identified, namely sarcosine, glutamate, nonaylcarnitine (C9), 5 long chain lysophosphatidylcholine acyls, one sphingolipid, 12 phosphatidylcholine diacyls and 11 phosphatidylcholine acyl-akyls, all showing increased concentrations in NT1. Metabolite concentrations significantly affected by NT1 (n = 42) and BMI category (n = 40) showed little overlap (n = 5). Quantitative enrichment analysis revealed common metabolic pathways that were implicated in the NT1/control differences, in both normal BMI and obese comparisons, namely glycine and serine, arachidonic acid, and tryptophan metabolisms. The metabolites driving these differences were glutamate, sarcosine and ornithine (glycine and serine metabolism), glutamate and PC aa C34:4 (arachidonic acid metabolism) and glutamate, serotonin and tryptophan (tryptophan metabolism). Linear metabolite-endophenotype regression analyses highlight that as part of the NT1 metabolic phenotype, most of the relationships between the sleep parameters (i.e. slow wave sleep duration, sleep latency and periodic leg movement) and metabolite concentrations seen in the controls were lost. DISCUSSION These results represented the most comprehensive metabolic profiling of patients with NT1 as a function of BMI and propose some metabolic diagnostic biomarkers for NT1, namely glutamate, sarcosine, serotonin, tryptophan, nonaylcarnitine and some phosphatidylcholines. The metabolic pathways identified offer, if confirmed, possible targets for treatment of obesity in NT1. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that a distinct metabolic profile can differentiate patients with Narcolepsy Type 1 from patients without the disorder.
Collapse
Affiliation(s)
- Dauvilliers Yves
- National Reference Centre for Orphan Diseases, Narcolepsy - Rare hypersomnias, Sleep Unit, Department of Neurology, CHU Montpellier, Univ Montpellier, Montpellier, France .,Institute for Neurosciences of Montpellier INM, Univ Montpellier, INSERM, Montpellier, France
| | - Lucie Barateau
- National Reference Centre for Orphan Diseases, Narcolepsy - Rare hypersomnias, Sleep Unit, Department of Neurology, CHU Montpellier, Univ Montpellier, Montpellier, France.,Institute for Neurosciences of Montpellier INM, Univ Montpellier, INSERM, Montpellier, France
| | - Benita Middleton
- Chronobiology, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, United Kingdom
| | - Daan Van Der Veen
- Chronobiology, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, United Kingdom
| | - Debra J Skene
- Chronobiology, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, United Kingdom
| |
Collapse
|
96
|
Osorio-Forero A, Cardis R, Vantomme G, Guillaume-Gentil A, Katsioudi G, Devenoges C, Fernandez LMJ, Lüthi A. Noradrenergic circuit control of non-REM sleep substates. Curr Biol 2021; 31:5009-5023.e7. [PMID: 34648731 DOI: 10.1016/j.cub.2021.09.041] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/09/2021] [Accepted: 09/15/2021] [Indexed: 12/13/2022]
Abstract
To understand what makes sleep vulnerable in disease, it is useful to look at how wake-promoting mechanisms affect healthy sleep. Wake-promoting neuronal activity is inhibited during non-rapid-eye-movement sleep (NREMS). However, sensory vigilance persists in NREMS in animals and humans, suggesting that wake promotion could remain functional. Here, we demonstrate that consolidated mouse NREMS is a brain state with recurrent fluctuations of the wake-promoting neurotransmitter noradrenaline on the ∼50-s timescale in the thalamus. These fluctuations occurred around mean noradrenaline levels greater than the ones of quiet wakefulness, while noradrenaline (NA) levels declined steeply in REMS. They coincided with a clustering of sleep spindle rhythms in the forebrain and with heart-rate variations, both of which are correlates of sensory arousability. We addressed the origins of these fluctuations by using closed-loop optogenetic locus coeruleus (LC) activation or inhibition timed to moments of low and high spindle activity during NREMS. We could suppress, lock, or entrain sleep-spindle clustering and heart-rate variations, suggesting that both fore- and hindbrain-projecting LC neurons show coordinated infraslow activity variations in natural NREMS. Noradrenergic modulation of thalamic, but not cortical, circuits was required for sleep-spindle clustering and involved NA release into primary sensory and reticular thalamic nuclei that activated both α1- and β-adrenergic receptors to cause slowly decaying membrane depolarizations. Noradrenergic signaling by LC constitutes a vigilance-promoting mechanism that renders mammalian NREMS vulnerable to disruption on the close-to-minute timescale through sustaining thalamocortical and autonomic sensory arousability. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Alejandro Osorio-Forero
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, Switzerland
| | - Romain Cardis
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, Switzerland
| | - Gil Vantomme
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, Switzerland
| | - Aurélie Guillaume-Gentil
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, Switzerland
| | - Georgia Katsioudi
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, Switzerland
| | - Christiane Devenoges
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, Switzerland
| | - Laura M J Fernandez
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, Switzerland
| | - Anita Lüthi
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, Switzerland.
| |
Collapse
|
97
|
Brain Clocks, Sleep, and Mood. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021. [PMID: 34773227 DOI: 10.1007/978-3-030-81147-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
The suprachiasmatic nucleus houses the master clock, but the genes which encode the circadian clock components are also expressed throughout the brain. Here, we review how circadian clock transcription factors regulate neuromodulator systems such as histamine, dopamine, and orexin that promote arousal. These circadian transcription factors all lead to repression of the histamine, dopamine, and orexin systems during the sleep period, so ensuring integration with the ecology of the animal. If these transcription factors are deleted or mutated, in addition to the global disturbances in circadian rhythms, this causes a chronic up-regulation of neuromodulators leading to hyperactivity, elevated mood, and reduced sleep, which have been suggested to be states resembling mania.
Collapse
|
98
|
Gagnon K, Labrosse M, Gingras MA, Godbout R. Sleep Instability Correlates with Attentional Impairment in Boys with Attention Deficit Hyperactivity Disorder. Brain Sci 2021; 11:1425. [PMID: 34827422 PMCID: PMC8615536 DOI: 10.3390/brainsci11111425] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 10/19/2021] [Accepted: 10/22/2021] [Indexed: 11/16/2022] Open
Abstract
Theoretical models of sleep and attention deficit hyperactivity disorder (ADHD) suggest that symptoms of ADHD are associated with daytime sleepiness, but it has received little support. The present study aimed at testing an alternative model involving the association of attentional instability with sleep instability, i.e., sleep stage transitions and arousals. Twelve ADHD and 15 healthy control (HC) boys aged between 8 and 12 years old underwent polysomnography recording and attentional testing. The microarousal index, the number of awakenings, and the number of stage shifts between stages 1, 2, 3, 4 and REM sleep throughout the night were computed as sleep stability parameters. Attentional functioning was assessed using the Continuous Performance Test-II. We found significantly higher sleep instability in ADHD compared to HC. Sleep arousals and stage transitions (micro arousal index, stage 4/3 and 2/4 transitions) in ADHD significantly correlated with lower attentional scores. No association whatsoever was found between sleep instability and attentional functioning in HC. The results show that sleep instability is associated with lower attentional performance in boys with ADHD, but not in HC. This could be compatible with a model according to which attention and sleep stability share a common neural substrate in ADHD.
Collapse
Affiliation(s)
- Katia Gagnon
- Sleep Laboratory and Clinic, Hôpital en Santé mentale Rivière-des-Prairies, Montréal, QC H1E 1A4, Canada; (K.G.); (M.L.); (M.-A.G.)
- Department of Psychiatry, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Mélanie Labrosse
- Sleep Laboratory and Clinic, Hôpital en Santé mentale Rivière-des-Prairies, Montréal, QC H1E 1A4, Canada; (K.G.); (M.L.); (M.-A.G.)
| | - Marc-André Gingras
- Sleep Laboratory and Clinic, Hôpital en Santé mentale Rivière-des-Prairies, Montréal, QC H1E 1A4, Canada; (K.G.); (M.L.); (M.-A.G.)
| | - Roger Godbout
- Sleep Laboratory and Clinic, Hôpital en Santé mentale Rivière-des-Prairies, Montréal, QC H1E 1A4, Canada; (K.G.); (M.L.); (M.-A.G.)
- Department of Psychiatry, Université de Montréal, Montréal, QC H3T 1J4, Canada
| |
Collapse
|
99
|
Different Peas in the Same Pod: The Histaminergic Neuronal Heterogeneity. Curr Top Behav Neurosci 2021; 59:303-327. [PMID: 34455575 DOI: 10.1007/7854_2021_241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The histaminergic neuronal system is recently receiving increasing attention, as much has been learned over the past 25 years about histamine role as a neurotransmitter. Indeed, this amine is crucial in maintaining arousal and provides important contributions to regulate circadian rhythms, energy, endocrine homeostasis, motor behavior, and cognition. The extent to which these distinct physiological functions are operated by independent histamine neuronal subpopulation is unclear. In the rat brain histamine neuronal cell bodies are grouped within the tuberomamillary nucleus of the posterior hypothalamus in five clusters, E1-E5, each sending overlapping axons throughout the entire central nervous system with no strict topographical pattern. These features lead to the concept that histamine regulation of a wide range of functions in the central nervous system is achieved by the histaminergic neuronal system as a whole. However, increasing experimental evidence suggesting that the histaminergic system is organized into distinct pathways modulated by selective mechanisms challenges this view. In this review, we summarized experimental evidence supporting the heterogeneity of histamine neurons, and their organization in functionally distinct circuits impinging on separate brain regions and displaying selective control mechanisms. This implies independent functions of subsets of histaminergic neurons according to their respective origin and terminal projections with relevant consequences for the development of specific compounds that affect only subsets of histamine neurons, thus increasing the target specificity.
Collapse
|
100
|
Deurveilher S, Antonchuk M, Saumure BSC, Baldin A, Semba K. No loss of orexin/hypocretin, melanin-concentrating hormone or locus coeruleus noradrenergic neurons in a rat model of chronic sleep restriction. Eur J Neurosci 2021; 54:6027-6043. [PMID: 34355453 DOI: 10.1111/ejn.15412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/13/2021] [Accepted: 07/29/2021] [Indexed: 12/31/2022]
Abstract
Chronic sleep restriction (CSR) is common in modern society, adversely affecting cognitive performance and health. Yet how it impacts neurons regulating sleep remains unclear. Several studies using mice reported substantial losses of wake-active orexin/hypocretin and locus coeruleus (LC) noradrenergic neurons, but not rapid eye movement sleep-active melanin-concentrating hormone (MCH) neurons, following CSR. Here, we used immunohistochemistry and stereology to examine orexin, MCH and LC noradrenergic neurons in a rat model of CSR that uses programmed wheel rotation (3 h on/1 h off; '3/1' protocol). Adult male Wistar rats underwent one or four cycles of the 4-day 3/1 CSR protocol, with 2-day recovery between cycles in home cages. Time-matched control rats were housed in locked wheels/home cages. We found no significant differences in the numbers of orexin, MCH and LC noradrenergic neurons following either one- or four-cycle CSR protocol compared to respective controls. Similarly, the four-cycle CSR protocol had no effect on the densities of orexin axon terminals in the LC, noradrenergic dendrites in the LC and noradrenergic axon terminals in the frontal cortex. Body weights, however, decreased after one cycle of CSR and then increased with diminishing slope over the next three cycles. Thus, we found no evidence for loss of orexin or LC noradrenergic neurons following one and four cycles of the 4-day 3/1 CSR protocol in rats. Differences in CSR protocols and/or possible species differences in neuronal vulnerability to sleep loss may account for the discrepancy between the current results in rats and previous findings in mice.
Collapse
Affiliation(s)
- Samuel Deurveilher
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Michael Antonchuk
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Brock St C Saumure
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Andrew Baldin
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kazue Semba
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|