1
|
Ralston M, Ehlen JC, Paul K. Reproductive hormones and sex chromosomes drive sex differences in the sleep-wake cycle. Front Neurosci 2024; 18:1478820. [PMID: 39544910 PMCID: PMC11561190 DOI: 10.3389/fnins.2024.1478820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 10/11/2024] [Indexed: 11/17/2024] Open
Abstract
There are well-documented gender differences in the risk and severity of sleep disorders and associated comorbidities. While fundamental sex differences in sleep regulatory mechanisms may contribute to gender disparities, biological responses to sleep loss and stress may underlie many of the risks for sleep disorders in women and men. Some of these sex differences appear to be dependent on sex chromosome complement (XX or XY) and the organizational effects of reproductive hormones. Reproductive development plays a critical role in the ability of sex chromosomes and reproductive hormones to produce sex differences in sleep and wakefulness. Rodent models reveal that reproductive hormones drive many but not all sex differences in sleep-wake architecture. The ability of reproductive hormones to alter sleep are often dependent on responses to sleep loss and stress. However, in the absence of reproductive hormones (in gonadectomized rodents) sex differences in sleep amount and the ability to recover from sleep loss persist. The suprachiasmatic nucleus (SCN) and the ventrolateral preoptic nucleus (VLPO) of the hypothalamus play crucial regulatory roles in mediating the effects of reproductive hormones on the sleep-wake cycle. Taken together, the work reviewed here reveals that the reproductive hormone environment and sex chromosome complement may underlie gender disparities in sleep patterns and the risk for sleep disorders.
Collapse
Affiliation(s)
- Micah Ralston
- Department of Integrative Biology and Physiology, College of Life Sciences, University of California, Los Angeles, Los Angeles, CA, United States
| | - J. Christopher Ehlen
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Ketema Paul
- Department of Integrative Biology and Physiology, College of Life Sciences, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
2
|
Deng Q, Li Y, Sun Z, Gao X, Zhou J, Ma G, Qu WM, Li R. Sleep disturbance in rodent models and its sex-specific implications. Neurosci Biobehav Rev 2024; 164:105810. [PMID: 39009293 DOI: 10.1016/j.neubiorev.2024.105810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/17/2024]
Abstract
Sleep disturbances, encompassing altered sleep physiology or disorders like insomnia and sleep apnea, profoundly impact physiological functions and elevate disease risk. Despite extensive research, the underlying mechanisms and sex-specific differences in sleep disorders remain elusive. While polysomnography serves as a cornerstone for human sleep studies, animal models provide invaluable insights into sleep mechanisms. However, the availability of animal models of sleep disorders is limited, with each model often representing a specific sleep issue or mechanism. Therefore, selecting appropriate animal models for sleep research is critical. Given the significant sex differences in sleep patterns and disorders, incorporating both male and female subjects in studies is essential for uncovering sex-specific mechanisms with clinical relevance. This review provides a comprehensive overview of various rodent models of sleep disturbance, including sleep deprivation, sleep fragmentation, and circadian rhythm dysfunction. We evaluate the advantages and disadvantages of each model and discuss sex differences in sleep and sleep disorders, along with potential mechanisms. We aim to advance our understanding of sleep disorders and facilitate sex-specific interventions.
Collapse
Affiliation(s)
- Qi Deng
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Yuhong Li
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Zuoli Sun
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Xiang Gao
- Shanxi Bethune Hospital, Shanxi, China
| | | | - Guangwei Ma
- Peking University Sixth Hospital, Beijing, China
| | - Wei-Min Qu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China; Department of Pharmacology, School of Basic Medical Sciences, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Rena Li
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
3
|
Gao Z, Guan J, Yin S, Liu F. The role of ATP in sleep-wake regulation: In adenosine-dependent and -independent manner. Sleep Med 2024; 119:147-154. [PMID: 38678758 DOI: 10.1016/j.sleep.2024.04.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 03/31/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
ATP plays a crucial role as an energy currency in the body's various physiological functions, including the regulation of the sleep-wake cycle. Evidence from genetics and pharmacology demonstrates a strong association between ATP metabolism and sleep. With the advent of new technologies such as optogenetics, genetically encoded biosensors, and novel ATP detection methods, the dynamic changes in ATP levels between different sleep states have been further uncovered. The classic mechanism for regulating sleep by ATP involves its conversion to adenosine, which increases sleep pressure when accumulated extracellularly. However, emerging evidence suggests that ATP can directly bind to P2 receptors and influence sleep-wake regulation through both adenosine-dependent and independent pathways. The outcome depends on the brain region where ATP acts and the expression type of P2 receptors. This review summarizes the experimental evidence on the relationship between ATP levels and changes in sleep states and outlines the mechanisms by which ATP is involved in regulating the sleep-wake cycle through both adenosine-dependent and independent pathways. Hopefully, this review will provide a comprehensive understanding of the current research basis and progress in this field and promote further investigations into the specific mechanisms of ATP in regulating sleep.
Collapse
Affiliation(s)
- Zhenfei Gao
- Shanghai Key Laboratory of Sleep Disordered Breathing, Department of Otolaryngology-Head and Neck Surgery, Otolaryngology Institute of Shanghai Jiaotong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jian Guan
- Shanghai Key Laboratory of Sleep Disordered Breathing, Department of Otolaryngology-Head and Neck Surgery, Otolaryngology Institute of Shanghai Jiaotong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Shankai Yin
- Shanghai Key Laboratory of Sleep Disordered Breathing, Department of Otolaryngology-Head and Neck Surgery, Otolaryngology Institute of Shanghai Jiaotong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Feng Liu
- Shanghai Key Laboratory of Sleep Disordered Breathing, Department of Otolaryngology-Head and Neck Surgery, Otolaryngology Institute of Shanghai Jiaotong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
4
|
Tiwari J, Sur S, Yadav A, Kumar R, Rai N, Rani S, Malik S. Photoperiod-driven concurrent changes in hypothalamic and brainstem transcription of sleep and immune genes in migratory redheaded bunting. Proc Biol Sci 2023; 290:20222374. [PMID: 36750197 PMCID: PMC9904947 DOI: 10.1098/rspb.2022.2374] [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: 11/25/2022] [Accepted: 01/16/2023] [Indexed: 02/09/2023] Open
Abstract
The molecular regulation of sleep in avian migrants is still obscure. We thus investigated this in migratory redheaded buntings, where four life-history states (LHS; i.e. non-migratory, pre-migratory, migratory and refractory states) were induced. There was increased night-time activity (i.e. Zugunruhe) during the migratory state with reduced daytime activity. The recordings of the sleep-wake cycle in buntings showed increased night-time active wakefulness coupled with drastically reduced front and back sleep during migratory phase. Interestingly, we found the buntings to feed and drink even after lights-off during migration. Gene expression studies revealed increased hypothalamic expression of glucocorticoid receptor (nr3c1), and pro-inflammatory cytokines (il1b and il6) in pre-migratory and migratory states, respectively, whereas in brainstem Ca2+/calmodulin-dependent protein kinase 2 (camk2) was upregulated during the migratory state. This suggested a heightened pro-inflammatory state during migration which is a feature of chronic sleep loss, and a possible role of Ca2+ signalling in promoting wakefulness. In both the hypothalamus and brainstem, the expression of melatonin receptors (mel1a and mel1b) was increased in the pre-migratory state, and growth hormone-releasing hormone (ghrh, known to induce sleep) was reduced during the migratory state. The current results demonstrate key molecules involved in the regulation of sleep-wake cycle across LHS in migratory songbirds.
Collapse
Affiliation(s)
- Jyoti Tiwari
- Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh 226007, India
| | - Sayantan Sur
- Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh 226007, India
| | - Anupama Yadav
- Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh 226007, India
| | - Raj Kumar
- Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh 226007, India
| | - Niraj Rai
- Ancient DNA Lab, Birbal Sahni Institute of Palaeosciences, Lucknow, Uttar Pradesh 226007, India
| | - Sangeeta Rani
- Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh 226007, India
| | - Shalie Malik
- Department of Zoology, University of Lucknow, Lucknow, Uttar Pradesh 226007, India
| |
Collapse
|
5
|
Fernandes GL, Araujo P, Tufik S, Andersen ML. SLEEPINESS PROFILES IN MICE SUBMITTED TO ACUTE AND CHRONIC SLEEP DEPRIVATION. Behav Processes 2022; 200:104661. [PMID: 35618241 DOI: 10.1016/j.beproc.2022.104661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/09/2022] [Accepted: 05/16/2022] [Indexed: 11/29/2022]
Abstract
Sleepiness is a behavioural consequence of sleep pressure, which shows interindividual variation, a characteristic possibly related to central sleep mechanisms. However, there is a lack of evidence linking progressive sleep need and sleepiness with factors of individual variability, which could be tested by total acute and chronic sleep deprivation. Thus, the objective of the study was to investigate the development of sleepiness in sleep deprived mice. Male C57BL/6J mice were distributed in sleep deprivation, sleep rebound and control groups. Animals underwent acute sleep deprivation for 3, 6, 9 or 12hours or chronic sleep deprivation for 6hours for 5 consecutive days. Sleep rebound groups had a sleep opportunity for 1, 2, 3, or 4hours after acute sleep deprivation or 24hours after chronic sleep deprivation. During the protocols, sleep attempts were counted to calculate a sleepiness index. After euthanasia, blood was collected for corticosterone assessment. Using the average of group sleep attempts, it was possible to differentiate between sleepy (mean>group average) and resistant animals (mean<group average). Resistant mice were more frequent in all protocols. Individual variation accounted for 52% of sleepiness variance during chronic sleep deprivation and extended wakefulness explained 68% of sleepiness variance during acute sleep deprivation. A normal corticosterone peak was observed at the start of the dark phase, independent of sleep deprivation. Different profiles of sleepiness emerged in sleep deprived mice. Sleep deprivation was the main factor for sleepiness during acute sleep deprivation whereas in chronic deprivation individual variation was more relevant.
Collapse
Affiliation(s)
| | - Paula Araujo
- Departamento de Psicobiologia - Universidade Federal de São Paulo - São Paulo, Brazil; Departamento de Ciências Fisiológicas - Escola de Ciências Médicas, Santa Casa de São Paulo - São Paulo, Brazil
| | - Sergio Tufik
- Departamento de Psicobiologia - Universidade Federal de São Paulo - São Paulo, Brazil
| | - Monica Levy Andersen
- Departamento de Psicobiologia - Universidade Federal de São Paulo - São Paulo, Brazil.
| |
Collapse
|
6
|
Translational Approaches to Influence Sleep and Arousal. Brain Res Bull 2022; 185:140-161. [PMID: 35550156 PMCID: PMC9554922 DOI: 10.1016/j.brainresbull.2022.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 04/27/2022] [Accepted: 05/03/2022] [Indexed: 12/16/2022]
Abstract
Sleep disorders are widespread in society and are prevalent in military personnel and in Veterans. Disturbances of sleep and arousal mechanisms are common in neuropsychiatric disorders such as schizophrenia, post-traumatic stress disorder, anxiety and affective disorders, traumatic brain injury, dementia, and substance use disorders. Sleep disturbances exacerbate suicidal ideation, a major concern for Veterans and in the general population. These disturbances impair quality of life, affect interpersonal relationships, reduce work productivity, exacerbate clinical features of other disorders, and impair recovery. Thus, approaches to improve sleep and modulate arousal are needed. Basic science research on the brain circuitry controlling sleep and arousal led to the recent approval of new drugs targeting the orexin/hypocretin and histamine systems, complementing existing drugs which affect GABAA receptors and monoaminergic systems. Non-invasive brain stimulation techniques to modulate sleep and arousal are safe and show potential but require further development to be widely applicable. Invasive viral vector and deep brain stimulation approaches are also in their infancy but may be used to modulate sleep and arousal in severe neurological and psychiatric conditions. Behavioral, pharmacological, non-invasive brain stimulation and cell-specific invasive approaches covered here suggest the potential to selectively influence arousal, sleep initiation, sleep maintenance or sleep-stage specific phenomena such as sleep spindles or slow wave activity. These manipulations can positively impact the treatment of a wide range of neurological and psychiatric disorders by promoting the restorative effects of sleep on memory consolidation, clearance of toxic metabolites, metabolism, and immune function and by decreasing hyperarousal.
Collapse
|
7
|
Zielinski MR, Gibbons AJ. Neuroinflammation, Sleep, and Circadian Rhythms. Front Cell Infect Microbiol 2022; 12:853096. [PMID: 35392608 PMCID: PMC8981587 DOI: 10.3389/fcimb.2022.853096] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/24/2022] [Indexed: 12/14/2022] Open
Abstract
Molecules involved in innate immunity affect sleep and circadian oscillators and vice versa. Sleep-inducing inflammatory molecules are activated by increased waking activity and pathogens. Pathologies that alter inflammatory molecules, such as traumatic brain injury, cancer, cardiovascular disease, and stroke often are associated with disturbed sleep and electroencephalogram power spectra. Moreover, sleep disorders, such as insomnia and sleep disordered breathing, are associated with increased dysregulation of inflammatory processes. Inflammatory molecules in both the central nervous system and periphery can alter sleep. Inflammation can also modulate cerebral vascular hemodynamics which is associated with alterations in electroencephalogram power spectra. However, further research is needed to determine the interactions of sleep regulatory inflammatory molecules and circadian clocks. The purpose of this review is to: 1) describe the role of the inflammatory cytokines interleukin-1 beta and tumor necrosis factor-alpha and nucleotide-binding domain and leucine-rich repeat protein-3 inflammasomes in sleep regulation, 2) to discuss the relationship between the vagus nerve in translating inflammatory signals between the periphery and central nervous system to alter sleep, and 3) to present information about the relationship between cerebral vascular hemodynamics and the electroencephalogram during sleep.
Collapse
Affiliation(s)
- Mark R. Zielinski
- Veterans Affairs (VA) Boston Healthcare System, West Roxbury, MA, United States,Harvard Medical School, West Roxbury, MA, United States,*Correspondence: Mark R. Zielinski,
| | - Allison J. Gibbons
- Veterans Affairs (VA) Boston Healthcare System, West Roxbury, MA, United States
| |
Collapse
|
8
|
Cellular Effects of Rhynchophylline and Relevance to Sleep Regulation. Clocks Sleep 2021; 3:312-341. [PMID: 34207633 PMCID: PMC8293156 DOI: 10.3390/clockssleep3020020] [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: 04/02/2021] [Revised: 05/25/2021] [Accepted: 06/03/2021] [Indexed: 01/06/2023] Open
Abstract
Uncaria rhynchophylla is a plant highly used in the traditional Chinese and Japanese medicines. It has numerous health benefits, which are often attributed to its alkaloid components. Recent studies in humans show that drugs containing Uncaria ameliorate sleep quality and increase sleep time, both in physiological and pathological conditions. Rhynchophylline (Rhy) is one of the principal alkaloids in Uncaria species. Although treatment with Rhy alone has not been tested in humans, observations in rodents show that Rhy increases sleep time. However, the mechanisms by which Rhy could modulate sleep have not been comprehensively described. In this review, we are highlighting cellular pathways that are shown to be targeted by Rhy and which are also known for their implications in the regulation of wakefulness and sleep. We conclude that Rhy can impact sleep through mechanisms involving ion channels, N-methyl-d-aspartate (NMDA) receptors, tyrosine kinase receptors, extracellular signal-regulated kinases (ERK)/mitogen-activated protein kinases (MAPK), phosphoinositide 3-kinase (PI3K)/RAC serine/threonine-protein kinase (AKT), and nuclear factor-kappa B (NF-κB) pathways. In modulating multiple cellular responses, Rhy impacts neuronal communication in a way that could have substantial effects on sleep phenotypes. Thus, understanding the mechanisms of action of Rhy will have implications for sleep pharmacology.
Collapse
|
9
|
Mamelak M. Sleep, Narcolepsy, and Sodium Oxybate. Curr Neuropharmacol 2021; 20:272-291. [PMID: 33827411 PMCID: PMC9413790 DOI: 10.2174/1570159x19666210407151227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 11/23/2022] Open
Abstract
Sodium oxybate (SO) has been in use for many decades to treat narcolepsy with cataplexy. It functions as a weak GABAB agonist but also as an energy source for the brain as a result of its metabolism to succinate and as a powerful antioxidant because of its capacity to induce the formation of NADPH. Its actions at thalamic GABAB receptors can induce slow-wave activity, while its actions at GABAB receptors on monoaminergic neurons can induce or delay REM sleep. By altering the balance between monoaminergic and cholinergic neuronal activity, SO uniquely can induce and prevent cataplexy. The formation of NADPH may enhance sleep’s restorative process by accelerating the removal of the reactive oxygen species (ROS), which accumulate during wakefulness. SO improves alertness in normal subjects and in patients with narcolepsy. SO may allay severe psychological stress - an inflammatory state triggered by increased levels of ROS and characterized by cholinergic supersensitivity and monoaminergic deficiency. SO may be able to eliminate the inflammatory state and correct the cholinergic/ monoaminergic imbalance.
Collapse
Affiliation(s)
- Mortimer Mamelak
- Department of Psychiatry, Baycrest Hospital, University of Toronto, Toronto, Ontario. Canada
| |
Collapse
|
10
|
Murakawa-Hirachi T, Mizoguchi Y, Ohgidani M, Haraguchi Y, Monji A. Effect of memantine, an anti-Alzheimer's drug, on rodent microglial cells in vitro. Sci Rep 2021; 11:6151. [PMID: 33731780 PMCID: PMC7969939 DOI: 10.1038/s41598-021-85625-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 02/24/2021] [Indexed: 01/07/2023] Open
Abstract
The pathophysiology of Alzheimer's disease (AD) is related to neuroinflammatory responses mediated by microglia. Memantine, an antagonist of N-methyl-D-aspartate (NMDA) receptors used as an anti-Alzheimer's drug, protects from neuronal death accompanied by suppression of proliferation and activation of microglial cells in animal models of AD. However, it remains to be tested whether memantine can directly affect microglial cell function. In this study, we examined whether pretreatment with memantine affects intracellular NO and Ca2+ mobilization using DAF-2 and Fura-2 imaging, respectively, and tested the effects of memantine on phagocytic activity by human β-Amyloid (1-42) phagocytosis assay in rodent microglial cells. Pretreatment with memantine did not affect production of NO or intracellular Ca2+ elevation induced by TNF in rodent microglial cells. Pretreatment with memantine also did not affect the mRNA expression of pro-inflammatory (TNF, IL-1β, IL-6 and CD45) or anti-inflammatory (IL-10, TGF-β and arginase) phenotypes in rodent microglial cells. In addition, pretreatment with memantine did not affect the amount of human β-Amyloid (1-42) phagocytosed by rodent microglial cells. Moreover, we observed that pretreatment with memantine did not affect 11 major proteins, which mainly function in the phagocytosis and degradation of β-Amyloid (1-42), including TREM2, DAP12 and neprilysin in rodent microglial cells. To the best of our knowledge, this is the first report to suggest that memantine does not directly modulate intracellular NO and Ca2+ mobilization or phagocytic activity in rodent microglial cells. Considering the neuroinflammation hypothesis of AD, the results might be important to understand the effect of memantine in the brain.
Collapse
Affiliation(s)
- Toru Murakawa-Hirachi
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Yoshito Mizoguchi
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan.
| | - Masahiro Ohgidani
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
- Department of Integrative Anatomy, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshinori Haraguchi
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Akira Monji
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| |
Collapse
|
11
|
Sleep loss mediates the effect of stress on nitrergic signaling in female mice. Neurosci Lett 2020; 740:135362. [PMID: 33166635 PMCID: PMC10084941 DOI: 10.1016/j.neulet.2020.135362] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 08/12/2020] [Accepted: 09/03/2020] [Indexed: 01/04/2023]
Abstract
Nitric oxide (NO) has been implicated as an important neurotransmitter in stress responses and sleep regulatory processes. However, the role of NO in the relationship between stress and sleep remains unclear. The medial septum (MS) and vertical diagonal band (VDB), regions of the basal forebrain involved in sleep regulation, contain nitric oxide synthase (NOS) producing neurons. Additionally, NOS neurons in the dorsal raphe nucleus (DRN) encode information about stress duration. The role of nitrergic neurons in these regions in subserving sex-specific responses to stress and sleep loss has yet to be elucidated. In this study, NADPH-d, an index of NOS activity, was used to examine the effects of acute restraint stress and sleep loss on NOS activity in the MS, VDB, and DRN. We show that NOS activity in response to restraint stress, total sleep deprivation (TSD), and partial sleep restriction (PSR) differs based on sex and region. Initial analysis showed no effect of restraint stress or TSD on NOS activity in the basal forebrain. However, investigation of each sex separately revealed that restraint stress and TSD significantly decrease NOS activity in the MS of females, but not males. Interestingly, the difference in NOS activity between restraint stress and TSD in females was not significant. Furthermore, PSR was not sufficient to affect NOS activity in males or females. These data suggest that restraint stress and sleep loss regulate NOS activation in a sex-dependent manner, and that the NOS stress response in females may be mediated by sleep loss.
Collapse
|
12
|
Nollet M, Wisden W, Franks NP. Sleep deprivation and stress: a reciprocal relationship. Interface Focus 2020; 10:20190092. [PMID: 32382403 PMCID: PMC7202382 DOI: 10.1098/rsfs.2019.0092] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2020] [Indexed: 12/19/2022] Open
Abstract
Sleep is highly conserved across evolution, suggesting vital biological functions that are yet to be fully understood. Animals and humans experiencing partial sleep restriction usually exhibit detrimental physiological responses, while total and prolonged sleep loss could lead to death. The perturbation of sleep homeostasis is usually accompanied by an increase in hypothalamic–pituitary–adrenal (HPA) axis activity, leading to a rise in circulating levels of stress hormones (e.g. cortisol in humans, corticosterone in rodents). Such hormones follow a circadian release pattern under undisturbed conditions and participate in the regulation of sleep. The investigation of the consequences of sleep deprivation, from molecular changes to behavioural alterations, has been used to study the fundamental functions of sleep. However, the reciprocal relationship between sleep and the activity of the HPA axis is problematic when investigating sleep using traditional sleep-deprivation protocols that can induce stress per se. This is especially true in studies using rodents in which sleep deprivation is achieved by exogenous, and potentially stressful, sensory–motor stimulations that can undoubtedly confuse their conclusions. While more research is needed to explore the mechanisms underlying sleep loss and health, avoiding stress as a confounding factor in sleep-deprivation studies is therefore crucial. This review examines the evidence of the intricate links between sleep and stress in the context of experimental sleep deprivation, and proposes a more sophisticated research framework for sleep-deprivation procedures that could benefit from recent progress in biotechnological tools for precise neuromodulation, such as chemogenetics and optogenetics, as well as improved automated real-time sleep-scoring algorithms.
Collapse
Affiliation(s)
- Mathieu Nollet
- Department of Life Sciences, Imperial College London, London, UK.,UK Dementia Research Institute at Imperial College London, London, UK
| | - William Wisden
- Department of Life Sciences, Imperial College London, London, UK.,UK Dementia Research Institute at Imperial College London, London, UK.,Centre for Neurotechnology, Imperial College London, London, UK
| | - Nicholas P Franks
- Department of Life Sciences, Imperial College London, London, UK.,UK Dementia Research Institute at Imperial College London, London, UK.,Centre for Neurotechnology, Imperial College London, London, UK
| |
Collapse
|
13
|
Wang Z, Xie H, Gong Y, Ouyang Y, Deng F, Tang Y, Li J. Relationship between rosacea and sleep. J Dermatol 2020; 47:592-600. [PMID: 32291809 DOI: 10.1111/1346-8138.15339] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 03/09/2020] [Indexed: 01/18/2023]
Abstract
Rosacea is a chronic facial skin disease involved in neurovascular dysregulation and neurogenic inflammation. Behavioral factors such as stress, anxiety, depression and sleep were identified to be associated with other inflammatory skin diseases. Few studies have reported sleep status in rosacea. Aiming to investigate the relationship between rosacea and sleep, a case-control survey was conducted, enrolling 608 rosacea patients and 608 sex- and age-matched healthy controls. Sleep quality was assessed through the Pittsburgh Sleep Quality Index (PSQI) questionnaire. Diagnosis and severity grading of rosacea were evaluated under the standard guidelines of the National Rosacea Society. More rosacea patients (52.3%, n = 318) suffered poor sleep quality (PSQI, >5) than the healthy controls (24.0%, n = 146), displaying a much higher PSQI score (rosacea vs control, 6.20 vs 3.95). There was a strong association between sleep quality and rosacea (odds ratio [OR], 3.525; 95% confidence interval [CI], 2.759-4.519). Moreover, the severity of rosacea was also associated with sleep quality (OR, 1.847; 95% CI, 1.332-2.570). Single nucleotide polymorphisms in hydroxytryptamine receptor 2A and adrenoceptor-β1 genes, which are associated with sleep behaviour, were detected and revealed to be associated with rosacea. Furthermore, the LL-37-induced rosacea-like phenotype and sleep-deprivation mice models were applied, revealing that sleep deprivation aggravated the rosacea-like phenotype in mice, with higher expression of matrix metallopeptidase 9, Toll-like receptor 2, cathelicidin antimicrobial peptide and vascular endothelial growth factor. In conclusion, rosacea patients presented poorer sleep quality, as well as a higher propability of genetic background with sleep disturbance. In addition, poor sleep might aggravate rosacea through regulating inflammatory factors, contributing to a vicious cycle in the progression of disease.
Collapse
Affiliation(s)
- Zhiqiang Wang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Hongfu Xie
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yihuan Gong
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Yuyan Ouyang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Faming Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Yan Tang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China.,Department of Dermatology, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
14
|
Sleep deficiency and chronic pain: potential underlying mechanisms and clinical implications. Neuropsychopharmacology 2020; 45:205-216. [PMID: 31207606 PMCID: PMC6879497 DOI: 10.1038/s41386-019-0439-z] [Citation(s) in RCA: 233] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/10/2019] [Accepted: 06/05/2019] [Indexed: 11/09/2022]
Abstract
Pain can be both a cause and a consequence of sleep deficiency. This bidirectional relationship between sleep and pain has important implications for clinical management of patients, but also for chronic pain prevention and public health more broadly. The review that follows will provide an overview of the neurobiological evidence of mechanisms thought to be involved in the modulation of pain by sleep deficiency, including the opioid, monoaminergic, orexinergic, immune, melatonin, and endocannabinoid systems; the hypothalamus-pituitary-adrenal axis; and adenosine and nitric oxide signaling. In addition, it will provide a broad overview of pharmacological and non-pharmacological approaches for the management of chronic pain comorbid with sleep disturbances and for the management of postoperative pain, as well as discuss the effects of sleep-disturbing medications on pain amplification.
Collapse
|
15
|
Abstract
Sleep is a universal phenomenon occurring in all species studied thus far. Sleep loss results in adverse physiological effects at both the organismal and cellular levels suggesting an adaptive role for sleep in the maintenance of overall health. This review examines the bidirectional relationship between sleep and cellular stress. Cellular stress in this review refers to a shift in cellular homeostasis in response to an external stressor. Studies that illustrate the fact that sleep loss induces cellular stress and those that provide evidence that cellular stress in turn promotes sleep will be discussed.
Collapse
Affiliation(s)
- Julie A Williams
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Nirinjini Naidoo
- Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Division of Sleep Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
16
|
The Neuropeptide Galanin Is Required for Homeostatic Rebound Sleep following Increased Neuronal Activity. Neuron 2019; 104:370-384.e5. [PMID: 31537465 DOI: 10.1016/j.neuron.2019.08.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 06/04/2019] [Accepted: 08/03/2019] [Indexed: 01/19/2023]
Abstract
Sleep pressure increases during wake and dissipates during sleep, but the molecules and neurons that measure homeostatic sleep pressure remain poorly understood. We present a pharmacological assay in larval zebrafish that generates short-term increases in wakefulness followed by sustained rebound sleep after washout. The intensity of global neuronal activity during drug-induced wakefulness predicted the amount of subsequent rebound sleep. Whole-brain mapping with the neuronal activity marker phosphorylated extracellular signal-regulated kinase (pERK) identified preoptic Galanin (Galn)-expressing neurons as selectively active during rebound sleep, and the relative induction of galn transcripts was predictive of total rebound sleep time. Galn is required for sleep homeostasis, as galn mutants almost completely lacked rebound sleep following both pharmacologically induced neuronal activity and physical sleep deprivation. These results suggest that Galn plays a key role in responding to sleep pressure signals derived from neuronal activity and functions as an output arm of the vertebrate sleep homeostat.
Collapse
|
17
|
Acute and Chronic Sleep Deprivation-Related Changes in N-methyl-D-aspartate Receptor-Nitric Oxide Signalling in the Rat Cerebral Cortex with Reference to Aging and Brain Lateralization. Int J Mol Sci 2019; 20:ijms20133273. [PMID: 31277281 PMCID: PMC6651230 DOI: 10.3390/ijms20133273] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 06/13/2019] [Accepted: 06/27/2019] [Indexed: 12/12/2022] Open
Abstract
Aging and chronic sleep deprivation (SD) are well-recognized risk factors for Alzheimer’s disease (AD), with N-methyl-D-aspartate receptor (NMDA) and downstream nitric oxide (NO) signalling implicated in the process. Herein, we investigate the impact of the age- and acute or chronic SD-dependent changes on the expression of NMDA receptor subunits (NR1, NR2A, and NR2B) and on the activities of NO synthase (NOS) isoforms in the cortex of Wistar rats, with reference to cerebral lateralization. In young adult controls, somewhat lateralized seasonal variations in neuronal and endothelial NOS have been observed. In aged rats, overall decreases in NR1, NR2A, and NR2B expression and reduction in neuronal and endothelial NOS activities were found. The age-dependent changes in NR1 and NR2B significantly correlated with neuronal NOS in both hemispheres. Changes evoked by chronic SD (dysfunction of endothelial NOS and the increasing role of NR2A) differed from those evoked by acute SD (increase in inducible NOS in the right side). Collectively, these results demonstrate age-dependent regulation of the level of NMDA receptor subunits and downstream NOS isoforms throughout the rat brain, which could be partly mimicked by SD. As described herein, age and SD alterations in the prevalence of NMDA receptors and NOS could contribute towards cognitive decline in the elderly, as well as in the pathobiology of AD and the neurodegenerative process.
Collapse
|
18
|
Estrada C, Cuenca L, Cano-Fernandez L, Gil-Martinez AL, Sanchez-Rodrigo C, González-Cuello AM, Fernandez-Villalba E, Herrero MT. Voluntary exercise reduces plasma cortisol levels and improves transitory memory impairment in young and aged Octodon degus. Behav Brain Res 2019; 373:112066. [PMID: 31269420 DOI: 10.1016/j.bbr.2019.112066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 06/28/2019] [Accepted: 06/28/2019] [Indexed: 02/04/2023]
Abstract
Sleep deprivation (SD) has been reported to induce transient cognitive impairment in functional domains commonly affected in dementia, including memory. Indeed, sleep disturbance has been proposed as an early marker for Alzheimer's disease (AD). SD emulates many aging-related modifications, including important memory dysfunctions possibly caused by triggers of stress such as cortisol. Although exercise is widely assumed to be beneficial for overall health, only recently has the research community focused its attention on its possible effects on brain functions such as cognition. Octodon degus (O. degus) is a recent rodent model considered suitable for the study of neurodegenerative diseases, since it spontaneously develops several histopathological hallmarks observed in AD. We aimed to uncover the interaction between stress, exercise, age and transient memory impairments after SD insult. In this study, animals had free individual access to wheels to practice voluntary exercise. The Barnes Maze (BM) task was conducted with young and aged O. degus animals after combining voluntary exercise and either normal sleep or SD. Plasma cortisol levels were measured after each condition. SD impaired hippocampus-dependent memory in both young and old animals, while cortisol levels did not significantly differ between non-SD and SD animals. However, voluntary exercise for 45 days improved the cognitive impairment caused by SD compared with the control condition. Moreover, voluntary exercise decreased plasma cortisol levels in both conditions, independently of the age.
Collapse
Affiliation(s)
- Cristina Estrada
- Clinical & Experimental Neuroscience, Department of Human Anatomy & Psychobiology, Institute for Biomedical Research of Murcia (IMIB), School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, 30100, Spain; Institute for Aging Research, School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, 30100, Spain
| | - Lorena Cuenca
- Clinical & Experimental Neuroscience, Department of Human Anatomy & Psychobiology, Institute for Biomedical Research of Murcia (IMIB), School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, 30100, Spain; Institute for Aging Research, School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, 30100, Spain
| | - Lorena Cano-Fernandez
- Clinical & Experimental Neuroscience, Department of Human Anatomy & Psychobiology, Institute for Biomedical Research of Murcia (IMIB), School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, 30100, Spain; Institute for Aging Research, School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, 30100, Spain
| | - Ana Luisa Gil-Martinez
- Clinical & Experimental Neuroscience, Department of Human Anatomy & Psychobiology, Institute for Biomedical Research of Murcia (IMIB), School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, 30100, Spain; Institute for Aging Research, School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, 30100, Spain
| | - Consuelo Sanchez-Rodrigo
- Clinical & Experimental Neuroscience, Department of Human Anatomy & Psychobiology, Institute for Biomedical Research of Murcia (IMIB), School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, 30100, Spain; Institute for Aging Research, School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, 30100, Spain
| | - Ana Maria González-Cuello
- Clinical & Experimental Neuroscience, Department of Human Anatomy & Psychobiology, Institute for Biomedical Research of Murcia (IMIB), School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, 30100, Spain; Institute for Aging Research, School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, 30100, Spain
| | - Emiliano Fernandez-Villalba
- Clinical & Experimental Neuroscience, Department of Human Anatomy & Psychobiology, Institute for Biomedical Research of Murcia (IMIB), School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, 30100, Spain; Institute for Aging Research, School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, 30100, Spain
| | - Maria Trinidad Herrero
- Clinical & Experimental Neuroscience, Department of Human Anatomy & Psychobiology, Institute for Biomedical Research of Murcia (IMIB), School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, 30100, Spain; Institute for Aging Research, School of Medicine, Campus Mare Nostrum, University of Murcia, Murcia, 30100, Spain.
| |
Collapse
|
19
|
Harding EC, Franks NP, Wisden W. The Temperature Dependence of Sleep. Front Neurosci 2019; 13:336. [PMID: 31105512 PMCID: PMC6491889 DOI: 10.3389/fnins.2019.00336] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/22/2019] [Indexed: 12/18/2022] Open
Abstract
Mammals have evolved a range of behavioural and neurological mechanisms that coordinate cycles of thermoregulation and sleep. Whether diurnal or nocturnal, sleep onset and a reduction in core temperature occur together. Non-rapid eye movement (NREM) sleep episodes are also accompanied by core and brain cooling. Thermoregulatory behaviours, like nest building and curling up, accompany this circadian temperature decline in preparation for sleeping. This could be a matter of simply comfort as animals seek warmth to compensate for lower temperatures. However, in both humans and other mammals, direct skin warming can shorten sleep-latency and promote NREM sleep. We discuss the evidence that body cooling and sleep are more fundamentally connected and that thermoregulatory behaviours, prior to sleep, form warm microclimates that accelerate NREM directly through neuronal circuits. Paradoxically, this warmth might also induce vasodilation and body cooling. In this way, warmth seeking and nesting behaviour might enhance the circadian cycle by activating specific circuits that link NREM initiation to body cooling. We suggest that these circuits explain why NREM onset is most likely when core temperature is at its steepest rate of decline and why transitions to NREM are accompanied by a decrease in brain temperature. This connection may have implications for energy homeostasis and the function of sleep.
Collapse
Affiliation(s)
- Edward C Harding
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Nicholas P Franks
- Department of Life Sciences, Imperial College London, London, United Kingdom.,Centre for Neurotechnology, Imperial College London, London, United Kingdom.,UK Dementia Research Institute, Imperial College London, London, United Kingdom
| | - William Wisden
- Department of Life Sciences, Imperial College London, London, United Kingdom.,Centre for Neurotechnology, Imperial College London, London, United Kingdom.,UK Dementia Research Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
20
|
Yamakawa T, Kurauchi Y, Hisatsune A, Seki T, Katsuki H. Endogenous Nitric Oxide Inhibits, Whereas Awakening Stimuli Increase, the Activity of a Subset of Orexin Neurons. Biol Pharm Bull 2019; 41:1859-1865. [PMID: 30504686 DOI: 10.1248/bpb.b18-00633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The lateral hypothalamic area contains neurons expressing neuronal nitric oxide synthase (nNOS), in addition to orexin neurons. Here we examined whether the activity of orexin neurons was regulated by endogenous nitric oxide (NO) in male C57BL/6 mice. Caffeine (30 mg/kg, intraperitoneally (i.p.)) increased the number of orexin neurons positive for c-Fos, a marker of neuronal activity, and also increased the number of NOS/c-Fos-positive cells as identified by reduced nicotinamide adenine dinucleotide phosphate (NADPH) diaphorase histochemistry and c-Fos immunohistochemistry. Diphenhydramine hydrochloride (10 mg/kg. i.p.) decreased c-Fos-positive orexin neurons but had no significant effect on the number of c-Fos-positive NOS neurons. nNOS inhibitor 7-nitroindazole (25 mg/kg, i.p.) alone increased c-Fos-positive orexin neurons, and combined treatment with caffeine and 7-nitroindazole did not show additive effect in the number of c-Fos-positive orexin neurons. In contrast, 7-nitroindazole decreased c-Fos-positive NOS neurons and attenuated caffeine-induced increase in c-Fos-positive NOS neurons. Sleep deprivation increased c-Fos-positive cells in both orexin neurons and NOS neurons, and 7-nitroindazole did not show additive effect with sleep deprivation in the activation of orexin neurons. Together, these results suggest that endogenous NO negatively regulates the activity of a subset of orexin neurons, and this subset of orexin neurons overlaps with that activated by awakening stimuli.
Collapse
Affiliation(s)
- Takao Yamakawa
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Yuki Kurauchi
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Akinori Hisatsune
- Program for Leading Graduate Schools "HIGO (Health life science: Interdisciplinary and Glocal Oriented) Program," Priority Organization for Innovation and Excellence, Kumamoto University
| | - Takahiro Seki
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Hiroshi Katsuki
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University
| |
Collapse
|
21
|
Varanoske AN, Wells AJ, Kozlowski GJ, Gepner Y, Frosti CL, Boffey D, Coker NA, Harat I, Hoffman JR. Effects of β-alanine supplementation on physical performance, cognition, endocrine function, and inflammation during a 24 h simulated military operation. Physiol Rep 2018; 6:e13938. [PMID: 30565426 PMCID: PMC6299243 DOI: 10.14814/phy2.13938] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 10/30/2018] [Indexed: 11/24/2022] Open
Abstract
Sustained military operations (SUSOPs) are associated with performance decrements and cognitive dysfunction. β-Alanine (BA) supplementation may have a role in increasing soldier resiliency by enhancing muscle-buffering capacity and reducing oxidative stress. The purpose of this study was to examine the effects of BA on physical performance, cognition, endocrine function, and inflammation during a 24 h simulated SUSOP. Nineteen males were randomized into one of two groups: BA (n = 10) or placebo (n = 9; PLA) (12 g/day) for 14 days preceding the 24 h SUSOP. Assessments were performed at 0 h (0H), 12 h (12H), and 24 h (24H) during the SUSOP. No changes in visual tracking ability, jump power, or upper-body muscular endurance were observed between groups or time points (P's > 0.05). Increases in subjective feelings of soreness and fatigue were noted at 12H compared to 0H (P < 0.05) in PLA, but not in BA. Visual reaction time for PLA was slower at 24H compared to 0H (P = 0.035), and PLA made more errors on reaction time testing at 12H compared to BA (P = 0.048), but motor reaction time was faster (P = 0.016) for PLA. Simulated litter carry and 1 km run completion times increased at 24H compared to 0H in both groups (P < 0.05), however, PLA had a longer 1 km time compared to BA at 24H (P = 0.050). Increases in inflammatory and endocrine markers were observed over the SUSOP, with no differences between groups. BA supplementation appears to maintain some aspects of cognition and physical performance during a 24 h SUSOP, with no effects on endocrine function or inflammation.
Collapse
Affiliation(s)
- Alyssa N. Varanoske
- Institute of Exercise Physiology and Wellness, Educational and Human Sciences, Sport and Exercise ScienceUniversity of Central FloridaOrlandoFlorida
| | - Adam J. Wells
- Institute of Exercise Physiology and Wellness, Educational and Human Sciences, Sport and Exercise ScienceUniversity of Central FloridaOrlandoFlorida
| | - Gregory J. Kozlowski
- Institute of Exercise Physiology and Wellness, Educational and Human Sciences, Sport and Exercise ScienceUniversity of Central FloridaOrlandoFlorida
| | - Yftach Gepner
- Institute of Exercise Physiology and Wellness, Educational and Human Sciences, Sport and Exercise ScienceUniversity of Central FloridaOrlandoFlorida
| | - Cheyanne L. Frosti
- Institute of Exercise Physiology and Wellness, Educational and Human Sciences, Sport and Exercise ScienceUniversity of Central FloridaOrlandoFlorida
| | - David Boffey
- Institute of Exercise Physiology and Wellness, Educational and Human Sciences, Sport and Exercise ScienceUniversity of Central FloridaOrlandoFlorida
| | - Nicholas A. Coker
- Institute of Exercise Physiology and Wellness, Educational and Human Sciences, Sport and Exercise ScienceUniversity of Central FloridaOrlandoFlorida
| | - Idan Harat
- Institute of Exercise Physiology and Wellness, Educational and Human Sciences, Sport and Exercise ScienceUniversity of Central FloridaOrlandoFlorida
| | - Jay R. Hoffman
- Institute of Exercise Physiology and Wellness, Educational and Human Sciences, Sport and Exercise ScienceUniversity of Central FloridaOrlandoFlorida
| |
Collapse
|
22
|
Haraguchi Y, Mizoguchi Y, Ohgidani M, Imamura Y, Murakawa-Hirachi T, Nabeta H, Tateishi H, Kato TA, Monji A. Donepezil suppresses intracellular Ca 2+ mobilization through the PI3K pathway in rodent microglia. J Neuroinflammation 2017; 14:258. [PMID: 29273047 PMCID: PMC5741946 DOI: 10.1186/s12974-017-1033-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 12/11/2017] [Indexed: 12/17/2022] Open
Abstract
Background Microglia are resident innate immune cells which release many factors including proinflammatory cytokines or nitric oxide (NO) when they are activated in response to immunological stimuli. Pathophysiology of Alzheimer’s disease (AD) is related to the inflammatory responses mediated by microglia. Intracellular Ca2+ signaling is important for microglial functions such as release of NO and cytokines. In addition, alteration of intracellular Ca2+ signaling underlies the pathophysiology of AD, while it remains unclear how donepezil, an acetylcholinesterase inhibitor, affects intracellular Ca2+ mobilization in microglial cells. Methods We examined whether pretreatment with donepezil affects the intracellular Ca2+ mobilization using fura-2 imaging and tested the effects of donepezil on phagocytic activity by phagocytosis assay in rodent microglial cells. Results In this study, we observed that pretreatment with donepezil suppressed the TNFα-induced sustained intracellular Ca2+ elevation in both rat HAPI and mouse primary microglial cells. On the other hand, pretreatment with donepezil did not suppress the mRNA expression of both TNFR1 and TNFR2 in rodent microglia we used. Pretreatment with acetylcholine but not donepezil suppressed the TNFα-induced intracellular Ca2+ elevation through the nicotinic α7 receptors. In addition, sigma 1 receptors were not involved in the donepezil-induced suppression of the TNFα-mediated intracellular Ca2+ elevation. Pretreatment with donepezil suppressed the TNFα-induced intracellular Ca2+ elevation through the PI3K pathway in rodent microglial cells. Using DAF-2 imaging, we also found that pretreatment with donepezil suppressed the production of NO induced by TNFα treatment and the PI3K pathway could be important for the donepezil-induced suppression of NO production in rodent microglial cells. Finally, phagocytosis assay showed that pretreatment with donepezil promoted phagocytic activity of rodent microglial cells through the PI3K but not MAPK/ERK pathway. Conclusions These suggest that donepezil could directly modulate the microglial function through the PI3K pathway in the rodent brain, which might be important to understand the effect of donepezil in the brain. Electronic supplementary material The online version of this article (10.1186/s12974-017-1033-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yoshinori Haraguchi
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Yoshito Mizoguchi
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan.
| | - Masahiro Ohgidani
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshiomi Imamura
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Toru Murakawa-Hirachi
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Hiromi Nabeta
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Hiroshi Tateishi
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Takahiro A Kato
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Akira Monji
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| |
Collapse
|
23
|
Yujra VQ, Antunes HKM, Mônico‐Neto M, Pisani LP, Santamarina AB, Quintana HT, de Oliveira F, Oshima CTF, Ribeiro DA. Sleep deprivation induces pathological changes in rat masticatory muscles: Role of Toll like signaling pathway and atrophy. J Cell Biochem 2017; 119:2269-2277. [DOI: 10.1002/jcb.26389] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 08/23/2017] [Indexed: 11/10/2022]
Affiliation(s)
- Veronica Q. Yujra
- Department of PathologyFederal University of São Paulo, UNIFESPSao PauloBrazil
| | - Hanna K. M. Antunes
- Department of BiosciencesFederal University of São Paulo, UNIFESPSao PauloBrazil
| | - Marcos Mônico‐Neto
- Department of PshychobiologyFederal University of São Paulo, UNIFESPSao PauloBrazil
| | - Luciana P. Pisani
- Department of BiosciencesFederal University of São Paulo, UNIFESPSao PauloBrazil
| | - Aline B. Santamarina
- Department of BiosciencesFederal University of São Paulo, UNIFESPSao PauloBrazil
| | - Hananiah T. Quintana
- Department of BiosciencesFederal University of São Paulo, UNIFESPSao PauloBrazil
| | - Flavia de Oliveira
- Department of BiosciencesFederal University of São Paulo, UNIFESPSao PauloBrazil
| | - Celina T. F. Oshima
- Department of BiosciencesFederal University of São Paulo, UNIFESPSao PauloBrazil
| | - Daniel A. Ribeiro
- Department of BiosciencesFederal University of São Paulo, UNIFESPSao PauloBrazil
| |
Collapse
|
24
|
Marini S, Santangeli O, Saarelainen P, Middleton B, Chowdhury N, Skene DJ, Costa R, Porkka-Heiskanen T, Montagnese S. Abnormalities in the Polysomnographic, Adenosine and Metabolic Response to Sleep Deprivation in an Animal Model of Hyperammonemia. Front Physiol 2017; 8:636. [PMID: 28912724 PMCID: PMC5583967 DOI: 10.3389/fphys.2017.00636] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 08/14/2017] [Indexed: 12/17/2022] Open
Abstract
Patients with liver cirrhosis can develop hyperammonemia and hepatic encephalopathy (HE), accompanied by pronounced daytime sleepiness. Previous studies with healthy volunteers show that experimental increase in blood ammonium levels increases sleepiness and slows the waking electroencephalogram. As ammonium increases adenosine levels in vitro, and adenosine is a known regulator of sleep/wake homeostasis, we hypothesized that the sleepiness-inducing effect of ammonium is mediated by adenosine. Eight adult male Wistar rats were fed with an ammonium-enriched diet for 4 weeks; eight rats on standard diet served as controls. Each animal was implanted with electroencephalography/electromyography (EEG/EMG) electrodes and a microdialysis probe. Sleep EEG recording and cerebral microdialysis were carried out at baseline and after 6 h of sleep deprivation. Adenosine and metabolite levels were measured by high-performance liquid chromatography (HPLC) and targeted LC/MS metabolomics, respectively. Baseline adenosine and metabolite levels (12 of 16 amino acids, taurine, t4-hydroxy-proline, and acetylcarnitine) were lower in hyperammonemic animals, while putrescine was higher. After sleep deprivation, hyperammonemic animals exhibited a larger increase in adenosine levels, and a number of metabolites showed a different time-course in the two groups. In both groups the recovery period was characterized by a significant decrease in wakefulness/increase in NREM and REM sleep. However, while control animals exhibited a gradual compensatory effect, hyperammonemic animals showed a significantly shorter recovery phase. In conclusion, the adenosine/metabolite/EEG response to sleep deprivation was modulated by hyperammonemia, suggesting that ammonia affects homeostatic sleep regulation and its metabolic correlates.
Collapse
Affiliation(s)
- Selena Marini
- Department of Biology, University of PaduaPadua, Italy.,Department of Physiology, Institute of Biomedicine and Physiology, University of HelsinkiHelsinki, Finland
| | - Olena Santangeli
- Department of Physiology, Institute of Biomedicine and Physiology, University of HelsinkiHelsinki, Finland
| | - Pirjo Saarelainen
- Department of Physiology, Institute of Biomedicine and Physiology, University of HelsinkiHelsinki, Finland
| | - Benita Middleton
- Chronobiology, Faculty of Health and Medical Sciences, University of SurreyGuildford, United Kingdom
| | - Namrata Chowdhury
- Chronobiology, Faculty of Health and Medical Sciences, University of SurreyGuildford, United Kingdom
| | - Debra J Skene
- Chronobiology, Faculty of Health and Medical Sciences, University of SurreyGuildford, United Kingdom
| | - Rodolfo Costa
- Department of Biology, University of PaduaPadua, Italy
| | - Tarja Porkka-Heiskanen
- Department of Physiology, Institute of Biomedicine and Physiology, University of HelsinkiHelsinki, Finland
| | | |
Collapse
|
25
|
Serum and Brain Metabolomic Variations Reveal Perturbation of Sleep Deprivation on Rats and Ameliorate Effect of Total Ginsenoside Treatment. Int J Genomics 2017; 2017:5179271. [PMID: 28900617 PMCID: PMC5576418 DOI: 10.1155/2017/5179271] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/19/2017] [Indexed: 02/06/2023] Open
Abstract
Sleep loss or sleep deprivation (SD) refers to shorter sleep than average baseline need, and SD has been a serious problem of modern societies which affects health and well-being. Panax ginseng is a well-known traditional Chinese medicine (TCM). Our previous study has demonstrated that total ginsenosides (GS), the extracts from Panax ginseng, could effectively improve cognition and behavior on SD rats. However, little is known about its metabolomic study. In this study, serum and brain metabolomic method based on gas chromatography coupled with mass spectrometry (GC/MS) was employed to evaluate the efficacy and study the mechanism of GS on a rat model of SD. With pattern recognition analysis of serum and brain tissue metabolite profile, a clear separation of the model group and control group was acquired for serum and brain tissue samples; the MGS (model + GS) group showed a tendency of recovering when compared to control group, which was consistent with behavioral and biochemical parameters. 39 and 40 potential biomarkers of brain tissues and serum samples, respectively, were identified and employed to explore the possible mechanism. Our work revealed that GS has significant protective effects on SD, and metabolomics is a useful tool for evaluating efficacy and elucidating mechanism in TCM.
Collapse
|
26
|
Onaolapo JO, Onaolapo YA, Akanmu AM, Olayiwola G. Caffeine and sleep-deprivation mediated changes in open-field behaviours, stress response and antioxidant status in mice. Sleep Sci 2016; 9:236-243. [PMID: 28123668 PMCID: PMC5241623 DOI: 10.1016/j.slsci.2016.10.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/25/2016] [Accepted: 10/27/2016] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVES Effects of daily caffeine consumption on open-field behaviours, serum corticosterone and brain antioxidant levels were investigated after six hours of total sleep-deprivation in prepubertal mice. We tested the hypothesis that daily caffeine consumption may significantly alter behaviour, stress and antioxidative response of prepubertal mice to an acute episode of total sleep-deprivation. METHODS Prepubertal Swiss mice of both sexes were assigned to two main groups of 120 each (subdivided into 6 groups of 10 each, based on sex), and administered vehicle or graded oral doses of caffeine (10, 20, 40, 80 and 120 mg/kg/day) for 14 days. On day 14, a main group was subjected to 6 h of total sleep-deprivation by 'gentle-handling'. Open-field behaviours were then assessed in both groups, after which animals were euthanized, and levels of corticosterone, superoxide dismutase and glutathione peroxidase assayed. RESULTS Horizontal locomotion, rearing and grooming increased significantly, compared to control, with sleep-deprived (SD) mice showing stronger caffeine-driven responses at higher doses; and SD female mice showing sustained response to caffeine, compared to respective males. Plasma corticosterone increased with increasing doses of caffeine in both non sleep-deprived (NSD) and SD mice; although SD mice had higher corticosterone levels. Sleep-deprivation and/or higher doses of caffeine were associated with derangements in brain antioxidant levels. CONCLUSION Repeated caffeine consumption and/or acute sleep-deprivation led to significant changes in pattern of open-field behaviour and stress/antioxidant response in mice. Responses seen in the study are probably due to modulatory effects of caffeine on the total body response to stressful stimuli.
Collapse
Affiliation(s)
- J. Olakunle Onaolapo
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Ladoke Akintola University of Technology, Osogbo, Osun, Nigeria
| | - Y. Adejoke Onaolapo
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - A. Moses Akanmu
- Department of Pharmacology, Faculty of Pharmacy, Obafemi Awolowo University, Ile, Ife, Osun State, Nigeria
| | - Gbola Olayiwola
- Department of Clinical Pharmacy and Pharmacy Administration, Faculty of Pharmacy, Obafemi Awolowo University, Ile, Ife, Osun State, Nigeria
| |
Collapse
|
27
|
Abstract
Sleep is a complex physiological process that is regulated globally, regionally, and locally by both cellular and molecular mechanisms. It occurs to some extent in all animals, although sleep expression in lower animals may be co-extensive with rest. Sleep regulation plays an intrinsic part in many behavioral and physiological functions. Currently, all researchers agree there is no single physiological role sleep serves. Nevertheless, it is quite evident that sleep is essential for many vital functions including development, energy conservation, brain waste clearance, modulation of immune responses, cognition, performance, vigilance, disease, and psychological state. This review details the physiological processes involved in sleep regulation and the possible functions that sleep may serve. This description of the brain circuitry, cell types, and molecules involved in sleep regulation is intended to further the reader's understanding of the functions of sleep.
Collapse
Affiliation(s)
- Mark R. Zielinski
- Veterans Affairs Boston Healthcare System, West Roxbury, MA 02132, USA and Harvard Medical School, Department of Psychiatry
| | - James T. McKenna
- Veterans Affairs Boston Healthcare System, West Roxbury, MA 02132, USA and Harvard Medical School, Department of Psychiatry
| | - Robert W. McCarley
- Veterans Affairs Boston Healthcare System, Brockton, MA 02301, USA and Harvard Medical School, Department of Psychiatry
| |
Collapse
|
28
|
Manchanda S, Mishra R, Singh R, Kaur T, Kaur G. Aqueous Leaf Extract of Withania somnifera as a Potential Neuroprotective Agent in Sleep-deprived Rats: a Mechanistic Study. Mol Neurobiol 2016; 54:3050-3061. [PMID: 27037574 DOI: 10.1007/s12035-016-9883-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/22/2016] [Indexed: 10/22/2022]
Abstract
Modern lifestyle and sustained stress of professional commitments in the current societal set up often disrupts the normal sleep cycle and duration which is known to lead to cognitive impairments. In the present study, we report whether leaf extract of Withania somnifera (Ashwagandha) has potential neuroprotective role in acute stress of sleep deprivation. Experiments were performed on three groups of adult Wistar rats: group 1 (vehicle treated-undisturbed sleep [VUD]), group 2 (vehicle treated-sleep deprived [VSD]), and group 3 (ASH-WEX treated-sleep deprived [WSD]). Groups 1 and 2 received single oral feeding of vehicle and group 3 received ASH-WEX orally (140 mg/kg or 1 ml/250 g of body weight) for 15 consecutive days. Immediately after this regimen, animals from group 1 were allowed undisturbed sleep (between 6 a.m. and 6 p.m.), whereas rats of groups 2 and 3 were deprived of sleep during this period. We observed that WSD rats showed significant improvement in their performance in behavioral tests as compared to VSD group. At the molecular level, VSD rats showed acute change in the expression of proteins involved in synaptic plasticity, cell survival, and apoptosis in the hippocampus region of brain, which was suppressed by ASH-WEX treatment thus indicating decreased cellular stress and apoptosis in WSD group. This data suggest that Ashwagandha may be a potential agent to suppress the acute effects of sleep loss on learning and memory impairments and may emerge as a novel supplement to control SD-induced cognitive impairments.
Collapse
Affiliation(s)
- Shaffi Manchanda
- Medical Biotechnology Laboratory, Department of Biotechnology, Guru Nanak Dev University, Amritsar, 143005, Punjab, India
| | - Rachana Mishra
- Medical Biotechnology Laboratory, Department of Biotechnology, Guru Nanak Dev University, Amritsar, 143005, Punjab, India
| | - Rumani Singh
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Taranjeet Kaur
- Medical Biotechnology Laboratory, Department of Biotechnology, Guru Nanak Dev University, Amritsar, 143005, Punjab, India
| | - Gurcharan Kaur
- Medical Biotechnology Laboratory, Department of Biotechnology, Guru Nanak Dev University, Amritsar, 143005, Punjab, India.
| |
Collapse
|
29
|
H1N1 influenza virus induces narcolepsy-like sleep disruption and targets sleep-wake regulatory neurons in mice. Proc Natl Acad Sci U S A 2015; 113:E368-77. [PMID: 26668381 DOI: 10.1073/pnas.1521463112] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
An increased incidence in the sleep-disorder narcolepsy has been associated with the 2009-2010 pandemic of H1N1 influenza virus in China and with mass vaccination campaigns against influenza during the pandemic in Finland and Sweden. Pathogenetic mechanisms of narcolepsy have so far mainly focused on autoimmunity. We here tested an alternative working hypothesis involving a direct role of influenza virus infection in the pathogenesis of narcolepsy in susceptible subjects. We show that infection with H1N1 influenza virus in mice that lack B and T cells (Recombinant activating gene 1-deficient mice) can lead to narcoleptic-like sleep-wake fragmentation and sleep structure alterations. Interestingly, the infection targeted brainstem and hypothalamic neurons, including orexin/hypocretin-producing neurons that regulate sleep-wake stability and are affected in narcolepsy. Because changes occurred in the absence of adaptive autoimmune responses, the findings show that brain infections with H1N1 virus have the potential to cause per se narcoleptic-like sleep disruption.
Collapse
|
30
|
Disrupted sleep-wake regulation in type 1 equilibrative nucleoside transporter knockout mice. Neuroscience 2015; 303:211-9. [PMID: 26143012 DOI: 10.1016/j.neuroscience.2015.06.037] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 06/18/2015] [Accepted: 06/19/2015] [Indexed: 01/25/2023]
Abstract
The type 1 equilibrative nucleoside transporter (ENT1) is implicated in regulating levels of extracellular adenosine ([AD]ex). In the basal forebrain (BF) levels of [AD]ex increase during wakefulness and closely correspond to the increases in the electroencephalogram (EEG) delta (0.75-4.5Hz) activity (NRδ) during subsequent non-rapid eye movement sleep (NREMS). Thus in the BF, [AD]ex serves as a biochemical marker of sleep homeostasis. Waking EEG activity in theta range (5-9Hz, Wθ) is also described as a marker of sleep homeostasis. An hour-by-hour temporal relationship between the Wθ and NRδ is unclear. In this study we examined the relationship between these EEG markers of sleep homeostasis during spontaneous sleep-wakefulness and during sleep deprivation (SD) and recovery sleep in the ENT1 gene knockout (ENT1KO) mouse. We observed that baseline NREMS amount was decreased during the light period in ENT1KO mice, accompanied by a weak correlation between Wθ of each hour and NRδ of its subsequent hour when compared to their wild-type (WT) littermates. Perfusion of low dose of adenosine into BF not only strengthened the Wθ-NRδ relationship, but also increased NREMS to match with the WT littermates suggesting decreased [AD]ex in ENT1KO mice. However, the SD-induced [AD]ex increase in the BF and the linear correlation between the EEG markers of sleep homeostasis were unaffected in ENT1KO mice suggesting that during SD, sources other than ENT1 contribute to increase in [AD]ex. Our data provide evidence for a differential regulation of wakefulness-associated [AD]ex during spontaneous vs prolonged waking.
Collapse
|
31
|
Datta S, Knapp CM, Koul-Tiwari R, Barnes A. The homeostatic regulation of REM sleep: A role for localized expression of brain-derived neurotrophic factor in the brainstem. Behav Brain Res 2015; 292:381-92. [PMID: 26146031 DOI: 10.1016/j.bbr.2015.06.038] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 06/23/2015] [Accepted: 06/27/2015] [Indexed: 01/17/2023]
Abstract
Homeostatic regulation of REM sleep plays a key role in neural plasticity and deficits in this process are implicated in the development of many neuropsychiatric disorders. Little is known, however, about the molecular mechanisms that underlie this homeostatic regulation process. This study examined the hypothesis that, during selective REM sleep deprivation (RSD), increased brain-derived neurotrophic factor (BDNF) expression in REM sleep regulating areas is critical for the development of homeostatic drive for REM sleep, as measured by an increase in the number of REM sleep transitions. Rats were assigned to RSD, non-sleep deprived (BSL), or total sleep deprivation (TSD) groups. Physiological recordings were obtained from cortical, hippocampal, and pontine EEG electrodes over a 6h period, in which sleep deprivation occurred during the first 3h. In the RSD, but not the other conditions, homeostatic drive for REM sleep increased progressively. BDNF protein expression was significantly greater in the pedunculopontine tegmentum (PPT) and subcoeruleus nucleus (SubCD) in the RSD as compared to the TSD and BSL groups, areas that regulate REM sleep, but not in the medial preoptic area, which regulates non-REM sleep. There was a significant positive correlation between RSD-induced increases in number of REM sleep episodes and increased BDNF expression in the PPT and SubCD. These increases positively correlated with levels of homeostatic drive for REM sleep. These results, for the first time, suggest that selective RSD-induced increased expression of BDNF in the PPT and SubCD are determinant factors in the development of the homeostatic drive for REM sleep.
Collapse
Affiliation(s)
- Subimal Datta
- Department of Anesthesiology, Graduate School of Medicine, The University of Tennessee, 1924 Alcoa Highway, Knoxville, TN 37920, USA; Department of Psychology, College of Arts and Sciences, The University of Tennessee, 1404 Circle Drive, Knoxville, TN 37996, USA.
| | - Clifford M Knapp
- Department of Psychiatry, Boston University School of Medicine, 85 East Newton Street, Boston, MA 02118, USA
| | - Richa Koul-Tiwari
- Department of Anesthesiology, Graduate School of Medicine, The University of Tennessee, 1924 Alcoa Highway, Knoxville, TN 37920, USA
| | - Abigail Barnes
- Department of Anesthesiology, Graduate School of Medicine, The University of Tennessee, 1924 Alcoa Highway, Knoxville, TN 37920, USA
| |
Collapse
|
32
|
Huang ZL, Zhang Z, Qu WM. Roles of adenosine and its receptors in sleep-wake regulation. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 119:349-71. [PMID: 25175972 DOI: 10.1016/b978-0-12-801022-8.00014-3] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
This chapter summarizes the current knowledge about the role of adenosine in the sleep-wake regulation with a focus on adenosine in the brain, regulation of adenosine levels, adenosine receptors, and manipulations of the adenosine system by the use of pharmacological and molecular biological tools. Adenosine is neither stored nor released as a classical neurotransmitter and is thought to be formed inside cells or on their surface, mostly by breakdown of adenine nucleotides. The extracellular level of adenosine increases in the cortex and basal forebrain (BF) during prolonged wakefulness and decreases during the sleep-recovery period. Therefore, adenosine is proposed to act as a homeostatic regulator of sleep. The endogenous somnogen prostaglandin (PG) D2 increases the extracellular level of adenosine under the subarachnoid space of the BF and promotes physiological sleep. There are four adenosine receptor subtypes: adenosine A1 receptor (R, A1R), A2AR, A2BR, and A3R. Both the A1R and the A2AR have been reported to be involved in sleep induction. The A2AR plays an important role in the somnogenic effects of PGD2. Activation of A2AR by its agonist infused into the brain potently increases sleep and the arousal effect of caffeine, an A1R and A2AR antagonist, was shown to be dependent on the A2AR. On the other hand, inhibition of wake-promoting neurons via the A1R also mediates the sleep-inducing effects of adenosine, whereas activation of A1R in the lateral preoptic area induces wakefulness. These findings indicate that A2AR plays a predominant role in sleep induction, whereas A1R regulates the sleep-wake cycle in a site-dependent manner.
Collapse
Affiliation(s)
- Zhi-Li Huang
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology, Institute of Brain Science, Shanghai Medical College of Fudan University, Shanghai, China.
| | - Ze Zhang
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology, Institute of Brain Science, Shanghai Medical College of Fudan University, Shanghai, China
| | - Wei-Min Qu
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology, Institute of Brain Science, Shanghai Medical College of Fudan University, Shanghai, China.
| |
Collapse
|
33
|
Sleep deprivation and oxidative stress in animal models: a systematic review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:234952. [PMID: 25945148 PMCID: PMC4402503 DOI: 10.1155/2015/234952] [Citation(s) in RCA: 178] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/22/2015] [Accepted: 03/22/2015] [Indexed: 12/15/2022]
Abstract
Because the function and mechanisms of sleep are partially clear, here we applied a meta-analysis to address the issue whether sleep function includes antioxidative properties in mice and rats. Given the expansion of the knowledge in the sleep field, it is indeed ambitious to describe all mammals, or other animals, in which sleep shows an antioxidant function. However, in this paper we reviewed the current understanding from basic studies in two species to drive the hypothesis that sleep is a dynamic-resting state with antioxidative properties. We performed a systematic review of articles cited in Medline, Scopus, and Web of Science until March 2015 using the following search terms: Sleep or sleep deprivation and oxidative stress, lipid peroxidation, glutathione, nitric oxide, catalase or superoxide dismutase. We found a total of 266 studies. After inclusion and exclusion criteria, 44 articles were included, which are presented and discussed in this study. The complex relationship between sleep duration and oxidative stress is discussed. Further studies should consider molecular and genetic approaches to determine whether disrupted sleep promotes oxidative stress.
Collapse
|
34
|
Chittora R, Jain A, Suhalka P, Sharma C, Jaiswal N, Bhatnagar M. Sleep deprivation: Neural regulation and consequences. Sleep Biol Rhythms 2015. [DOI: 10.1111/sbr.12110] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Reena Chittora
- Department of Zoology; Mohan Lal Sukhadia University; Udaipur India
| | - Ayushi Jain
- Department of Zoology; Mohan Lal Sukhadia University; Udaipur India
| | - Pooja Suhalka
- Department of Zoology; Mohan Lal Sukhadia University; Udaipur India
| | - Chhavi Sharma
- Department of Zoology; Mohan Lal Sukhadia University; Udaipur India
| | - Neha Jaiswal
- Department of Zoology; Mohan Lal Sukhadia University; Udaipur India
| | - Maheep Bhatnagar
- Department of Zoology; Mohan Lal Sukhadia University; Udaipur India
| |
Collapse
|
35
|
Stern AL, Naidoo N. Wake-active neurons across aging and neurodegeneration: a potential role for sleep disturbances in promoting disease. SPRINGERPLUS 2015; 4:25. [PMID: 25635245 PMCID: PMC4306674 DOI: 10.1186/s40064-014-0777-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 12/23/2014] [Indexed: 12/13/2022]
Abstract
Sleep/wake disturbance is a feature of almost all common age-related neurodegenerative diseases. Although the reason for this is unknown, it is likely that this inability to maintain sleep and wake states is in large part due to declines in the number and function of wake-active neurons, populations of cells that fire only during waking and are silent during sleep. Consistent with this, many of the brain regions that are most susceptible to neurodegeneration are those that are necessary for wake maintenance and alertness. In the present review, these wake-active populations are systematically assessed in terms of their observed pathology across aging and several neurodegenerative diseases, with implications for future research relating sleep and wake disturbances to aging and age-related neurodegeneration.
Collapse
Affiliation(s)
- Anna L Stern
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Nirinjini Naidoo
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
36
|
Kalinchuk AV, Porkka-Heiskanen T, McCarley RW, Basheer R. Cholinergic neurons of the basal forebrain mediate biochemical and electrophysiological mechanisms underlying sleep homeostasis. Eur J Neurosci 2015; 41:182-95. [PMID: 25369989 PMCID: PMC4460789 DOI: 10.1111/ejn.12766] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 09/26/2014] [Accepted: 09/30/2014] [Indexed: 12/13/2022]
Abstract
The tight coordination of biochemical and electrophysiological mechanisms underlies the homeostatic sleep pressure (HSP) produced by sleep deprivation (SD). We have reported that during SD the levels of inducible nitric oxide synthase (iNOS), extracellular nitric oxide (NO), adenosine [AD]ex , lactate [Lac]ex and pyruvate [Pyr]ex increase in the basal forebrain (BF). However, it is not clear whether all of them contribute to HSP leading to increased electroencephalogram (EEG) delta activity during non-rapid eye movement (NREM) recovery sleep (RS) following SD. Previously, we showed that NREM delta increase evident during RS depends on the presence of BF cholinergic (ChBF) neurons. Here, we investigated the role of ChBF cells in coordination of biochemical and EEG changes seen during SD and RS in the rat. Increases in low-theta power (5-7 Hz), but not high-theta (7-9 Hz), during SD correlated with the increase in NREM delta power during RS, and with the changes in nitrate/nitrite [NOx ]ex and [AD]ex . Lesions of ChBF cells using IgG 192-saporin prevented increases in [NOx ]ex , [AD]ex and low-theta activity, during SD, but did not prevent increases in [Lac]ex and [Pyr]ex . Infusion of NO donor DETA NONOate into the saporin-treated BF failed to increase NREM RS and delta power, suggesting ChBF cells are important for mediating NO homeostatic effects. Finally, SD-induced iNOS was mostly expressed in ChBF cells, and the intensity of iNOS induction correlated with the increase in low-theta activity. Together, our data indicate ChBF cells are important in regulating the biochemical and EEG mechanisms that contribute to HSP.
Collapse
Affiliation(s)
- Anna V. Kalinchuk
- VA Boston Healthcare System and Harvard Medical School, 1400 V.F.W. Parkway, West Roxbury MA 02067
| | | | - Robert W. McCarley
- VA Boston Healthcare System and Harvard Medical School, 1400 V.F.W. Parkway, West Roxbury MA 02067
| | - Radhika Basheer
- VA Boston Healthcare System and Harvard Medical School, 1400 V.F.W. Parkway, West Roxbury MA 02067
| |
Collapse
|
37
|
Elliott AS, Huber JD, O'Callaghan JP, Rosen CL, Miller DB. A review of sleep deprivation studies evaluating the brain transcriptome. SPRINGERPLUS 2014; 3:728. [PMID: 25932362 PMCID: PMC4409616 DOI: 10.1186/2193-1801-3-728] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 11/25/2014] [Indexed: 12/14/2022]
Abstract
Epidemiological studies show a positive association between adequate sleep and good health. Further, disrupted sleep may increase the risk for CNS diseases, such as stroke and Alzheimer’s disease. However, there has been limited progress in determining how sleep is linked to brain health or how sleep disruption may increase susceptibility to brain insult and disease. Animal studies can aid in understanding these links. In reviewing the animal literature related to the effects of sleep disruption on the brain, we found most of the work was directed toward investigating and characterizing the role of various brain areas or structures in initiating and regulating sleep. In contrast, limited effort has been directed towards understanding how sleep disruption alters the brain’s health or susceptibility to insult. We also note many current studies have determined the changes in the brain following compromised sleep by examining, for example, the brain transcriptome or to a more limited extent the proteome. However, these studies have utilized almost exclusively total sleep deprivation (e.g., 24 out of 24 hours) paradigms or single short periods of limited acute sleep deprivation (e.g., 3 out of 24 hours). While such strategies are beneficial in understanding how sleep is controlled, they may not have much translational value for determining links between sleep and brain health or for determining how sleep disruption may increase brain susceptibility to insult. Surprisingly, few studies have determined how the duration and recurrence of sleep deprivation influence the effects seen after sleep deprivation. Our aim in this review was to identify relevant rodent studies from 1980 through 2012 and analyze those that use varying durations of sleep deprivation or restriction in their effort to evaluate the effects of sleep deprivation on the brain transcriptome and to a more limited extent the proteome. We examined how differences in the duration of sleep deprivation affect gene and protein expression to better understand the full consequences of repeated sleep disruption on the brain. Future research needs to consider and emphasize how the type and extent of the sleep deprivation exposure impacts the conclusions reached concerning the influence of sleep disruption on the brain. We identified relevant studies between 1980 and 2012 by searching the electronic databases of PubMed, Medline (Ovid), Embase (Ovid), and Web of Science using the terms “sleep” AND “disrupt”, “deprivation”, “restrict”, “fragment”, “loss”, “disturb”, “disorder”, “dysfunction”, “brain”, “cortex”, striatum”, hypothalamus”, “hippocampus”, “gene”, “protein”, “genomics”, “proteomics”, “polymerase chain reaction”, “pcr”, “microarray”, “molecular”, “rodent” “rat”, “rats”, “mouse”, “mice”. All searches were limited to rodent studies in English and the reference lists of retrieved articles were searched for additional pertinent studies.
Collapse
Affiliation(s)
- Alisa S Elliott
- School of Medicine, West Virginia University, Morgantown, WV USA
| | - Jason D Huber
- School of Pharmacy, West Virginia University, Morgantown, WV USA
| | - James P O'Callaghan
- Toxicology and Molecular Biology Branch, CDC-NIOSH, 1095 Willowdale Rd, Morgantown, WV 26505 USA
| | - Charles L Rosen
- School of Medicine, West Virginia University, Morgantown, WV USA
| | - Diane B Miller
- Toxicology and Molecular Biology Branch, CDC-NIOSH, 1095 Willowdale Rd, Morgantown, WV 26505 USA
| |
Collapse
|
38
|
Mizoguchi Y, Kato TA, Seki Y, Ohgidani M, Sagata N, Horikawa H, Yamauchi Y, Sato-Kasai M, Hayakawa K, Inoue R, Kanba S, Monji A. Brain-derived neurotrophic factor (BDNF) induces sustained intracellular Ca2+ elevation through the up-regulation of surface transient receptor potential 3 (TRPC3) channels in rodent microglia. J Biol Chem 2014; 289:18549-55. [PMID: 24811179 DOI: 10.1074/jbc.m114.555334] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Microglia are immune cells that release factors, including proinflammatory cytokines, nitric oxide (NO), and neurotrophins, following activation after disturbance in the brain. Elevation of intracellular Ca(2+) concentration ([Ca(2+)]i) is important for microglial functions such as the release of cytokines and NO from activated microglia. There is increasing evidence suggesting that pathophysiology of neuropsychiatric disorders is related to the inflammatory responses mediated by microglia. Brain-derived neurotrophic factor (BDNF) is a neurotrophin well known for its roles in the activation of microglia as well as in pathophysiology and/or treatment of neuropsychiatric disorders. In this study, we sought to examine the underlying mechanism of BDNF-induced sustained increase in [Ca(2+)]i in rodent microglial cells. We observed that canonical transient receptor potential 3 (TRPC3) channels contribute to the maintenance of BDNF-induced sustained intracellular Ca(2+) elevation. Immunocytochemical technique and flow cytometry also revealed that BDNF rapidly up-regulated the surface expression of TRPC3 channels in rodent microglial cells. In addition, pretreatment with BDNF suppressed the production of NO induced by tumor necrosis factor α (TNFα), which was prevented by co-adiministration of a selective TRPC3 inhibitor. These suggest that BDNF induces sustained intracellular Ca(2+) elevation through the up-regulation of surface TRPC3 channels and TRPC3 channels could be important for the BDNF-induced suppression of the NO production in activated microglia. We show that TRPC3 channels could also play important roles in microglial functions, which might be important for the regulation of inflammatory responses and may also be involved in the pathophysiology and/or the treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Yoshito Mizoguchi
- From the Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan,
| | - Takahiro A Kato
- the Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, the Innovation Center for Medical Redox Navigation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, and
| | - Yoshihiro Seki
- the Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masahiro Ohgidani
- the Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, the Innovation Center for Medical Redox Navigation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, and
| | - Noriaki Sagata
- the Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, the Innovation Center for Medical Redox Navigation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, and
| | - Hideki Horikawa
- the Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yusuke Yamauchi
- the Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Mina Sato-Kasai
- the Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kohei Hayakawa
- the Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Ryuji Inoue
- the Department of Physiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jyonan-ku, Fukuoka 812-0180, Japan
| | - Shigenobu Kanba
- the Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Akira Monji
- From the Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
| |
Collapse
|
39
|
Béchade C, Colasse S, Diana MA, Rouault M, Bessis A. NOS2 expression is restricted to neurons in the healthy brain but is triggered in microglia upon inflammation. Glia 2014; 62:956-63. [DOI: 10.1002/glia.22652] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 02/04/2014] [Accepted: 02/07/2014] [Indexed: 01/13/2023]
Affiliation(s)
- Catherine Béchade
- Institut de Biologie de l'Ecole Normale Supérieure; F-75005 Paris France
- Institut National de la Santé et de la Recherche Médicale U1024; F-75005 Paris France
- Centre National de la Recherche Scientifique; Unité Mixte de Recherche 8197 F-75005 France
| | - Sabrina Colasse
- Institut de Biologie de l'Ecole Normale Supérieure; F-75005 Paris France
- Institut National de la Santé et de la Recherche Médicale U1024; F-75005 Paris France
- Centre National de la Recherche Scientifique; Unité Mixte de Recherche 8197 F-75005 France
| | - Marco A. Diana
- Institut de Biologie de l'Ecole Normale Supérieure; F-75005 Paris France
- Institut National de la Santé et de la Recherche Médicale U1024; F-75005 Paris France
- Centre National de la Recherche Scientifique; Unité Mixte de Recherche 8197 F-75005 France
| | - Martin Rouault
- Institut de Biologie de l'Ecole Normale Supérieure; F-75005 Paris France
- Institut National de la Santé et de la Recherche Médicale U1024; F-75005 Paris France
- Centre National de la Recherche Scientifique; Unité Mixte de Recherche 8197 F-75005 France
| | - Alain Bessis
- Institut de Biologie de l'Ecole Normale Supérieure; F-75005 Paris France
- Institut National de la Santé et de la Recherche Médicale U1024; F-75005 Paris France
- Centre National de la Recherche Scientifique; Unité Mixte de Recherche 8197 F-75005 France
| |
Collapse
|
40
|
Lima AMA, de Bruin VMS, Rios ERV, de Bruin PFC. Differential effects of paradoxical sleep deprivation on memory and oxidative stress. Naunyn Schmiedebergs Arch Pharmacol 2014; 387:399-406. [PMID: 24424716 DOI: 10.1007/s00210-013-0955-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 12/25/2013] [Indexed: 02/06/2023]
Abstract
Sleep has important functions for every organ in the body and sleep deprivation (SD) leads to disorders that cause irreparable damage. The aim of this study was to investigate behavioral and brain structural alterations in mice deprived of paradoxical sleep for 48 and 72 h. Working memory, aversive memory as well as levels of nitric oxide (NO) and thiobarbituric acid reactive substances (TBARS) in the hippocampus, body striatum, and prefrontal cortex were evaluated. Working memory was affected in the 48- and 72-h SD groups while aversive memory was altered only in the 48-h SD group (p ≤ 0.05). Our findings showed that SD reduces NO levels in most brain areas (p < 0.05): NO levels were unaltered in the striatum of animals sleep-deprived for 48 h. Higher levels of TBARS were observed in all areas of the SD groups (p ≤ 0.05). Thus, we confirmed that SD has duration-dependent effects on behavior as well as on NO and TBARS levels in the brain. Preserved striatum NO levels suggest that this structure is less vulnerable to oxidative stress and is only affected by SD of longer duration. Increased TBARS and reduced NO levels in the hippocampus and prefrontal cortex confirm a central role for both these structures in working memory and aversive memory. Contextual fear conditioning was not affected by longer periods of SD. Thus, our findings suggest that shorter SD time may be more beneficial to avoid aversive memory where this may have implications for the management of posttraumatic stress.
Collapse
Affiliation(s)
- Alisson Menezes Araujo Lima
- Programa de Pós-Graduação em Ciências Médicas, Faculdade de Medicina, Universidade Federal do Ceará, R. Cel Nunes de Melo 1315, Rodolfo Teófilo, 60.430-270, Fortaleza, Ceará, Brazil
| | | | | | | |
Collapse
|
41
|
Sha L, Linden DR, Farrugia G, Szurszewski JH. Effect of endogenous hydrogen sulfide on the transwall gradient of the mouse colon circular smooth muscle. J Physiol 2013; 592:1077-89. [PMID: 24366262 DOI: 10.1113/jphysiol.2013.266841] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
A transwall gradient in resting membrane potential (RMP) exists across the circular muscle layer in the mouse colon. This gradient is dependent on endogenous generation of CO. H2S is also generated in muscle layers of the mouse colon. The effect of endogenously generated H2S on the transwall gradient is not known. The aim was to investigate the role of endogenous H2S. Our results showed that the CSE inhibitor dl-propargylglycine (PAG, 500 μm) had no effect on the transwall gradient. However, in preparations pretreated with the nitric oxide synthase inhibitor N-nitro-l-arginine (l-NNA, 200 μm) and in nNOS-knockout (KO) mouse preparations, PAG shifted the transwall gradient in the depolarizing direction. In CSE-KO-nNOS-KO mice, the gradient was shifted in the depolarizing direction. Endogenous generation of NO was significantly higher in muscle preparations of CSE-KO mice compared to wild-type (WT) mice. The amplitude of NO-mediated slow inhibitory junction potentials (S-IJPs) evoked by electric field stimulation was significantly higher in CSE-KO mouse preparations compared to the amplitude of S-IJPs in wild-type mouse preparations. CSE was present in all submucosal ganglion neurons and in almost all myenteric ganglion neurons. Eleven per cent of CSE positive neurons in the submucosal plexus and 50% of CSE positive neurons in the myenteric plexus also contained nNOS. Our results suggest that endogenously generated H2S acts as a stealth hyperpolarizing factor on smooth muscle cells to maintain the CO-dependent transwall gradient and inhibits NO production from nNOS.
Collapse
Affiliation(s)
- L Sha
- Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA. E-mail:
| | | | | | | |
Collapse
|
42
|
Saletin JM, van der Helm E, Walker MP. Structural brain correlates of human sleep oscillations. Neuroimage 2013; 83:658-68. [PMID: 23770411 PMCID: PMC4263481 DOI: 10.1016/j.neuroimage.2013.06.021] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Revised: 05/17/2013] [Accepted: 06/04/2013] [Indexed: 02/02/2023] Open
Abstract
Sleep is strongly conserved within species, yet marked and perplexing inter-individual differences in sleep physiology are observed. Combining EEG sleep recordings and high-resolution structural brain imaging, here we demonstrate that the morphology of the human brain offers one explanatory factor of such inter-individual variability. Gray matter volume in interoceptive and exteroceptive cortices correlated with the expression of slower NREM sleep spindle frequencies, supporting their proposed role in sleep protection against conscious perception. Conversely, and consistent with an involvement in declarative memory processing, gray matter volume in bilateral hippocampus was associated with faster NREM sleep spindle frequencies. In contrast to spindles, gray matter volume in the homeostatic sleep-regulating center of the basal forebrain/hypothalamus, together with the medial prefrontal cortex, accounted for individual differences in NREM slow wave oscillations. Together, such findings indicate that the qualitative and quantitative expression of human sleep physiology is significantly related to anatomically specific differences in macroscopic brain structure.
Collapse
Affiliation(s)
- Jared M. Saletin
- Sleep and Neuroimaging Laboratory, Department of Psychology, University of California, Berkeley, California 94720-1650, USA
| | - Els van der Helm
- Sleep and Neuroimaging Laboratory, Department of Psychology, University of California, Berkeley, California 94720-1650, USA
| | - Matthew P. Walker
- Sleep and Neuroimaging Laboratory, Department of Psychology, University of California, Berkeley, California 94720-1650, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720-1650, USA
| |
Collapse
|
43
|
A role for cortical nNOS/NK1 neurons in coupling homeostatic sleep drive to EEG slow wave activity. Proc Natl Acad Sci U S A 2013; 110:20272-7. [PMID: 24191004 DOI: 10.1073/pnas.1314762110] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Although the neural circuitry underlying homeostatic sleep regulation is little understood, cortical neurons immunoreactive for neuronal nitric oxide synthase (nNOS) and the neurokinin-1 receptor (NK1) have been proposed to be involved in this physiological process. By systematically manipulating the durations of sleep deprivation and subsequent recovery sleep, we show that activation of cortical nNOS/NK1 neurons is directly related to non-rapid eye movement (NREM) sleep time, NREM bout duration, and EEG δ power during NREM sleep, an index of preexisting homeostatic sleep drive. Conversely, nNOS knockout mice show reduced NREM sleep time, shorter NREM bouts, and decreased power in the low δ range during NREM sleep, despite constitutively elevated sleep drive. Cortical NK1 neurons are still activated in response to sleep deprivation in these mice but, in the absence of nNOS, they are unable to up-regulate NREM δ power appropriately. These findings support the hypothesis that cortical nNOS/NK1 neurons translate homeostatic sleep drive into up-regulation of NREM δ power through an NO-dependent mechanism.
Collapse
|
44
|
Nitric oxide mediates selective degeneration of hypothalamic orexin neurons through dysfunction of protein disulfide isomerase. J Neurosci 2013; 33:12557-68. [PMID: 23904594 DOI: 10.1523/jneurosci.0595-13.2013] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
We addressed the role of nitric oxide (NO) in orexin neuron degeneration that has been observed under various pathological conditions. Administration of an NO donor NOC18 (50 nmol) into the third ventricle of mice resulted in a significant decrease of orexin-immunoreactive (-IR) neurons, in contrast to a modest change in melanin-concentrating hormone-IR neurons. In addition, NOC18 promoted formation of orexin-A-IR aggregates within orexin neurons. An endoplasmic reticulum stress inducer tunicamycin replicated the effect of NOC18 with regard to decrease of orexin-IR neurons and formation of aggregates. We also found that NOC18 caused an increase in S-nitrosation of protein disulfide isomerase (PDI) and a decrease in PDI activity in hypothalamic tissues. Moreover, PDI inhibitors, such as cystamine and securinine, caused a selective decrease of orexin neurons and promoted formation of orexin-A-IR aggregates. Aggregate formation in orexin-IR neurons was also induced by local injection of small interfering RNA targeting PDI. Interestingly, sleep deprivation for 7 consecutive days induced a selective decrease of orexin-IR neurons, which was preceded by aggregate formation in orexin-IR neurons and an increase in S-nitrosated PDI in the hypothalamus. Activity of neuronal NO synthase (nNOS)-positive neurons in the lateral hypothalamus as assessed by c-Fos expression was elevated in response to sleep deprivation. Finally, sleep deprivation-induced decrease of orexin-IR neurons, formation of aggregates, and S-nitrosation of PDI were not observed in nNOS knock-out mice. These results indicate that nNOS-derived NO may mediate specific pathological events in orexin neurons, including neuropeptide misfolding via S-nitrosation and inactivation of PDI.
Collapse
|
45
|
Sahu S, Kauser H, Ray K, Kishore K, Kumar S, Panjwani U. Caffeine and modafinil promote adult neuronal cell proliferation during 48h of total sleep deprivation in rat dentate gyrus. Exp Neurol 2013; 248:470-81. [DOI: 10.1016/j.expneurol.2013.07.021] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Revised: 07/15/2013] [Accepted: 07/29/2013] [Indexed: 10/26/2022]
|
46
|
Kostin A, McGinty D, Szymusiak R, Alam MN. Sleep-wake and diurnal modulation of nitric oxide in the perifornical-lateral hypothalamic area: real-time detection in freely behaving rats. Neuroscience 2013; 254:275-84. [PMID: 24056193 DOI: 10.1016/j.neuroscience.2013.09.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 09/09/2013] [Accepted: 09/10/2013] [Indexed: 01/12/2023]
Abstract
Nitric oxide (NO) has been implicated in the regulation of sleep. The perifornical-lateral hypothalamic area (PF-LHA) is a key wake-promoting region and contains neurons that are active during behavioral or cortical activation. Recently, we found higher levels of NO metabolites (NOx), an indirect measure of NO levels, in the PF-LHA during prolonged waking (SD). However, NO is highly reactive and diffuses rapidly and the NOx assay is not sensitive enough to detect rapid-changes in NO levels across spontaneous sleep-waking states. We used a novel Nafion®-modified Platinum (NF-PT) electrode for real-time detection of NO levels in the PF-LHA across sleep-wake cycles, dark-light phases, and during SD. Sprague-Dawley male rats were surgically prepared for chronic sleep-wake recording and implantation of NF-PT electrode into the PF-LHA. Electroencephalogram (EEG), electromyogram (EMG), and electrochemical current generated by NF-PT electrode were continuously acquired for 5-7days including one day with 3h of SD. In the PF-LHA, NO levels exhibited a waking>rapid eye movement (REM)>non-rapid eye movement (nonREM) sleep pattern (0.56±0.03μM>0.47±0.02μM>0.42±0.02μM; p<0.01). NO levels were also higher during the dark- as compared to the light-phase (0.53±0.03μM vs. 0.44±0.02μM; p<0.01). NO levels increased during 3h of SD as compared to undisturbed control (0.58±0.04μM vs. 0.47±0.01μM; p<0.05). The findings indicate that in the PF-LHA, NO is produced during behavioral or cortical activation. Since elevated levels of NO inhibits most of the PF-LHA neurons that are active during cortical activation, these findings support a hypothesis that NO produced in conjunction with the activation of PF-LHA neurons during waking/SD, inhibits the same neuronal population to promote sleep.
Collapse
Affiliation(s)
- A Kostin
- Research Service, Veterans Affairs Greater Los Angeles Healthcare System, Sepulveda, CA 91343, USA
| | | | | | | |
Collapse
|
47
|
Bittman EL, Kilduff TS, Kriegsfeld LJ, Szymusiak R, Toth LA, Turek FW. Animal care practices in experiments on biological rhythms and sleep: report of the Joint Task Force of the Society for Research on Biological Rhythms and the Sleep Research Society. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2013; 52:437-443. [PMID: 23849440 PMCID: PMC3725927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 01/29/2013] [Accepted: 03/07/2013] [Indexed: 06/02/2023]
Abstract
Many physiological and molecular processes are strongly rhythmic and profoundly influenced by sleep. The continuing effort of biological, medical, and veterinary science to understand the temporal organization of cellular, physiological, behavioral and cognitive function holds great promise for the improvement of the welfare of animals and human beings. As a result, attending veterinarians and IACUC are often charged with the responsibility of evaluating experiments on such rhythms or the effects of sleep (or its deprivation) in vertebrate animals. To produce interpretable data, animals used in such research must often be maintained in carefully controlled (often constant) conditions with minimal disruption. The lighting environment must be strictly controlled, frequent changes of cages and bedding are undesirable, and daily visual checks are often not possible. Thus deviations from the standard housing procedures specified in the Guide for the Care and Use of Laboratory Animals are often necessary. This report reviews requirements for experiments on biological rhythms and sleep and discusses how scientific considerations can be reconciled with the recommendations of the Guide.
Collapse
Affiliation(s)
- Eric L Bittman
- Department of Biology and Program in Neuroscience and Behavior, University of Massachusetts, Amherst, MA, USA.
| | | | | | | | | | | |
Collapse
|
48
|
Porkka-Heiskanen T. Sleep homeostasis. Curr Opin Neurobiol 2013; 23:799-805. [PMID: 23510741 DOI: 10.1016/j.conb.2013.02.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 02/19/2013] [Accepted: 02/20/2013] [Indexed: 10/27/2022]
Abstract
Research on sleep homeostasis aims to answer the question: how does the brain measure the duration and intensity of previous wakefulness in order to increase the duration and intensity of subsequent sleep? The search of regulatory factors has identified a number of potential molecules that increase their concentration in waking and decrease it during sleep. These factors regulate many physiological functions, including energy metabolism, neural plasticity and immune functions and one molecule may participate in the regulation of all these functions. The method to study regulation of sleep homeostasis is experimental prolongation of waking, which is used also to address the question of physiological purpose of sleep: prolonging wakefulness provokes symptoms that tell us what goes wrong during lack of sleep. The interpretation of the role of each identified factor in the regulation of sleep/sleep homeostasis reflects the theoretical background concept of the research. Presently three main concepts are being actively studied: the energy (depletion) hypothesis, the neural plasticity hypothesis and the (immune) defense hypothesis.
Collapse
Affiliation(s)
- Tarja Porkka-Heiskanen
- University of Helsinki, Institute of Biomedicine, Department of Physiology, PO Box 63, 00014 University of Helsinki, Finland.
| |
Collapse
|
49
|
Bransfield RC. The psychoimmunology of lyme/tick-borne diseases and its association with neuropsychiatric symptoms. Open Neurol J 2012; 6:88-93. [PMID: 23091569 PMCID: PMC3474947 DOI: 10.2174/1874205x01206010088] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2012] [Revised: 06/22/2012] [Accepted: 07/02/2012] [Indexed: 11/22/2022] Open
Abstract
Disease progression of neuropsychiatric symptoms in Lyme/tick-borne diseases can be better understood by greater attention to psychoimmunology. Although there are multiple contributors that provoke and weaken the immune system, infections and persistent infections are significant causes of pathological immune reactions. Immune mediated ef-fects are a significant contributor to the pathophysiological processes and disease progression. These immune effects in-clude persistent inflammation with cytokine effects and molecular mimicry and both of these mechanisms may be present at the same time in persistent infections. Sickness syndrome associated with interferon treatment and autoimmune limbic encephalopathies are models to understand inflammatory and molecular mimicry effects upon neuropsychiatric symp-toms. Progressive inflammatory reactions have been proposed as a model to explain disease progression in depression, psychosis, dementia, epilepsy, autism and other mental illnesses and pathophysiological changes have been associated with oxidative stress, excitotoxicity, changes in homocysteine metabolism and altered tryptophan catabolism. Lyme dis-ease has been associated with the proinflammatory cytokines IL-6, IL-8, IL-12, IL-18 and interferon-gamma, the chemokines CXCL12 and CXCL13 and increased levels proinflammatory lipoproteins. Borrelia burgdorferi surface gly-colipids and flagella antibodies appear to elicit anti-neuronal antibodies and anti-neuronal antibodies and Borrelia burgdorferi lipoproteins can disseminate from the periphery to inflame the brain. Autism spectrum disorders associated with Lyme/tick-borne diseases may be mediated by a combination of inflammatory and molecular mimicry mechanisms. Greater interaction is needed between infectious disease specialists, immunologists and psychiatrists to benefit from this awareness and to further understand these mechanisms.
Collapse
|
50
|
Kelly JM, Bianchi MT. Mammalian sleep genetics. Neurogenetics 2012; 13:287-326. [DOI: 10.1007/s10048-012-0341-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Accepted: 08/10/2012] [Indexed: 10/27/2022]
|